51
|
Jouffroy R, Hergault H, Antero J, Vieillard Baron A, Mansencal N. Relationship between echocardiographic characteristics and cardiac biomarkers during long-distance trail running. Front Cardiovasc Med 2022; 9:954032. [PMID: 36051277 PMCID: PMC9424639 DOI: 10.3389/fcvm.2022.954032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 07/18/2022] [Indexed: 11/15/2022] Open
Abstract
Background Even if the beneficial cardiovascular effects of moderate exercise are recognized, effects of prolonged and intense exercise are still debated. This study aims to detect cardiovascular changes associated with long endurance running by assessing the relationship between echocardiographic parameters and cardiac biomarkers during long-distance trail running. Methods We performed a prospective observational study that included 20 participants who were all amateur runners (median age of 41 years old, still alive after a 7-year clinical follow-up) from 80-km trail running. All the participants underwent an echocardiographic examination and venous blood sampling before the race, at the intermediate refreshment checkpoints of the race (21st and 53rd km), and within 10 min after arrival. Results Mitral E/A velocity ratio and mitral TDI e’ wave were significantly decreased at the 21st km to arrival (p < 0.05). Mitral S wave and global longitudinal strain (GLS) were significantly decreased from the 53rd km to arrival (p < 0.05 for 53rd and 80th km). As compared to baseline, T-troponin and NT-proBNP were significantly increased at the 21st km in all the participants, but T-troponin values were systematically increased above the significative threshold. Diastolic echocardiographic abnormalities were mainly observed among participants with highest NT-proBNP (> 77 ng.l–1) values at the 21st km. As compared to baseline, mitral e’ wave was significantly decreased (–35%) in participants with highest values of NT-proBNP. Similarly, GLS was also depressed among participants with highest troponin values at the 53rd km (p = 0.01 for 53rd km and p = 0.04 for arrival). Conclusion During the long-distance trail running, the early LV decrease in diastolic echocardiographic parameters is associated with increase in NT pro-BNP blood levels, and the decrease in LV systolic echocardiographic parameters later is associated with increase in T-troponin blood levels.
Collapse
Affiliation(s)
- Romain Jouffroy
- Intensive Care Unit, Ambroise Paré Hospital, Assistance Publique—Hôpitaux de Paris (AP-HP), Université de Versailles-Saint Quentin (UVSQ), Boulogne-Billancourt, France
- IRMES - Institute for Research in Medicine and Epidemiology of Sport, INSEP, Paris, France
- INSERM U-1018, CESP, Clinical Epidemiology, Université de Versailles-Saint Quentin (UVSQ), Villejuif, France
- *Correspondence: Romain Jouffroy,
| | - Hélène Hergault
- INSERM U-1018, CESP, Clinical Epidemiology, Université de Versailles-Saint Quentin (UVSQ), Villejuif, France
- Department of Cardiology, Ambroise Paré Hospital, AP-HP, Centre de Référence des Cardiomyopathies et des Troubles du Rythme Cardiaque Héréditaires ou Rares, Université de Versailles-Saint Quentin (UVSQ), Boulogne-Billancourt, France
| | - Juliana Antero
- IRMES - Institute for Research in Medicine and Epidemiology of Sport, INSEP, Paris, France
| | - Antoine Vieillard Baron
- Intensive Care Unit, Ambroise Paré Hospital, Assistance Publique—Hôpitaux de Paris (AP-HP), Université de Versailles-Saint Quentin (UVSQ), Boulogne-Billancourt, France
- INSERM U-1018, CESP, Clinical Epidemiology, Université de Versailles-Saint Quentin (UVSQ), Villejuif, France
| | - Nicolas Mansencal
- INSERM U-1018, CESP, Clinical Epidemiology, Université de Versailles-Saint Quentin (UVSQ), Villejuif, France
- Department of Cardiology, Ambroise Paré Hospital, AP-HP, Centre de Référence des Cardiomyopathies et des Troubles du Rythme Cardiaque Héréditaires ou Rares, Université de Versailles-Saint Quentin (UVSQ), Boulogne-Billancourt, France
| |
Collapse
|
52
|
Zou HX, Qiu BQ, Zhang ZY, Hu T, Wan L, Liu JC, Huang H, Lai SQ. Dysregulated autophagy-related genes in septic cardiomyopathy: Comprehensive bioinformatics analysis based on the human transcriptomes and experimental validation. Front Cardiovasc Med 2022; 9:923066. [PMID: 35983185 PMCID: PMC9378994 DOI: 10.3389/fcvm.2022.923066] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 07/14/2022] [Indexed: 11/13/2022] Open
Abstract
