1
|
Geng J, Zheng Z, Li L, Ren Z, Tian G, Qin J, Zhao T, Feng X. Apigenin attenuated sepsis induced acute lung injury via polarizing macrophage towards M2 by blocking miR-146a →TLR7 interaction. Int Immunopharmacol 2025; 152:114446. [PMID: 40088874 DOI: 10.1016/j.intimp.2025.114446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 01/19/2025] [Accepted: 03/06/2025] [Indexed: 03/17/2025]
Abstract
TLR7 (Toll-like receptor 7) has been indicated as an important sensor for single -stranded RNA contributes to systemic inflammation and mortality in acute lung injury (ALI), which is an acute diffuse inflammatory lung injury. Cumulative results show that macrophages contribute to the development and progression of ALI through the secretion of inflammatory cytokines/chemokines. Here we show that macrophage polarizes towards M1 phenotype and TLR7 signaling is activated in septic mice. Moreover, TLR7 deficiency promotes macrophage polarized towards M2 phenotype and attenuates ALI. Strikingly, the natural product of flavone apigenin (Xu et al., 2017 [1]) significantly improves sepsis-induced lung inflammation and lung injury via inhibiting inflammatory macrophages in a TLR7-dependent manner. Mechanically, Api blocked the binding of TLR7 with its agonist miR-146a. This finding reveals TLR7 is an important therapeutic target and Api as a modulator of TLR7 is a potential lead compound for treatment of septic diseases and inflammation related diseases.
Collapse
Affiliation(s)
- Jiafeng Geng
- Shandong Province University Clinical Immunology Translational Medicine Laboratory, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jingshi Road 16766, Jinan, Shandong 250014, China; School of Clinical and Basic Medical Sciences, Shandong First Medical University& Shandong Academy of Medical Sciences, Jinan 250117, Shandong, China
| | - Zhihuan Zheng
- Shandong Province University Clinical Immunology Translational Medicine Laboratory, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jingshi Road 16766, Jinan, Shandong 250014, China; School of Clinical and Basic Medical Sciences, Shandong First Medical University& Shandong Academy of Medical Sciences, Jinan 250117, Shandong, China
| | - Liangge Li
- Shandong Province University Clinical Immunology Translational Medicine Laboratory, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jingshi Road 16766, Jinan, Shandong 250014, China; School of Clinical and Basic Medical Sciences, Shandong First Medical University& Shandong Academy of Medical Sciences, Jinan 250117, Shandong, China
| | - Zixuan Ren
- Shandong Province University Clinical Immunology Translational Medicine Laboratory, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jingshi Road 16766, Jinan, Shandong 250014, China; School of Clinical and Basic Medical Sciences, Shandong First Medical University& Shandong Academy of Medical Sciences, Jinan 250117, Shandong, China
| | - Ge Tian
- School of Life Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong 271000, China
| | - Jing Qin
- School of Clinical and Basic Medical Sciences, Shandong First Medical University& Shandong Academy of Medical Sciences, Jinan 250117, Shandong, China
| | - Tong Zhao
- Shandong Province University Clinical Immunology Translational Medicine Laboratory, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jingshi Road 16766, Jinan, Shandong 250014, China; School of Clinical and Basic Medical Sciences, Shandong First Medical University& Shandong Academy of Medical Sciences, Jinan 250117, Shandong, China
| | - Xiujing Feng
- Shandong Province University Clinical Immunology Translational Medicine Laboratory, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jingshi Road 16766, Jinan, Shandong 250014, China; School of Clinical and Basic Medical Sciences, Shandong First Medical University& Shandong Academy of Medical Sciences, Jinan 250117, Shandong, China; Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China.
| |
Collapse
|
2
|
Ma Z, Liu L, Tian J, Tu C, Zhang D, Zhang M, Zhang H, An Z, Sun M, Zhang H, Song X. Causal Relationships between Lymphocyte Subsets and Risk of Coronary Artery Disease: A Two-Sample Mendelian Randomization Study. Rev Cardiovasc Med 2024; 25:326. [PMID: 39355583 PMCID: PMC11440411 DOI: 10.31083/j.rcm2509326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 04/08/2024] [Accepted: 04/15/2024] [Indexed: 10/03/2024] Open
Abstract
Background Currently, the causal relationship between lymphocyte subsets and coronary artery disease (CAD) remains unclear. Therefore, we utilized Mendelian randomization (MR) to assess the association between lymphocyte subsets and CAD. Methods We performed a two-sample MR analysis using publicly available genome-wide association studies (GWAS) datasets. The primary method of analysis to comprehensively evaluate causal effects was the inverse variance-weighted (IVW) method. The four additional MR approaches were MR-Egger, weighted median, simple mode, and weighted mode. Sensitivity analysis incorporated Cochran's Q and MR-Egger intercept tests to identify residual heterogeneity and potential horizontal pleiotropy, respectively. The MR-PRESSO distortion test was applied to identify potential pleiotropic outliers. Leave-one-out analysis confirmed that no single single-nucleotide polymorphism (SNP) significantly affected the MR estimate. We conducted reverse MR analysis to investigate the impact of variables correlated with outcomes in forward MR analysis. Results The IVW method revealed a significant positive association between B cell count and CAD (odds ratio (OR) = 1.08 (95% CI: 1.04, 1.11), p = 2.67 × 10-5). A similar association was observed between B cell count and myocardial infarction (MI) (OR = 1.07 (95% CI: 1.03, 1.11), p = 5.69 × 10-4). Sensitivity analyses detected no outliers, heterogeneity, or pleiotropy. The reverse MR analysis was conducted to investigate the impact of CAD and MI on B cell count, and the IVW results showed no statistical significance. Conclusions Our study suggests that a higher absolute B cell count is linked to an increased risk of CAD and MI.
Collapse
Affiliation(s)
- Zhao Ma
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, 100029 Beijing, China
| | - Libo Liu
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, 100029 Beijing, China
| | - Jinfan Tian
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, 100029 Beijing, China
| | - Chenchen Tu
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, 100029 Beijing, China
| | - Dongfeng Zhang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, 100029 Beijing, China
| | - Mingduo Zhang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, 100029 Beijing, China
| | - Huan Zhang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, 100029 Beijing, China
| | - Ziyu An
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, 100029 Beijing, China
| | - Meichen Sun
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, 100029 Beijing, China
| | - Hongjia Zhang
- Department of Cardiovascular Surgery, Beijing Anzhen Hospital, Capital Medical University, 100029 Beijing, China
| | - Xiantao Song
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, 100029 Beijing, China
| |
Collapse
|
3
|
Gu Y, Feng J, Shi J, Xiao G, Zhang W, Shao S, Liu B, Guo H. Global Research Trends on Exosome in Cardiovascular Diseases: A Bibliometric-Based Visual Analysis. Vasc Health Risk Manag 2024; 20:377-402. [PMID: 39188326 PMCID: PMC11346494 DOI: 10.2147/vhrm.s473520] [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: 05/09/2024] [Accepted: 08/10/2024] [Indexed: 08/28/2024] Open
Abstract
Background Exosomes in cardiovascular diseases (CVDs) have attracted huge attention with substantial value and potential. Our bibliometrics is based on literature from the field of cardiovascular exosomes over the past 30 years, which has been visualized to display the development process, research hotspots, and cutting-edge trends of clinical practices, mechanisms, and management strategies related to psych cardiology. Methods We selected articles and reviews on exosomes in CVDs from the core collection of Web of Science, and generated visual charts by using CiteSpace and VOSviewer software. Results Our research included 1613 publications. The number of exosome articles in CVD fluctuates slightly, but overall shows an increasing trend. The main research institutions were Tongji University and Nanjing Medical University. The International Journal of Molecular Sciences has the highest publication volume, while the Journal of Cellular and Molecular Medicine has the highest citation count. Among all the authors, Eduardo Marban ranks first in terms of publication volume and H-index. The most common keywords are exosome, extracellular vesicles, and angiogenesis. Conclusion This is a bibliometric study on the research hotspots and trends of exosomes in CVD. Exosome research in the field of cardiovascular medicine is on the rise. Some exosome treatment methods may become the focus of future research.
Collapse
Affiliation(s)
- Yunxiao Gu
- Department of Anatomy, School of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Jiaming Feng
- Department of Anatomy, School of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Jiayi Shi
- Department of Anatomy, School of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Guanyi Xiao
- Department of Anatomy, School of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Weiwei Zhang
- Department of Anatomy, School of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Shuijin Shao
- Department of Anatomy, School of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Baonian Liu
- Department of Anatomy, School of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Haidong Guo
- Department of Anatomy, School of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| |
Collapse
|
4
|
Kiełbowski K, Skórka P, Plewa P, Bakinowska E, Pawlik A. The Role of Alarmins in the Pathogenesis of Atherosclerosis and Myocardial Infarction. Curr Issues Mol Biol 2024; 46:8995-9015. [PMID: 39194749 DOI: 10.3390/cimb46080532] [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: 06/29/2024] [Revised: 07/30/2024] [Accepted: 08/15/2024] [Indexed: 08/29/2024] Open
Abstract
Atherosclerosis is a condition that is associated with lipid accumulation in the arterial intima. Consequently, the enlarging lesion, which is also known as an atherosclerotic plaque, may close the blood vessel lumen, thus leading to organ ischaemia. Furthermore, the plaque may rupture and initiate the formation of a thrombus, which can cause acute ischaemia. Atherosclerosis is a background pathological condition that can eventually lead to major cardiovascular diseases such as acute coronary syndrome or ischaemic stroke. The disorder is associated with an altered profile of alarmins, stress response molecules that are secreted due to cell injury or death and that induce inflammatory responses. High-mobility group box 1 (HMGB1), S100 proteins, interleukin-33, and heat shock proteins (HSPs) also affect the behaviour of endothelial cells and vascular smooth muscle cells (VSMCs). Thus, alarmins control the inflammatory responses of endothelial cells and proliferation of VSMCs, two important processes implicated in the pathogenesis of atherosclerosis. In this review, we will discuss the role of alarmins in the pathophysiology of atherosclerosis and myocardial infarction.
Collapse
Affiliation(s)
- Kajetan Kiełbowski
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland
| | - Patryk Skórka
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland
| | - Paulina Plewa
- Institute of Biology, University of Szczecin, 71-412 Szczecin, Poland
| | - Estera Bakinowska
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland
| | - Andrzej Pawlik
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland
| |
Collapse
|
5
|
Hazra J, Vijayakumar A, Mahapatra NR. Emerging role of heat shock proteins in cardiovascular diseases. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2023; 134:271-306. [PMID: 36858739 DOI: 10.1016/bs.apcsb.2022.10.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Heat Shock Proteins (HSPs) are evolutionarily conserved proteins from prokaryotes to eukaryotes. They are ubiquitous proteins involved in key physiological and cellular pathways (viz. inflammation, immunity and apoptosis). Indeed, the survivability of the cells under various stressful conditions depends on appropriate levels of HSP expression. There is a growing line of evidence for the role of HSPs in regulating cardiovascular diseases (CVDs) (viz. hypertension, atherosclerosis, atrial fibrillation, cardiomyopathy and heart failure). Furthermore, studies indicate that a higher concentration of circulatory HSP antibodies correlate to CVDs; some are even potential markers for CVDs. The multifaceted roles of HSPs in regulating cellular signaling necessitate unraveling their links to pathophysiology of CVDs. This review aims to consolidate our understanding of transcriptional (via multiple transcription factors including HSF-1, NF-κB, CREB and STAT3) and post-transcriptional (via microRNAs including miR-1, miR-21 and miR-24) regulation of HSPs. The cytoprotective nature of HSPs catapults them to the limelight as modulators of cell survival. Yet another attractive prospect is the development of new therapeutic strategies against cardiovascular diseases (from hypertension to heart failure) by targeting the regulation of HSPs. Moreover, this review provides insights into how genetic variation of HSPs can contribute to the manifestation of CVDs. It would also offer a bird's eye view of the evolving role of different HSPs in the modulation and manifestation of cardiovascular disease.
Collapse
Affiliation(s)
- Joyita Hazra
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, India
| | - Anupama Vijayakumar
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, India
| | - Nitish R Mahapatra
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, India.
| |
Collapse
|
6
|
Ginsenoside Re inhibits myocardial fibrosis by regulating miR-489/myd88/NF-κB pathway. J Ginseng Res 2023; 47:218-227. [PMID: 36926602 PMCID: PMC10014187 DOI: 10.1016/j.jgr.2021.11.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 11/23/2021] [Accepted: 11/29/2021] [Indexed: 12/17/2022] Open
Abstract
Background Myocardial fibrosis (MF) is an advanced pathological manifestation of many cardiovascular diseases, which can induce heart failure and malignant arrhythmias. However, the current treatment of MF lacks specific drugs. Ginsenoside Re has anti-MF effect in rat, but its mechanism is still not clear. Therefore, we investigated the anti-MF effect of ginsenoside Re by constructing mouse acute myocardial infarction (AMI) model and AngⅡ induced cardiac fibroblasts (CFs) model. Methods The anti-MF effect of miR-489 was investigated by transfection of miR-489 mimic and inhibitor in CFs. Effect of ginsenoside Re on MF and its related mechanisms were investigated by ultrasonographic, ELISA, histopathologic staining, transwell test, immunofluorescence, Western blot and qPCR in the mouse model of AMI and the AngⅡ-induced CFs model. Results MiR-489 decreased the expression of α-SMA, collagenⅠ, collagen Ⅲ and myd88, and inhibited the phosphorylation of NF-κB p65 in normal CFs and CFs treated with AngⅡ. Ginsenoside Re could improve cardiac function, inhibit collagen deposition and CFs migration, promote the transcription of miR-489, and reduce the expression of myd88 and the phosphorylation of NF-κB p65. Conclusion MiR-489 can effectively inhibit the pathological process of MF, and the mechanism is at least partly related to the regulation of myd88/NF-κB pathway. Ginsenoside Re can ameliorate AMI and AngⅡ induced MF, and the mechanism is at least partially related to the regulation of miR-489/myd88/NF-κB signaling pathway. Therefore, miR-489 may be a potential target of anti-MF and ginsenoside Re may be an effective drug for the treatment of MF.
Collapse
|
7
|
Benković V, Milić M, Oršolić N, Horvat Knežević A, Brozović G, Borojević N. Brain DNA damaging effects of volatile anesthetics and 1 and 2 Gy gamma irradiation in vivo: Preliminary results. Toxicol Ind Health 2023; 39:67-80. [PMID: 36602468 DOI: 10.1177/07482337221145599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Although both can cause DNA damage, the combined impact of volatile anesthetics halothane/sevoflurane/isoflurane and radiotherapeutic exposure on sensitive brain cells in vivo has not been previously analyzed. Healthy Swiss albino male mice (240 in total, 48 groups) were exposed to either halothane/sevoflurane/isoflurane therapeutic doses alone (2 h); 1 or 2 gray of gamma radiation alone; or combined exposure. Frontal lobe brain samples from five animals were taken immediately and 2, 6, and 24 h after exposure. DNA damage and cellular repair index were analyzed using the alkaline comet assay and the tail intensity parameter. Elevated tail intensity levels for sevoflurane/halothane were the highest at 6 h and returned to baseline within 24 h for sevoflurane, but not for halothane, while isoflurane treatment caused lower tail intensity than control values. Combined exposure demonstrated a slightly halothane/sevoflurane protective and isoflurane protective effect, which was stronger for 2 than for 1 gray. Cellular repair indices and tail intensity histograms indicated different modes of action in DNA damage creation. Isoflurane/sevoflurane/halothane preconditioning demonstrated protective effects in sensitive brain cells in vivo. Owing to the constant increases in the combined use of radiotherapy and volatile anesthetics, further studies should explore the mechanisms behind these effects, including longer and multiple exposure treatments and in vivo brain tumor models.
