101
|
Nishikido T, Oyama JI, Shiraki A, Tsukamoto I, Igarashi J, Node K. COA-Cl (2-Cl-C.OXT-A) can promote coronary collateral development following acute myocardial infarction in mice. Sci Rep 2019; 9:2533. [PMID: 30796271 PMCID: PMC6385273 DOI: 10.1038/s41598-019-39222-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 01/11/2019] [Indexed: 01/04/2023] Open
Abstract
2-Cl-C.OXT-A (COA-Cl) is a novel nucleic acid analogue that promotes tube-forming activity of human umbilical vein endothelial cells (HUVEC) through vascular endothelial growth factor (VEGF). The development of coronary collateral circulation is critical to rescue the ischemic myocardium and to prevent subsequent irreversible ischemic injury. We evaluated whether COA-Cl can promote angiogenesis in ischemic tissue, reduce infarct size and preserve cardiac contractility in vivo. Mice received COA-Cl or placebo daily for three days after myocardial infarction (MI) by coronary ligation. The degree of angiogenesis in ischemic myocardium was assessed by staining endothelial cells and vascular smooth muscle cells, and measuring infarct size/area-at-risk. In mice treated with COA-Cl, enhanced angiogenesis and smaller infarct size were recognized, even given a similar area at risk. We observed increases in the protein expression levels of VEGF and in the protein phosphorylation level of eNOS. In addition, the heart weight to body weight ratio and myocardial fibrosis in COA-Cl mice were decreased on Day 7. Administration of COA-Cl after MI promotes angiogenesis, which is associated with reduced infarct size and attenuated cardiac remodeling. This may help to prevent heart failure due to cardiac dysfunction after MI.
Collapse
Affiliation(s)
- Toshiyuki Nishikido
- Department of Cardiovascular of Medicine, Saga University Hospital, Saga, Japan
| | - Jun-Ichi Oyama
- Department of Cardiovascular of Medicine, Saga University Hospital, Saga, Japan.
| | - Aya Shiraki
- Department of Cardiovascular of Medicine, Saga University Hospital, Saga, Japan
| | - Ikuko Tsukamoto
- Department of Pharmaco-Bio-Informatics, Kagawa University, Kagawa, Japan
| | - Junsuke Igarashi
- Graduate School of Health Sciences, Morinomiya University of Medical Sciences, Osaka, Japan
| | - Koichi Node
- Department of Cardiovascular of Medicine, Saga University Hospital, Saga, Japan
| |
Collapse
|
102
|
Jiang X, Kong B, Shuai W, Shen C, Yang F, Fu H, Huang H. Loss of MD1 exacerbates myocardial ischemia/reperfusion injury and susceptibility to ventricular arrhythmia. Eur J Pharmacol 2019; 844:79-86. [DOI: 10.1016/j.ejphar.2018.11.025] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Revised: 11/13/2018] [Accepted: 11/16/2018] [Indexed: 12/23/2022]
|
103
|
Wu RN, Yu TY, Zhou JC, Li M, Gao HK, Zhao C, Dong RQ, Peng D, Hu ZW, Zhang XW, Wu YQ. Targeting HMGB1 ameliorates cardiac fibrosis through restoring TLR2-mediated autophagy suppression in myocardial fibroblasts. Int J Cardiol 2019; 267:156-162. [PMID: 29957254 DOI: 10.1016/j.ijcard.2018.04.103] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 04/19/2018] [Accepted: 04/23/2018] [Indexed: 01/14/2023]
Abstract
BACKGROUND Extracellular high-mobility group box 1 (HMGB1) has been identified as playing a critical role in the pathogenesis of tissue fibrosis. However, the underlying mechanism of its involvement in cardiac fibrosis is still not well-defined. Here, we aim to investigate whether toll-like receptor 2 (TLR2) contributes to the extracellular HMGB1-mediated development and progression of cardiac fibrosis. METHODS A mouse model of cardiac fibrosis was induced by subcutaneous injection of isoproterenol (ISO). Glycyrrhizic acid (GA), an inhibitor of HMGB1 derived from natural products, was simultaneously administered by intraperitoneal injection. Echocardiography, H&E and Sirius red staining were used to evaluate cardiac function and fibrosis. The myocardial expression of autophagy-associated proteins was examined using immunoblotting. Cardiac fibroblasts were treated with different concentrations of HMGB1 to examine the expression levels of α-SMA, collagen I and autophagy markers. Interactions of HMGB1/TLR2 and α-SMA/p62 were examined by immunoprecipitation and immunofluorescence. RESULTS ISO-treated mice showed characteristic cardiac fibrosis, increased expression and co-localization of HMGB1 and TLR2, as well as impaired autophagic signals in myocardial tissues, which could be prevented by silencing TLR2. Exogenous administration of HMGB1 blocked the autophagic flux in fibroblasts, which caused extensive accumulation of collagen I and α-SMA. In addition, cardiac fibrosis was alleviated by GA treatment through abrogating the interaction between HMGB1 and TLR2. CONCLUSIONS Our study suggests that the interaction between TLR2 and HMGB1 contributes to the pathogenesis of cardiac fibrosis via suppressing fibroblast autophagy, and that inhibiting HMGB1 with GA provides therapeutic benefits for the treatment of fibroproliferative heart diseases.
Collapse
Affiliation(s)
- Ri-Na Wu
- Department of Cardiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China; Department of Cardiology, Baotou Central Hospital, Inner Mongolia, China
| | - Tian-Yu Yu
- Department of Cardiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Ji-Chao Zhou
- Molecular Immunology and Pharmacology Group, State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Meng Li
- Department of Cardiology, Baotou Central Hospital, Inner Mongolia, China
| | - Hui-Kuan Gao
- Department of Cardiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Can Zhao
- Department of Cardiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Rui-Qing Dong
- Department of Cardiology, Hangzhou First People's Hospital, Nanjing Medical University, Zhejiang, China
| | - Dian Peng
- Molecular Immunology and Pharmacology Group, State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Zhuo-Wei Hu
- Molecular Immunology and Pharmacology Group, State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xiao-Wei Zhang
- Molecular Immunology and Pharmacology Group, State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| | - Yong-Quan Wu
- Department of Cardiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
104
|
The Guizhi Gancao Decoction Attenuates Myocardial Ischemia-Reperfusion Injury by Suppressing Inflammation and Cardiomyocyte Apoptosis. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 2019:1947465. [PMID: 30800167 PMCID: PMC6360628 DOI: 10.1155/2019/1947465] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 12/10/2018] [Accepted: 12/20/2018] [Indexed: 12/25/2022]
Abstract
Guizhi Gancao Decoction (GGD) is a well-known traditional Chinese herbal medicine for the treatment of various cardiovascular diseases, such as myocardial ischemia-reperfusion (I/R) injury and arrhythmia. However, the mechanism by which GGD contributes to the amelioration of cardiac injury remains unclear. The aim of this study was to investigate the potential protective role of GGD against myocardial I/R injury and its possible mechanism. Consistent with the effect of the positive drug (Trimetazidine, TMZ), we subsequently validated that GGD could ameliorate myocardial I/R injury as evidenced by histopathological examination and triphenyltetrazolium chloride (TTC) staining. Moreover, terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay demonstrated that GGD suppressed myocardial apoptosis, which may be related to the upregulation of Bcl-2, PPARα, and PPARγ and downregulation of Bax, caspase-3, and caspase-9. Pretreatment with GGD attenuated the levels of proinflammatory cytokines including tumor necrosis factor-α (TNF-α), interleukin- (IL-) 6, and IL-1β in serum by inhibiting Toll-like receptor 4 (TLR4)/NF-κB signaling pathway. These results indicated that GGD exhibits cardioprotective effects on myocardial I/R injury through inhibition of the TLR4/NF-κB signaling pathway, which led to reduced inflammatory response and the subsequent cardiomyocyte apoptosis.
Collapse
|
105
|
Dexmedetomidine Preconditioning Ameliorates Inflammation and Blood-Spinal Cord Barrier Damage After Spinal Cord Ischemia-Reperfusion Injury by Down-Regulation High Mobility Group Box 1-Toll-Like Receptor 4-Nuclear Factor κB Signaling Pathway. Spine (Phila Pa 1976) 2019; 44:E74-E81. [PMID: 29975331 DOI: 10.1097/brs.0000000000002772] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
STUDY DESIGN To evaluate the effect of Dexmedetomidine (Dex) on the inflammatory response and the integrity of blood-spinal cord barrier (BSCB) after spinal cord ischemia-reperfusion injury (SCIRI). OBJECTIVE To investigate the role of Dex in spinal cord I/R, particularly in the high mobility group box 1-toll-like receptor 4-nuclear factor κB (HMGB1-TLR4-NF-κB) pathway and the integrity of BSCB. SUMMARY OF BACKGROUND DATA High mobility group box 1 (HMGB1) has been identified as a key mediator for the inflammatory response after spinal cord injury. Toll-like receptor 4-nuclear factor κB (TLR4-NF-κB) signaling pathway is the downstream of HMGB1. Dex preconditioning could protect the spinal cord from I/R injury by inhibiting HMGB1 and stabilizing the integrity of BSCB. But its underlying mechanism is not fully understood. METHODS Forty-eight male Japanese white rabbits were randomly assigned to three groups (16 rabbits/group): sham, I/R, and Dex + I/R. The hind-limb motor function was assessed at 12 hours intervals for 48 hours after reperfusion using the modified Tarlov scale score. The expression of HMGB1, TLR4, NF-κB, and tumor necrosis factor α (TNF-α) was evaluated by real-time polymerase chain reaction (RT-PCR) and Western blot. The permeability of BSCB was examined via Evans blue (EB) extravasation. RESULTS Compared with sham group, spinal cord I/R increased the expression of HMGB1, TLR4, NF-κB, and TNF-α as well as the permeability of BSCB (P < 0.05). Spinal cord I/R induced the decline of the score of hind-limb motor function (P < 0.01). Preconditioning with Dex attenuated the up-regulation of the express of HMGB1, TLR4, NF-κB, TNF-α, and stabilized the permeability of BSCB (P < 0.05). Dex preconditioning also improved the hiatopathological outcome and the motor function (P < 0.01). CONCLUSION Dex preconditioning may inhibit the inflammatory response and stabilize the integrity of BSCB at least partially by inhibiting the HMGB1-TLR4-NF-κB signaling pathway to protect spinal cord from ischemia/reperfusion injury. LEVEL OF EVIDENCE 2.
Collapse
|
106
|
Oyama JI, Node K. Gut microbiota and hypertension. Hypertens Res 2019; 42:741-743. [DOI: 10.1038/s41440-018-0203-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 12/16/2018] [Accepted: 12/19/2018] [Indexed: 12/20/2022]
|
107
|
Jung M, Dodsworth M, Thum T. Inflammatory cells and their non-coding RNAs as targets for treating myocardial infarction. Basic Res Cardiol 2018; 114:4. [PMID: 30523422 PMCID: PMC6290728 DOI: 10.1007/s00395-018-0712-z] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Accepted: 11/29/2018] [Indexed: 12/22/2022]
Abstract
Myocardial infarction triggers infiltration of several types of immune cells that coordinate both innate and adaptive immune responses. These play a dual role in post-infarction cardiac remodeling by initiating and resolving inflammatory processes, which needs to occur in a timely and well-orchestrated way to ensure a reestablishment of normalized cardiac functions. Thus, therapeutic modulation of immune responses might have benefits for infarct patients. While such strategies have shown great potential in treating cancer, applications in the post-infarction context have been disappointing. One challenge has been the complexity and plasticity of immune cells and their functions in cardiac regulation and healing. The types appear in patterns that are temporally and spatially distinct, while influencing each other and the surrounding tissue. A comprehensive understanding of the immune cell repertoire and their regulatory functions following infarction is sorely needed. Processes of cardiac remodeling trigger additional genetic changes that may also play critical roles in the aftermath of cardiovascular disease. Some of these changes involve non-coding RNAs that play crucial roles in the regulation of immune cells and may, therefore, be of therapeutic interest. This review summarizes what is currently known about the functions of immune cells and non-coding RNAs during post-infarction wound healing. We address some of the challenges that remain and describe novel therapeutic approaches under development that are based on regulating immune responses through non-coding RNAs in the aftermath of the disease.
Collapse
Affiliation(s)
- Mira Jung
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Michael Dodsworth
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.