Septic cardiomyopathy (SCM) is severe organ dysfunction caused by sepsis that is associated with poor prognosis, and its pathobiological mechanisms remain unclear. Autophagy is a biological process that has recently been focused on SCM, yet the current understanding of the role of dysregulated autophagy in the pathogenesis of SCM remains limited and uncertain. Exploring the molecular mechanisms of disease based on the transcriptomes of human pathological samples may bring the closest insights. In this study, we analyzed the differential expression of autophagy-related genes in SCM based on the transcriptomes of human septic hearts, and further explored their potential crosstalk and functional pathways. Key functional module and hub genes were identified by constructing a protein–protein interaction network. Eight key genes (CCL2, MYC, TP53, SOD2, HIF1A, CTNNB1, CAT, and ADIPOQ) that regulate autophagy in SCM were identified after validation in a lipopolysaccharide (LPS)-induced H9c2 cardiomyoblast injury model, as well as the autophagic characteristic features. Furthermore, we found that key genes were associated with abnormal immune infiltration in septic hearts and have the potential to serve as biomarkers. Finally, we predicted drugs that may play a protective role in SCM by regulating autophagy based on our results. Our study provides evidence and new insights into the role of autophagy in SCM based on human septic heart transcriptomes, which would be of great benefit to reveal the molecular pathological mechanisms and explore the diagnostic and therapeutic targets for SCM.
Collapse
Affiliation(s)
- Hua-Xi Zou
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
- Institute of Cardiovascular Diseases, Jiangxi Academy of Clinical Medical Sciences, The First Affiliated Hospital of Nanchang University, Nanchang, China
- Department of Cardiovascular Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Bai-Quan Qiu
- Institute of Cardiovascular Diseases, Jiangxi Academy of Clinical Medical Sciences, The First Affiliated Hospital of Nanchang University, Nanchang, China
- Department of Cardiovascular Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Ze-Yu Zhang
- Institute of Nanchang University Trauma Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Tie Hu
- Institute of Cardiovascular Diseases, Jiangxi Academy of Clinical Medical Sciences, The First Affiliated Hospital of Nanchang University, Nanchang, China
- Department of Cardiovascular Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Li Wan
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
- Institute of Cardiovascular Diseases, Jiangxi Academy of Clinical Medical Sciences, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Ji-Chun Liu
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
- Institute of Cardiovascular Diseases, Jiangxi Academy of Clinical Medical Sciences, The First Affiliated Hospital of Nanchang University, Nanchang, China
- Department of Cardiovascular Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Huang Huang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
- Institute of Cardiovascular Diseases, Jiangxi Academy of Clinical Medical Sciences, The First Affiliated Hospital of Nanchang University, Nanchang, China
- Huang Huang,
| | - Song-Qing Lai
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
- Institute of Cardiovascular Diseases, Jiangxi Academy of Clinical Medical Sciences, The First Affiliated Hospital of Nanchang University, Nanchang, China
- *Correspondence: Song-Qing Lai,
| |
Collapse
|
53
|
Mir T, Uddin M, Qureshi WT, Abohashem S, Alqalyoobi S, Sheikh M, Soubani A, Saydain G, Albertson TE. ST-Elevation Myocardial Infarction Among Septic Shock and Coronary Interventions: A National Emergency Database Study. J Intensive Care Med 2022; 37:1094-1100. [PMID: 34812084 DOI: 10.1177/08850666211061731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
OBJECTIVE To study coronary interventions and mortality among patients with ST-elevated myocardial infarction (STEMI) who were admitted with septic shock. METHODS Data from the national emergency department sample (NEDS) that constitutes 20% sample of hospital-owned emergency departments in the United States was analyzed for the septic shock related visits from 2016 to 2018. Septic shock was defined by the ICD codes. RESULTS Out of 1 375 507 adult septic shock patients, 521 300 had a primary diagnosis of septic shock (mean age 67.41±15.67 years, 51.1% females) in the national emergency database for the years 2016 to 2018. Of these patients, 2768 (0.53%) had STEMI recorded during the hospitalization. Mortality rates for STEMI patients were higher than patients without STEMI (52.3% vs 23.5%). Mortality rates improved with PCI among STEMI patients (43.8% vs 56.2%). Coronary angiography was performed among 16% of patients of which percutaneous coronary intervention (PCI) rates were 7.7% among patients with STEMI septic shock. PCI numerically improved mortality, however, had no significant difference than patients without PCI on multivariate logistic regression and univariate logistic regression post coarsened exact matching of baseline characteristics among STEMI patients. Among the predictors, STEMI was a significant predictor of mortality in septic shock patients (OR 2.87, 95% CI 2.37-3.49; P<.001). Age, peripheral vascular disease, were predominant predictors of mortality in STEMI with septic shock subgroup (P <.001). Pneumonia was the predominant underlying infection among STEMI (36.4%) and without STEMI group (29.5%). CONCLUSION STEMI complicating septic shock worsens mortality. PCI and coronary angiography numerically improved mortality, however, had no significant difference from patients without PCI. More research will be needed to improve mortality in such a critically ill subgroup of patients.