Collapse
Affiliation(s)
- Vesna Benković
- Faculty of Science, 117036University of Zagreb, Zagreb, Croatia
| | - Mirta Milić
- Mutagenesis Unit, 118938Institute for Medical Research and Occupational Health, Zagreb, Croatia
| | - Nada Oršolić
- Faculty of Science, 117036University of Zagreb, Zagreb, Croatia
| | | | - Gordana Brozović
- Department of Anesthesiology, Reanimatology and ICU, University Hospital for Tumors, 499232Sestre Milosrdnice University Hospital Centre, Zagreb, Croatia.,Faculty of Dental Medicine and Health, 84992University of Osijek, Osijek, Croatia
| | - Nikola Borojević
- 8256Warrington and Halton Teaching Hospitals NHS Foundation Trust, Warrington, UK
| |
Collapse
|
8
|
Abstract
ABSTRACT Sepsis and trauma remain the leading causes of morbidity and mortality. Our understanding of the molecular pathogenesis in the development of multiple organ dysfunction in sepsis and trauma has evolved as more focus is on secondary injury from innate immunity, inflammation, and the potential role of endogenous danger molecules. Studies of the past several decades have generated evidence for extracellular RNAs (exRNAs) as biologically active mediators in health and disease. Here, we review studies on plasma exRNA profiling in mice and humans with sepsis and trauma, the role and mode of action by exRNAs, such as ex-micro(mi)RNAs, in host innate immune response, and their potential implications in various organ injury during sepsis and trauma.
Collapse
Affiliation(s)
- Williams Brittney
- Translational Research Program, Department of Anesthesiology, and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Rosemary Kozar
- Shock Trauma Center and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Chao Wei
- Translational Research Program, Department of Anesthesiology, and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
9
|
Zhan X, Cheng L, Huo N, Yu L, Liu C, Liu T, Li G, Fu H. Sodium-glucose cotransporter-2 inhibitor alleviated atrial remodeling in STZ-induced diabetic rats by targeting TLR4 pathway. Front Cardiovasc Med 2022; 9:908037. [PMID: 36148071 PMCID: PMC9485554 DOI: 10.3389/fcvm.2022.908037] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 08/05/2022] [Indexed: 11/23/2022] Open
Abstract
PURPOSE The mechanism of sodium-glucose cotransporter-2 inhibitor (SGLT-2i) reducing the incidence of atrial fibrillation remains unclear. We hypothesize that sodium-glucose cotransporter-2 inhibitor alleviated atrial remodeling in STZ-induced diabetic rats by targeting TLR4 pathway. METHODS A total of 42 rats were randomly assigned into three groups: control group (CON group); diabetes group (DM group): diabetes mellitus rats were established by 65 mg/kg streptozotocin (STZ) intraperitoneal injection; and diabetes + dapagliflozin group (DM + DAPA group): diabetic rats were given DAPA gavage administration (DAPA 2mg/kg/d for 4 weeks by gavage administration), 14 rats in each group. Epicardial multiple-lead recording and intracardiac electrophysiology studies were performed to investigate the electrical remodeling in the heart and the atrial fibrillation inducibility in each group. Western blot analysis and real-time PCR were used to determine the protein and mRNA expression of toll-like receptor 4 (TLR4), interleukin receptor-associated kinase 1 (IRAK1), tumor necrosis factor receptor-associated factor 6 (TRAF6), nuclear factor-kappa B (NF-κB), and type I collagen (collagen I). RESULTS Compared with rats in CON group, rats in DM group showed marked myocardial fibrosis, ectopic pacing excitement, reduced conduction velocity, decreased cardiac function. TLR4/IRAK1/TRAF6/NF-κB, collagen I proteins expressions and incidence of atrial fibrillation (27.3%) were increased in DM group. Parts of these changes were reversed by treatment of DAPA. Incidence of atrial fibrillation was decreased in DM + DAPA group (2.8%). CONCLUSIONS SGLT-2i dapagliflozin may prevent diabetic rats' atrial remodeling and reduce the inducibility of atrial fibrillation partly by targeting TLR4/IRAK1/TRAF6/NF-κB inflammatory pathway.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Huaying Fu
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin, China
| |
Collapse
|
10
|
Chen F, Wang Y, Wang J, Hu L, Huang S, Cao Y, Yu Y. Exosome biomarkers in cardiovascular diseases and their prospective forensic application in the identification of sudden cardiac death. JOURNAL OF FORENSIC SCIENCE AND MEDICINE 2022. [DOI: 10.4103/jfsm.jfsm_118_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
|
11
|
Chen F, Zou L, Williams B, Chao W. Targeting Toll-Like Receptors in Sepsis: From Bench to Clinical Trials. Antioxid Redox Signal 2021; 35:1324-1339. [PMID: 33588628 PMCID: PMC8817700 DOI: 10.1089/ars.2021.0005] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Significance: Sepsis is a critical clinical syndrome with life-threatening organ dysfunction induced by a dysregulated host response to infection. Despite decades of intensive research, sepsis remains a leading cause of in-hospital mortality with few specific treatments. Recent Advances: Toll-like receptors (TLRs) are a part of the innate immune system and play an important role in host defense against invading pathogens such as bacteria, virus, and fungi. Using a combination of genetically modified animal models and pharmacological agents, numerous preclinical studies during the past two decades have demonstrated that dysregulated TLR signaling may contribute to sepsis pathogenesis. However, many clinical trials targeting inflammation and innate immunity such as TLR4 have yielded mixed results. Critical Issues: Here we review various TLRs and the specific molecules these TLRs sense-both the pathogen-associated and host-derived stress molecules, and their converging signaling pathways. We critically analyze preclinical investigations into the role of TLRs in animal sepsis, the complexity of targeting TLRs for sepsis intervention, and the disappointing clinical trials of the TLR4 antagonist eritoran. Future Directions: Future sepsis treatments will depend on better understanding the complex biological mechanisms of sepsis pathogenesis, the high heterogeneity of septic humans as defined by clinical presentations and unique immunological biomarkers, and improved stratifications for targeted interventions.
Collapse
Affiliation(s)
- Fengqian Chen
- Translational Research Program, Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Lin Zou
- Translational Research Program, Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Brittney Williams
- Translational Research Program, Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Wei Chao
- Translational Research Program, Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
12
|
Chakafana G, Spracklen TF, Kamuli S, Zininga T, Shonhai A, Ntusi NAB, Sliwa K. Heat Shock Proteins: Potential Modulators and Candidate Biomarkers of Peripartum Cardiomyopathy. Front Cardiovasc Med 2021; 8:633013. [PMID: 34222357 PMCID: PMC8241919 DOI: 10.3389/fcvm.2021.633013] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 05/06/2021] [Indexed: 12/31/2022] Open
Abstract
Peripartum cardiomyopathy (PPCM) is a potentially life-threatening condition in which heart failure and systolic dysfunction occur late in pregnancy or within months following delivery. To date, no reliable biomarkers or therapeutic interventions for the condition exist, thus necessitating an urgent need for identification of novel PPCM drug targets and candidate biomarkers. Leads for novel treatments and biomarkers are therefore being investigated worldwide. Pregnancy is generally accompanied by dramatic hemodynamic changes, including a reduced afterload and a 50% increase in cardiac output. These increased cardiac stresses during pregnancy potentially impair protein folding processes within the cardiac tissue. The accumulation of misfolded proteins results in increased toxicity and cardiac insults that trigger heart failure. Under stress conditions, molecular chaperones such as heat shock proteins (Hsps) play crucial roles in maintaining cellular proteostasis. Here, we critically assess the potential role of Hsps in PPCM. We further predict specific associations between the Hsp types Hsp70, Hsp90 and small Hsps with several proteins implicated in PPCM pathophysiology. Furthermore, we explore the possibility of select Hsps as novel candidate PPCM biomarkers and drug targets. A better understanding of how these Hsps modulate PPCM pathogenesis holds promise in improving treatment, prognosis and management of the condition, and possibly other forms of acute heart failure.
Collapse
Affiliation(s)
- Graham Chakafana
- Department of Medicine, Faculty of Health Sciences, Cape Heart Institute, University of Cape Town, Cape Town, South Africa.,Division of Cardiology, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Timothy F Spracklen
- Department of Medicine, Faculty of Health Sciences, Cape Heart Institute, University of Cape Town, Cape Town, South Africa.,Division of Cardiology, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Stephen Kamuli
- Department of Medicine, Faculty of Health Sciences, Cape Heart Institute, University of Cape Town, Cape Town, South Africa.,Division of Cardiology, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Tawanda Zininga
- Department of Biochemistry, Stellenbosch University, Stellenbosch, South Africa
| | - Addmore Shonhai
- Department of Biochemistry, University of Venda, Thohoyandou, South Africa
| | - Ntobeko A B Ntusi
- Department of Medicine, Faculty of Health Sciences, Cape Heart Institute, University of Cape Town, Cape Town, South Africa.,Division of Cardiology, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.,Cape Universities Body Imaging Centre, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Karen Sliwa
- Department of Medicine, Faculty of Health Sciences, Cape Heart Institute, University of Cape Town, Cape Town, South Africa.,Division of Cardiology, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
13
|
Soni UK, Chadchan SB, Gupta RK, Kumar V, Kumar Jha R. miRNA-149 targets PARP-2 in endometrial epithelial and stromal cells to regulate the trophoblast attachment process. Mol Hum Reprod 2021; 27:6288493. [PMID: 34051087 DOI: 10.1093/molehr/gaab039] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 04/26/2021] [Indexed: 11/12/2022] Open
Abstract
Embryo implantation is a highly complex process involving many regulatory factors, including several micro RNAs (miRNAs/miRs). One miRNA present in the stromal cells of normal endometrium is miR-149, which targets poly (ADP-ribose) polymerase 2 (PARP-2), a gene involved in endometrial receptivity for trophoblast implantation. However, the precise role of miR-149 in the endometrial receptivity during blastocyst implantation is still unknown. We studied miR-149-dependent PARP-2 regulation during trophoblast attachment to endometrial epithelial cells. Using FISH, we found that miR-149 is expressed in mouse endometrial epithelial and stromal cells at implantation and inter-implantation sites. Endometrial receptivity for embryo implantation and attachment is inhibited by the upregulation of miR-149 in the endometrium. Our RT-PCR analysis revealed downregulation of miR-149 in the implantation region of the uterus during the receptive stage (Day 5, 0500 h, p.c.) in the mouse. Under in-vitro conditions, miR-149 overexpression in human endometrial epithelial cells (hEECs) abrogated the human trophoblastic cells spheroid and mouse blastocyst attachment. Subsequently, miR-149 also regulates transformed human endometrial stromal cell (T-hESCs) decidualization by downregulating PARP-2 and upregulating caspase-8 proteins. Overexpression of miR-149 in hEECs and downregulated PARP-2 protein expression, reconfirming that PARP-2 is a downstream target of miR-149 in endometrial cells as well. miR-149 is also able to alter the expression of caspase-8, another PARP-2 regulator. In conclusion, our data indicate that miR-149 is one of the regulators of endometrial receptivity and decidualization for trophoblast implantation, and it exerts the effects by acting on the downstream targets PARP-2 and caspase-8.
Collapse
Affiliation(s)
- Upendra Kumar Soni
- Female Reproductive Biology Laboratory, Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, India
| | - Sangappa Basanna Chadchan
- Female Reproductive Biology Laboratory, Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, India
| | - Rakesh Kumar Gupta
- Female Reproductive Biology Laboratory, Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, India
| | - Vijay Kumar
- Female Reproductive Biology Laboratory, Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, India
| | - Rajesh Kumar Jha
- Female Reproductive Biology Laboratory, Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, U.P., India
| |
Collapse
|
14
|
Kyaw T, Loveland P, Kanellakis P, Cao A, Kallies A, Huang AL, Peter K, Toh BH, Bobik A. Alarmin-activated B cells accelerate murine atherosclerosis after myocardial infarction via plasma cell-immunoglobulin-dependent mechanisms. Eur Heart J 2021; 42:938-947. [PMID: 33338208 DOI: 10.1093/eurheartj/ehaa995] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 07/30/2020] [Accepted: 11/29/2020] [Indexed: 12/25/2022] Open
Abstract
AIMS Myocardial infarction (MI) accelerates atherosclerosis and greatly increases the risk of recurrent cardiovascular events for many years, in particular, strokes and MIs. Because B cell-derived autoantibodies produced in response to MI also persist for years, we investigated the role of B cells in adaptive immune responses to MI. METHODS AND RESULTS We used an apolipoprotein-E-deficient (ApoE-/-) mouse model of MI-accelerated atherosclerosis to assess the importance of B cells. One week after inducing MI in atherosclerotic mice, we depleted B cells using an anti-CD20 antibody. This treatment prevented subsequent immunoglobulin G accumulation in plaques and MI-induced accelerated atherosclerosis. In gain of function experiments, we purified spleen B cells from mice 1 week after inducing MI and transferred these cells into atherosclerotic ApoE-/- mice, which greatly increased immunoglobulin G (IgG) accumulation in plaque and accelerated atherosclerosis. These B cells expressed many cytokines that promote humoural immunity and in addition, they formed germinal centres within the spleen where they differentiated into antibody-producing plasma cells. Specifically deleting Blimp-1 in B cells, the transcriptional regulator that drives their terminal differentiation into antibody-producing plasma cells prevented MI-accelerated atherosclerosis. Alarmins released from infarcted hearts were responsible for activating B cells via toll-like receptors and deleting MyD88, the canonical adaptor protein for inflammatory signalling downstream of toll-like receptors, prevented B-cell activation and MI-accelerated atherosclerosis. CONCLUSION Our data implicate early B-cell activation and autoantibodies as a central cause for accelerated atherosclerosis post-MI and identifies novel therapeutic strategies towards preventing recurrent cardiovascular events such as MI and stroke.