- National Heart and Lung Institute, Imperial College London, London, UK.
| |
Collapse
|
108
|
Xiong L, Sun L, Liu S, Zhu X, Teng Z, Yan J. The Protective Roles of Urinary Trypsin Inhibitor in Brain Injury Following Fat Embolism Syndrome in a Rat Model. Cell Transplant 2018; 28:704-712. [PMID: 30449147 PMCID: PMC6686433 DOI: 10.1177/0963689718814766] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Fat embolism syndrome (FES) is a common complication following long bone fracture; fat droplets are released into the blood circulation and form embolisms, mainly in lung and brain. However, the potential mechanisms involved remain to be clarified. In this study, the mechanism of brain injury following FES and the protective effects of urinary trypsin inhibitor (UTI)—a serine protease inhibitor—were investigated. Sixty male Sprague-Dawley rats were divided randomly into sham, FES and FES+UTI treatment groups. The FES model was established using tail vein injection of glycerol trioleate, and UTI was administered by intraperitoneal injection immediately following FES. Brain/lung water content evaluation, Evans blue content and magnetic resonance imaging examination were used to assess the effects of UTI. Furthermore, immunohistochemistry and western blot were also applied to explore the protective mechanism of UTI following FES. The results of oil red O staining indicated that the FES model was successfully established. UTI could significantly attenuate blood-brain-barrier (BBB) disruption, as seen through brain edema evaluation and Evans blue content examination. Immunofluorescence staining results indicated that the TLR4-JNK pathway was involved in brain injury after FES; this effect could be quenched by UTI treatment. Furthermore, UTI could decrease the levels of downstream target proteins of the TLR4-JNK pathway, phosphorylated-NF- κB (p65) and p53 in brain. Our results showed that UTI could alleviate BBB injury after FES through blocking activity of the TLR4-JNK pathway.
Collapse
Affiliation(s)
- Lili Xiong
- 1 West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Linlin Sun
- 2 Department of Anatomy and Histology, School of Basic Medical Sciences, Peking University, Beijing, China.,3 Beijing Key Lab of Magnetic Resonance Imaging Technology, Beijing, China
| | - Shanshan Liu
- 2 Department of Anatomy and Histology, School of Basic Medical Sciences, Peking University, Beijing, China.,3 Beijing Key Lab of Magnetic Resonance Imaging Technology, Beijing, China
| | - Xingyun Zhu
- 2 Department of Anatomy and Histology, School of Basic Medical Sciences, Peking University, Beijing, China.,3 Beijing Key Lab of Magnetic Resonance Imaging Technology, Beijing, China
| | - Ze Teng
- 3 Beijing Key Lab of Magnetic Resonance Imaging Technology, Beijing, China
| | - Junhao Yan
- 2 Department of Anatomy and Histology, School of Basic Medical Sciences, Peking University, Beijing, China.,3 Beijing Key Lab of Magnetic Resonance Imaging Technology, Beijing, China
| |
Collapse
|
109
|
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: 1.5] [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
|
110
|
Preoperative systemic inflammation and perioperative myocardial injury: prospective observational multicentre cohort study of patients undergoing non-cardiac surgery. Br J Anaesth 2018; 122:180-187. [PMID: 30686303 DOI: 10.1016/j.bja.2018.09.002] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 08/26/2018] [Accepted: 09/03/2018] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Systemic inflammation is pivotal in the pathogenesis of cardiovascular disease. As inflammation can directly cause cardiomyocyte injury, we hypothesised that established systemic inflammation, as reflected by elevated preoperative neutrophil-lymphocyte ratio (NLR) >4, predisposes patients to perioperative myocardial injury. METHODS We prospectively recruited 1652 patients aged ≥45 yr who underwent non-cardiac surgery in two UK centres. Serum high sensitivity troponin T (hsTnT) concentrations were measured on the first three postoperative days. Clinicians and investigators were blinded to the troponin results. The primary outcome was perioperative myocardial injury, defined as hsTnT≥14 ng L-1 within 3 days after surgery. We assessed whether myocardial injury was associated with preoperative NLR>4, activated reactive oxygen species (ROS) generation in circulating monocytes, or both. Multivariable logistic regression analysis explored associations between age, sex, NLR, Revised Cardiac Risk Index, individual leukocyte subsets, and myocardial injury. Flow cytometric quantification of ROS was done in 21 patients. Data are presented as n (%) or odds ratio (OR) with 95% confidence intervals. RESULTS Preoperative NLR>4 was present in 239/1652 (14.5%) patients. Myocardial injury occurred in 405/1652 (24.5%) patients and was more common in patients with preoperative NLR>4 [OR: 2.56 (1.92-3.41); P<0.0001]. Myocardial injury was independently associated with lower absolute preoperative lymphocyte count [OR 1.80 (1.50-2.17); P<0.0001] and higher absolute preoperative monocyte count [OR 1.93 (1.12-3.30); P=0.017]. Monocyte ROS generation correlated with NLR (r=0.47; P=0.03). CONCLUSIONS Preoperative NLR>4 is associated with perioperative myocardial injury, independent of conventional risk factors. Systemic inflammation may contribute to the development of perioperative myocardial injury. CLINICAL TRIAL REGISTRATION NCT01842568.
Collapse
|
111
|
Yang CF. Clinical manifestations and basic mechanisms of myocardial ischemia/reperfusion injury. Tzu Chi Med J 2018; 30:209-215. [PMID: 30305783 PMCID: PMC6172894 DOI: 10.4103/tcmj.tcmj_33_18] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 03/01/2018] [Accepted: 03/12/2018] [Indexed: 12/21/2022] Open
Abstract
Acute myocardial ischemia/reperfusion (I/R) injury is a significant, unsolved clinical puzzle. In the disease context of acute myocardial infarction, reperfusion remains the only effective strategy to salvage ischemic myocardium, but it also causes additional damage. Myocardial I/R injury is composed of four types of damage, and these events attenuate the benefits of reperfusion therapy. Thus, inventing new strategies to conquer I/R injury is an unmet clinical need. A variety of pathological processes and mediators, including changes in the pH, generation of reactive oxygen radicals, and intracellular calcium overload, are proposed to be crucial in I/R-related cell injury. Among the intracellular events that occur during I/R, we stress the importance of protein phosphorylation signaling and elaborate its regulation. A variety of protein kinase pathways could be activated in I/R, including reperfusion injury salvage kinase and survivor-activating factor enhancement pathways, which are critical to cardiomyocyte survival. In addition to serine/threonine phosphorylation signaling, protein tyrosine phosphorylation is also critical in multiple cell functions and survival. However, the roles of protein kinases and phosphatases in I/R have not been extensively studied yet. By better understanding the mechanisms of I/R injury, we may have a better chance to develop new strategies for I/R injury and apply them in the clinical patient care.
Collapse
Affiliation(s)
- Chiu-Fen Yang
- Department of Cardiology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Doctoral Degree Program in Translation Medicine, Tzu Chi University and Academia Sinica, Hualien, Taiwan
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
112
|
Liu L, Jiang Y, Steinle JJ. Epac1 protects the retina against ischemia/reperfusion-induced neuronal and vascular damage. PLoS One 2018; 13:e0204346. [PMID: 30235337 PMCID: PMC6147720 DOI: 10.1371/journal.pone.0204346] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 09/05/2018] [Indexed: 12/18/2022] Open
Abstract
We had previously reported that exchange protein for cAMP 1 (Epac1) reduced inflammatory mediators in the retina of mice and in retinal endothelial cells (REC). Since ischemia can induce retinal damage potentially through activation of inflammatory cascades, we hypothesized that Epac1 would protect the retina against neuronal and vascular damage after exposure to ischemia/reperfusion (I/R). We used Epac1 floxed and endothelial cell specific Epac1 knockout mice for this work. We exposed them to ischemia for 90 minutes followed by reperfusion. One day after I/R, some mice were used for fluorescein angiography imaging or Evan’s blue measurements of permeability. Mice were sacrificed at 2 days for neuronal measurements and at 10 days for measurements of degenerate capillaries. Data show increased leakage in the Epac1 Cre-Lox (Epac1 EC-KO) mice exposed to I/R when compared to Epac1 floxed mice with the same treatment. I/R also increased numbers of degenerate capillaries and cell loss in all retinal layers of Epac1 EC-KO mice. Retinal thickness was reduced more significantly in the Epac1 EC-KO mice compared to Epac1 floxed mice after I/R. Taken together, the data suggest that Epac1 is protective against both neuronal and vascular damage to the retina after exposure to I/R.
Collapse
Affiliation(s)
- Li Liu
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit MI, United States of America
| | - Youde Jiang
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit MI, United States of America
| | - Jena J Steinle
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit MI, United States of America
| |
Collapse
|
113
|
Chorawala MR, Prakash P, Doddapattar P, Jain M, Dhanesha N, Chauhan AK. Deletion of Extra Domain A of Fibronectin Reduces Acute Myocardial Ischaemia/Reperfusion Injury in Hyperlipidaemic Mice by Limiting Thrombo-Inflammation. Thromb Haemost 2018; 118:1450-1460. [PMID: 29960272 DOI: 10.1055/s-0038-1661353] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Fibronectin splicing variant containing extra domain A (Fn-EDA), which is an endogenous ligand for Toll-like receptor 4 (TLR4), is present in negligible amounts in the plasma of healthy humans, but markedly elevated in patients with co-morbid conditions including diabetes and hyperlipidaemia, which are risk factors for myocardial infarction (MI). Very little is known about the role of Fn-EDA in the pathophysiology of acute MI under these co-morbid conditions. MATERIALS AND METHODS We determined the role of Fn-EDA in myocardial ischaemia/reperfusion (I/R) injury in the hyperlipidaemic apolipoprotein E-deficient (ApoE-/-) mice. Infarct size, plasma cardiac troponin I (cTnI) levels, intravascular thrombosis (CD41-positive), neutrophil infiltration (Ly6 B.2-positive), neutrophil extracellular traps (citrullinated H3-positive) and myocyte apoptosis (terminal deoxynucleotidyl transferase-mediated dUTP nick-end labelling-positive) were assessed in myocardial I/R injury model (1-hour ischaemia/23 hours of reperfusion). RESULTS Irrespective of gender, Fn-EDA-/-ApoE-/- mice exhibited smaller infarct size and decreased cTnI levels concomitant with reduced post-ischaemic intra-vascular thrombi, neutrophils influx, neutrophil extracellular traps and myocyte apoptosis (p < 0.05 vs. ApoE-/- mice). Genetic deletion of TLR4 attenuated myocardial I/R injury in ApoE-/- mice (p < 0.05 vs. ApoE-/- mice), but did not further reduce in Fn-EDA-/- ApoE-/- mice suggesting that Fn-EDA requires TLR4 to mediate myocardial I/R injury. Bone marrow transplantation experiments revealed that Fn-EDA exacerbates myocardial I/R injury through TLR4 expressed on the haematopoietic cells. Infusion of a specific inhibitor of Fn-EDA, 15 minutes post-reperfusion, into ApoE-/- mice attenuated myocardial I/R injury. CONCLUSION Fn-EDA exacerbates TLR4-dependent myocardial I/R injury by promoting post-ischaemic thrombo-inflammatory response. Targeting Fn-EDA may reduce cardiac damage following coronary artery re-canalization after acute MI.
Collapse
Affiliation(s)
- Mehul R Chorawala
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, United States
| | - Prem Prakash
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, United States
| | - Prakash Doddapattar
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, United States
| | - Manish Jain
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, United States
| | - Nirav Dhanesha
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, United States
| | - Anil K Chauhan
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, United States
| |
Collapse
|
114
|
Exosomes and cardiovascular cell–cell communication. Essays Biochem 2018; 62:193-204. [DOI: 10.1042/ebc20170081] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 03/26/2018] [Accepted: 03/29/2018] [Indexed: 12/21/2022]
Abstract
Exosomes have become an important player in intercellular signaling. These lipid microvesicles can stably transfer miRNA, protein, and other molecules between cells and circulate throughout the body. Exosomes are released by almost all cell types and are present in most if not all biological fluids. The biologically active cargo carried by exosomes can alter the phenotype of recipient cells. Exosomes increasingly are recognized as having an important role in the progression and treatment of cardiac disease states. Injured cardiac cells can release exosomes with important pathological effects on surrounding tissue, in addition to effecting other organs. But of equal interest is the possible benefit(s) conferred by exosomes released from stem cells for use in treatment and possible repair of cardiac damage.