Collapse
Affiliation(s)
- Tanveer Mir
- 2954Wayne State University, School of Medicine, MI, USA
| | | | - Waqas T Qureshi
- 12262University of Massachusetts School of Medicine, Worcester, MA, USA
| | - Shady Abohashem
- 273161Harvard Medical School Massachusetts General Hospital, Boston, USA
| | | | | | - Ayman Soubani
- 2954Wayne State University, School of Medicine, MI, USA
| | | | | |
Collapse
|
54
|
Xu H, Ye W, Shi B. LncRNA MALAT1 Regulates USP22 Expression Through EZH2-Mediated H3K27me3 Modification to Accentuate Sepsis-Induced Myocardial Dysfunction. Cardiovasc Toxicol 2022; 22:813-830. [PMID: 35726125 DOI: 10.1007/s12012-022-09758-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 06/03/2022] [Indexed: 11/29/2022]
Abstract
Metastasis-associated lung adenocarcinoma transcript 1 (MALAT1), a long non-coding RNA (lncRNA), has been confirmed to recruit enhancer of zeste 2 polycomb repressive complex 2 subunit (EZH2) to regulate cardiomyocyte apoptosis in diabetic cardiomyopathy. However, whether the similar regulatory axis exists in sepsis-induced myocardial dysfunction (SIMD) has not been clearly established. The current study sought to define the mechanism governing MALAT1-mediated EZH2 in SIMD. MALAT1 was significantly upregulated in lipopolysaccharide-induced cardiomyocytes. Depletion of MALAT1 by caudal vein injection of small interfering RNA targeting MALAT1 alleviated myocardial injury in SIMD rats, restored cardiac function, reduced oxidative stress production and fibrosis, and inhibited inflammatory factors and apoptosis in myocardial tissues. Moreover, MALAT1 bound to EZH2 and promoted EZH2 activity in the nucleus of cardiomyocytes. EZH2 repressed ubiquitin-specific peptidase 22 (USP22) expression through H3K27me3 modification. EZH2 elevation aggravated the cardiac injury in SIMD rats, while USP22 upregulation inhibited the effect of EZH2, which reduced the cardiac injury in SIMD rats. Taken together, MALAT1 decreased USP22 expression by interacting with EZH2, thereby worsening SIMD, highlighting an attractive therapeutic strategy for SIMD.
Collapse
Affiliation(s)
- Hong Xu
- Department of Cardiovascular Division, Shandong Provincial Third Hospital, Jinan, 250031, Shandong, People's Republic of China
| | - Wei Ye
- Department of Respiratory Medicine, Shandong Provincial Third Hospital, Jinan, 250031, Shandong, People's Republic of China
| | - Baochang Shi
- Department of Hepatobiliary Surgery, Shandong Provincial Third Hospital, No. 12, WuYingShan Middle Road, Tianqiao District, Jinan, 250031, Shandong, People's Republic of China.