Collapse
Affiliation(s)
- Tin Kyaw
- Vascular Biology and Atherosclerosis, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia.,Centre for Inflammatory Diseases, Department of Medicine, Medical Centre, 246 Clayton Road, Clayton, VIC 3168, Australia
| | - Paula Loveland
- Vascular Biology and Atherosclerosis, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
| | - Peter Kanellakis
- Vascular Biology and Atherosclerosis, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
| | - Anh Cao
- Vascular Biology and Atherosclerosis, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia.,Centre for Inflammatory Diseases, Department of Medicine, Medical Centre, 246 Clayton Road, Clayton, VIC 3168, Australia
| | - Axel Kallies
- Department of Microbiology and Immunology, University of Melbourne, 792 Elizabeth Street, Melbourne, Vic 3000, Australia
| | - Alex L Huang
- Atherothrombosis and Vascular Biology, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia.,Department of Cardiology, Alfred Hospital, 55 Commercial Rd, Melbourne, VIC 3004, Australia
| | - Karlheinz Peter
- Atherothrombosis and Vascular Biology, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia.,Department of Cardiology, Alfred Hospital, 55 Commercial Rd, Melbourne, VIC 3004, Australia.,Department of Immunology, Central Clinical School, 99 Commercial Rd, Melbourne, VIC 3004, Australia
| | - Ban-Hock Toh
- Centre for Inflammatory Diseases, Department of Medicine, Medical Centre, 246 Clayton Road, Clayton, VIC 3168, Australia
| | - Alex Bobik
- Vascular Biology and Atherosclerosis, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia.,Centre for Inflammatory Diseases, Department of Medicine, Medical Centre, 246 Clayton Road, Clayton, VIC 3168, Australia.,Department of Immunology, Central Clinical School, 99 Commercial Rd, Melbourne, VIC 3004, Australia
| |
Collapse
|
15
|
Schwartz RE, Shokhirev MN, Andrade LR, Gutkind JS, Iglesias-Bartolome R, Shadel GS. Insights into epithelial cell senescence from transcriptome and secretome analysis of human oral keratinocytes. Aging (Albany NY) 2021; 13:4747-4777. [PMID: 33601339 PMCID: PMC7950289 DOI: 10.18632/aging.202658] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 01/21/2021] [Indexed: 01/08/2023]
Abstract
Senescent cells produce chronic inflammation that contributes to the diseases and debilities of aging. How this process is orchestrated in epithelial cells, the origin of human carcinomas, is poorly understood. We used human normal oral keratinocytes (NOKs) to elucidate senescence programs in a prototype primary mucosal epithelial cell that senesces spontaneously. While NOKs exhibit several typical facets of senescence, they also display distinct characteristics. These include expression of p21WAF1/CIP1 at early passages, making this common marker of senescence unreliable in NOKs. Transcriptome analysis by RNA-seq revealed specific commonalities with and differences from cancer cells, explicating the tumor avoidance role of senescence. Repression of DNA repair genes that correlated with downregulation of E2F1 mRNA and protein was observed for two donors; a divergent result was seen for the third. Using proteomic profiling of soluble (non-vesicular) and extracellular vesicle (EV) associated secretions, we propose additions to the senescence associated secretory phenotype, including HSP60, which localizes to the surface of EVs. Finally, EVs from senescent NOKs activate interferon pathway signaling in THP-1 monocytes in a STING-dependent manner and associate with mitochondrial and nuclear DNA. Our results highlight senescence changes in epithelial cells and how they might contribute to chronic inflammation and age-related diseases.
Collapse
Affiliation(s)
- Rachael E Schwartz
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies and University of California - San Diego, La Jolla, CA 92037, USA
| | - Maxim N Shokhirev
- Razavi Newman Integrative Genomics and Bioinformatics Core, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Leonardo R Andrade
- Waitt Advanced Biophotonics Core, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - J Silvio Gutkind
- Department of Pharmacology and Moores Cancer Center, University of California - San Diego, La Jolla, CA 92093, USA
| | - Ramiro Iglesias-Bartolome
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Gerald S Shadel
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies and University of California - San Diego, La Jolla, CA 92037, USA
| |
Collapse
|
16
|
Li L, Dai W, Li W, Zhang Y, Wu Y, Guan C, Zhang A, Huang H, Li Y. Integrated Network Pharmacology and Metabonomics to Reveal the Myocardial Protection Effect of Huang-Lian-Jie-Du-Tang on Myocardial Ischemia. Front Pharmacol 2021; 11:589175. [PMID: 33613277 PMCID: PMC7890363 DOI: 10.3389/fphar.2020.589175] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 12/02/2020] [Indexed: 12/14/2022] Open
Abstract
Myocardial ischemia (MI) is one of the most common cardiovascular diseases with high incidence and mortality. Huang-Lian-Jie-Du-Tang (HLJDT) is a classic traditional Chinese prescription to clear “heat” and “poison”. In this study, we used a deliberate strategy integrating the methods of network pharmacology, pharmacodynamics, and metabonomics to investigate the molecular mechanism and potential targets of HLJDT in the treatment of MI. Firstly, by a network pharmacology approach, a global view of the potential compound-target-pathway network based on network pharmacology was constructed to provide a preliminary understanding of bioactive compounds and related targets of HLJDT for elucidating its molecular mechanisms in MI. Subsequently, in vivo efficacy of HLJDT was validated in a rat model. Meanwhile, the corresponding metabonomic profiles were used to explore differentially induced metabolic markers thus providing the metabolic mechanism of HLJDT in treating MI. The results demonstrated the myocardial protection effect of HLJDT on ischemia by a multicomponent-multitarget mode. This study highlights the reliability and effectiveness of a network pharmacology-based approach that identifies and validates the complex of natural compounds in HLJDT for illustrating the mechanism for the treatment of MI.
Collapse
Affiliation(s)
- Li Li
- Department of Pharmacy, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Weixing Dai
- Department of Chemical and Biological Engineering, Hong Kong University of Science and Technology, Kowloon, China
| | - Wenting Li
- Department of Pharmacy, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Yumao Zhang
- Department of Pharmacy, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Yanqin Wu
- Department of Pharmacy, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Chenfeng Guan
- Department of Pharmacy, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Anye Zhang
- Department of Gastroenterology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Hui Huang
- Department of Cardiovascular, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Yuzhen Li
- Department of Pharmacy, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| |
Collapse
|
17
|
Zhang K, Yan H, Liu R, Xiang P, Zhang J, Deng K, Huang P, Wang Z. The Use of Gas Chromatography Coupled with High-Resolution Mass Spectrometry-Based Untargeted Metabolomics to Discover Metabolic Changes and Help in the Determination of Complex Causes of Death: A Preliminary Study. ACS OMEGA 2021; 6:2100-2109. [PMID: 33521449 PMCID: PMC7841927 DOI: 10.1021/acsomega.0c05178] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Accepted: 12/29/2020] [Indexed: 05/10/2023]
Abstract
The determination of cause of death (COD) is one of the most important tasks in forensic practice and is mainly based on macroscopical and microscopical morphological signatures. However, some CODs are hard to determine because the significant morphological signatures can be nonspecific, variable, subjective, or even absent in the real world. In this study, gas chromatography coupled with high-resolution mass spectrometry (GC-HRMS)-based untargeted metabolomics was employed to obtain plasma metabolic profiles of rats that died from anaphylactic shock (AS), mechanical asphyxia (MA), or sudden cardiac death (SCD). The metabolic alterations of each COD group compared to the control group were investigated using a principal component analysis, partial least-squares discriminant analysis, the Wilcoxon test, and fold change analysis. A range of differential features was screened, and 11, 8, and 7 differential metabolites were finally verified for the AS, MA, and SCD groups, respectively. We proposed some explanations that may account for these metabolic differences, including glucose metabolism, the tricarboxylic acid cycle, glycolysis, lipid metabolism, creatinine catabolism, and purine metabolism. Next, for each COD, we used its differential metabolites, which were obtained through comparisons of each COD group to the control group and represented the metabolic changes of the individual COD, to perform a receiver operating characteristic (ROC) analysis to preliminarily evaluate their ability to discriminate each COD group from the other COD groups. We found that creatinine in the AS group and malic acid and uric acid in the MA group might represent some specific metabolic changes for the relevant COD with high areas under the curve in the ROC curve analysis. Moreover, the combination panel for AS or MA also showed a good ability to discriminate it from the others. However, SCD had fewer metabolic signatures and was relatively harder to discriminate from the other CODs in our work. The preliminary study demonstrates the feasibility of GC-HRMS-based untargeted metabolomics as a promising tool to reveal metabolic changes in different death processes and to determine the complex CODs.
Collapse
Affiliation(s)
- Kai Zhang
- Department
of Forensic Pathology, College of Forensic Medicine, Xi’an Jiaotong University, Xi’an 710061, P. R. China
| | - Hui Yan
- Shanghai
Key Laboratory of Forensic Medicine, Shanghai Forensic Service Platform, Academy of Forensic Science, Shanghai 200063, P. R. China
| | - Ruina Liu
- Department
of Forensic Pathology, College of Forensic Medicine, Xi’an Jiaotong University, Xi’an 710061, P. R. China
| | - Ping Xiang
- Shanghai
Key Laboratory of Forensic Medicine, Shanghai Forensic Service Platform, Academy of Forensic Science, Shanghai 200063, P. R. China
| | - Ji Zhang
- Shanghai
Key Laboratory of Forensic Medicine, Shanghai Forensic Service Platform, Academy of Forensic Science, Shanghai 200063, P. R. China
| | - Kaifei Deng
- Shanghai
Key Laboratory of Forensic Medicine, Shanghai Forensic Service Platform, Academy of Forensic Science, Shanghai 200063, P. R. China
| | - Ping Huang
- Shanghai
Key Laboratory of Forensic Medicine, Shanghai Forensic Service Platform, Academy of Forensic Science, Shanghai 200063, P. R. China
- . Tel.: +86-021-52367986
| | - Zhenyuan Wang
- Department
of Forensic Pathology, College of Forensic Medicine, Xi’an Jiaotong University, Xi’an 710061, P. R. China
- . Tel.: +86-029-82655472
| |
Collapse
|
18
|
Silvis MJM, Kaffka genaamd Dengler SE, Odille CA, Mishra M, van der Kaaij NP, Doevendans PA, Sluijter JPG, de Kleijn DPV, de Jager SCA, Bosch L, van Hout GPJ. Damage-Associated Molecular Patterns in Myocardial Infarction and Heart Transplantation: The Road to Translational Success. Front Immunol 2020; 11:599511. [PMID: 33363540 PMCID: PMC7752942 DOI: 10.3389/fimmu.2020.599511] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 11/03/2020] [Indexed: 12/23/2022] Open
Abstract
In the setting of myocardial infarction (MI), ischemia reperfusion injury (IRI) occurs due to occlusion (ischemia) and subsequent re-establishment of blood flow (reperfusion) of a coronary artery. A similar phenomenon is observed in heart transplantation (HTx) when, after cold storage, the donor heart is connected to the recipient's circulation. Although reperfusion is essential for the survival of cardiomyocytes, it paradoxically leads to additional myocardial damage in experimental MI and HTx models. Damage (or danger)-associated molecular patterns (DAMPs) are endogenous molecules released after cellular damage or stress such as myocardial IRI. DAMPs activate pattern recognition receptors (PRRs), and set in motion a complex signaling cascade resulting in the release of cytokines and a profound inflammatory reaction. This inflammatory response is thought to function as a double-edged sword. Although it enables removal of cell debris and promotes wound healing, DAMP mediated signalling can also exacerbate the inflammatory state in a disproportional matter, thereby leading to additional tissue damage. Upon MI, this leads to expansion of the infarcted area and deterioration of cardiac function in preclinical models. Eventually this culminates in adverse myocardial remodeling; a process that leads to increased myocardial fibrosis, gradual further loss of cardiomyocytes, left ventricular dilation and heart failure. Upon HTx, DAMPs aggravate ischemic damage, which results in more pronounced reperfusion injury that impacts cardiac function and increases the occurrence of primary graft dysfunction and graft rejection via cytokine release, cardiac edema, enhanced myocardial/endothelial damage and allograft fibrosis. Therapies targeting DAMPs or PRRs have predominantly been investigated in experimental models and are potentially cardioprotective. To date, however, none of these interventions have reached the clinical arena. In this review we summarize the current evidence of involvement of DAMPs and PRRs in the inflammatory response after MI and HTx. Furthermore, we will discuss various current therapeutic approaches targeting this complex interplay and provide possible reasons why clinical translation still fails.
Collapse
Affiliation(s)
- Max J. M. Silvis
- Department of Cardiology, University Medical Center Utrecht, Utrecht, Netherlands
| | | | - Clémence A. Odille
- Department of Cardiology, Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Mudit Mishra
- Department of Cardiothoracic Surgery, University Medical Center Utrecht, Utrecht, Netherlands
| | - Niels P. van der Kaaij
- Department of Cardiothoracic Surgery, University Medical Center Utrecht, Utrecht, Netherlands
| | - Pieter A. Doevendans
- Department of Cardiology, University Medical Center Utrecht, Utrecht, Netherlands
- Central Military Hospital, Utrecht, University Medical Center Utrecht, Utrecht, Netherlands
- Netherlands Heart Institute, Utrecht, The Netherlands
| | - Joost P. G. Sluijter
- Department of Cardiology, Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht, Netherlands
- UMC Utrecht Regenerative Medicine Center, Circulatory Health Laboratory, University Utrecht, University Medical Center Utrecht, Utrecht, Netherlands
| | | | - Saskia C. A. de Jager
- Department of Cardiology, Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht, Netherlands
- Center for Translational Immunology, University Medical Center Utrecht, Netherlands
| | - Lena Bosch
- Department of Cardiology, University Medical Center Utrecht, Utrecht, Netherlands
- Department of Cardiology, Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Gerardus P. J. van Hout
- Department of Cardiology, University Medical Center Utrecht, Utrecht, Netherlands
- Department of Cardiology, Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht, Netherlands
| |
Collapse
|
19
|
Soni UK, Chadchan SB, Joshi A, Kumar V, Maurya VK, Verma RK, Jha RK. Poly(ADP-ribose) polymerase-2 is essential for endometrial receptivity and blastocyst implantation, and regulated by caspase-8. Mol Cell Endocrinol 2020; 518:110946. [PMID: 32679243 DOI: 10.1016/j.mce.2020.110946] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 06/29/2020] [Accepted: 07/07/2020] [Indexed: 12/21/2022]
Abstract
Embryo implantation is a very complex process and several factors play important roles. Using a mouse model, we investigated the functions of PARP-2 and caspase-8 during endometrial receptivity for blastocyst implantation. We found that PARP-2 was upregulated at the receptive stage's implantation region and predominantly expressed in the endometrial stromal region, but downregulated during pregnancy failure and pseudopregnancy. To reinforce the necessity of PARP-2 for embryo implantation, we pharmacologically inhibited PARP-2 'before' & 'after' embryo arrival and observed a reduction in blastocyst implantation. Conversely, elevated caspase-8 expression and activity during pseudopregnancy, delayed implantation, and embryo implantation failure conditions and decreased levels in the decidualization exhibited an inverse pattern with PARP-2, suggesting caspase-8 as a negative regulator for embryo implantation. In vitro caspase-8 downregulates the PARP-2 activity in the mouse endometrial epithelial and stromal cells. These data suggest that PARP-2 and its negative regulation by caspase-8 constitute a crucial step in embryo implantation.