Collapse
|
115
|
Mouton AJ, Rivera OJ, Lindsey ML. Myocardial infarction remodeling that progresses to heart failure: a signaling misunderstanding. Am J Physiol Heart Circ Physiol 2018; 315:H71-H79. [PMID: 29600895 PMCID: PMC6087773 DOI: 10.1152/ajpheart.00131.2018] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
After myocardial infarction, remodeling of the left ventricle involves a wound-healing orchestra involving a variety of cell types. In order for wound healing to be optimal, appropriate communication must occur; these cells all need to come in at the right time, be activated at the right time in the right amount, and know when to exit at the right time. When this occurs, a new homeostasis is obtained within the infarct, such that infarct scar size and quality are sufficient to maintain left ventricular size and shape. The ideal scenario does not always occur in reality. Often, miscommunication can occur between infarct and remote spaces, across the temporal wound-healing spectrum, and across organs. When miscommunication occurs, adverse remodeling can progress to heart failure. This review discusses current knowledge gaps and recent development of the roles of inflammation and the extracellular matrix in myocardial infarction remodeling. In particular, the macrophage is one cell type that provides direct and indirect regulation of both the inflammatory and scar-forming responses. We summarize current research efforts focused on identifying biomarker indicators that reflect the status of each component of the wound-healing process to better predict outcomes.
Collapse
Affiliation(s)
- Alan J Mouton
- Mississippi Center for Heart Research, Department of Physiology and Biophysics, University of Mississippi Medical Center , Jackson, Mississippi
| | - Osvaldo J Rivera
- Mississippi Center for Heart Research, Department of Physiology and Biophysics, University of Mississippi Medical Center , Jackson, Mississippi
| | - Merry L Lindsey
- Mississippi Center for Heart Research, Department of Physiology and Biophysics, University of Mississippi Medical Center , Jackson, Mississippi.,Research Service, G. V. (Sonny) Montgomery Veterans Affairs Medical Center , Jackson, Mississippi
| |
Collapse
|
116
|
Veeranki S, Tyagi SC. Mdivi-1 induced acute changes in the angiogenic profile after ischemia-reperfusion injury in female mice. Physiol Rep 2018; 5:5/11/e13298. [PMID: 28576854 PMCID: PMC5471437 DOI: 10.14814/phy2.13298] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 05/03/2017] [Accepted: 05/04/2017] [Indexed: 01/04/2023] Open
Abstract
The aim of this study is to determine the effects of mitochondrial division inhibitor 1 (Mdivi‐1), the mitochondrial fission inhibitor, on the angiogenic profiles after the ischemia reperfusion injury (IR injury) in female mice. Female mice were treated with Mdivi‐1 inhibitor, 2 days prior, on the day of IR injury and 2 days after IR injury, for a period of 5 days. Both control and treatment groups underwent 30 min of ischemia and 72 h of reperfusion. On the day 3, mice were sacrificed and the ischemic and nonischemic portions of heart tissue were collected. Relative levels of 53 angiogenesis‐related proteins were quantified simultaneously using Angiogenic arrays. Heart function was evaluated before and after 72 h of IR injury. Mdivi‐1 treatment ameliorated IR induced functional deterioration with positive angiogenic profile. The seminal changes include suppression of Matrix metalloproteinase (MMP3), tissue inhibitor of metalloproteases (TIMP1) and chemokine (C‐X‐C motif) ligand 10 (CXCL10) levels and prevention of connexin 43 (Cx43) loss and downregulation in the antioxidant enzyme levels. These changes are correlated with enhanced endothelial progenitor cell marker (cluster of differentiation (CD31), endothelial‐specific receptor tyrosine kinase (Tek), fMS‐like tyrosine kinase 4 (Flt4) and kinase insert domain protein receptor (Kdr)) presence. Our study is the first to report the role of mitochondrial dynamics in regulation of myocardial IR‐induced angiogenic responses. Inhibition of excessive mitochondrial fission after IR injury ameliorated heart dysfunction and conferred positive angiogenic response. In addition, there were improvements in the preservation of Cx43 levels and oxidative stress handling along with suppression of apoptosis activation. The findings will aid in shaping the rational drug development process for the prevention of ischemic heart disease, especially in females.
Collapse
Affiliation(s)
- Sudhakar Veeranki
- Department of Physiology and Biophysics, University of Louisville School of Medicine, Louisville, Kentucky, 40202
| | - Suresh C Tyagi
- Department of Physiology and Biophysics, University of Louisville School of Medicine, Louisville, Kentucky, 40202
| |
Collapse
|
117
|
Abstract
Heart diseases are major causes of mortality. Cardiac hypertrophy, myocardial infarction (MI), viral cardiomyopathy, ischemic and reperfusion (I/R) heart injury finally lead to heart failure and death. Insulin and IGF1 signal pathways play key roles in normal cardiomyocyte growth and physiological cardiac hypertrophy while inflammatory signal pathway is associated with pathological cardiac hypertrophy, MI, viral cardiomyopathy, I/R heart injury, and heart failure. Adapter proteins are the major family proteins, which transduce signals from insulin, IGF1, or cytokine receptors to the downstream pathways and have been shown to regulate variety of heart diseases. Here, we summarized the recent advances in understanding the physiological and pathological roles of adapter proteins in heart failure.
Collapse
Affiliation(s)
- Li Tao
- Cardiovascular Center, 305 Hospital of People's Liberation Army, Beijing, 100017, China
| | - Linna Jia
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), School of Life Sciences, Northeast Normal University, Changchun, 130024, Jilin, China
| | - Yuntian Li
- Cardiovascular Center, 305 Hospital of People's Liberation Army, Beijing, 100017, China
| | - Chengyun Song
- Cardiovascular Center, 305 Hospital of People's Liberation Army, Beijing, 100017, China.
| | - Zheng Chen
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), School of Life Sciences, Northeast Normal University, Changchun, 130024, Jilin, China.
| |
Collapse
|
118
|
Olivares-Silva F, Landaeta R, Aránguiz P, Bolivar S, Humeres C, Anfossi R, Vivar R, Boza P, Muñoz C, Pardo-Jiménez V, Peiró C, Sánchez-Ferrer CF, Díaz-Araya G. Heparan sulfate potentiates leukocyte adhesion on cardiac fibroblast by enhancing Vcam-1 and Icam-1 expression. Biochim Biophys Acta Mol Basis Dis 2018; 1864:831-842. [DOI: 10.1016/j.bbadis.2017.12.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 11/30/2017] [Accepted: 12/04/2017] [Indexed: 12/14/2022]
|
119
|
Lewis AJM, Miller JJ, Lau AZ, Curtis MK, Rider OJ, Choudhury RP, Neubauer S, Cunningham CH, Carr CA, Tyler DJ. Noninvasive Immunometabolic Cardiac Inflammation Imaging Using Hyperpolarized Magnetic Resonance. Circ Res 2018; 122:1084-1093. [PMID: 29440071 PMCID: PMC5908252 DOI: 10.1161/circresaha.117.312535] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2017] [Revised: 02/04/2018] [Accepted: 02/12/2018] [Indexed: 12/12/2022]
Abstract
RATIONALE Current cardiovascular clinical imaging techniques offer only limited assessment of innate immune cell-driven inflammation, which is a potential therapeutic target in myocardial infarction (MI) and other diseases. Hyperpolarized magnetic resonance (MR) is an emerging imaging technology that generates contrast agents with 10- to 20 000-fold improvements in MR signal, enabling cardiac metabolite mapping. OBJECTIVE To determine whether hyperpolarized MR using [1-13C]pyruvate can assess the local cardiac inflammatory response after MI. METHODS AND RESULTS We performed hyperpolarized [1-13C]pyruvate MR studies in small and large animal models of MI and in macrophage-like cell lines and measured the resulting [1-13C]lactate signals. MI caused intense [1-13C]lactate signal in healing myocardial segments at both day 3 and 7 after rodent MI, which was normalized at both time points after monocyte/macrophage depletion. A near-identical [1-13C]lactate signature was also seen at day 7 after experimental MI in pigs. Hyperpolarized [1-13C]pyruvate MR spectroscopy in macrophage-like cell suspensions demonstrated that macrophage activation and polarization with lipopolysaccharide almost doubled hyperpolarized lactate label flux rates in vitro; blockade of glycolysis with 2-deoxyglucose in activated cells normalized lactate label flux rates and markedly inhibited the production of key proinflammatory cytokines. Systemic administration of 2-deoxyglucose after rodent MI normalized the hyperpolarized [1-13C]lactate signal in healing myocardial segments at day 3 and also caused dose-dependent improvement in IL (interleukin)-1β expression in infarct tissue without impairing the production of key reparative cytokines. Cine MRI demonstrated improvements in systolic function in 2-DG (2-deoxyglucose)-treated rats at 3 months. CONCLUSIONS Hyperpolarized MR using [1-13C]pyruvate provides a novel method for the assessment of innate immune cell-driven inflammation in the heart after MI, with broad potential applicability across other cardiovascular disease states and suitability for early clinical translation.
Collapse
Affiliation(s)
- Andrew J M Lewis
- From the Department of Physiology, Anatomy, and Genetics (A.J.M.L., J.J.M., M.K.C., C.A.C., D.J.T.), Department of Physics, Clarendon Laboratory (J.J.M.), Radcliffe Department of Medicine (A.J.M.L., O.J.R., R.P.C., S.N.), and Acute Vascular Imaging Centre (R.P.C.), Radcliffe Department of Medicine, University of Oxford, United Kingdom; and Department of Medical Biophysics, University of Toronto, Ontario, Canada (A.Z.L., C.H.C.)
| | - Jack J Miller
- From the Department of Physiology, Anatomy, and Genetics (A.J.M.L., J.J.M., M.K.C., C.A.C., D.J.T.), Department of Physics, Clarendon Laboratory (J.J.M.), Radcliffe Department of Medicine (A.J.M.L., O.J.R., R.P.C., S.N.), and Acute Vascular Imaging Centre (R.P.C.), Radcliffe Department of Medicine, University of Oxford, United Kingdom; and Department of Medical Biophysics, University of Toronto, Ontario, Canada (A.Z.L., C.H.C.)
| | - Angus Z Lau
- From the Department of Physiology, Anatomy, and Genetics (A.J.M.L., J.J.M., M.K.C., C.A.C., D.J.T.), Department of Physics, Clarendon Laboratory (J.J.M.), Radcliffe Department of Medicine (A.J.M.L., O.J.R., R.P.C., S.N.), and Acute Vascular Imaging Centre (R.P.C.), Radcliffe Department of Medicine, University of Oxford, United Kingdom; and Department of Medical Biophysics, University of Toronto, Ontario, Canada (A.Z.L., C.H.C.)
| | - Mary K Curtis
- From the Department of Physiology, Anatomy, and Genetics (A.J.M.L., J.J.M., M.K.C., C.A.C., D.J.T.), Department of Physics, Clarendon Laboratory (J.J.M.), Radcliffe Department of Medicine (A.J.M.L., O.J.R., R.P.C., S.N.), and Acute Vascular Imaging Centre (R.P.C.), Radcliffe Department of Medicine, University of Oxford, United Kingdom; and Department of Medical Biophysics, University of Toronto, Ontario, Canada (A.Z.L., C.H.C.)
| | - Oliver J Rider
- From the Department of Physiology, Anatomy, and Genetics (A.J.M.L., J.J.M., M.K.C., C.A.C., D.J.T.), Department of Physics, Clarendon Laboratory (J.J.M.), Radcliffe Department of Medicine (A.J.M.L., O.J.R., R.P.C., S.N.), and Acute Vascular Imaging Centre (R.P.C.), Radcliffe Department of Medicine, University of Oxford, United Kingdom; and Department of Medical Biophysics, University of Toronto, Ontario, Canada (A.Z.L., C.H.C.)
| | - Robin P Choudhury
- From the Department of Physiology, Anatomy, and Genetics (A.J.M.L., J.J.M., M.K.C., C.A.C., D.J.T.), Department of Physics, Clarendon Laboratory (J.J.M.), Radcliffe Department of Medicine (A.J.M.L., O.J.R., R.P.C., S.N.), and Acute Vascular Imaging Centre (R.P.C.), Radcliffe Department of Medicine, University of Oxford, United Kingdom; and Department of Medical Biophysics, University of Toronto, Ontario, Canada (A.Z.L., C.H.C.)
| | - Stefan Neubauer
- From the Department of Physiology, Anatomy, and Genetics (A.J.M.L., J.J.M., M.K.C., C.A.C., D.J.T.), Department of Physics, Clarendon Laboratory (J.J.M.), Radcliffe Department of Medicine (A.J.M.L., O.J.R., R.P.C., S.N.), and Acute Vascular Imaging Centre (R.P.C.), Radcliffe Department of Medicine, University of Oxford, United Kingdom; and Department of Medical Biophysics, University of Toronto, Ontario, Canada (A.Z.L., C.H.C.)