| |
Collapse
|
55
|
Teng Y, Li N, Wang Y, Sun S, Hou J, Chen Y, Pan H. NRF2 Inhibits Cardiomyocyte Pyroptosis Via Regulating CTRP1 in Sepsis-Induced Myocardial Injury. Shock 2022; 57:590-599. [PMID: 34907120 DOI: 10.1097/shk.0000000000001901] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
ABSTRACT C1q/tumor necrosis factor-related protein 1 (CTRP1) has been demonstrated as a crucial regulator in myocardial injury (MI). The present study aims to evaluate the mechanism of CTRP1 in sepsis-induced MI. The septic mouse model was established via cecal ligation and puncture and the in vitro cell model was established via lipopolysaccharide treatment. The mouse survival rate within 96 h was recorded. Morphologic changes of cardiomyocytes were observed and cell viability and cardiac functions were detected. CTRP1 and nuclear factor erythroid 2-related factor (Nrf2) expressions, creatine troponin-T, and creatine phosphokinase isoenzyme levels, and expressions of pyroptotic markers were determined. The binding relationship between Nrf2 and the CTRP1 promotor was predicted and verified. Rescue experiments were designed to confirm the role of CTRP1. CTRP1 was poorly expressed in septic mice. CTRP1 overexpression inhibited cardiomyocyte pyroptosis and improved cardiac functions, MI, and survival rate in septic mice. Nrf2was decreased in cecal ligation and puncture -treated mice. Nrf2 overexpression promoted CTRP1 expression via binding to the CTRP1 promotor and suppressed cardiomyocyte pyroptosis. CTRP1 downregulation abolished the inhibitory effect of Nrf2 overexpression on cardiomyocyte pyroptosis. Overall, Nrf2 promoted CTRP1 expression via binding to the CTRP1 promotor to inhibit cardiomyocyte pyroptosis, thereby alleviating MI in septic mice.
Collapse
Affiliation(s)
- Yan Teng
- Department of Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an City, Shaanxi Province, PR China
| | - Ningjun Li
- Department of Intensive Care Unit, The Fifth Affiliated Hospital of SUN YAT-SEN University, Zhuhai City, Guangdong Province, PR China
| | - Yi Wang
- Department of Intensive Care Unit, The Fifth Affiliated Hospital of SUN YAT-SEN University, Zhuhai City, Guangdong Province, PR China
| | - Shuling Sun
- Department of Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an City, Shaanxi Province, PR China
| | - Junxia Hou
- Department of Critical Care Medicine, Chang'an District Hospital of the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an City, Shaanxi Province, PR China
| | - Yahui Chen
- Department of Critical Care Medicine, Chang'an District Hospital of the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an City, Shaanxi Province, PR China
| | - Haiyan Pan
- Department of Intensive Care Unit, The Fifth Affiliated Hospital of SUN YAT-SEN University, Zhuhai City, Guangdong Province, PR China
| |
Collapse
|
56
|
Yin Z, Zou Y, Wang D, Huang X, Xiong S, Cao L, Zhang Y, Sun Y, Zhang N. Regulation of the Tec family of non-receptor tyrosine kinases in cardiovascular disease. Cell Death Discov 2022; 8:119. [PMID: 35296647 PMCID: PMC8927484 DOI: 10.1038/s41420-022-00927-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 02/14/2022] [Accepted: 02/25/2022] [Indexed: 12/04/2022] Open
Abstract
Tyrosine phosphorylation by protein tyrosine kinases (PTKs) is a type of post-translational modification. Tec kinases, which are a subfamily of non-receptor PTKs, were originally discovered in the hematopoietic system and include five members: Tec, Btk, Itk/Emt/Tsk, Etk/Bmx, and Txk/Rlk. With the progression of modern research, certain members of the Tec family of kinases have been found to be expressed outside the hematopoietic system and are involved in the development and progression of a variety of diseases. The role of Tec family kinases in cardiovascular disease is receiving increasing attention. Tec kinases are involved in the occurrence and progression of ischemic heart disease, atherosclerosis, cardiac dysfunction associated with sepsis, atrial fibrillation, myocardial hypertrophy, coronary atherosclerotic heart disease, and myocardial infarction and post-myocardial. However, no reviews have comprehensively clarified the role of Tec kinases in the cardiovascular system. Therefore, this review summarizes research on the role of Tec kinases in cardiovascular disease, providing new insights into the prevention and treatment of cardiovascular disease.