Collapse
Affiliation(s)
- Upendra Kumar Soni
- Female Reproductive Biology Laboratory, Endocrinology Division, CSIR-Central Drug Research Institute, B.S. 10/1, Sector-10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India
| | - Sangappa Basanna Chadchan
- Female Reproductive Biology Laboratory, Endocrinology Division, CSIR-Central Drug Research Institute, B.S. 10/1, Sector-10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India
| | - Anubha Joshi
- Female Reproductive Biology Laboratory, Endocrinology Division, CSIR-Central Drug Research Institute, B.S. 10/1, Sector-10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India
| | - Vijay Kumar
- Female Reproductive Biology Laboratory, Endocrinology Division, CSIR-Central Drug Research Institute, B.S. 10/1, Sector-10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India
| | - Vineet Kumar Maurya
- Female Reproductive Biology Laboratory, Endocrinology Division, CSIR-Central Drug Research Institute, B.S. 10/1, Sector-10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India
| | - Raj Kumar Verma
- Female Reproductive Biology Laboratory, Endocrinology Division, CSIR-Central Drug Research Institute, B.S. 10/1, Sector-10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India
| | - Rajesh Kumar Jha
- Female Reproductive Biology Laboratory, Endocrinology Division, CSIR-Central Drug Research Institute, B.S. 10/1, Sector-10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India.
| |
Collapse
|
20
|
Park JC, Kim DH, Lee Y, Lee MC, Kim TK, Yim JH, Lee JS. Genome-wide identification and structural analysis of heat shock protein gene families in the marine rotifer Brachionus spp.: Potential application in molecular ecotoxicology. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY D-GENOMICS & PROTEOMICS 2020; 36:100749. [PMID: 33065474 DOI: 10.1016/j.cbd.2020.100749] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 09/26/2020] [Accepted: 09/26/2020] [Indexed: 01/07/2023]
Abstract
Heat shock proteins (Hsp) are class of conserved and ubiquitous stress proteins present in all living organisms from primitive to higher level. Various studies have demonstrated multiple cellular functions of Hsp in living organisms as an important biomarker in response to abiotic and biotic stressors including temperature, salinity, pH, hypoxia, environmental pollutants, and pathogens. However, full understanding on the mechanism and pathway involved in the induction of Hsp still remains challenging, especially in aquatic invertebrates. In this study, the entire Hsp family and subfamily members in the marine rotifers Brachionus spp., one of the cosmopolitan ecotoxicological model organisms, have been genome-widely identified. In Brachionus spp. Hsp family was comprised of Hsp10, small hsp (sHsp), Hsp40, Hsp60, Hsp70/105, and Hsp90, with highest number of genes found within Hsp40 DnaJ homolog subfamily C members. Also, the differences in the orientation of the conserved motifs within Hsp family may have induced differences in transcriptional gene modulation in response to thermal stress in Brachionus koreanus. Overall, Hsp family-specific domains were highly conserved in all three Brachionus spp., relative to Homo sapiens and across other animal taxa and these findings will be helpful for future ecotoxicological studies focusing on Hsps.
Collapse
Affiliation(s)
- Jun Chul Park
- Department of Biological Sciences, College of Science, Sungkyunkwan University, Suwon 16419, South Korea
| | - Duck-Hyun Kim
- Department of Biological Sciences, College of Science, Sungkyunkwan University, Suwon 16419, South Korea
| | - Yoseop Lee
- Department of Biological Sciences, College of Science, Sungkyunkwan University, Suwon 16419, South Korea
| | - Min-Chul Lee
- Department of Biological Sciences, College of Science, Sungkyunkwan University, Suwon 16419, South Korea
| | - Tai Kyoung Kim
- Division of Polar Life Science, Korea Polar Research Institute, Incheon 21990, South Korea
| | - Joung Han Yim
- Division of Polar Life Science, Korea Polar Research Institute, Incheon 21990, South Korea
| | - Jae-Seong Lee
- Department of Biological Sciences, College of Science, Sungkyunkwan University, Suwon 16419, South Korea.
| |
Collapse
|
21
|
Wu J, Chen S, Liu Y, Liu Z, Wang D, Cheng Y. Therapeutic perspectives of heat shock proteins and their protein-protein interactions in myocardial infarction. Pharmacol Res 2020; 160:105162. [DOI: 10.1016/j.phrs.2020.105162] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 08/03/2020] [Accepted: 08/17/2020] [Indexed: 12/26/2022]
|
22
|
Krishnan-Sivadoss I, Mijares-Rojas IA, Villarreal-Leal RA, Torre-Amione G, Knowlton AA, Guerrero-Beltrán CE. Heat shock protein 60 and cardiovascular diseases: An intricate love-hate story. Med Res Rev 2020; 41:29-71. [PMID: 32808366 PMCID: PMC9290735 DOI: 10.1002/med.21723] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 08/03/2020] [Accepted: 08/05/2020] [Indexed: 12/23/2022]
Abstract
Cardiovascular diseases (CVDs) are the result of complex pathophysiological processes in the tissues comprising the heart and blood vessels. Inflammation is the main culprit for the development of cardiovascular dysfunction, and it may be traced to cellular stress events including apoptosis, oxidative and shear stress, and cellular and humoral immune responses, all of which impair the system's structure and function. An intracellular chaperone, heat shock protein 60 (HSP60) is an intriguing example of a protein that may both be an ally and a foe for cardiovascular homeostasis; on one hand providing protection against cellular injury, and on the other triggering damaging responses through innate and adaptive immunity. In this review we will discuss the functions of HSP60 and its effects on cells and the immune system regulation, only to later address its implications in the development and progression of CVD. Lastly, we summarize the outcome of various studies targeting HSP60 as a potential therapeutic strategy for cardiovascular and other diseases.
Collapse
Affiliation(s)
- Indumathi Krishnan-Sivadoss
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Medicina Cardiovascular y Metabolómica, Monterrey, Nuevo León, México
| | - Iván A Mijares-Rojas
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Medicina Cardiovascular y Metabolómica, Monterrey, Nuevo León, México
| | - Ramiro A Villarreal-Leal
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Medicina Cardiovascular y Metabolómica, Monterrey, Nuevo León, México
| | - Guillermo Torre-Amione
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Medicina Cardiovascular y Metabolómica, Monterrey, Nuevo León, México.,Methodist DeBakey Heart and Vascular Center, The Methodist Hospital, Houston, Texas
| | - Anne A Knowlton
- Veterans Affairs Medical Center, Sacramento, California, USA.,Department of Internal Medicine, Molecular and Cellular Cardiology, Cardiovascular Division, University of California, Davis, California, USA.,Department of Pharmacology, University of California, Davis, California, USA
| | - C Enrique Guerrero-Beltrán
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Medicina Cardiovascular y Metabolómica, Monterrey, Nuevo León, México.,Tecnologico de Monterrey, Hospital Zambrano Hellion, TecSalud, Centro de Investigación Biomédica, San Pedro Garza García, Nuevo León, México
| |
Collapse
|
23
|
Dutta P, Bishayi B. Neutralization of TNF-α and IL-1β Regulates CXCL8 Production through CXCL8/CXCR1 Axis in Macrophages during Staphylococcus aureus Infection. Immunol Invest 2020; 50:700-725. [PMID: 32602757 DOI: 10.1080/08820139.2020.1787436] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Anti-cytokine therapy is widely acknowledged as an anti-inflammatory technique to treat varied infectious diseases. TNF-α and IL-1β are major cytokines that regulate every aspect of the inflammatory process. However, the effects of single or dual cytokine neutralization on S. aureus mediated CXCL8 secretion and CXCR1 expression in murine peritoneal macrophages remained noninvestigated. Thus we aimed to explore the effects of kinetic-dose dependent neutralization of TNF-α and IL-1β using specific anti-cytokine antibodies and its influential impact on the CXCL8/CXCR1 axis at different stages of S. aureus (30, 60, and 90 min) infection. The murine peritoneal macrophages were isolated and infected with viable S. aureus followed by subsequent addition of anti-TNF-α and anti-IL-1β into the medium. The treated cells were centrifuged and lysate and supernatant collected for various experiments. The ROS generation was measured and cytokine production was estimated by ELISA. The expression of TNFR1, IL-1R, CXCR1, signaling molecules (NF-κB and JNK) were evaluated by Western blot. The role of single or dual cytokine neutralization on intracellular bacterial phagocytosis had also been analyzed by confocal microscopy. Dual cytokine neutralization significantly suppressed ROS, cytokines, CXCL8 secretion, and intracellular bacterial count compared to single cytokine neutralization and it was more apparent at 90 min post S. aureus infection. There was a drastic reduction in TNFR1, IL-1R, and CXCR1 expression on macrophage surface due to reduced expression of downstream signaling molecules, NF-κB and JNK. Hence dual cytokine neutralization was more effectual compared to single cytokine neutralization in the downregulation of S. aureus induced CXCR1 expression.
Collapse
Affiliation(s)
- Puja Dutta
- Department of Physiology, Immunology laboratory, University of Calcutta, University Colleges of Science and Technology, Calcutta, India
| | - Biswadev Bishayi
- Department of Physiology, Immunology laboratory, University of Calcutta, University Colleges of Science and Technology, Calcutta, India
| |
Collapse
|
24
|
Seclì L, Sorge M, Morotti A, Brancaccio M. Blocking Extracellular Chaperones to Improve Cardiac Regeneration. Front Bioeng Biotechnol 2020; 8:411. [PMID: 32528937 PMCID: PMC7264090 DOI: 10.3389/fbioe.2020.00411] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 04/14/2020] [Indexed: 12/24/2022] Open
Abstract
Chronic or acute insults to the myocardium are responsible for the onset of cardiomyopathy and heart failure. Due to the poor regenerative ability of the human adult heart, the survival of cardiomyocytes is a prerequisite to support heart function. Chaperone proteins, by regulating sarcomeric protein folding, function, and turnover in the challenging environment of the beating heart, play a fundamental role in myocardial physiology. Nevertheless, a number of evidences indicate that, under stress conditions or during cell damage, myocardial cells release chaperone proteins that, from the extracellular milieu, play a detrimental function, by perpetuating inflammation and inducing cardiomyocyte apoptosis. Blocking the activity of extracellular chaperones has been proven to have beneficial effects on heart function in preclinical models of myocardial infarction and cardiomyopathy. The application of this approach in combination with tissue engineering strategies may represent a future innovation in cardiac regenerative medicine.
Collapse
Affiliation(s)
- Laura Seclì
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Matteo Sorge
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Alessandro Morotti
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Mara Brancaccio
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| |
Collapse
|
25
|
Abstract
In the infarcted myocardium, cardiomyocyte necrosis triggers an intense inflammatory reaction that not only is critical for cardiac repair, but also contributes to adverse remodeling and to the pathogenesis of heart failure. Both CC and CXC chemokines are markedly induced in the infarcted heart, bind to endothelial glycosaminoglycans, and regulate leukocyte trafficking and function. ELR+ CXC chemokines (such as CXCL8) control neutrophil infiltration, whereas CC chemokines (such as CCL2) mediate recruitment of mononuclear cells. Moreover, some members of the chemokine family (such as CXCL10 and CXCL12) may mediate leukocyte-independent actions, directly modulating fibroblast and vascular cell function. This review manuscript discusses our understanding of the role of the chemokines in regulation of injury, repair, and remodeling following myocardial infarction. Although several chemokines may be promising therapeutic targets in patients with myocardial infarction, clinical implementation of chemokine-based therapeutics is hampered by the broad effects of the chemokines in both injury and repair.
Collapse
|
26
|
Duan Y, Tang H, Mitchell-Silbaugh K, Fang X, Han Z, Ouyang K. Heat Shock Protein 60 in Cardiovascular Physiology and Diseases. Front Mol Biosci 2020; 7:73. [PMID: 32426370 PMCID: PMC7203681 DOI: 10.3389/fmolb.2020.00073] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 03/31/2020] [Indexed: 01/01/2023] Open
Abstract
Heat shock protein 60 (HSP60) is a highly conserved protein abundantly expressed in both prokaryotic and eukaryotic cells. In mammals, HSP60 has been primarily considered to reside in the mitochondria, where HSP60 and HSP10 form a complex and facilitate mitochondrial protein folding. However, HSP60 is also observed in the cytoplasm, the plasma membrane, and the extracellular space. HSP60 regulates a broad spectrum of cellular events including protein trafficking, peptide hormone signaling, cell survival, cell proliferation, inflammation, and immunization. In the cardiovascular system, growing evidence indicates that HSP60 could not only play an important role under physiological conditions, but also regulate the initiation and progression of heart failure and atherosclerosis. In this review, we focus on recent progress in understanding the function of HSP60 in cardiomyocytes, endothelial cells, and vascular smooth muscle cells (VSMCs), respectively, and discuss the related signaling pathways that have been found in these cells, so as to illustrate the role of HSP60 in the development of cardiovascular disease.
Collapse
Affiliation(s)
- Yaoyun Duan
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Huayuan Tang
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Kali Mitchell-Silbaugh
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Xi Fang
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Zhen Han
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Kunfu Ouyang
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| |
Collapse
|
27
|
Vadivel S, Vincent P, Sekaran S, Visaga Ambi S, Muralidar S, Selvaraj V, Palaniappan B, Thirumalai D. Inflammation in myocardial injury- Stem cells as potential immunomodulators for myocardial regeneration and restoration. Life Sci 2020; 250:117582. [PMID: 32222465 DOI: 10.1016/j.lfs.2020.117582] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Revised: 03/14/2020] [Accepted: 03/20/2020] [Indexed: 12/11/2022]
Abstract
The ineffective immunosuppressant's and targeted strategies to neutralize inflammatory mediators have worsened the scenario of heart failure and have opened many questions for debate. Stem cell therapy has proven to be a promising approach for treating heart following myocardial infarction (MI). Adult stem cells, induced pluripotent stem cells and embryonic stem cells are possible cell types and have successfully shown to regenerate damaged myocardial tissue in pre-clinical and clinical studies. Current implications of using mesenchymal stem cells (MSCs) owing to their immunomodulatory functions and paracrine effects could serve as an effective alternative treatment option for rejuvenating the heart post MI. The major setback associated with the use of MSCs is reduced cell retention, engraftment and decreased effectiveness. With a few reports on understanding the role of inflammation and its dual effects on the structure and function of heart, this review focuses on these missing insights and further exemplifies the role of MSCs as an alternative therapy in treating the pathological consequences in myocardial infarction (MI).