| | - Charles H Cunningham
- From the Department of Physiology, Anatomy, and Genetics (A.J.M.L., J.J.M., M.K.C., C.A.C., D.J.T.), Department of Physics, Clarendon Laboratory (J.J.M.), Radcliffe Department of Medicine (A.J.M.L., O.J.R., R.P.C., S.N.), and Acute Vascular Imaging Centre (R.P.C.), Radcliffe Department of Medicine, University of Oxford, United Kingdom; and Department of Medical Biophysics, University of Toronto, Ontario, Canada (A.Z.L., C.H.C.)
| | - Carolyn A Carr
- From the Department of Physiology, Anatomy, and Genetics (A.J.M.L., J.J.M., M.K.C., C.A.C., D.J.T.), Department of Physics, Clarendon Laboratory (J.J.M.), Radcliffe Department of Medicine (A.J.M.L., O.J.R., R.P.C., S.N.), and Acute Vascular Imaging Centre (R.P.C.), Radcliffe Department of Medicine, University of Oxford, United Kingdom; and Department of Medical Biophysics, University of Toronto, Ontario, Canada (A.Z.L., C.H.C.)
| | - Damian J Tyler
- From the Department of Physiology, Anatomy, and Genetics (A.J.M.L., J.J.M., M.K.C., C.A.C., D.J.T.), Department of Physics, Clarendon Laboratory (J.J.M.), Radcliffe Department of Medicine (A.J.M.L., O.J.R., R.P.C., S.N.), and Acute Vascular Imaging Centre (R.P.C.), Radcliffe Department of Medicine, University of Oxford, United Kingdom; and Department of Medical Biophysics, University of Toronto, Ontario, Canada (A.Z.L., C.H.C.).
| |
Collapse
|
120
|
Yu L, Feng Z. The Role of Toll-Like Receptor Signaling in the Progression of Heart Failure. Mediators Inflamm 2018; 2018:9874109. [PMID: 29576748 PMCID: PMC5822798 DOI: 10.1155/2018/9874109] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 11/28/2017] [Accepted: 12/14/2017] [Indexed: 12/14/2022] Open
Abstract
Medical systems worldwide are being faced with a growing need to understand mechanisms behind the pathogenesis of heart failure (HF) that is considered as a leading cause of morbidity and mortality around the world. Elevated levels of inflammatory mediators have been identified in patients with HF, which are primarily manifestations of innate immune responses mediated by pattern recognition receptors (PRRs). Toll-like receptors (TLRs), which belong to PRRs, are subjected to the release of pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs) to generate innate immune responses. More and more emerging data indicate that TLR signaling pathway molecules are involved in the progression of HF. Herein, we present new data with regard to the activation of TLRs in the failing heart, focusing on TLR2, TLR3, TLR4, and TLR9, and suggest the potential use of TLRs in target therapy.
Collapse
Affiliation(s)
- Lili Yu
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan 453003, China
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA 70808, USA
- Henan Key Laboratory of immunology and Targeted Drugs, Xinxiang, Henan 453003, China
- Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang, Henan 453003, China
| | - Zhiwei Feng
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan 453003, China
| |
Collapse
|
121
|
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
|
122
|
Inflammation following acute myocardial infarction: Multiple players, dynamic roles, and novel therapeutic opportunities. Pharmacol Ther 2018; 186:73-87. [PMID: 29330085 PMCID: PMC5981007 DOI: 10.1016/j.pharmthera.2018.01.001] [Citation(s) in RCA: 531] [Impact Index Per Article: 88.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Acute myocardial infarction (AMI) and the heart failure that often follows, are major causes of death and disability worldwide. As such, new therapies are required to limit myocardial infarct (MI) size, prevent adverse left ventricular (LV) remodeling, and reduce the onset of heart failure following AMI. The inflammatory response to AMI, plays a critical role in determining MI size, and a persistent pro-inflammatory reaction can contribute to adverse post-MI LV remodeling, making inflammation an important therapeutic target for improving outcomes following AMI. In this article, we provide an overview of the multiple players (and their dynamic roles) involved in the complex inflammatory response to AMI and subsequent LV remodeling, and highlight future opportunities for targeting inflammation as a therapeutic strategy for limiting MI size, preventing adverse LV remodeling, and reducing heart failure in AMI patients.
Collapse
|
123
|
Seo EH, Song GY, Namgung JH, Oh CS, Lee SH, Kim SH. Receptor for activated C kinase 1 in rats with ischemia-reperfusion injury: intravenous versus inhalation anaesthetic agents. Int J Med Sci 2018; 15:352-358. [PMID: 29511370 PMCID: PMC5835705 DOI: 10.7150/ijms.22591] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 01/12/2018] [Indexed: 01/04/2023] Open
Abstract
Background: The study examined the difference in the expression of the receptor for activated C kinase 1 (RACK1) between anaesthesia with propofol and isoflurane in rats with myocardial ischemia-reperfusion injury (IRI). Methods: Male Sprague-Dawley rats were studied. Anaesthesia was induced with xylazine 20 µg/g by intraperitoneal injection and maintained with propofol or isoflurane. Myocardial IRI was induced by ligating the left anterior descending artery for 1 hour. Reactive oxygen species (ROS), cardiomyocyte apoptosis, the expression of RACK1 and toll-like receptor 4 (TLR4), and the heart injury score were compared between the two groups. Results: Cardiomyocyte apoptosis with ROS was significantly lower in the propofol group than in the isoflurane group. The propofol group had significantly higher RACK1 expression and lower TLR4 expression, compared with the isoflurane group (RACK1, 1970.50 ± 120.50 vs. 1350.20 ± 250.30, p<0.05; TLR4, 980.50 ± 110.75 vs. 1275.50 ± 75.35, p<0.05). However, the heart injury scores in the two groups did not differ significantly (3.56 ± 0.29 vs. 4.33 ± 0.23 in the propofol and isoflurane groups, respectively, p=0.33). Conclusion: There were significant differences in inflammation and apoptosis, including the expression of RACK1 and TLR4, after myocardial IRI between the propofol and isoflurane groups. However, both groups had similar heart injury scores.
Collapse
Affiliation(s)
- Eun-Hye Seo
- BK21 Plus, Department of Cellular and Molecular Medicine, Konkuk University School of Medicine, Seoul, Korea
| | - Ga-Yun Song
- Department of Microbiology, Konkuk University School of Medicine, Seoul, Korea
| | - Ji Hyeon Namgung
- Department of Microbiology, Konkuk University School of Medicine, Seoul, Korea
| | - Chung-Sik Oh
- Department of Medicine, Institute of Biomedical Science and Technology, Konkuk University School of Medicine, Seoul, Korea
| | - Seung Hyun Lee
- Department of Microbiology, Konkuk University School of Medicine, Seoul, Korea.,Department of Medicine, Institute of Biomedical Science and Technology, Konkuk University School of Medicine, Seoul, Korea
| | - Seong-Hyop Kim
- Department of Medicine, Institute of Biomedical Science and Technology, Konkuk University School of Medicine, Seoul, Korea.,Department of Anesthesiology and Pain medicine, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul, Korea.,Department of Infection and Immunology, Konkuk University School of Medicine, Seoul, Korea
| |
Collapse
|
124
|
Saquinavir Ameliorates Liver Warm Ischemia-Reperfusion-Induced Lung Injury via HMGB-1- and P38/JNK-Mediated TLR-4-Dependent Signaling Pathways. Mediators Inflamm 2017; 2017:7083528. [PMID: 29440779 PMCID: PMC5758951 DOI: 10.1155/2017/7083528] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 11/13/2017] [Accepted: 11/22/2017] [Indexed: 12/11/2022] Open
Abstract
Liver ischemia and reperfusion (I/R) induce local and distant tissue injuries, contributing to morbidity and mortality in a wider range of pathologies. This is especially seen under uncontrolled aseptic inflammatory conditions, leading to injury of remote organs, such as lung injury, and even failure. Saquinavir (SQV) is a kind of HIV protease inhibitor that possesses an anti-inflammatory property. In this study, we investigated whether SQV suppresses Toll-like receptor 4- (TLR4-) dependent signaling pathways of high-mobility group box 1 (HMGB1) and P38/JNK, conferring protection against murine liver I/R-induced lung injury. To investigate our hypothesis, C57BL/6 mice and TLR4 knockout mice (TLR4−/−) were used to perform the study. SQV administration markedly attenuated remote lung tissue injury after 1-hour ischemia and 6-hour reperfusion of the liver. To our expectation, SQV attenuated I/R-induced lung edema, hyperpermeability, and pathological injury. The beneficial effects of SQV were associated with decreased levels of circulating and lung tissue inflammatory cytokines, such as IL-6, IL-1β, TNF-α, and iNOS. The protective effect of SQV was also associated with decreased lung tissue expression of HMGB1, TLR-4, and p-P38/JNK, but not p-ERK in wild-type liver I/R mice. Overall, this study demonstrated a new role of SQV, facilitating negative regulation of HMGB1- and P38/JNK-mediated TLR-4-dependent signaling pathways, conferring protection against liver I/R-induced lung injury.
Collapse
|
125
|
Ellenbroek GHJM, de Haan JJ, van Klarenbosch BR, Brans MAD, van de Weg SM, Smeets MB, de Jong S, Arslan F, Timmers L, Goumans MJTH, Hoefer IE, Doevendans PA, Pasterkamp G, Meyaard L, de Jager SCA. Leukocyte-Associated Immunoglobulin-like Receptor-1 is regulated in human myocardial infarction but its absence does not affect infarct size in mice. Sci Rep 2017; 7:18039. [PMID: 29269840 PMCID: PMC5740066 DOI: 10.1038/s41598-017-13678-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 09/27/2017] [Indexed: 01/04/2023] Open
Abstract
Heart failure after myocardial infarction (MI) depends on infarct size and adverse left ventricular (LV) remodelling, both influenced by the inflammatory response. Leukocyte-associated immunoglobulin-like receptor 1 (LAIR-1) is an inhibitory receptor of ITAM-dependent cell activation, present on almost all immune cells. We investigated regulation of LAIR-1 leukocyte expression after MI in patients and hypothesized that its absence in a mouse model of MI would increase infarct size and adverse remodelling. In patients, LAIR-1 expression was increased 3 days compared to 6 weeks after MI on circulating monocytes (24.8 ± 5.3 vs. 21.2 ± 5.1 MFI, p = 0.008) and neutrophils (12.9 ± 4.7 vs. 10.6 ± 3.1 MFI, p = 0.046). In WT and LAIR-1-/- mice, infarct size after ischemia-reperfusion injury was comparable (37.0 ± 14.5 in WT vs. 39.4 ± 12.2% of the area at risk in LAIR-1-/-, p = 0.63). Remodelling after permanent left coronary artery ligation did not differ between WT and LAIR-1-/- mice (end-diastolic volume 133.3 ± 19.3 vs. 132.1 ± 27.9 μL, p = 0.91 and end-systolic volume 112.1 ± 22.2 vs. 106.9 ± 33.5 μL, p = 0.68). Similarly, no differences were observed in inflammatory cell influx or fibrosis. In conclusion, LAIR-1 expression on monocytes and neutrophils is increased in the acute phase after MI in patients, but the absence of LAIR-1 in mice does not influence infarct size, inflammation, fibrosis or adverse cardiac remodelling.
Collapse
Affiliation(s)
| | - Judith J de Haan
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Bas R van Klarenbosch
- Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Maike A D Brans
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Sander M van de Weg
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Mirjam B Smeets
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Sanne de Jong
- Department of Medical Physiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Fatih Arslan
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands.,Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Leo Timmers
- Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Marie-José T H Goumans
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Imo E Hoefer
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands.,Department of Clinical Chemistry and Haematology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Pieter A Doevendans
- Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands.,Netherlands Heart Institute, Utrecht, The Netherlands
| | - Gerard Pasterkamp
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands.,Department of Clinical Chemistry and Haematology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Linde Meyaard
- Laboratory of Translational Immunology, Department of Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Saskia C A de Jager
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands. .,Laboratory of Translational Immunology, Department of Immunology, University Medical Center Utrecht, Utrecht, The Netherlands.
| |
Collapse
|
126
|
Chen C, Chen W, Li Y, Dong Y, Teng X, Nong Z, Pan X, Lv L, Gao Y, Wu G. Hyperbaric oxygen protects against myocardial reperfusion injury via the inhibition of inflammation and the modulation of autophagy. Oncotarget 2017; 8:111522-111534. [PMID: 29340072 PMCID: PMC5762340 DOI: 10.18632/oncotarget.22869] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 08/27/2017] [Indexed: 01/01/2023] Open
Abstract
Our previous study demonstrated that hyperbaric oxygen (HBO) preconditioning protected against myocardial ischemia reperfusion injury (MIRI) and improved myocardial infarction. However, HBO’s effect on MIRI-induced inflammation and autophagy remains unclear. In this study, we investigate the potential impact and underlying mechanism of HBO preconditioning on an MIRI-induced inflammatory response and autophagy using a ligation of the left anterior descending (LAD) coronary artery rat model. Our results showed that HBO restored myocardial enzyme levels and decreased the apoptosis of cardiomyocytes, which were induced by MIRI. Moreover, HBO significantly suppressed MIRI-induced inflammatory cytokines. This effect was associated with the inhibition of the TLR4-nuclear factor kappa-B (NF-κB) pathway. Interestingly, lower expression levels of microtubule-associated protein 1 light chain 3B (LC3B) and Beclin-1 were observed in the HBO-treatment group. Furthermore, we observed that HBO reduced excessive autophagy by activating the mammalian target of the rapamycin (mTOR) pathway, as evidenced by higher expression levels of threonine protein kinase (Akt) and phosphorylated-mTOR. In conclusion, HBO protected cardiomocytes during MIRI by attenuating inflammation and autophagy. Our results provide a new mechanistic insight into the cardioprotective role of HBO against MIRI.