Collapse
Affiliation(s)
- Zeyu Yin
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yuanming Zou
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Dong Wang
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xinyue Huang
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Shengjun Xiong
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Liu Cao
- Key Laboratory of Medical Cell Biology, Ministry of Education; Institute of Translational Medicine, China Medical University; Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis and Treatment and Prevention, Shenyang, Liaoning, China
| | - Ying Zhang
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, China.
| | - Yingxian Sun
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, China.
| | - Naijin Zhang
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, China.
| |
Collapse
|
57
|
Kim M, Nikouee A, Sun Y, Zhang QJ, Liu ZP, Zang QS. Evaluation of Parkin in the Regulation of Myocardial Mitochondria-Associated Membranes and Cardiomyopathy During Endotoxemia. Front Cell Dev Biol 2022; 10:796061. [PMID: 35265609 PMCID: PMC8898903 DOI: 10.3389/fcell.2022.796061] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 01/14/2022] [Indexed: 11/16/2022] Open
Abstract
Background: Mitochondrial deficiency is a known pathology in sepsis-induced organ failure. We previously found that mitochondria-associated membranes (MAMs), a subcellular domain supporting mitochondrial status, are impaired in the heart during endotoxemia, suggesting a mechanism of mitochondrial damage occurred in sepsis. Mitophagy pathway via E3 ubiquitin ligase Parkin and PTEN-induced kinase 1 (PINK1) controls mitochondrial quality. Studies described here examined the impact of Parkin on cardiac MAMs and endotoxemia-induced cardiomyopathy. Additionally, point mutation W403A in Parkin was previously identified as a constitutively active mutation in vitro. In vivo effects of forced expression of this mutation were evaluated in the endotoxemia model. Methods: Mice of wild type (WT), Parkin-deficiency (Park2−/−), and knock-in expression of Parkin W402A (human Parkin W403A) were given lipopolysaccharide (LPS) challenge. Cardiac function was evaluated by echocardiography. In the harvested heart tissue, MAM fractions were isolated by ultracentrifugation, and their amount and function were quantified. Ultrastructure of MAMs and mitochondria was examined by electron microscopy. Mitochondrial respiratory activities were measured by enzyme assays. Myocardial inflammation was estimated by levels of pro-inflammatory cytokine IL-6. Myocardial mitophagy was assessed by levels of mitophagy factors associated with mitochondria and degrees of mitochondria-lysosome co-localization. Parkin activation, signified by phosphorylation on serine 65 of Parkin, was also evaluated. Results: Compared with WT, Park2−/− mice showed more severely impaired cardiac MAMs during endotoxemia, characterized by disrupted structure, reduced quantity, and weakened transporting function. Endotoxemia-induced cardiomyopathy was intensified in Park2−/− mice, shown by worsened cardiac contractility and higher production of IL-6. Mitochondria from the Park2−/− hearts were more deteriorated, indicated by losses in both structural integrity and respiration function. Unexpectedly, mice carrying Parkin W402A showed similar levels of cardiomyopathy and mitochondrial damage when compared with their WT counterparts. Further, Parkin W402A mutation neither enhanced mitophagy nor increased Parkin activation in myocardium under the challenge of endotoxemia. Conclusion: our results suggest that Parkin/PINK1 mitophagy participates in the regulation of cardiac MAMs during endotoxemia. Point mutation W402A (human W403A) in Parkin is not sufficient to alleviate cardiomyopathy induced by endotoxemia in vivo.
Collapse
Affiliation(s)
- Matthew Kim
- Department of Surgery, Burn & Shock Trauma Research Institute, Loyola University Chicago Stritch School of Medicine, Maywood, IL, United States
| | - Azadeh Nikouee
- Department of Surgery, Burn & Shock Trauma Research Institute, Loyola University Chicago Stritch School of Medicine, Maywood, IL, United States
| | - Yuxiao Sun
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Qing-Jun Zhang
- Internal Medicine-Cardiology Division, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Zhi-Ping Liu
- Internal Medicine-Cardiology Division, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Qun Sophia Zang
- Department of Surgery, Burn & Shock Trauma Research Institute, Loyola University Chicago Stritch School of Medicine, Maywood, IL, United States
| |
Collapse
|
58
|
Neuropsychological Outcome of Critically Ill Patients with Severe Infection. Biomedicines 2022; 10:biomedicines10030526. [PMID: 35327328 PMCID: PMC8945835 DOI: 10.3390/biomedicines10030526] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 02/17/2022] [Accepted: 02/20/2022] [Indexed: 12/29/2022] Open
Abstract
Sepsis and septic shock represent important burdens of disease around the world. Sepsis-associated neurological consequences have a great impact on patients, both in the acute phase and in the long term. Sepsis-associated encephalopathy (SAE) is a severe brain dysfunction that may contribute to long-term cognitive impairment. Its pathophysiology recognizes the following two main mechanisms: neuroinflammation and hemodynamic impairment. Clinical manifestations include different forms of altered mental status, from agitation and restlessness to delirium and deep coma. A definite diagnosis is difficult because of the absence of specific radiological and biological criteria; clinical management is restricted to the treatment of sepsis, focusing on early detection of the infection source, maintenance of hemodynamic homeostasis, and avoidance of metabolic disturbances or neurotoxic drugs.