Collapse
Affiliation(s)
- Sajini Vadivel
- School of Chemical and Biotechnology, SASTRA Deemed-to-be-University, Thanjavur 613 401, Tamil Nadu, India
| | - Preethi Vincent
- School of Chemical and Biotechnology, SASTRA Deemed-to-be-University, Thanjavur 613 401, Tamil Nadu, India
| | - Saravanan Sekaran
- School of Chemical and Biotechnology, SASTRA Deemed-to-be-University, Thanjavur 613 401, Tamil Nadu, India.
| | - Senthil Visaga Ambi
- School of Chemical and Biotechnology, SASTRA Deemed-to-be-University, Thanjavur 613 401, Tamil Nadu, India.
| | - Shibi Muralidar
- School of Chemical and Biotechnology, SASTRA Deemed-to-be-University, Thanjavur 613 401, Tamil Nadu, India
| | - Vimalraj Selvaraj
- Centre for Biotechnology, Anna University, Chennai 600 025, Tamil Nadu, India
| | - Balamurugan Palaniappan
- School of Chemical and Biotechnology, SASTRA Deemed-to-be-University, Thanjavur 613 401, Tamil Nadu, India
| | - Diraviyam Thirumalai
- School of Chemical and Biotechnology, SASTRA Deemed-to-be-University, Thanjavur 613 401, Tamil Nadu, India
| |
Collapse
|
28
|
Yang M, Hua T, Yang Z, Chen L, Zou Y, Huang X, Li J. The Protective Effect of rhBNP on Postresuscitation Myocardial Dysfunction in a Rat Cardiac Arrest Model. BIOMED RESEARCH INTERNATIONAL 2020; 2020:6969053. [PMID: 32149124 PMCID: PMC7049428 DOI: 10.1155/2020/6969053] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 12/23/2019] [Accepted: 01/20/2020] [Indexed: 12/25/2022]
Abstract
PURPOSE We investigated the protective effects and the underlying mechanisms through which recombinant human brain natriuretic peptide (rhBNP) acts on postresuscitation myocardial dysfunction (PRMD) in the cardiac arrest (CA) model. METHODS Ventricular fibrillation was induced and untreated for 6 min. And the time of cardiopulmonary resuscitation was 8 min, after which defibrillation was attempted in this rat model. 24 Sprague Dawley rats (450-550g) were randomized into cardiopulmonary resuscitation (CPR) + rhBNP and CPR + placebo groups after restoration of spontaneous circulation (ROSC). rhBNP was infused at PR 30 min (loading dose: 1.5 µg/kg, 3 min; maintenance dose: 0.01 µg/kg, 3 min; maintenance dose: 0.01 α (TNF-α (TNF-α (TNF-κB (NF-κB (NF. RESULTS The administration of rhBNP attenuated the severity of PRMD and myocardial tissue injuries, with improvement of MAP (mean arterial blood pressure), ETCO2 (end-tidal CO2), serum level of NT-proBNP, EF, CO, and MPI values. The serum levels and protein expression levels in myocardial tissue of IL-6 and TNF-α (TNF-κB (NF. CONCLUSION Our research demonstrated that the administration of rhBNP attenuated the severity of PRMD and myocardial tissue injuries and increased the 24 h survival rate in this CA model. rhBNP administration also reduced the serum and myocardial tissue levels of IL-6 and TNF-α after ROSC, likely due to the suppression of the TLR4/NF-κB signaling pathway and the regulation of inflammatory mediator secretion.α (TNF-κB (NF.
Collapse
Affiliation(s)
- Min Yang
- Department of Basic and Clinical Pharmacology, School of Pharmacy, Anhui Medical University, Hefei, China
- Department of Intensive Care Unit, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Cardiopulmonary Cerebral Resuscitation, Sun Yat-sen University, Guangzhou, China
| | - Tianfeng Hua
- Department of Intensive Care Unit, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Cardiopulmonary Cerebral Resuscitation, Sun Yat-sen University, Guangzhou, China
| | - Zhengfei Yang
- Institute of Cardiopulmonary Cerebral Resuscitation, Sun Yat-sen University, Guangzhou, China
| | - Limin Chen
- Department of Intensive Care Unit, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yangyang Zou
- Department of Intensive Care Unit, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xiaohui Huang
- Department of Basic and Clinical Pharmacology, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Jun Li
- Department of Basic and Clinical Pharmacology, School of Pharmacy, Anhui Medical University, Hefei, China
| |
Collapse
|
29
|
Ferenčić A, Cuculić D, Stemberga V, Šešo B, Arbanas S, Jakovac H. Left ventricular hypertrophy is associated with overexpression of HSP60, TLR2, and TLR4 in the myocardium. Scand J Clin Lab Invest 2020; 80:236-246. [PMID: 32057259 DOI: 10.1080/00365513.2020.1725977] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Left ventricular hypertrophy is a common adaptive response to increased cardiac workload. Cardiomyocytes growth and increase in contractile force are conditioned by sufficient energy production, which implies appropriate mitochondrial function. The 60 kDa heat shock protein (HSP60) is a chaperone essential for mitochondrial proteostasis, but when translocates from mitochondria, it can also act as a potent inflammatory mediator binding to toll-like receptors (TLRs). In this study, we aimed to compare the expression pattern of HSP60, TLR2, and TLR4 in hypertrophic vs non-hypertrophic, normal human myocardium. We further examined whether HSP60 in situ binds to TLRs in hypertrophic myocardial tissue. In addition, expression of activated downstream targets of TLR 2/4 pathways was also evaluated.For this purpose, immunohistochemical expression analyses were performed on myocardial tissue samples obtained during the autopsy of human subjects in which left ventricular hypertrophy was the only cardiopathological finding and had died from sudden cardiac death, as well as from the subjects without any cardiac pathology, that died by unnatural death (accident or suicide). Double immunofluorescence was used to examine HSP60 translocation, while proximity ligation assay (PLA) was performed to assess HSP60 and TLRs interactions.Hypertrophic myocardium showed significantly higher expression of HSP60, TLR2, and TLR4 compared to normal myocardium. Furthermore, in hypertrophic cardiomyocytes, we found membrane translocation of HSP60 and signs of HSP60/TLR interactions.Conclusion: The obtained data point to an important supportive role of HSP60 in adaptive cardiomyocytes growth, while concomitant induction of TLR2 and TLR4 candidates HSP60-TLRs interactions as an early events during pathogenesis of secondary complications consequently to the left ventricular hypertrophy.
Collapse
Affiliation(s)
- Antun Ferenčić
- Department of Forensic Medicine and Criminalistics, University of Rijeka, Faculty of Medicine, Rijeka, Croatia
| | - Dražen Cuculić
- Department of Forensic Medicine and Criminalistics, University of Rijeka, Faculty of Medicine, Rijeka, Croatia
| | - Valter Stemberga
- Department of Forensic Medicine and Criminalistics, University of Rijeka, Faculty of Medicine, Rijeka, Croatia
| | - Bernard Šešo
- Department of Clinical, Health and Organisational Psychology, Clinical Hospital Centre Rijeka, Rijeka, Croatia
| | - Silvia Arbanas
- Department of Forensic Medicine and Criminalistics, University of Rijeka, Faculty of Medicine, Rijeka, Croatia
| | - Hrvoje Jakovac
- Department of Physiology and Immunology, University of Rijeka, Faculty of Medicine, Rijeka, Croatia
| |
Collapse
|
30
|
Dutta P, Sultana S, Dey R, Bishayi B. Regulation of Staphylococcus aureus-induced CXCR1 expression via inhibition of receptor mobilization and receptor shedding during dual receptor (TNFR1 and IL-1R) neutralization. Immunol Res 2020; 67:241-260. [PMID: 31290001 DOI: 10.1007/s12026-019-09083-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Our earlier studies proposed a radically new idea suggesting interdependency between TNF-α/TNFR1 and IL-1β/IL-1R pathways in modulation of Staphylococcus aureus-induced CXCL8/CXCR1 axis. However, the effects of inhibition of cytokine receptor mobilization at intracellular level and surface TNFR1 and IL-1R shedding on S. aureus-induced CXCR1 expression have not been studied so far in peritoneal macrophages. This study aimed to investigate the role of inhibition of receptor mobilization from the intracellular pool (using brefeldin A) and surface receptor shedding (using TAPI-1) on CXCR1 expression during dual receptor (TNFR1 plus IL-1R) neutralization in peritoneal macrophages isolated from wild-type Swiss Albino mice. Release of superoxide anion, nitric oxide, and hydrogen peroxide was measured and cytokine production was done by ELISA. Expression of surface receptors (TNFR1, IL-1R, and CXCR1) and inflammatory mediators was studied by Western blot. It was observed that S. aureus-infected macrophages showed elevated ROS production, secretion of TNF-α, IL-1β, and CXCL8, along with increased expression of surface receptors (TNFR1, IL-1R, and CXCR1), and inflammatory markers (iNOS and COX-2) compared with control or treated groups (p < 0.05). However, prior treatment of macrophages with BFA or TAPI-1 in the presence of anti-TNFR1 antibody and IRAP during S. aureus infection showed significant reduction of all these parameters (p < 0.05). We can conclude that targeting of TNFR1 and IL-1R (with major focus on surface expression study) either through blockage of intracellular receptor trafficking pathway or via surface receptor shedding diminishes TNFR1/IL-1R interaction and consequently downregulates CXCR1 expression along with inflammatory signalling pathways during bacterial infections.
Collapse
Affiliation(s)
- Puja Dutta
- Department of Physiology, Immunology Laboratory, University of Calcutta, University Colleges of Science and Technology, 92 APC Road, Calcutta, West Bengal, 700009, India
| | - Sahin Sultana
- Department of Physiology, Immunology Laboratory, University of Calcutta, University Colleges of Science and Technology, 92 APC Road, Calcutta, West Bengal, 700009, India
| | - Rajen Dey
- Department of Physiology, Immunology Laboratory, University of Calcutta, University Colleges of Science and Technology, 92 APC Road, Calcutta, West Bengal, 700009, India
| | - Biswadev Bishayi
- Department of Physiology, Immunology Laboratory, University of Calcutta, University Colleges of Science and Technology, 92 APC Road, Calcutta, West Bengal, 700009, India.
| |
Collapse
|
31
|
MPMBP down-regulates Toll-like receptor (TLR) 2 ligand-induced proinflammatory cytokine production by inhibiting NF-κB but not AP-1 activation. Int Immunopharmacol 2020; 79:106085. [DOI: 10.1016/j.intimp.2019.106085] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 11/23/2019] [Accepted: 11/25/2019] [Indexed: 12/21/2022]
|
32
|
Song L, Wang Q, Chai Z, Zhang C, Wang J, Ma J, Guo J, Yu J, Xiao B, Han G, Huang J, Ma C. Anhydrosafflor Yellow B alleviates brain injury of acute permanent cerebral ischemia in rats by anti-inflammatory mechanism. ALL LIFE 2020. [DOI: 10.1080/26895293.2020.1750492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
Affiliation(s)
- Lijuan Song
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine, Research Center of Neurobiology, Shanxi University of Chinese Medicine, Jinzhong, People’s Republic of China
- Department of Physiology, Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Qing Wang
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine, Research Center of Neurobiology, Shanxi University of Chinese Medicine, Jinzhong, People’s Republic of China
| | - Zhi Chai
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine, Research Center of Neurobiology, Shanxi University of Chinese Medicine, Jinzhong, People’s Republic of China
| | - Ce Zhang
- Department of Physiology, Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Jing Wang
- Department of Neurology, First Affiliated Hospital, Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Jingping Ma
- Department of Neurology, First Affiliated Hospital, Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Junhong Guo
- Department of Neurology, First Affiliated Hospital, Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Jiezhong Yu
- Institute of Brain Science, Shanxi Key Laboratory of Inflammatory Neurodegenerative Diseases, Department of Neurology, First Affiliated Hospital, Shanxi Datong University, Datong, People’s Republic of China
| | - Baoguo Xiao
- Institute of Neurology, Huashan Hospital, Fudan University, Shanghai, People’s Republic of China
| | - Guangyuan Han
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine, Research Center of Neurobiology, Shanxi University of Chinese Medicine, Jinzhong, People’s Republic of China
| | - Jianjun Huang
- Department of Neurosurgery, General Hospital, Datong Coalmine Group, Datong, People’s Republic of China
| | - Cungen Ma
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine, Research Center of Neurobiology, Shanxi University of Chinese Medicine, Jinzhong, People’s Republic of China
- Department of Neurology, First Affiliated Hospital, Shanxi Medical University, Taiyuan, People’s Republic of China
- Institute of Brain Science, Shanxi Key Laboratory of Inflammatory Neurodegenerative Diseases, Department of Neurology, First Affiliated Hospital, Shanxi Datong University, Datong, People’s Republic of China
| |
Collapse
|
33
|
Mitsui K, Ikedo T, Kamio Y, Furukawa H, Lawton MT, Hashimoto T. TLR4 (Toll-Like Receptor 4) Mediates the Development of Intracranial Aneurysm Rupture. Hypertension 2019; 75:468-476. [PMID: 31865791 DOI: 10.1161/hypertensionaha.118.12595] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Inflammation is emerging as a critical factor in the pathophysiology of intracranial aneurysm. TLR4 (toll-like receptor 4) contributes not only to the innate immune responses but also to the inflammatory processes associated with vascular disease. Therefore, we examined the contribution of the TLR4 pathway to the development of the rupture of intracranial aneurysm. We used a mouse model of intracranial aneurysm. TLR4 inhibition significantly reduced the development of aneurysmal rupture. In addition, the rupture rate and levels of proinflammatory cytokines were lower in TLR4 knockout mice than the control littermates. Macrophage/monocyte-specific TLR4 knockout mice had a lower rupture rate than the control littermate mice. Moreover, the deficiency of MyD88 (myeloid differentiation primary-response protein 88), a key mediator of TLR4, reduced the rupture rate. These findings suggest that the TLR4 pathway promotes the development of intracranial aneurysmal rupture by accelerating inflammation in aneurysmal walls. Inhibition of the TLR4 pathway in inflammatory cells may be a promising approach for the prevention of aneurysmal rupture and subsequent subarachnoid hemorrhage.
Collapse
Affiliation(s)
- Kazuha Mitsui
- From the Department of Neurosurgery and Neurobiology, Barrow Aneurysm and AVM Research Center, Barrow Neurological Institute, Phoenix, AZ
| | - Taichi Ikedo
- From the Department of Neurosurgery and Neurobiology, Barrow Aneurysm and AVM Research Center, Barrow Neurological Institute, Phoenix, AZ
| | - Yoshinobu Kamio
- From the Department of Neurosurgery and Neurobiology, Barrow Aneurysm and AVM Research Center, Barrow Neurological Institute, Phoenix, AZ
| | - Hajime Furukawa
- From the Department of Neurosurgery and Neurobiology, Barrow Aneurysm and AVM Research Center, Barrow Neurological Institute, Phoenix, AZ
| | - Michael T Lawton
- From the Department of Neurosurgery and Neurobiology, Barrow Aneurysm and AVM Research Center, Barrow Neurological Institute, Phoenix, AZ
| | - Tomoki Hashimoto
- From the Department of Neurosurgery and Neurobiology, Barrow Aneurysm and AVM Research Center, Barrow Neurological Institute, Phoenix, AZ
| |
Collapse
|
34
|
Finger S, Knorr M, Molitor M, Schüler R, Garlapati V, Waisman A, Brandt M, Münzel T, Bopp T, Kossmann S, Karbach S, Wenzel P. A sequential interferon gamma directed chemotactic cellular immune response determines survival and cardiac function post-myocardial infarction. Cardiovasc Res 2019; 115:1907-1917. [PMID: 30949687 DOI: 10.1093/cvr/cvz092] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 03/15/2019] [Accepted: 04/02/2019] [Indexed: 12/17/2023] Open
Abstract
AIMS Myelomonocytic cells are critical in injury and healing post-myocardial infarction (MI). Mechanisms of regulation, however, are incompletely understood. The aim of the study was to elucidate the role of interferon gamma (IFN-γ) in the orchestrated inflammatory response in a murine model of MI. METHODS AND RESULTS MI was induced in 8- to 12-week-old male mice (C57BL/6 background) by permanent ligation of the left anterior descending (LAD) coronary artery. Lysozyme M (LysM)+ cell-depleted LysMiDTR transgenic mice displayed a reduced influx of CD45.2+/CD3-/CD11b+/Gr-1high neutrophils into infarcted myocardium 1 day post-MI compared with infarcted controls, paralleled by decreased cardiac mRNA levels of IFN-γ and tumour necrosis factor alpha (TNF-α). Mortality after MI was significantly increased in LysM+ cell-depleted mice within 28 days post-MI. To more specifically address the role of neutrophils, we depleted C57BL/6 mice with a monoclonal anti-Gr-1 antibody and found increased mortality, deteriorated cardiac function as well as decreased cardiac IFN-γ mRNA expression early after MI. Ccl2, Cxcl1, Cx3cl1, and Il12b mRNA were reduced 3 days after MI, as was the amount of CD11b+/Ly-6G-/Ly-6Chigh inflammatory monocytes. LAD-ligated Cramp-/- mice lacking cathelicidin important in neutrophil-dependent monocyte chemotaxis as well as IFNγ-/- and TNFα-/- mice phenocopied Gr-1+ cell-depleted mice, supporting a regulatory role of IFN-γ impacting on both the sequence of inflammatory cell invasion and cardiac outcome early after MI. The use of conditional IFN-γ receptor deficient mice indicated a direct effect of IFN-γ on LysM+ cells in cardiac injury post-MI. Using IFN-γ reporter mice and flow cytometry, we identified cardiac lymphoid cells (CD4+ and CD8+ T cells and natural killer cells) as primary source of this cytokine in the cardiac inflammatory response post-MI. CONCLUSION IFN-γ directs a sequential chemotactic cellular immune response and determines survival and cardiac function post-MI.