Collapse
Affiliation(s)
- Chunxia Chen
- Department of Hyperbaric Oxygen, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, P. R. China
| | - Wan Chen
- Department of Emergency, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, P. R. China
| | - Yaoxuan Li
- Department of Neurology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, P. R. China
| | - Yanling Dong
- Department of Neurology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, P. R. China
| | - Xiaoming Teng
- Department of Neurology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, P. R. China
| | - Zhihuan Nong
- Department of Pharmacology, Guangxi Institute of Chinese Medicine and Pharmaceutical Science, Nanning, Guangxi 530022, P. R. China
| | - Xiaorong Pan
- Department of Hyperbaric Oxygen, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, P. R. China
| | - Liwen Lv
- Department of Emergency, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, P. R. China
| | - Ying Gao
- Department of Biology and Tennessee Center for Botanical Medicine Research, Middle Tennessee State University, Murfreesboro, TN 37132, USA
| | - Guangwei Wu
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, P. R. China
| |
Collapse
|
127
|
Suppression of Stim1 reduced intracellular calcium concentration and attenuated hypoxia/reoxygenation induced apoptosis in H9C2 cells. Biosci Rep 2017; 37:BSR20171249. [PMID: 29089467 PMCID: PMC5700273 DOI: 10.1042/bsr20171249] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 10/23/2017] [Accepted: 10/26/2017] [Indexed: 01/04/2023] Open
Abstract
Objective: Previous studies have demonstrated Stromal interaction molecule 1 (STIM1)-mediated store-operated Ca2+ entry (SOCE) contributes to intracellular Ca2+ accumulation. The present study aimed to investigate the expression of STIM1 and its downstream molecules Orai1/TRPC1 in the context of myocardial ischemia/reperfusion injury (MIRI) and the effect of STIM1 inhibition on Ca2+ accumulation and apoptosis in H9c2 cardiomyocytes subjected to hypoxia/reoxygenation (H/R). Methods: Expression of STIM1/Orai1/TRPC1 was determined by RT-PCR and Western blot in mice subjected to MIRI and H9C2 cardiomyocytes subjected to H/R. To knock-down STIM1, H9C2 cardiomyocytes was transfected with Stealth SiRNA. Apoptosis was analyzed by both flow cytometry and TUNEL assay. Cell viability was measured by MTT assay. Intracellular Ca2+ concentration was detected by laser scanning confocal microscopy using Fluo-3/AM probe. Furthermore, the opening of mitochondrial permeability transition pore (mPTP) was assessed by coloading with calcein AM and CoCl2, while ROS generation was evaluated using the dye DCFH-DA in H9C2 cardiomyocytes. Results: Expression of STIM1/Orai1/TRPC1 significantly increased in transcript and translation level after MIRI in vivo and H/R in vitro. In H9C2 cardiomyocytes subjected to H/R, intracellular Ca2+ accumulation significantly increased compared with control group, along with enhanced mPTP opening and elevated ROS generation. However, suppression of STIM1 by SiRNA significantly decreased apoptosis and intracellular Ca2+ accumulation induced by H/R in H9C2 cardiomyocytes, accompanied by attenuated mPTP opening and decreased ROS generation. In addition, suppression of STIM1 increased the Bcl-2/Bax ratio, decreased Orai1/TRPC1, and cleaved caspase-3 expression. Conclusion: Suppression of STIM1 reduced intracellular calcium level and attenuated hypoxia/reoxygenation induced apoptosis in H9C2 cardiomyocytes. Our findings provide a new perspective in understanding STIM1-mediated calcium overload in the setting of MIRI.
Collapse
|
128
|
The Role of Toll-Like Receptors and Vitamin D in Cardiovascular Diseases-A Review. Int J Mol Sci 2017; 18:ijms18112252. [PMID: 29077004 PMCID: PMC5713222 DOI: 10.3390/ijms18112252] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Revised: 10/24/2017] [Accepted: 10/25/2017] [Indexed: 12/17/2022] Open
Abstract
Cardiovascular diseases are the leading cause of mortality worldwide. Therefore, a better understanding of their pathomechanisms and the subsequent implementation of optimal prophylactic and therapeutic strategies are of utmost importance. A growing body of evidence states that low-grade inflammation is a common feature for most of the cardiovascular diseases in which the contributing factors are the activation of toll-like receptors (TLRs) and vitamin D deficiency. In this article, available data concerning the association of cardiovascular diseases with TLRs and vitamin D status are reviewed, followed by a discussion of new possible approaches to cardiovascular disease management.
Collapse
|
129
|
Hung YC, Kuo YJ, Huang SS, Huang TF. Trimucrin, an Arg-Gly-Asp containing disintegrin, attenuates myocardial ischemia-reperfusion injury in murine by inhibiting platelet function. Eur J Pharmacol 2017; 813:24-32. [DOI: 10.1016/j.ejphar.2017.07.039] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Revised: 07/19/2017] [Accepted: 07/20/2017] [Indexed: 01/04/2023]
|
130
|
Liu J, Sui H, Zhao J, Wang Y. Osmotin Protects H9c2 Cells from Simulated Ischemia-Reperfusion Injury through AdipoR1/PI3K/AKT Signaling Pathway. Front Physiol 2017; 8:611. [PMID: 28993734 PMCID: PMC5622187 DOI: 10.3389/fphys.2017.00611] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 08/09/2017] [Indexed: 01/04/2023] Open
Abstract
Objective: This study aimed to investigate the effect of osmotin on myocardial ischemia/reperfusion (I/R), as well as the underlying mechanisms. Methods:In vitro I/R injury model was established on rat cardiac myoblast H9c2 cells by oxygen and glucose deprivation followed by reperfusion (OGD/R). Cells were administrated with osmotin, and transfected with small interfering RNAs (siRNAs) which specifically target adiponectin receptor 1 or 2 (AdipoR1/2). Besides, the cells were incubated with or without LY294002 as inhibitor of phosphatidylinositol 3-kinase (PI3K) under OGD/R condition. Cell viability, apoptosis, expressions of apoptosis-related proteins and inflammatory factors were analyzed. Results: The results showed that osmotin significantly increased H9c2 cells viability compared with the cells treated with vehicle (P < 0.05), and decreased H9c2 cells apoptosis by regulating expressions of apoptosis-related proteins. Moreover, we observed that osmotin statistically reduced the release of proinflammatory factors and increased the release of anti-inflammatory factors in H9c2 cells (P < 0.05). However, these effects were markedly reversed by AdipoR1 silence but not AdipoR2. Furthermore, osmotin dramatically upregulated the phosphorylation levels of PI3K, AKT, ERK, and downregulated the phosphorylation level of NF-κB (P < 0.05). While administration of LY294002 reduced cell viability, increased cell apoptosis, and aggravated inflammatory response (P < 0.05). Conclusion: Our results suggested that the protective effect of osmotin on the simulated OGD/R injured H9c2 cells might be associated with AdipoR1/PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Jianhua Liu
- Department of Cardiology, Xinxiang Central HospitalXinxiang, Henan, China
| | - Hua Sui
- Department of Endocrinology, Xinxiang Central HospitalXinxiang, Henan, China
| | - Jianlin Zhao
- Department of Endocrinology, Xinxiang Central HospitalXinxiang, Henan, China
| | - Yan Wang
- Department of Cardiology, Xinxiang Central HospitalXinxiang, Henan, China
| |
Collapse
|
131
|
Yang YF, Peng K, Liu H, Meng XW, Zhang JJ, Ji FH. Dexmedetomidine preconditioning for myocardial protection in ischaemia-reperfusion injury in rats by downregulation of the high mobility group box 1-toll-like receptor 4-nuclear factor κB signalling pathway. Clin Exp Pharmacol Physiol 2017; 44:353-361. [PMID: 27998004 DOI: 10.1111/1440-1681.12711] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Revised: 12/02/2016] [Accepted: 12/08/2016] [Indexed: 01/04/2023]
Abstract
Pharmacological preconditioning reduces myocardial infarct size in ischaemia-reperfusion (I-R) injury. Dexmedetomidine, a selective α2 -adrenoceptor agonist, has a proven cardioprotective effect when administered prior to I-R, although the underlying mechanisms for this effect are not fully understood. We evaluated whether dexmedetomidine preconditioning could induce a myocardio-protective effect against I-R injury by inhibiting associated inflammatory processes through downregulation of the high mobility group box 1 (HMGB1)-toll-like receptor 4 (TLR4)-nuclear factor κB (NF-κB) signalling pathway. Seventy rats were randomly assigned to seven groups: a control and six test groups, involving I-R for 30 and 120 minutes, respectively, in isolated rat hearts and different pretreatment protocols with dexmedetomidine (10 nmol/L) as well as yohimbine (1 μmol/L) and recombinant HMGB1 peptide (rHMGB1; 20 μg/L), alone or in combination with dexmedetomidine. Cardiac function was recorded; myocardial HMGB1, TLR4, and NF-κB activities and levels of tumour necrosis factor-α (TNF-α) and interleukin-6 (IL-6) were measured as were lactate dehydrogenase (LDH) and creatine kinase (CK) in coronary outflow. Dexmedetomidine preconditioning significantly improved cardiac function (P<.05), downregulated the expression of HMGB1-TLR4-NF-κB, reduced levels of TNF-α and IL-6 in isolated ventricles during I-R injury, and significantly reduced CK and LDH levels in coronary outflow (P<.05). All of these effects were partially reversed by yohimbine (P<.05) or rHMGB1 (P<.05). Dexmedetomidine preconditioning alleviated myocardial I-R injury in rats through inhibition of inflammatory processes associated with downregulation of the HMGB1-TLR4-NF-κB signalling pathway via activation at α2 -adrenergic receptors.
Collapse
Affiliation(s)
- Yu-Fan Yang
- Department of Anaesthesiology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Ke Peng
- Department of Anaesthesiology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Hong Liu
- Department of Anaesthesiology and Pain Medicine, University of California Davis Health System, Sacramento, CA, USA
| | - Xiao-Wen Meng
- Department of Anaesthesiology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Jing-Jing Zhang
- Department of Anaesthesiology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Fu-Hai Ji
- Department of Anaesthesiology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
132
|
Ren Z, Zhang R, Li Y, Li Y, Yang Z, Yang H. Ferulic acid exerts neuroprotective effects against cerebral ischemia/reperfusion-induced injury via antioxidant and anti-apoptotic mechanisms in vitro and in vivo. Int J Mol Med 2017; 40:1444-1456. [PMID: 28901374 PMCID: PMC5627889 DOI: 10.3892/ijmm.2017.3127] [Citation(s) in RCA: 128] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 08/24/2017] [Indexed: 12/21/2022] Open
Abstract
Ferulic acid (FA) is a derivative of cinnamic acid. It is used in the treatment of heart head blood-vessel disease and exerts protective effects against hypoxia/ischemia-induced cell injury in the brain. This study investigated the potential neuroprotective effects of FA against ischemia/reperfusion (I/R)-induced brain injury in vivo and in vitro through hematoxylin and eosin (H&E) and Nissl staining assays, flow cytometry, Hoechst 33258 staining, quantitative PCR, western blot analysis and fluorescence microscopic analysis. In this study, models of cerebral I/R injury were established using rats and pheochromocytoma (PC-12) cells. The results revealed that treatment with FA significantly attenuated memory impairment, and reduced hippocampal neuronal apoptosis and oxidative stress in a dose-dependent manner. The results from in vitro experiments also indicated that FA protected the PC-12 cells against I/R-induced reactive oxygen species (ROS) generation and apoptosis by inhibiting apoptosis, Ca2+ influx, superoxide anion (O2-), malondialdehyde (MDA) and glutathione peroxidase (GSH-Px) production in a concentration-dependent manner. Moreover, FA inactivated the Toll-like receptor (TLR)/myeloid differentiation factor 88 (MyD88) pathway. MyD88 overexpression abolished the neuroprotective effects of FA. On the whole, we found that FA attenuated memory dysfunction and exerted protective effects against oxidative stress and apoptosis induced by I/R injury by inhibiting the TLR4/MyD88 signaling pathway. This study supports the view that FA may be a promising neuroprotective agent for use in the treatment of cerebral ischemia.