Collapse
|
59
|
Zhu Z, Zhang G, Li D, Yin X, Wang T. Silencing of specificity protein 1 protects H9c2 cells against lipopolysaccharide-induced injury via binding to the promoter of chemokine CXC receptor 4 and suppressing NF-κB signaling. Bioengineered 2022; 13:3395-3409. [PMID: 35048778 PMCID: PMC8973921 DOI: 10.1080/21655979.2022.2026548] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
G protein-coupled protein receptor CXC chemokine receptor 4 (CXCR4) has been shown to be involved in the development of sepsis; however, it remains unclear whether CXCR4 participates in the septic myocardial injury. In our study, treatment with lipopolysaccharide (LPS) increased the expression of specificity protein 1 (SP1) and CXCR4 in H9c2 cells. Notably, a positive association between SP1 and CXCR4 expression was observed in LPS-treated H9c2 cells, and SP1 positively regulated CXCR4 expression in H9c2 cells. Moreover, silencing of SP1 or CXCR4 suppressed LPS-induced inflammation and cell apoptosis in H9c2 cells, as evidenced by the increase in cell viability and decrease in lactate dehydrogenase release, interleukin (IL)-6, IL-8, and tumor necrosis factor (TNF)-α levels, and caspase-3 activity. Additionally, overexpression of CXCR4 abolished the protective effects of SP1 silencing on LPS-induced injury in H9c2 cells. SP1 was also shown to enhance the promoter activity of CXCR4 by directly binding with the binding motif site – 109/–100 in CXCR4 promoter. Besides, downregulation of SP1 or CXCR4 blocked LPS-induced activation of the NF-кB signaling in H9c2 cells. Furthermore, inhibition of NF-кB signaling by DHMEQ abolished LPS-induced myocardial inflammation and apoptosis. In conclusion, silencing of SP1 protected H9c2 cells against LPS-induced injury by binding to the promoter of CXCR4 and suppressing the NF-κB signaling pathway. Hence, our findings provide evidence that manipulation of SP1 or CXCR4 may be an effective approach to promote prevention or recovery of septic myocardial injury, and thereby, may serve as a potential therapeutic strategy for sepsis.
Collapse
Affiliation(s)
- Zhao Zhu
- Department of Emergency, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, 471003 China
| | - Guoxiu Zhang
- Department of Emergency, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, 471003 China
| | - Dahuan Li
- Department of Emergency, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, 471003 China
| | - Xiaojun Yin
- Department of Emergency, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, 471003 China
| | - Tianzhong Wang
- Department of Emergency, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, 471003 China
| |
Collapse
|
60
|
Falach R, Goldvaser M, Halpern P, Rosner A, Sapoznikov A, Gal Y, Goren O, Sabo T, Kronman C, Katalan S. Pathophysiological profile of awake and anesthetized pigs following systemic exposure to the highly lethal ricin toxin. Clin Toxicol (Phila) 2022; 60:76-82. [PMID: 34080504 DOI: 10.1080/15563650.2021.1933513] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/27/2021] [Accepted: 05/18/2021] [Indexed: 10/21/2022]
Abstract
Ricin, a plant-derived toxin originating from the seeds of Ricinus communis (castor bean plant), is one of the most lethal toxins known. To date, no in-depth study of systemic exposure to ricin in a standardized large animal model has been reported. This study details for the first time the pathophysiological hemodynamic profile following systemic/intramuscular exposure to the ricin toxin in a porcine model by comprehensive cardiorespiratory monitoring of awake and anesthetized pigs. Unlike respiratory exposure to ricin, which is characterized by the development of acute respiratory distress syndrome, following intramuscular exposure to ricin respiratory parameters were grossly unaffected, however the hemodynamics of both awake and anesthetize pigs were unsustainably compromised. We show that in the early phase until approximately 24 h post-exposure, cardiac output is not impaired although one of its components, stroke volume, is relatively low. This is due to compensatory increase in heart rate, which eventually becomes insufficient. Later, distributive shock develops, characterized by severe vasodilatation (decreased systemic vascular resistance), low central venous oxygen saturation and elevation of venous-to-arterial carbon dioxide difference indicating increase in tissue oxygen demand not met by cardiac supply. These findings serve as a basis for further studies to evaluate the ability of supportive treatments such as vasoactive and inotropic drugs, to postpone the hemodynamic deterioration and thus expand the therapeutic window for the anti-ricin treatment. Such studies are of crucial importance for judicious treatment of victims of acts of bioterrorism or of intentional self-poisoning.