Collapse
Affiliation(s)
- Stefanie Finger
- Center for Cardiology-Cardiology I, University Medical Center Mainz, Langenbeckstraße 1, Mainz, Germany
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Langenbeckstraße 1, Mainz, Germany
- German Center for Cardiovascular Research (DZHK)-Partner site RheinMain, University Medical Center Mainz, Langenbeckstraße 1, Mainz, Germany
| | - Maike Knorr
- Center for Cardiology-Cardiology I, University Medical Center Mainz, Langenbeckstraße 1, Mainz, Germany
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Langenbeckstraße 1, Mainz, Germany
| | - Michael Molitor
- Center for Cardiology-Cardiology I, University Medical Center Mainz, Langenbeckstraße 1, Mainz, Germany
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Langenbeckstraße 1, Mainz, Germany
- German Center for Cardiovascular Research (DZHK)-Partner site RheinMain, University Medical Center Mainz, Langenbeckstraße 1, Mainz, Germany
| | - Rebecca Schüler
- Center for Cardiology-Cardiology I, University Medical Center Mainz, Langenbeckstraße 1, Mainz, Germany
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Langenbeckstraße 1, Mainz, Germany
- Institute for Molecular Medicine, University Medical Center Mainz, Langenbeckstraße 1, Mainz, Germany
| | - Venkata Garlapati
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Langenbeckstraße 1, Mainz, Germany
- German Center for Cardiovascular Research (DZHK)-Partner site RheinMain, University Medical Center Mainz, Langenbeckstraße 1, Mainz, Germany
| | - Ari Waisman
- Institute for Molecular Medicine, University Medical Center Mainz, Langenbeckstraße 1, Mainz, Germany
| | - Moritz Brandt
- Center for Cardiology-Cardiology I, University Medical Center Mainz, Langenbeckstraße 1, Mainz, Germany
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Langenbeckstraße 1, Mainz, Germany
| | - Thomas Münzel
- Center for Cardiology-Cardiology I, University Medical Center Mainz, Langenbeckstraße 1, Mainz, Germany
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Langenbeckstraße 1, Mainz, Germany
- German Center for Cardiovascular Research (DZHK)-Partner site RheinMain, University Medical Center Mainz, Langenbeckstraße 1, Mainz, Germany
| | - Tobias Bopp
- Institute for Immunology, University Medical Center Mainz, Langenbeckstraße, 1, Mainz, Germany
| | - Sabine Kossmann
- Center for Cardiology-Cardiology I, University Medical Center Mainz, Langenbeckstraße 1, Mainz, Germany
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Langenbeckstraße 1, Mainz, Germany
- The Heart Research Institute, 7 Eliza Street, Newtown, NSW, Australia
| | - Susanne Karbach
- Center for Cardiology-Cardiology I, University Medical Center Mainz, Langenbeckstraße 1, Mainz, Germany
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Langenbeckstraße 1, Mainz, Germany
- German Center for Cardiovascular Research (DZHK)-Partner site RheinMain, University Medical Center Mainz, Langenbeckstraße 1, Mainz, Germany
| | - Philip Wenzel
- Center for Cardiology-Cardiology I, University Medical Center Mainz, Langenbeckstraße 1, Mainz, Germany
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Langenbeckstraße 1, Mainz, Germany
- German Center for Cardiovascular Research (DZHK)-Partner site RheinMain, University Medical Center Mainz, Langenbeckstraße 1, Mainz, Germany
| |
Collapse
|
35
|
Williams B, Neder J, Cui P, Suen A, Tanaka K, Zou L, Chao W. Toll-like receptors 2 and 7 mediate coagulation activation and coagulopathy in murine sepsis. J Thromb Haemost 2019; 17:1683-1693. [PMID: 31211901 PMCID: PMC7197442 DOI: 10.1111/jth.14543] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 06/10/2019] [Indexed: 01/10/2023]
Abstract
BACKGROUND Sepsis is a life-threatening condition often manifested as marked inflammation and severe coagulopathy. Toll-like receptors (TLRs) play a pivotal role in inflammation, organ dysfunction and mortality in animal sepsis. OBJECTIVES To investigate the role of TLR signaling in mediating sepsis-induced coagulopathy (SIC) in a mouse model. METHODS Polymicrobial sepsis was created by cecal ligation and puncture (CLP) or fecal slurry peritoneal injection. To quantify global clotting function, two viscoelastic assays were performed with rotational thromboelastometry, and the results were presented as maximum clot firmness (MCF): (a) EXTEM to test tissue factor (TF)-initiated clot formation; and (b) FIBTEM to test EXTEM in the presence of a platelet inhibitor, cytochalasin D. Plasma coagulation factors were quantified with ELISA. TF gene expression and protein expression were determined with real-time quantitative reverse transcription PCR and flow cytometry, respectively. RESULTS Between 4 and 24 hours after CLP surgery, wild-type mice showed significant MCF reduction in both EXTEM and FIBTEM tests. This was accompanied by marked thrombocytopenia and a significant increase in the levels of plasminogen activator inhibitor-1, plasma TF, and D-dimer. In comparison, TLR2-/- and TLR7-/- CLP mice showed preserved MCF and platelet counts, and near-normal plasma TF levels. Bone marrow-derived macrophages treated with a TLR2 agonist Pam3cys-Ser-(Lys)4 (Pam3cys) or a TLR7 agonist (R837) showed marked increases in TF gene expression and protein expression. MicroRNA-146a, a newly identified proinflammatory mediator that is upregulated during sepsis, induced TF production via a TLR7-dependent mechanism. CONCLUSIONS Murine sepsis leads to an increased procoagulant response, thrombocytopenia, and global coagulopathy. TLR2 and TLR7 play an important role in procoagulant production and in SIC.
Collapse
Affiliation(s)
- Brittney Williams
- Translational Research Program, Department of Anesthesiology & Center for Shock Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, Maryland
| | - Jessica Neder
- Translational Research Program, Department of Anesthesiology & Center for Shock Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, Maryland
| | - Ping Cui
- Translational Research Program, Department of Anesthesiology & Center for Shock Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, Maryland
| | - Andrew Suen
- Translational Research Program, Department of Anesthesiology & Center for Shock Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, Maryland
| | - Kenichi Tanaka
- Translational Research Program, Department of Anesthesiology & Center for Shock Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, Maryland
| | - Lin Zou
- Translational Research Program, Department of Anesthesiology & Center for Shock Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, Maryland
| | - Wei Chao
- Translational Research Program, Department of Anesthesiology & Center for Shock Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, Maryland
| |
Collapse
|
36
|
Rotter Sopasakis V, Sandstedt J, Johansson M, Lundqvist A, Bergström G, Jeppsson A, Mattsson Hultén L. Toll-like receptor-mediated inflammation markers are strongly induced in heart tissue in patients with cardiac disease under both ischemic and non-ischemic conditions. Int J Cardiol 2019; 293:238-247. [PMID: 31230935 DOI: 10.1016/j.ijcard.2019.06.033] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 05/29/2019] [Accepted: 06/12/2019] [Indexed: 01/04/2023]
Abstract
BACKGROUND A sustained low grade inflammatory state is a recognized feature of various diseases, including cardiovascular disease. This state of chronic inflammation involves activation of Toll-like receptor (TLR) signaling. However, little is known regarding the genetic profile of TLR components in cardiac tissue from patients with cardiac disease. METHODS In this study we investigated the genetic profile of 84 TLR markers in a unique set of cardiac tissue from patients that had undergone either coronary artery bypass grafting (CABG) or aortic valve replacement (AVR). In addition, we compared the gene data from the cardiac tissue with the same gene profile in blood as well as circulating cytokines to elucidate possible targets in blood that could be used to estimate the inflammatory state of the heart in cardiac disease. RESULTS We found a marked upregulation of TLR-induced inflammation in cardiac tissue from both patient groups compared to healthy controls. The inflammation appeared to be primarily mediated through TLR1, 3, 7, 8 and 10, resulting in a marked induction of mediators of the innate immune response. Furthermore, the gene expression data in combination with unbiased multivariate analysis suggested a difference in inflammatory response in ischemic cardiac tissue compared to non-ischemic cardiac tissue. Serum levels of IL-13 were significantly elevated in both CABG and AVR patients compared to controls, whereas other cytokines did not appear to coincide with cardiac TLR-induced inflammation. CONCLUSIONS We propose that cardiac disease in humans may be mediated by local cardiac TLR signaling under both ischemic and non-ischemic conditions.
Collapse
Affiliation(s)
- Victoria Rotter Sopasakis
- Department of Clinical Chemistry, Sahlgrenska University Hospital and Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, SE-413 45 Gothenburg, Sweden.
| | - Joakim Sandstedt
- Department of Clinical Chemistry, Sahlgrenska University Hospital and Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, SE-413 45 Gothenburg, Sweden
| | - Michaela Johansson
- Department of Clinical Chemistry, Sahlgrenska University Hospital and Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, SE-413 45 Gothenburg, Sweden
| | - Annika Lundqvist
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, SE-413 45 Gothenburg, Sweden
| | - Göran Bergström
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, SE-413 45 Gothenburg, Sweden
| | - Anders Jeppsson
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, SE-413 45 Gothenburg, Sweden; Department of Cardiothoracic Surgery, Sahlgrenska University Hospital, SE-413 45 Gothenburg, Sweden
| | - Lillemor Mattsson Hultén
- Department of Clinical Chemistry, Sahlgrenska University Hospital and Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, SE-413 45 Gothenburg, Sweden; Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, SE-413 45 Gothenburg, Sweden
| |
Collapse
|
37
|
Younis NS, Mohamed ME. β-Caryophyllene as a Potential Protective Agent Against Myocardial Injury: The Role of Toll-Like Receptors. Molecules 2019; 24:molecules24101929. [PMID: 31109132 PMCID: PMC6572120 DOI: 10.3390/molecules24101929] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 05/10/2019] [Accepted: 05/16/2019] [Indexed: 12/22/2022] Open
Abstract
Myocardial infarction (MI) remains one of the major causes of mortality around the world. A possible mechanism involved in myocardial infarction is the engagement of Toll-like receptors (TLRs). This study was intended to discover the prospective cardioprotective actions of β-caryophyllene, a natural sesquiterpene, to ameliorate isoproterenol (ISO)-induced myocardial infarction through HSP-60/TLR/MyD88/NFκB pathway. β-Caryophyllene (100 or 200 mg/kg/day orally) was administered for 21 days then MI was induced via ISO (85 mg/kg, subcutaneous) on 20th and 21st days. The results indicated that ISO induced a significant infarcted area associated with several alterations in the electrocardiogram (ECG) and blood pressure (BP) indices and caused an increase in numerous cardiac indicators such as creatine phosphokinase (CPK), creatine kinase-myocardial bound (CK-MB), lactate dehydrogenase (LDH), and cardiac tropinine T (cTnT). In addition, ISO significantly amplified heat shock protein 60 (HSP-60) and other inflammatory markers, such as TNF-α, IL-Iβ, and NFκB, and affected TLR2 and TLR4 expression and their adaptor proteins; Myeloid differentiation primary response 88 (MYD88), and TIR-domain-containing adapter-inducing interferon-β (TRIF). On the other hand, consumption of β-caryophyllene significantly reversed the infarcted size, ECG and BP alterations, ameliorated the ISO elevation in cardiac indicators; it also notably diminished HSP-60, and subsequently TLR2, TLR4, MYD88, and TRIF expression, with a substantial reduction in inflammatory mediator levels. This study revealed the cardioprotective effect of β-caryophyllene against MI through inhibiting HSP-60/TLR/MyD88/NFκB signaling pathways.
Collapse
Affiliation(s)
- Nancy S Younis
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, 31982 Al-Ahsa, Saudi Arabia.
- Department of Pharmacology, Zagazig University, Zagazig 44519, Egypt.
| | - Maged E Mohamed
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, 31982 Al-Ahsa, Saudi Arabia.
- Department of Pharmacognosy, College of Pharmacy, Zagazig University, Zagazig 44519, Egypt.
| |
Collapse
|
38
|
Jewhurst K, McLaughlin KA. Recovery of the Xenopus laevis heart from ROS-induced stress utilizes conserved pathways of cardiac regeneration. Dev Growth Differ 2019; 61:212-227. [PMID: 30924142 DOI: 10.1111/dgd.12602] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 01/31/2019] [Accepted: 02/12/2019] [Indexed: 01/22/2023]
Abstract
Urodele amphibians and some fish are capable of regenerating up to a quarter of their heart tissue after cardiac injury. While many anuran amphibians like Xenopus laevis are not capable of such feats, they are able to repair lesser levels of cardiac damage, such as that caused by oxidative stress, to a far greater degree than mammals. Using an optogenetic stress induction model that utilizes the protein KillerRed, we have investigated the extent to which mechanisms of cardiac regeneration are conserved during the restoration of normal heart morphology post oxidative stress in X. laevis tadpoles. We focused particularly on the processes of cardiomyocyte proliferation and dedifferentiation, as well as the pathways that facilitate the regulation of these processes. The cardiac response to KillerRed-induced injury in X. laevis tadpole hearts consists of a phase dominated by indicators of cardiac stress, followed by a repair-like phase with characteristics similar to mechanisms of cardiac regeneration in urodeles and fish. In the latter phase, we found markers associated with partial dedifferentiation and cardiomyocyte proliferation in the injured tadpole heart, which, unlike in regenerating hearts, are not dependent on Notch or retinoic acid signaling. Ultimately, the X. laevis cardiac response to KillerRed-induced oxidative stress shares characteristics with both mammalian and urodele/fish repair mechanisms, but is nonetheless a unique form of recovery, occupying an intermediate place on the spectrum of cardiac regenerative ability. An understanding of how Xenopus repairs cardiac damage can help bridge the gap between mammals and urodeles and contribute to new methods of treating heart disease.