Collapse
Affiliation(s)
- Zhongkun Ren
- Department of Neurosurgery, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Rongping Zhang
- Biomedical Engineering Center, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Yuanyuan Li
- Department of Neurosurgery, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Yu Li
- Department of Neurosurgery, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Zhiyong Yang
- Department of Neurosurgery, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Hui Yang
- Biomedical Engineering Center, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| |
Collapse
|
133
|
Poe AJ, Knowlton AA. Exosomes as agents of change in the cardiovascular system. J Mol Cell Cardiol 2017; 111:40-50. [PMID: 28782514 DOI: 10.1016/j.yjmcc.2017.08.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 07/21/2017] [Accepted: 08/02/2017] [Indexed: 12/17/2022]
Abstract
Exosomes have an evolving role in paracrine and autocrine signaling, which is enhanced because these lipid vesicles are quite stable and can deliver miRNA, DNA, protein and other molecules to cells throughout the body. Most cell types release exosomes, and exosomes are found in all biological fluids, making them accessible biomarkers. Significantly, exosomes can carry a biologically potent cargo, which can alter the phenotype of recipient cells. In the cardiovascular system exosomes have been primarily studied for their role in mediating the beneficial effects of mesenchymal stem cells after myocardial injury. Exosomes released by cardiac cells in disease states, such as myocardial ischemia, can potentially have important pathophysiologic effects on other cardiac cells as well as on distant organs.
Collapse
Affiliation(s)
- A J Poe
- Molecular & Cellular Cardiology, Cardiovascular Division, Department of Medicine, University of California-Davis, Davis, CA, United States
| | - A A Knowlton
- VA Medical Center Sacramento, University of California-Davis, Davis, CA, United States; Molecular & Cellular Cardiology, Cardiovascular Division, Department of Medicine, University of California-Davis, Davis, CA, United States; Pharmacology Department, University of California-Davis, Davis, CA, United States.
| |
Collapse
|
134
|
PRDX2 in Myocyte Hypertrophy and Survival is Mediated by TLR4 in Acute Infarcted Myocardium. Sci Rep 2017; 7:6970. [PMID: 28765537 PMCID: PMC5539327 DOI: 10.1038/s41598-017-06718-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 06/16/2017] [Indexed: 01/04/2023] Open
Abstract
Peroxiredoxin 2 (PRDX2) is an antioxidant and molecular chaperone that can be secreted from tumor cells. But the role of PRDX2 in acute myocardial infarction (AMI) is not clear. In the current study, we demonstrate the role of PRDX2 from clinical trials, H9c2 cells and in a mouse model. ELISA analysis shows that serum concentrations of VEGF and inflammatory factor IL-1β, TNF-α and IL-6 were increased in AMI patients compared to a control group. The expression of PRDX2 was also upregulated. In vivo experiments show that the expression of PRDX2 inhibits hypoxia-induced oxidative stress injury to H9c2 cells. However, PRDX2 expression promotes TLR4 mediated inflammatory factor expression and VEGF expression under hypoxia conditions. PRDX2 overexpression in H9c2 cells also promotes human endothelial cell migration, vasculogenic mimicry formation and myocardial hypertrophy related protein expression. The overexpression of PRDX2 inhibits ROS level and myocardial injury after AMI but promotes inflammatory responses in vivo. Immunocytochemistry and immunofluorescence analysis show that overexpression of PRDX2 promotes angiogenesis and myocardial hypertrophy. Taken together, our results indicate that PRDX2 plays two roles in acute infarction – the promotion of cell survival and inflammatory myocardial hypertrophy.
Collapse
|
135
|
Zhang Y, Huang Z, Li H. Insights into innate immune signalling in controlling cardiac remodelling. Cardiovasc Res 2017; 113:1538-1550. [DOI: 10.1093/cvr/cvx130] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 06/29/2017] [Indexed: 01/22/2023] Open
Affiliation(s)
- Yaxing Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuchang District, Wuhan 430060, People’s Republic of China
- Institute of Model Animal of Wuhan University, Donghu Road 115, Wuchang District, Wuhan 430071, People’s Republic of China
- Medical Research Institute, School of Medicine, Wuhan University, Donghu Road 115, Wuchang District, Wuhan 430071, People’s Republic of China
| | - Zan Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuchang District, Wuhan 430060, People’s Republic of China
- Institute of Model Animal of Wuhan University, Donghu Road 115, Wuchang District, Wuhan 430071, People’s Republic of China
- Medical Research Institute, School of Medicine, Wuhan University, Donghu Road 115, Wuchang District, Wuhan 430071, People’s Republic of China
- College of Life Sciences, Wuhan University, Wuhan 430072, People’s Republic of China
| | - Hongliang Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuchang District, Wuhan 430060, People’s Republic of China
- Institute of Model Animal of Wuhan University, Donghu Road 115, Wuchang District, Wuhan 430071, People’s Republic of China
- Medical Research Institute, School of Medicine, Wuhan University, Donghu Road 115, Wuchang District, Wuhan 430071, People’s Republic of China
| |
Collapse
|
136
|
The protective effect of Luteolin on myocardial ischemia/reperfusion (I/R) injury through TLR4/NF-κB/NLRP3 inflammasome pathway. Biomed Pharmacother 2017; 91:1042-1052. [DOI: 10.1016/j.biopha.2017.05.033] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2016] [Revised: 04/30/2017] [Accepted: 05/06/2017] [Indexed: 11/22/2022] Open
|
137
|
Donndorf P, Abubaker S, Vollmar B, Rimmbach C, Steinhoff G, Kaminski A. Therapeutic progenitor cell application for tissue regeneration: Analyzing the impact of toll-like receptor signaling on c-kit + cell migration following ischemia-reperfusion injury in vivo. Microvasc Res 2017; 112:87-92. [DOI: 10.1016/j.mvr.2017.03.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Revised: 02/23/2017] [Accepted: 03/27/2017] [Indexed: 02/08/2023]
|
138
|
Lee SM, Hutchinson M, Saint DA. The role of Toll-like receptor 4 (TLR4) in cardiac ischaemic-reperfusion injury, cardioprotection and preconditioning. Clin Exp Pharmacol Physiol 2017; 43:864-71. [PMID: 27249055 DOI: 10.1111/1440-1681.12602] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2016] [Revised: 05/26/2016] [Accepted: 05/30/2016] [Indexed: 01/04/2023]
Abstract
Cardiac ischaemic-reperfusion injury (IRI) remains the primary cause of mortality throughout the developed world. Molecular mechanisms underlying IRI are complex and are often interlinked with each other driving a synergistic response. Toll-like receptor 4 (TLR4), an immunosurveillance receptor, is known to enhance tissue injury during IRI by enhancing the inflammatory response. The release of endogenous components during IRI bind onto TLR4 leading to the activation of multiple signalling kinases. Once this event occurs these proteins are defined as danger associated molecular patterns molecules (DAMPs) or alarmins. Examples include heat shock proteins, high mobility group box one (HMGB1) and extracellular matrix proteins, all of which are involved in IRI. However, literature in the last two decades suggests that transient stimulation of TLR4 may suppress IRI and thus improve cardiac recovery. Furthermore, it remains to be seen what role TLR4 plays during ischaemic-preconditioning where acute bouts of ischaemia, preceding a harmful bout of ischaemic-reperfusion, is cardioprotective. The other question which also needs to be considered is that if transient TLR4 signalling drives a preconditioning response then what are the ligands which drive this? Hence the second part of this review explores the possible TLR4 ligands which may promote cardioprotection against IRI.
Collapse
Affiliation(s)
- Sam Man Lee
- School of Medicine, University of Adelaide, Adelaide, SA, Australia
| | - Mark Hutchinson
- School of Medicine, University of Adelaide, Adelaide, SA, Australia.,Centre for Nanoscale Biophotonics, University of Adelaide, Adelaide, SA, Australia
| | - David A Saint
- School of Medicine, University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
139
|
Katare PB, Bagul PK, Dinda AK, Banerjee SK. Toll-Like Receptor 4 Inhibition Improves Oxidative Stress and Mitochondrial Health in Isoproterenol-Induced Cardiac Hypertrophy in Rats. Front Immunol 2017; 8:719. [PMID: 28690610 PMCID: PMC5479928 DOI: 10.3389/fimmu.2017.00719] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 06/06/2017] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Inflammation remains a crucial factor for progression of cardiac diseases and cardiac hypertrophy remains an important cause of cardiac failure over all age groups. As a key regulator of inflammation, toll-like receptor 4 (TLR4) plays an important role in pathogenesis of cardiac diseases. Being an important regulator of innate immunity, the precise pathway of TLR4-mediated cardiac complications is yet to be established. Therefore, the primary objective of the present study was to find the role of TLR4 in cardiac hypertrophy and the molecular mechanism thereof. METHODS Cardiac hypertrophy was induced with administration of isoproterenol (5 mg/kg/day, sc). TLR4 receptor inhibitor RS-LPS (lipopolysaccharide from the photosynthetic bacterium Rhodobacter sphaeroides; 5 μg/day) and agonist lipopolysaccharide (LPS) (from Escherichia coli; 3.12 μg/day) were administered through osmotic pump along with isoproterenol. Cardiac hypertrophy as well as oxidative stress and mitochondrial parameters were evaluated. RESULTS Cardiac hypertrophy was confirmed with increased heart weight/body weight ratio as well as assessment of hypertrophic markers in heart. There was a marked increase in the TLR4 expression and oxidative stress along with mitochondrial dysfunction in ISO group. TLR4 inhibition significantly decreased heart weight/body weight ratio and ANP, collagen, and β-MHC expression and restored the disturbed cellular antioxidant flux. The mitochondrial perturbations that were observed in hypertrophy heart was normalized after administration of TLR4 inhibitor but not with the agonist. TLR4 agonism further exaggerated the oxidative stress in heart and hence accelerated the disease development and progression. CONCLUSION Our data show that increased TLR4 ligand pool in cardiac hypertrophy may exaggerate the disease progression. However, inhibition of TLR4 attenuated cardiac hypertrophy through reduced cardiac redox imbalance and mitochondrial dysfunction.
Collapse
Affiliation(s)
- Parmeshwar B Katare
- Drug Discovery Research Center (DDRC), Translational Health Science and Technology Institute (THSTI), Faridabad, India
| | - Pankaj K Bagul
- Drug Discovery Research Center (DDRC), Translational Health Science and Technology Institute (THSTI), Faridabad, India
| | - Amit K Dinda
- Department of Pathology, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Sanjay K Banerjee
- Drug Discovery Research Center (DDRC), Translational Health Science and Technology Institute (THSTI), Faridabad, India
| |
Collapse
|
140
|
Toll-Like Receptor 4 Deficiency Improves Short-term Renal Function but not Long-term Graft Survival in a Fully MHC-Mismatched Murine Model of Renal Allograft Transplantation. Transplantation 2017; 100:1219-27. [PMID: 27077596 DOI: 10.1097/tp.0000000000001168] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND We have previously demonstrated that absence of myeloid differentiation primary response gene 88 (MyD88) induced donor-specific kidney allograft tolerance. The upstream pathways of MyD88 that mediate this process, however, remain unclear. Toll-like receptor 4 (TLR4) is an innate immune receptor that is dependent upon MyD88 for activity of its dominant signaling pathway. Here, we investigated the role of TLR4 in kidney allograft rejection using a fully major histocompatibility complex-mismatched, life-sustaining, murine model of renal allograft rejection. METHODS Donor (BALB/c) and recipient (C57BL/6) mice either both deficient or sufficient for TLR4 underwent heterotopic renal allograft transplantation, with an additional group of mice receiving renal isografts as controls. Survival was assessed up to 100 days posttransplantation. Animals were also sacrificed 14 days posttransplantation for assessment of the acute allograft rejection response. RESULTS Both wild-type (WT) and TLR4 allografts showed inferior survival compared to isografts, with no difference in survival between the allograft groups. Serum creatinine was lower in TLR4 allografts at day 14 posttransplantation compared with WT allografts, but this was not sustained by day 100. At day 14 posttransplant, increased CD11c dendritic cell accumulation, expression of IL-2 and indoleamine 2,3-dioxygenase were evident in TLR4 compared with WT allografts, whereas expression of inducible nitric oxide synthase was decreased. CONCLUSIONS Acute kidney allograft rejection was modestly attenuated in TLR4 mice; however, long-term allograft survival and function were not affected in our model. Protection against acute rejection may involve increased accumulation of CD11c cells and indoleamine 2,3-dioxygenase expression.