Collapse
Affiliation(s)
- Kyle Jewhurst
- Department of Biology, Allen Discovery Center at Tufts University, Medford, Massachusetts
| | - Kelly A McLaughlin
- Department of Biology, Allen Discovery Center at Tufts University, Medford, Massachusetts
| |
Collapse
|
39
|
Li W, Feng G, Gauthier JM, Lokshina I, Higashikubo R, Evans S, Liu X, Hassan A, Tanaka S, Cicka M, Hsiao HM, Ruiz-Perez D, Bredemeyer A, Gross RW, Mann DL, Tyurina YY, Gelman AE, Kagan VE, Linkermann A, Lavine KJ, Kreisel D. Ferroptotic cell death and TLR4/Trif signaling initiate neutrophil recruitment after heart transplantation. J Clin Invest 2019; 129:2293-2304. [PMID: 30830879 DOI: 10.1172/jci126428] [Citation(s) in RCA: 341] [Impact Index Per Article: 56.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Non-apoptotic forms of cell death can trigger sterile inflammation through the release of danger-associated molecular patterns, which are recognized by innate immune receptors. However, despite years of investigation the mechanisms which initiate inflammatory responses after heart transplantation remain elusive. Here, we demonstrate that ferrostatin-1 (Fer-1), a specific inhibitor of ferroptosis, decreases the level of pro-ferroptotic hydroperoxy-arachidonoyl-phosphatidylethanolamine, reduces cardiomyocyte cell death and blocks neutrophil recruitment following heart transplantation. Inhibition of necroptosis had no effect on neutrophil trafficking in cardiac grafts. We extend these observations to a model of coronary artery ligation-induced myocardial ischemia reperfusion injury where inhibition of ferroptosis resulted in reduced infarct size, improved left ventricular systolic function, and reduced left ventricular remodeling. Using intravital imaging of cardiac transplants, we uncover that ferroptosis orchestrates neutrophil recruitment to injured myocardium by promoting adhesion of neutrophils to coronary vascular endothelial cells through a TLR4/TRIF/type I IFN signaling pathway. Thus, we have discovered that inflammatory responses after cardiac transplantation are initiated through ferroptotic cell death and TLR4/Trif-dependent signaling in graft endothelial cells. These findings provide a platform for the development of therapeutic strategies for heart transplant recipients and patients, who are vulnerable to ischemia reperfusion injury following restoration of coronary blood flow.
Collapse
Affiliation(s)
| | - Guoshuai Feng
- Department of Medicine, Washington University, Saint Louis, Missouri, USA
| | | | - Inessa Lokshina
- Department of Medicine, Washington University, Saint Louis, Missouri, USA
| | | | - Sarah Evans
- Department of Medicine, Washington University, Saint Louis, Missouri, USA
| | - Xinping Liu
- Department of Medicine, Washington University, Saint Louis, Missouri, USA
| | | | | | - Markus Cicka
- Department of Medicine, Washington University, Saint Louis, Missouri, USA
| | | | | | - Andrea Bredemeyer
- Department of Medicine, Washington University, Saint Louis, Missouri, USA
| | - Richard W Gross
- Department of Medicine, Washington University, Saint Louis, Missouri, USA
| | - Douglas L Mann
- Department of Medicine, Washington University, Saint Louis, Missouri, USA
| | - Yulia Y Tyurina
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Andrew E Gelman
- Department of Surgery and.,Department of Pathology and Immunology, Washington University, Saint Louis, Missouri, USA
| | - Valerian E Kagan
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Laboratory of Navigational Redox Lipidomics, I.M. Sechenov Moscow State Medical University, Moscow, Russia
| | - Andreas Linkermann
- Department of Internal Medicine III, Division of Nephrology, University Hospital Carl Gustav Carus at Technische Universität Dresden, Dresden, Germany
| | - Kory J Lavine
- Department of Medicine, Washington University, Saint Louis, Missouri, USA.,Department of Pathology and Immunology, Washington University, Saint Louis, Missouri, USA.,Department of Developmental Biology, Washington University, Saint Louis, Missouri, USA
| | - Daniel Kreisel
- Department of Surgery and.,Department of Pathology and Immunology, Washington University, Saint Louis, Missouri, USA
| |
Collapse
|
40
|
Fan Q, Tao R, Zhang H, Xie H, Lu L, Wang T, Su M, Hu J, Zhang Q, Chen Q, Iwakura Y, Shen W, Zhang R, Yan X. Dectin-1 Contributes to Myocardial Ischemia/Reperfusion Injury by Regulating Macrophage Polarization and Neutrophil Infiltration. Circulation 2019; 139:663-678. [PMID: 30586706 DOI: 10.1161/circulationaha.118.036044] [Citation(s) in RCA: 176] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Macrophage-associated immune response plays an important role in myocardial ischemia/reperfusion (IR) injury. Dectin-1, expressed mainly on activated myeloid cells, is crucial for the regulation of immune homeostasis as a pattern recognition receptor. However, its effects and roles during the myocardial IR injury remain unknown. METHODS Genetic ablation, antibody blockade, or Dectin-1 activation, along with the adoptive bone marrow transfer chimeric model, was used to determine the functional significance of Dectin-1 in myocardial IR injury. Immune cell filtration and inflammation were examined by flow cytometry, quantitative real-time polymerase chain reaction, and immunohistochemistry. Moreover, Dectin-1+ cells were analyzed by flow cytometry in the blood of patients with ST-segment-elevation myocardial infarction and stable patients with normal coronary artery (control). RESULTS We demonstrated that Dectin-1 expression observed on the bone marrow-derived macrophages is increased in the heart during the early phase after IR injury. Dectin-1 deficiency and antibody-mediated Dectin-1 inhibition led to a considerable improvement in cardiac function, accompanied by a reduction in cardiomyocyte apoptosis, which was associated with a decrease in M1 macrophage polarization and Ly-6C+ monocyte and neutrophil infiltration. Activation of Dectin-1 with its agonist had the opposite effects. Furthermore, Dectin-1 contributed to neutrophil recruitment through the regulation of Cxcl1 and granulocyte colony-stimulating factor expression. In addition, Dectin-1-dependent interleukin-23/interleukin-1β production was shown to be essential for interleukin-17A expression by γδT cells, leading to neutrophil recruitment and myocardial IR injury. Furthermore, we demonstrated that circulating Dectin-1+CD14++CD16- and Dectin-1+CD14++CD16+ monocyte levels were significantly higher in patients with ST-segment-elevation myocardial infarction than in controls and positively correlated with the severity of cardiac dysfunction. CONCLUSIONS Our results reveal a crucial role of Dectin-1 in the process of mouse myocardial IR injury and provide a new, clinically significant therapeutic target.
Collapse
Affiliation(s)
- Qin Fan
- Departments of Cardiology (Q.F., R.T., H.Z., H.X., L.L., J.H., Q.Z., W.S., R.Z., X.Y.), Shanghai Jiaotong University School of Medicine, PR China
- Rui Jin Hospital, and Institute of Cardiovascular Diseases (Q.F., H.Z., H.X., L.L., Q.C., R.Z., X.Y.), Shanghai Jiaotong University School of Medicine, PR China
| | - Rong Tao
- Departments of Cardiology (Q.F., R.T., H.Z., H.X., L.L., J.H., Q.Z., W.S., R.Z., X.Y.), Shanghai Jiaotong University School of Medicine, PR China
| | - Hang Zhang
- Departments of Cardiology (Q.F., R.T., H.Z., H.X., L.L., J.H., Q.Z., W.S., R.Z., X.Y.), Shanghai Jiaotong University School of Medicine, PR China
- Rui Jin Hospital, and Institute of Cardiovascular Diseases (Q.F., H.Z., H.X., L.L., Q.C., R.Z., X.Y.), Shanghai Jiaotong University School of Medicine, PR China
| | - Hongyang Xie
- Departments of Cardiology (Q.F., R.T., H.Z., H.X., L.L., J.H., Q.Z., W.S., R.Z., X.Y.), Shanghai Jiaotong University School of Medicine, PR China
- Rui Jin Hospital, and Institute of Cardiovascular Diseases (Q.F., H.Z., H.X., L.L., Q.C., R.Z., X.Y.), Shanghai Jiaotong University School of Medicine, PR China
| | - Lin Lu
- Departments of Cardiology (Q.F., R.T., H.Z., H.X., L.L., J.H., Q.Z., W.S., R.Z., X.Y.), Shanghai Jiaotong University School of Medicine, PR China
- Rui Jin Hospital, and Institute of Cardiovascular Diseases (Q.F., H.Z., H.X., L.L., Q.C., R.Z., X.Y.), Shanghai Jiaotong University School of Medicine, PR China
| | - Ting Wang
- Pathology (T.W.), Shanghai Jiaotong University School of Medicine, PR China
| | - Min Su
- Institute of Clinical Pathology, Department of Pathology, Shantou University Medical College, Guangdong, PR China (M.S.)
| | - Jian Hu
- Departments of Cardiology (Q.F., R.T., H.Z., H.X., L.L., J.H., Q.Z., W.S., R.Z., X.Y.), Shanghai Jiaotong University School of Medicine, PR China
| | - Qi Zhang
- Departments of Cardiology (Q.F., R.T., H.Z., H.X., L.L., J.H., Q.Z., W.S., R.Z., X.Y.), Shanghai Jiaotong University School of Medicine, PR China
| | - Qiujing Chen
- Rui Jin Hospital, and Institute of Cardiovascular Diseases (Q.F., H.Z., H.X., L.L., Q.C., R.Z., X.Y.), Shanghai Jiaotong University School of Medicine, PR China
| | - Yoichiro Iwakura
- Division of Experimental Animal Immunology, Center for Animal Disease Models, Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba, Japan (Y.I.)
| | - Weifeng Shen
- Departments of Cardiology (Q.F., R.T., H.Z., H.X., L.L., J.H., Q.Z., W.S., R.Z., X.Y.), Shanghai Jiaotong University School of Medicine, PR China
| | - Ruiyan Zhang
- Departments of Cardiology (Q.F., R.T., H.Z., H.X., L.L., J.H., Q.Z., W.S., R.Z., X.Y.), Shanghai Jiaotong University School of Medicine, PR China
- Rui Jin Hospital, and Institute of Cardiovascular Diseases (Q.F., H.Z., H.X., L.L., Q.C., R.Z., X.Y.), Shanghai Jiaotong University School of Medicine, PR China
| | - Xiaoxiang Yan
- Departments of Cardiology (Q.F., R.T., H.Z., H.X., L.L., J.H., Q.Z., W.S., R.Z., X.Y.), Shanghai Jiaotong University School of Medicine, PR China
- Rui Jin Hospital, and Institute of Cardiovascular Diseases (Q.F., H.Z., H.X., L.L., Q.C., R.Z., X.Y.), Shanghai Jiaotong University School of Medicine, PR China
| |
Collapse
|
41
|
Inhibition of glycogen synthase kinase-3β is involved in cardioprotection by α7nAChR agonist and limb remote ischemic postconditionings. Biosci Rep 2018; 38:BSR20181315. [PMID: 30249754 PMCID: PMC6435451 DOI: 10.1042/bsr20181315] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 09/13/2018] [Accepted: 09/21/2018] [Indexed: 12/17/2022] Open
Abstract
The present study was designed to determine whether glycogen synthase kinase-3β (GSK-3β) was involved in the cardioprotection by α7 nicotinic acetylcholine receptor (α7nAChR) agonist and limb remote ischemic postconditionings. Forty male Sprague-Dawley rats were randomly divided equally into control (C), α7nAChR agonist postconditioning (P), limb remote ischemic postconditioning (L), combined α7nAChR agonist and limb remote ischemic postconditioning (P+L) groups. At the end of experiment, serum cTnI, creatine kinase-MB (CK-MB), tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), high mobility group protein (HMGB1) and interleukin-10 (IL-10) levels were measured; infarct size (IS), myocardial expressions of GSK-3β, p-GSK-3β (Ser9), nuclear factor-κB (NF-κB) and p-NF-κB (Ser536) in the ischemic area were assessed. The results showed that compared with group C, IS, serum cTnI and CK-MB levels obviously decreased in groups P, L and P+L. Compared with groups P and L, IS, serum cTnI and CK-MB levels significantly decreased in group P+L. Compared with group C, serum TNF-α, IL-6 and HMGB1 levels, and myocardial expression of p-NF-κBp65 (Ser536) evidently decreased, and myocardial expression of p-GSK-3β (Ser9) obviously increased in groups P, L and P+L. Compared with group P, serum TNF-α, IL-6 and HMGB1 levels and myocardial expression of p-NF-κBp65 (Ser536) significantly increased, and myocardial expression of p-GSK-3β (Ser9) evidently decreased in group L. Compared with group L, serum TNF-α, IL-6, HMGB1 levels, and myocardial expression of p-NF-κBp65 (Ser536) significantly decreased, and myocardial expression of p-GSK-3β (Ser9) obviously increased in group P+L. In conclusion, our findings indicate that inhibition of GSK-3β to decrease NF-κB transcription is one of cardioprotective mechanisms of α7nAChR agonist and limb remote ischemic postconditionings by anti-inflammation, but improved cardioprotection by combined two interventions is not completely attributable to an enhanced anti-inflammatory mechanism.
Collapse
|
42
|
Tian Y, Charles EJ, Yan Z, Wu D, French BA, Kron IL, Yang Z. The myocardial infarct-exacerbating effect of cell-free DNA is mediated by the high-mobility group box 1-receptor for advanced glycation end products-Toll-like receptor 9 pathway. J Thorac Cardiovasc Surg 2018; 157:2256-2269.e3. [PMID: 30401529 DOI: 10.1016/j.jtcvs.2018.09.043] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Revised: 08/13/2018] [Accepted: 09/12/2018] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Damage-associated molecular patterns, such as high-mobility group box 1 (HMGB1) and cell-free DNA (cfDNA), play critical roles in mediating ischemia-reperfusion injury (IRI). HMGB1 activates RAGE to exacerbate IRI, but the mechanism underlying cfDNA-induced myocardial IRI remains unknown. We hypothesized that the infarct-exacerbating effect of cfDNA is mediated by HMGB1 and receptor for advanced glycation end products (RAGE). METHODS C57BL/6 wild type mice, RAGE knockout (KO), and Toll-like receptor 9 KO mice underwent 20- or 40-minute occlusions of the left coronary artery followed by up to 60 minutes of reperfusion. Cardiac coronary perfusate was acquired from ischemic hearts without reperfusion. Exogenous mitochondrial DNA was acquired from livers of normal C57BL/6 mice. Myocardial infarct size (IS) was reported as percent risk region, as measured by 2,3,5-triphenyltetrazolium chloride and Phthalo blue (Heucotech, Fairless Hill, Pa) staining. cfDNA levels were measured by Sytox Green assay (Thermo Fisher Scientific, Waltham, Mass) and/or spectrophotometer. RESULTS Free HMGB1 and cfDNA levels were increased in the ischemic myocardium during prolonged ischemia and subsequently in the plasma during reperfusion. In C57BL/6 mice undergoing 40'/60' IRI, deoxyribonuclease I, or anti-HMGB1 monoclonal antibody reduced IS by approximately half to 29.0% ± 5.2% and 24.3% ± 3.5% (P < .05 vs control 54.3% ± 3.4%). However, combined treatment with deoxyribonuclease I + anti-HMGB1 monoclonal antibody did not further attenuate IS (29.3% ± 4.9%). In C57BL/6 mice undergoing 20'/60' IRI, injection of 40'/5' plasma upon reperfusion increased IS by more than 3-fold (to 19.9 ± 4.3; P < .05). This IS exacerbation was abolished by pretreating the plasma with deoxyribonuclease I or by depleting the HMGB1 by immunoprecipitation, or by splenectomy. The infarct-exacerbating effect also disappeared in RAGE KO mice and Toll-like receptor 9 KO mice. Injection of 40'/0' coronary perfusate upon reperfusion similarly increased IS. The levels of HMGB1 and cfDNA were significantly elevated in the 40'/0' coronary perfusate and 40'/reperfusion (min) plasma but not in those with 10' ischemia. In C57BL/6 mice without IRI, 40'/5' plasma significantly increased the interleukin-1β protein and messenger RNA expression in the spleen by 30 minutes after injection. Intravenous bolus injection of recombinant HMGB1 (0.1 μg/g) or mitochondrial DNA (0.5 μg/g) 5 minutes before reperfusion did not exacerbate IS (P = not significant vs control). However, combined administration of recombinant HMGB1 + mitochondrial DNA significantly increased IS (P < .05 vs individual treated groups) and this infarct-exacerbating effect disappeared in RAGE KO mice and splenectomized C57BL/6 mice. The accumulation of cfDNA in the spleen after combined recombinant HMGB1 + mitochondrial DNA treatment was significantly more elevated in C57BL/6 mice than in RAGE KO mice. CONCLUSIONS Both HMGB1 and cfDNA are released from the heart upon reperfusion after prolonged ischemia and both contribute importantly and interdependently to post-IRI by a common RAGE-Toll-like receptor 9-dependent mechanism. Depleting either of these 2 damage-associated molecular patterns suffices to significantly reduce IS by approximately 50%.
Collapse
Affiliation(s)
- Yikui Tian
- Department of Surgery, University of Virginia, Charlottesville, Va; Department of Cardiovascular Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Eric J Charles
- Department of Surgery, University of Virginia, Charlottesville, Va
| | - Zhen Yan
- Department of Cardiovascular Medicine, University of Virginia, Charlottesville, Va
| | - Di Wu
- Department of Surgery, University of Virginia, Charlottesville, Va
| | - Brent A French
- Department of Cardiovascular Medicine, University of Virginia, Charlottesville, Va; Department of Biomedical Engineering, University of Virginia, Charlottesville, Va
| | - Irving L Kron
- Department of Surgery, University of Virginia, Charlottesville, Va
| | - Zequan Yang
- Department of Surgery, University of Virginia, Charlottesville, Va; Department of Biomedical Engineering, University of Virginia, Charlottesville, Va.
| |
Collapse
|
43
|
Cai J, Xu M, Zhang X, Li H. Innate Immune Signaling in Nonalcoholic Fatty Liver Disease and Cardiovascular Diseases. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2018; 14:153-184. [PMID: 30230967 DOI: 10.1146/annurev-pathmechdis-012418-013003] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The physiological significance of innate immune signaling lies primarily in its role in host defense against invading pathogens. It is becoming increasingly clear that innate immune signaling also modulates the development of metabolic diseases, especially nonalcoholic fatty liver disease and cardiovascular diseases, which are characterized by chronic, low-grade inflammation due to a disarrangement of innate immune signaling. Notably, recent studies indicate that in addition to regulating canonical innate immune-mediated inflammatory responses (or immune-dependent signaling-induced responses), molecules of the innate immune system regulate pathophysiological responses in multiple organs during metabolic disturbances (termed immune-independent signaling-induced responses), including the disruption of metabolic homeostasis, tissue repair, and cell survival. In addition, emerging evidence from the study of immunometabolism indicates that the systemic metabolic status may have profound effects on cellular immune function and phenotypes through the alteration of cell-intrinsic metabolism. We summarize how the innate immune system interacts with metabolic disturbances to trigger immune-dependent and immune-independent pathogenesis in the context of nonalcoholic fatty liver disease, as representative of metabolic diseases, and cardiovascular diseases.
Collapse
Affiliation(s)
- Jingjing Cai
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; .,Institute of Model Animals of Wuhan University, Wuhan 430072, China.,Basic Medical School, Wuhan University, Wuhan 430071, China.,Department of Cardiology, The Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Meng Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; .,Institute of Model Animals of Wuhan University, Wuhan 430072, China.,Basic Medical School, Wuhan University, Wuhan 430071, China
| | - Xiaojing Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; .,Institute of Model Animals of Wuhan University, Wuhan 430072, China.,Basic Medical School, Wuhan University, Wuhan 430071, China
| | - Hongliang Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; .,Institute of Model Animals of Wuhan University, Wuhan 430072, China.,Basic Medical School, Wuhan University, Wuhan 430071, China
| |
Collapse
|
44
|
Cipolla GA, de Oliveira JC, Salviano-Silva A, Lobo-Alves SC, Lemos DS, Oliveira LC, Jucoski TS, Mathias C, Pedroso GA, Zambalde EP, Gradia DF. Long Non-Coding RNAs in Multifactorial Diseases: Another Layer of Complexity. Noncoding RNA 2018; 4:E13. [PMID: 29751665 PMCID: PMC6027498 DOI: 10.3390/ncrna4020013] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 04/13/2018] [Accepted: 05/04/2018] [Indexed: 02/06/2023] Open
Abstract
Multifactorial diseases such as cancer, cardiovascular conditions and neurological, immunological and metabolic disorders are a group of diseases caused by the combination of genetic and environmental factors. High-throughput RNA sequencing (RNA-seq) technologies have revealed that less than 2% of the genome corresponds to protein-coding genes, although most of the human genome is transcribed. The other transcripts include a large variety of non-coding RNAs (ncRNAs), and the continuous generation of RNA-seq data shows that ncRNAs are strongly deregulated and may be important players in pathological processes. A specific class of ncRNAs, the long non-coding RNAs (lncRNAs), has been intensively studied in human diseases. For clinical purposes, lncRNAs may have advantages mainly because of their specificity and differential expression patterns, as well as their ideal qualities for diagnosis and therapeutics. Multifactorial diseases are the major cause of death worldwide and many aspects of their development are not fully understood. Recent data about lncRNAs has improved our knowledge and helped risk assessment and prognosis of these pathologies. This review summarizes the involvement of some lncRNAs in the most common multifactorial diseases, with a focus on those with published functional data.
Collapse
Affiliation(s)
- Gabriel A Cipolla
- Department of Genetics, Federal University of Parana, Curitiba 81531-980, Brazil.
| | | | | | - Sara C Lobo-Alves
- Department of Genetics, Federal University of Parana, Curitiba 81531-980, Brazil.
| | - Debora S Lemos
- Department of Genetics, Federal University of Parana, Curitiba 81531-980, Brazil.
| | - Luana C Oliveira
- Department of Genetics, Federal University of Parana, Curitiba 81531-980, Brazil.
| | - Tayana S Jucoski
- Department of Genetics, Federal University of Parana, Curitiba 81531-980, Brazil.
| | - Carolina Mathias
- Department of Genetics, Federal University of Parana, Curitiba 81531-980, Brazil.
| | - Gabrielle A Pedroso
- Department of Genetics, Federal University of Parana, Curitiba 81531-980, Brazil.
| | - Erika P Zambalde
- Department of Genetics, Federal University of Parana, Curitiba 81531-980, Brazil.
| | - Daniela F Gradia
- Department of Genetics, Federal University of Parana, Curitiba 81531-980, Brazil.
| |
Collapse
|
45
|
Abstract
Mitochondrial dysfunction underlines a multitude of pathologies; however, studies are scarce that rescue the mitochondria for cellular resuscitation. Exploration into the protective role of mitochondrial transcription factor A (TFAM) and its mitochondrial functions respective to cardiomyocyte death are in need of further investigation. TFAM is a gene regulator that acts to mitigate calcium mishandling and ROS production by wrapping around mitochondrial DNA (mtDNA) complexes. TFAM's regulatory functions over serca2a, NFAT, and Lon protease contribute to cardiomyocyte stability. Calcium- and ROS-dependent proteases, calpains, and matrix metalloproteinases (MMPs) are abundantly found upregulated in the failing heart. TFAM's regulatory role over ROS production and calcium mishandling leads to further investigation into the cardioprotective role of exogenous TFAM. In an effort to restabilize physiological and contractile activity of cardiomyocytes in HF models, we propose that TFAM-packed exosomes (TFAM-PE) will act therapeutically by mitigating mitochondrial dysfunction. Notably, this is the first mention of exosomal delivery of transcription factors in the literature. Here we elucidate the role of TFAM in mitochondrial rescue and focus on its therapeutic potential.
Collapse
Affiliation(s)
- George H Kunkel
- Department of Physiology and Biophysics, Health Sciences Centre, 1216, School of Medicine, University of Louisville, 500, South Preston Street, Louisville, KY, 40202, USA
| | - Pankaj Chaturvedi
- Department of Physiology and Biophysics, Health Sciences Centre, 1216, School of Medicine, University of Louisville, 500, South Preston Street, Louisville, KY, 40202, USA.
| | - Suresh C Tyagi
- Department of Physiology and Biophysics, Health Sciences Centre, 1216, School of Medicine, University of Louisville, 500, South Preston Street, Louisville, KY, 40202, USA
| |
Collapse
|
46
|
Chen B, Frangogiannis NG. Immune cells in repair of the infarcted myocardium. Microcirculation 2018; 24. [PMID: 27542099 DOI: 10.1111/micc.12305] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 08/17/2016] [Indexed: 12/14/2022]
Abstract
The immune system plays a critical role in both repair and remodeling of the infarcted myocardium. Danger signals released by dying cardiomyocytes mobilize, recruit, and activate immune cells, triggering an inflammatory reaction. CXC chemokines containing the ELR motif attract neutrophils, while CC chemokines mediate recruitment of mononuclear cell subpopulations, contributing to clearance of the infarct from dead cells and matrix debris. Immune cell subsets also participate in suppression and containment of the postinfarction inflammatory response by secreting anti-inflammatory mediators, such as IL-10 and TGF-β. As proinflammatory signaling is suppressed, macrophage subpopulations, mast cells and lymphocytes, activate fibrogenic and angiogenic responses, contributing to scar formation. In the viable remodeling myocardium, chronic activation of immune cells may promote fibrosis and hypertrophy. This review discusses the role of immune cells in repair and remodeling of the infarcted myocardium. Understanding the role of immune cells in myocardial infarction is critical for the development of therapeutic strategies aimed at protecting the infarcted heart from adverse remodeling. Moreover, modulation of immune cell phenotype may be required in order to achieve the visionary goal of myocardial regeneration.
Collapse
Affiliation(s)
- Bijun Chen
- Department of Medicine (Cardiology), The Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY
| | - Nikolaos G Frangogiannis
- Department of Medicine (Cardiology), The Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY
| |
Collapse
|
47
|
Abstract
The occlusion of a coronary artery by a thrombus generated on a ruptured atherosclerotic plaque has been pursued in the last decades as a determining event for the clinical outcome after myocardial infarction (MI). Yet, MI causes a cell death wave front, which triggers an inflammatory response to clear cellular debris, and which in excess can double the myocardial lesion and influence the clinical prognosis in the short and long term. Accordingly, proper, timely regulated inflammatory response has now been considered a second pivotal player in cardiac recovery after MI justifying the search for pharmacological strategies to modulate inflammatory effectors. This chapter reviews the key events and the main effectors of inflammation after myocardial ischemic insult, as well as the contribution of this phenomenon to the progression of atherosclerosis.
Collapse
Affiliation(s)
- Joaquim B Oliveira
- Laboratory of Atherosclerosis and Vascular Biology, State University of Campinas, Campinas, Brazil
| | - Alexandre A S M Soares
- Laboratory of Atherosclerosis and Vascular Biology, State University of Campinas, Campinas, Brazil
| | - Andrei C Sposito
- Laboratory of Atherosclerosis and Vascular Biology, State University of Campinas, Campinas, Brazil.
| |
Collapse
|
48
|
Aljakna A, Fracasso T, Sabatasso S. Molecular tissue changes in early myocardial ischemia: from pathophysiology to the identification of new diagnostic markers. Int J Legal Med 2018; 132:425-438. [DOI: 10.1007/s00414-017-1750-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 11/20/2017] [Indexed: 02/06/2023]
|
49
|
Bishayi B, Adhikary R, Sultana S, Dey R, Nandi A. Altered expression of CXCR1 (IL-8R) in macrophages utilizing cell surface TNFR1 and IL-1 receptor during Staphylococcus aureus infection. Microb Pathog 2017; 113:460-471. [DOI: 10.1016/j.micpath.2017.11.028] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2017] [Revised: 11/04/2017] [Accepted: 11/18/2017] [Indexed: 01/28/2023]
|
50
|
Ouyang MZ, Zhou D, Zhu Y, Zhang M, Li L. The inhibition of MyD88 and TRIF signaling serve equivalent roles in attenuating myocardial deterioration due to acute severe inflammation. Int J Mol Med 2017; 41:399-408. [PMID: 29115392 DOI: 10.3892/ijmm.2017.3239] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 10/24/2017] [Indexed: 11/06/2022] Open
Abstract
Myeloid differentiation factor 88 (MyD88) and Toll or interleukin-1 receptor-domain-containing adaptor-inducing interferon-β (IFN-β) (TRIF) are two pivotal downstream adaptors of Toll-like receptors. Activation of MyD88 or TRIF signaling in cardiac immune pathology of severe inflammation negatively influences heart function. In the present study, severe septic cardiac injury was induced in C57BL/6 mice by cecum ligation and puncture (CLP). A total of 64 mice were divided randomly into the following four groups (n=16/group; 8 for observation of survival rate, 8 for heart sample analysis): Sham, CLP, anti-MyD88-CLP and anti-TRIF-CLP. Anti-MyD88 and anti-TRIF antibodies were administered to the respective mice through the tail veins 2 h before CLP. Measurements of cardiac function, including M-modes, velocity vector imaging and cardiac troponin I, were performed. Myocardial inflammatory cytokines were examined by reverse transcription-polymerase chain reaction (RT-PCR), myocardial neutrophil infiltration was measured by a myeloperoxidase activity assay, intracellular adhesion molecule and vascular cell adhesion molecule mRNA expression levels were investigated, and histopathological characteristics were evaluated. Levels of mRNA transcripts encoding genes for apoptosis production and MyD88, TRIF, nuclear factor-κB and IFN regulatory factor 3 were investigated by RT-PCR. Mice challenged with CLP demonstrated deleterious cardiac function, increased levels of interleukin-1β (IL-1β), IL-6β, and tumor necrosis factor-α mRNA, increased neutrophil infiltration, and increased apoptosis. In contrast, mice in the anti-MyD88 CLP and anti-TRIF CLP groups retained cardiac function with reduced cytokine release, decreased neutrophil infiltration, and reduced apoptosis. In addition, there was no significant difference between the anti-MyD88 CLP and anti-TRIF CLP groups. Thus, the present study indicated that MyD88 and TRIF blockades serve notable and equivalent roles in protecting cardiac deterioration from severe sepsis by attenuating cytokine release, reducing neutrophil infiltration and alleviating apoptosis.
Collapse
Affiliation(s)
- Min-Zhi Ouyang
- Department of Ultrasound Diagnosis, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| | - Dan Zhou
- Department of Ultrasound Diagnosis, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| | - Yun Zhu
- Department of Ultrasound Diagnosis, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| | - Ming Zhang
- Department of Ultrasound Diagnosis, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| | - Ling Li
- Medical Basic Teaching Experiment Center, College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, P.R. China
| |
Collapse
|