Collapse
|
141
|
Peng J, Zheng H, Wang X, Cheng Z. Upregulation of TLR4 via PKC activation contributes to impaired wound healing in high-glucose-treated kidney proximal tubular cells. PLoS One 2017; 12:e0178147. [PMID: 28542370 PMCID: PMC5443579 DOI: 10.1371/journal.pone.0178147] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 05/09/2017] [Indexed: 12/15/2022] Open
Abstract
Acute kidney injury (AKI) leads to a worse prognosis in diabetic patients compared with prognoses in non-diabetic patients, but whether and how diabetes affects kidney repair after AKI remains unknown. Here, we used scratch-wound healing and transwell migration models to examine whether and how wound healing is affected by high glucose levels in cultured kidney proximal tubular cells (RPTC). The results show that scratch-wound healing and transwell migration were significantly slower in high-glucose-treated kidney tubular cells (30 mM glucose) than in low-glucose-treated cells (5.5 mM). Toll-like receptor 4 (TLR4), MyD88, phospho-protein kinase C (PKC), phospho-p38 MAPK and monocyte chemoattractant protein-1 (MCP-1) mRNA levels were upregulated after high glucose treatments. Staurosporine, a selective PKC inhibitor, inhibited TLR4, MyD88 and p-p38 upregulation in the high-glucose-treated cells, indicating the involvement of PKC in high-glucose-induced TLR4 upregulation. The pharmacological inhibition of TLR4 or shRNA-mediated TLR4 knockdown improved wound healing and transwell migration in high-glucose-treated RPTC. In contrast, the overexpression of TLR4 in low-glucose-treated RPTC suppressed wound healing, mimicking the effects of high glucose levels. These results suggest that the upregulation of TLR4 expression via PKC activation contributes to defective wound healing in high-glucose-treated kidney tubular cells.
Collapse
Affiliation(s)
- Jianping Peng
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Hang Zheng
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
- * E-mail:
| | - Xia Wang
- Department of Internal Medicine, Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Zhixiang Cheng
- Department of General Surgery, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| |
Collapse
|
142
|
Meng X, Li J, Yu M, Yang J, Zheng M, Zhang J, Sun C, Liang H, Liu L. Transplantation of mesenchymal stem cells overexpressing IL10 attenuates cardiac impairments in rats with myocardial infarction. J Cell Physiol 2017; 233:587-595. [DOI: 10.1002/jcp.25919] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2017] [Accepted: 03/17/2017] [Indexed: 01/14/2023]
Affiliation(s)
- Xin Meng
- Department of UltrasonographyXijing HospitalFourth Military Medical UniversityXi'anShaanxiChina
| | - Jianping Li
- Department of Radiation OncologyXijing HospitalFourth Military Medical UniversityXi'anShaanxiChina
| | - Ming Yu
- Department of UltrasonographyXijing HospitalFourth Military Medical UniversityXi'anShaanxiChina
| | - Jian Yang
- Department of Cardiovascular SurgeryXijing HospitalFourth Military Medical UniversityXi'anShaanxiChina
| | - Minjuan Zheng
- Department of UltrasonographyXijing HospitalFourth Military Medical UniversityXi'anShaanxiChina
| | - Jinzhou Zhang
- Department of Cardiovascular SurgeryXijing HospitalFourth Military Medical UniversityXi'anShaanxiChina
| | - Chao Sun
- Department of UltrasonographyXijing HospitalFourth Military Medical UniversityXi'anShaanxiChina
| | - Hongliang Liang
- Department of Cardiovascular SurgeryXijing HospitalFourth Military Medical UniversityXi'anShaanxiChina
| | - Liwen Liu
- Department of UltrasonographyXijing HospitalFourth Military Medical UniversityXi'anShaanxiChina
| |
Collapse
|
143
|
Prabhu SD, Frangogiannis NG. The Biological Basis for Cardiac Repair After Myocardial Infarction: From Inflammation to Fibrosis. Circ Res 2017; 119:91-112. [PMID: 27340270 DOI: 10.1161/circresaha.116.303577] [Citation(s) in RCA: 1393] [Impact Index Per Article: 199.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2016] [Accepted: 04/15/2016] [Indexed: 12/14/2022]
Abstract
In adult mammals, massive sudden loss of cardiomyocytes after infarction overwhelms the limited regenerative capacity of the myocardium, resulting in the formation of a collagen-based scar. Necrotic cells release danger signals, activating innate immune pathways and triggering an intense inflammatory response. Stimulation of toll-like receptor signaling and complement activation induces expression of proinflammatory cytokines (such as interleukin-1 and tumor necrosis factor-α) and chemokines (such as monocyte chemoattractant protein-1/ chemokine (C-C motif) ligand 2 [CCL2]). Inflammatory signals promote adhesive interactions between leukocytes and endothelial cells, leading to extravasation of neutrophils and monocytes. As infiltrating leukocytes clear the infarct from dead cells, mediators repressing inflammation are released, and anti-inflammatory mononuclear cell subsets predominate. Suppression of the inflammatory response is associated with activation of reparative cells. Fibroblasts proliferate, undergo myofibroblast transdifferentiation, and deposit large amounts of extracellular matrix proteins maintaining the structural integrity of the infarcted ventricle. The renin-angiotensin-aldosterone system and members of the transforming growth factor-β family play an important role in activation of infarct myofibroblasts. Maturation of the scar follows, as a network of cross-linked collagenous matrix is formed and granulation tissue cells become apoptotic. This review discusses the cellular effectors and molecular signals regulating the inflammatory and reparative response after myocardial infarction. Dysregulation of immune pathways, impaired suppression of postinfarction inflammation, perturbed spatial containment of the inflammatory response, and overactive fibrosis may cause adverse remodeling in patients with infarction contributing to the pathogenesis of heart failure. Therapeutic modulation of the inflammatory and reparative response may hold promise for the prevention of postinfarction heart failure.
Collapse
Affiliation(s)
- Sumanth D Prabhu
- From the Division of Cardiovascular Disease, University of Alabama at Birmingham, and Medical Service, Birmingham VAMC (S.D.P.); and Department of Medicine, The Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY (N.G.F.)
| | - Nikolaos G Frangogiannis
- From the Division of Cardiovascular Disease, University of Alabama at Birmingham, and Medical Service, Birmingham VAMC (S.D.P.); and Department of Medicine, The Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY (N.G.F.).
| |
Collapse
|
144
|
Boag SE, Andreano E, Spyridopoulos I. Lymphocyte Communication in Myocardial Ischemia/Reperfusion Injury. Antioxid Redox Signal 2017; 26:660-675. [PMID: 28006953 DOI: 10.1089/ars.2016.6940] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
SIGNIFICANCE Myocardial ischemia/reperfusion (I/R) is an important complication of reperfusion therapy for myocardial infarction (MI). It is a complex process involving metabolic and immunological factors. To date, no effective treatment has been identified. Recent Advances: Previous research has focused on the role of innate immune cells in I/R injury. In recent years, increasing evidence has accumulated for an important role for adaptive immune cells, particularly T lymphocytes. Data from ST elevation MI patients have identified prognostic significance for lymphocyte counts, particularly postreperfusion lymphopenia. Dynamic changes in circulating CD4+ T cell subsets occurring early after reperfusion are associated with development of I/R injury in the form of microvascular obstruction. Transcoronary gradients in cell counts suggest sequestration of these cells into the reperfused myocardium. These findings support existing data from mouse models indicating a role for CD4+ T cells in I/R injury. It is clear, however, the effects of lymphocytes in the ischemic myocardium are time and subset specific, with some having protective effects, while others are pathogenic. CRITICAL ISSUES An understanding of the cellular events that lead to accumulation of lymphocytes in the myocardium, and their actions once there, is key to manipulating this process. Chemokines produced in response to ischemia and cellular injury have an important role, while lymphocyte-derived cytokines are critical in the balance between inflammation and healing. FUTURE DIRECTIONS Further research into the involvement of lymphocytes in myocardial I/R injury may allow development of targeted therapies, opening a new avenue of considerable therapeutic potential. Antioxid. Redox Signal. 26, 660-675.
Collapse
Affiliation(s)
- Stephen E Boag
- 1 Institute of Genetic Medicine, Newcastle University , Newcastle upon Tyne, United Kingdom .,2 Regional Department of Clinical Immunology, Royal Victoria Infirmary, Newcastle upon Tyne, United Kingdom
| | - Emanuele Andreano
- 1 Institute of Genetic Medicine, Newcastle University , Newcastle upon Tyne, United Kingdom
| | - Ioakim Spyridopoulos
- 1 Institute of Genetic Medicine, Newcastle University , Newcastle upon Tyne, United Kingdom
| |
Collapse
|
145
|
Julier Z, Park AJ, Briquez PS, Martino MM. Promoting tissue regeneration by modulating the immune system. Acta Biomater 2017; 53:13-28. [PMID: 28119112 DOI: 10.1016/j.actbio.2017.01.056] [Citation(s) in RCA: 464] [Impact Index Per Article: 66.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 01/03/2017] [Accepted: 01/20/2017] [Indexed: 02/07/2023]
Abstract
The immune system plays a central role in tissue repair and regeneration. Indeed, the immune response to tissue injury is crucial in determining the speed and the outcome of the healing process, including the extent of scarring and the restoration of organ function. Therefore, controlling immune components via biomaterials and drug delivery systems is becoming an attractive approach in regenerative medicine, since therapies based on stem cells and growth factors have not yet proven to be broadly effective in the clinic. To integrate the immune system into regenerative strategies, one of the first challenges is to understand the precise functions of the different immune components during the tissue healing process. While remarkable progress has been made, the immune mechanisms involved are still elusive, and there is indication for both negative and positive roles depending on the tissue type or organ and life stage. It is well recognized that the innate immune response comprising danger signals, neutrophils and macrophages modulates tissue healing. In addition, it is becoming evident that the adaptive immune response, in particular T cell subset activities, plays a critical role. In this review, we first present an overview of the basic immune mechanisms involved in tissue repair and regeneration. Then, we highlight various approaches based on biomaterials and drug delivery systems that aim at modulating these mechanisms to limit fibrosis and promote regeneration. We propose that the next generation of regenerative therapies may evolve from typical biomaterial-, stem cell-, or growth factor-centric approaches to an immune-centric approach. STATEMENT OF SIGNIFICANCE Most regenerative strategies have not yet proven to be safe or reasonably efficient in the clinic. In addition to stem cells and growth factors, the immune system plays a crucial role in the tissue healing process. Here, we propose that controlling the immune-mediated mechanisms of tissue repair and regeneration may support existing regenerative strategies or could be an alternative to using stem cells and growth factors. The first part of this review we highlight key immune mechanisms involved in the tissue healing process and marks them as potential target for designing regenerative strategies. In the second part, we discuss various approaches using biomaterials and drug delivery systems that aim at modulating the components of the immune system to promote tissue regeneration.
Collapse
Affiliation(s)
- Ziad Julier
- European Molecular Biology Laboratory Australia, Australian Regenerative Medicine Institute, Monash University, Victoria 3800, Australia
| | - Anthony J Park
- European Molecular Biology Laboratory Australia, Australian Regenerative Medicine Institute, Monash University, Victoria 3800, Australia
| | - Priscilla S Briquez
- Institute for Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Mikaël M Martino
- European Molecular Biology Laboratory Australia, Australian Regenerative Medicine Institute, Monash University, Victoria 3800, Australia.
| |
Collapse
|
146
|
On the value of therapeutic interventions targeting the complement system in acute myocardial infarction. Transl Res 2017; 182:103-122. [PMID: 27810412 DOI: 10.1016/j.trsl.2016.10.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 10/05/2016] [Accepted: 10/06/2016] [Indexed: 01/12/2023]
Abstract
The complement system plays an important role in the inflammatory response subsequent to acute myocardial infarction (AMI). The aim of this study is to create a systematic overview of studies that have investigated therapeutic administration of complement inhibitors in both AMI animal models and human clinical trials. To enable extrapolation of observations from included animal studies toward post-AMI clinical trials, ex vivo studies on isolated hearts and proof-of-principle studies on inhibitor administration before experimental AMI induction were excluded. Positive therapeutic effects in AMI animal models have been described for cobra venom factor, soluble complement receptor 1, C1-esterase inhibitor (C1-inh), FUT-175, C1s-inhibitor, anti-C5, ADC-1004, clusterin, and glycosaminoglycans. Two types of complement inhibitors have been tested in clinical trials, being C1-inh and anti-C5. Pexelizumab (anti-C5) did not result in reproducible beneficial effects for AMI patients. Beneficial effects were reported in AMI patients for C1-inhibitor, albeit in small patient groups. In general, despite the absence of consistent positive effects in clinical trials thus far, the complement system remains a potentially interesting target for therapy in AMI patients. Based on the study designs of previous animal studies and clinical trials, we discuss several issues which require attention in the design of future studies: adjustment of clinical trial design to precise mechanism of action of administered inhibitor, optimizing the duration of therapy, and optimization of time point(s) on which therapeutic effects will be evaluated.
Collapse
|
147
|
Abstract
Excessive feeding is associated with an increase in the incidence of chronic metabolic diseases, such as obesity, insulin resistance, and type 2 diabetes. Metabolic disturbance induces chronic low-grade inflammation in metabolically-important organs, such as the liver and adipose tissue. Many of the inflammatory signalling pathways are directly triggered by nutrients. The pro-inflammatory mediators in adipocytes and macrophages infiltrating adipose tissue promote both local and systemic pro-inflammatory status. Metabolic cardiomyopathy is a chronic metabolic disease characterized by structural and functional alterations and interstitial fibrosis without coronary artery disease or hypertension. In the early stage of metabolic cardiomyopathy, metabolic disturbance is not accompanied by substantial changes in myocardial structure and cardiac function. However, metabolic disturbance induces subcellular low-grade inflammation in the heart, and in turn, subcellular component abnormalities, such as oxidative stress, mitochondrial dysfunction, endoplasmic reticulum stress, and impaired calcium handling, leading to impaired myocardial relaxation. In the advanced stage, the vicious cycle of subcellular component abnormalities, inflammatory cell infiltration, and neurohumoral activation induces cardiomyocyte injury and death, and cardiac fibrosis, resulting in impairment of both diastolic and systolic functions. This review discusses some recent advances in understanding involvement of inflammation in metabolic cardiomyopathy.
Collapse
|
148
|
Zhang LN, Wang XH, Wu L, Huang L, Zhao CG, Peng QY, Ai YH. Diagnostic and Predictive Levels of Calcium-binding Protein A8 and Tumor Necrosis Factor Receptor-associated Factor 6 in Sepsis-associated Encephalopathy: A Prospective Observational Study. Chin Med J (Engl) 2017; 129:1674-81. [PMID: 27411454 PMCID: PMC4960956 DOI: 10.4103/0366-6999.185860] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Background: Despite its high prevalence, morbidity, and mortality, sepsis-associated encephalopathy (SAE) is still poorly understood. The aim of this prospective and observational study was to investigate the clinical significance of calcium-binding protein A8 (S100A8) in serum and tumor necrosis factor receptor-associated factor 6 (TRAF6) in peripheral blood mononuclear cells (PBMCs) in diagnosing SAE and predicting its prognosis. Methods: Data of septic patients were collected within 24 h after Intensive Care Unit admission from July 2014 to March 2015. Healthy medical personnel served as the control group. SAE was defined as cerebral dysfunction in the presence of sepsis that fulfilled the exclusion criteria. The biochemical indicators, Glasgow Coma Scale, Acute Physiology and Chronic Health Evaluation score II, TRAF6 in PBMC, serum S100A8, S100β, and neuron-specific enolase were evaluated in SAE patients afresh. TRAF6 and S100A8 were also measured in the control group. Results: Of the 57 enrolled patients, 29 were diagnosed with SAE. The S100A8 and TRAF6 concentrations in SAE patients were both significantly higher than that in no-encephalopathy (NE) patients, and higher in NE than that in controls (3.74 ± 3.13 vs. 1.08 ± 0.75 vs. 0.37 ± 0.14 ng/ml, P < 0.01; 3.18 ± 1.55 vs. 1.02 ± 0.63 vs. 0.47 ± 0.10, P < 0.01). S100A8 levels of 1.93 ng/ml were diagnostic of SAE with 92.90% specificity and 69.00% sensitivity in the receiver operating characteristic (ROC) curve, and the area under the curve was 0.86 (95% confidence interval [CI]: 0.76–0.95). TRAF6-relative levels of 1.44 were diagnostic of SAE with 85.70% specificity and 86.20% sensitivity, and the area under the curve was 0.94 (95% CI: 0.88–0.99). In addition, S100A8 levels of 2.41 ng/ml predicted 28-day mortality of SAE with 90.00% specificity and 73.70% sensitivity in the ROC curve, and the area under the curve was 0.88. TRAF6 relative levels of 2.94 predicted 28-day mortality of SAE with 80.00% specificity and 68.40% sensitivity, and the area under the curve was 0.77. Compared with TRAF6, the specificity of serum S100A8 in diagnosing SAE and predicting mortality was higher, although the sensitivity was low. In contrast, the TRAF6 had higher sensitivity for diagnosis. Conclusions: Peripheral blood levels of S100A8 and TRAF6 in SAE patients were elevated and might be related to the severity of SAE and predict the outcome of SAE. The efficacy and specificity of S100A8 for SAE diagnosis were superior, despite its weak sensitivity. S100A8 might be a better biomarker for diagnosis of SAE and predicting prognosis.
Collapse
Affiliation(s)
- Li-Na Zhang
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Xiao-Hong Wang
- Department of Anesthesiology and Critical Care Medicine, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan 410008, China
| | - Long Wu
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Li Huang
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Chun-Guang Zhao
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Qian-Yi Peng
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Yu-Hang Ai
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| |
Collapse
|
149
|
Dexmedetomidine preconditioning may attenuate myocardial ischemia/reperfusion injury by down-regulating the HMGB1-TLR4-MyD88-NF-кB signaling pathway. PLoS One 2017; 12:e0172006. [PMID: 28222157 PMCID: PMC5319750 DOI: 10.1371/journal.pone.0172006] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 01/30/2017] [Indexed: 12/21/2022] Open
Abstract
Aims To investigate whether dexmedetomidine (DEX) preconditioning could alleviate the inflammation caused by myocardial ischemia/reperfusion (I/R) injury by reducing HMGB1-TLR4-MyD88-NF-кB signaling. Methods Seventy rats were randomly assigned into five groups: sham group, myocardial I/R group (I/R), DEX+I/R group (DEX), DEX+yohimbine+I/R group (DEX/YOH), and yohimbine+I/R group (YOH). Animals were subjected to 30 min of ischemia induced by occluding the left anterior descending artery followed by 120 min of reperfusion. Myocardial infarct size and histological scores were evaluated. The levels of IL-6 and TNF-α in serum and myocardium were quantified by enzyme-linked immunosorbent assay, and expression of HMGB1, TLR4, MyD88, IκB and NF-κB in the myocardial I/R area were determined with Western blot and immunocytochemistry. Results Myocardial infarct sizes, histological scores, levels of circulating and myocardial IL-6 and TNF-α, the expression of HMGB1, TLR4, MyD88 and NF-κB, and the degradation of IκB were significantly increased in the I/R group compared with the sham group (P<0.01). DEX preconditioning significantly reduced the myocardial infarct size and histological scores (P<0.01 vs. I/R group). Similarly, the serum and myocardial levels of IL-6 and TNF-α, the expression of HMGB1, TLR4, MyD88 and NF-κB, and the degradation of IκB were significantly reduced in the DEX group (P<0.01 vs. I/R group). These effects were partly reversed by yohimbine, a selective α2-adrenergic receptor antagonist, while yohimbine alone had no significant effect on any of the above indicators. Conclusion DEX preconditioning reduces myocardial I/R injury in part by attenuating inflammation, which may be attributed to the downregulation of the HMGB1-TLR4-MyD88-NF-кB signaling pathway mediated by the α2-adrenergic receptor activation.
Collapse
|
150
|
Xiong X, Liu Y, Mei Y, Peng J, Wang Z, Kong B, Zhong P, Xiong L, Quan D, Li Q, Wang G, Huang H. Novel Protective Role of Myeloid Differentiation 1 in Pathological Cardiac Remodelling. Sci Rep 2017; 7:41857. [PMID: 28165494 PMCID: PMC5292962 DOI: 10.1038/srep41857] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 12/29/2016] [Indexed: 12/22/2022] Open
Abstract
Myeloid differentiation 1 (MD-1), a secreted protein interacting with radioprotective 105 (RP105), plays an important role in Toll-like receptor 4 (TLR4) signalling pathway. Previous studies showed that MD-1 may be restricted in the immune system. In this study, we demonstrated for the first time that MD-1 was highly expressed in both human and animal hearts. We also discovered that cardiac-specific overexpression of MD-1 significantly attenuated pressure overload-induced cardiac hypertrophy, fibrosis, and dysfunction, whereas loss of MD-1 had the opposite effects. Similar results were observed for in vitro angiotensin II-induced neonatal rat cardiomyocyte hypertrophy. The antihypertrophic effects of MD-1 under hypertrophic stimuli were associated with the blockage of MEK-ERK 1/2 and NF-κB signalling. Blocking MEK-ERK 1/2 signalling with a pharmacological inhibitor (U0126) greatly attenuated the detrimental effects observed in MD-1 knockout cardiomyocytes exposed to angiotensin II stimuli. Similar results were observed by blocking NF-κB signalling with a pharmacological inhibitor (BAY11–7082). Our data indicate that MD-1 inhibits cardiac hypertrophy and suppresses cardiac dysfunction during the remodelling process, which is dependent on its modulation of the MEK-ERK 1/2 and NF-κB signalling pathways. Thus, MD-1 might be a novel target for the treatment of pathological cardiac hypertrophy.
Collapse
Affiliation(s)
- Xiaojv Xiong
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, PR China.,Cardiovascular Research Institute of Wuhan University, Wuhan, Hubei Province, PR China.,Hubei Key Laboratory of Cardiology, Wuhan 430060, Hubei Province, PR China
| | - Yu Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, PR China.,Cardiovascular Research Institute of Wuhan University, Wuhan, Hubei Province, PR China.,Hubei Key Laboratory of Cardiology, Wuhan 430060, Hubei Province, PR China
| | - Yang Mei
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, PR China.,Cardiovascular Research Institute of Wuhan University, Wuhan, Hubei Province, PR China.,Hubei Key Laboratory of Cardiology, Wuhan 430060, Hubei Province, PR China
| | - Jianye Peng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, PR China.,Cardiovascular Research Institute of Wuhan University, Wuhan, Hubei Province, PR China.,Hubei Key Laboratory of Cardiology, Wuhan 430060, Hubei Province, PR China
| | - Zhiqiang Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, PR China.,Cardiovascular Research Institute of Wuhan University, Wuhan, Hubei Province, PR China.,Hubei Key Laboratory of Cardiology, Wuhan 430060, Hubei Province, PR China
| | - Bin Kong
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, PR China.,Cardiovascular Research Institute of Wuhan University, Wuhan, Hubei Province, PR China.,Hubei Key Laboratory of Cardiology, Wuhan 430060, Hubei Province, PR China
| | - Peng Zhong
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, PR China.,Cardiovascular Research Institute of Wuhan University, Wuhan, Hubei Province, PR China.,Hubei Key Laboratory of Cardiology, Wuhan 430060, Hubei Province, PR China
| | - Liang Xiong
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, PR China.,Cardiovascular Research Institute of Wuhan University, Wuhan, Hubei Province, PR China.,Hubei Key Laboratory of Cardiology, Wuhan 430060, Hubei Province, PR China
| | - Dajun Quan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, PR China.,Cardiovascular Research Institute of Wuhan University, Wuhan, Hubei Province, PR China.,Hubei Key Laboratory of Cardiology, Wuhan 430060, Hubei Province, PR China
| | - Qi Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, PR China.,Cardiovascular Research Institute of Wuhan University, Wuhan, Hubei Province, PR China.,Hubei Key Laboratory of Cardiology, Wuhan 430060, Hubei Province, PR China
| | - Guangji Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, PR China.,Cardiovascular Research Institute of Wuhan University, Wuhan, Hubei Province, PR China.,Hubei Key Laboratory of Cardiology, Wuhan 430060, Hubei Province, PR China
| | - He Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, PR China.,Cardiovascular Research Institute of Wuhan University, Wuhan, Hubei Province, PR China.,Hubei Key Laboratory of Cardiology, Wuhan 430060, Hubei Province, PR China
| |
Collapse
|