Collapse
Affiliation(s)
- Reut Falach
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness Ziona, Israel
| | - Michael Goldvaser
- Division of Medicinal Chemistry, Israel Institute for Biological Research, Ness Ziona, Israel
| | | | - Amir Rosner
- Veterinary Center for Preclinical Research, Israel Institute for Biological Research, Ness Ziona, Israel
| | - Anita Sapoznikov
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness Ziona, Israel
| | - Yoav Gal
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness Ziona, Israel
| | - Orr Goren
- Anesthesia, Pain and Intensive Care Division, Tel-Aviv Medical Center, Tel-Aviv University, Tel Aviv, Israel
| | - Tamar Sabo
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness Ziona, Israel
| | - Chanoch Kronman
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness Ziona, Israel
| | - Shahaf Katalan
- Division of Medicinal Chemistry, Israel Institute for Biological Research, Ness Ziona, Israel
| |
Collapse
|
61
|
Irisin Protects Against LPS-Stressed Cardiac Damage Through Inhibiting Inflammation, Apoptosis, and Pyroptosis. Shock 2021; 56:1009-1018. [PMID: 34779800 DOI: 10.1097/shk.0000000000001775] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
ABSTRACT Septic cardiac dysfunction remains a clinical problem due to its high morbidity and mortality. Uncontrolled cell death and excessive inflammatory response are closely related to sepsis-induced cardiac dysfunction. Irisin has been found to play cardioprotective roles in sepsis. However, there is enough uncertainty in the mechanism of irisin-mediated cardioprotection. We hypothesized that irisin may ameliorate myocardial dysfunction via reducing cardiac apoptosis, pyroptosis, and inflammation during LPS-induced sepsis. Mice were subjected to LPS with or without irisin treatment. After stimuli of LPS, the function of myocardium was distinctly impaired, which was closely related to increased level of apoptosis (decreased expression of Bcl-2 and elevated expression of Caspase-3 and Bax), pyroptosis (increased expression of Caspase1, NLR family pyrin domain containing 3 (NLRP3), and gasdermin D) and inflammatory mediators (increased level of IL-1β, TNF-α, and IL-6). This process is consistent with increased toll-like receptor 4 (TLR4)/nuclear factor-kappa B signal, apoptotic signal, and NLRP3-mediated pyroptotic signal. Activation of apoptosis and pyroptosis enhanced the expression of proinflammatory cytokines and further exacerbated septic myocardial damage. However, irisin can inhibit the expression of TLR4 and its downstream signaling molecules and also lower the level of apoptosis and pyroptosis. Besides, similar results were also found in vitro model of LPS-induced H9c2 cardiomyocyte injury. In general, irisin suppressed inflammation, apoptosis, and pyroptosis by blocking the TLR4 and NLRP3 inflammasome signalings to mitigate myocardial dysfunction in sepsis.
Collapse
|
62
|
MiR-702-3p inhibits the inflammatory injury in septic H9c2 cells by regulating NOD1. Transpl Immunol 2021; 70:101493. [PMID: 34774740 DOI: 10.1016/j.trim.2021.101493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 10/27/2021] [Accepted: 11/06/2021] [Indexed: 11/24/2022]
Abstract
BACKGROUND Cardiac insufficiency is a common complication of sepsis and septic shock and is the most common cause of death in critically ill patients. Recent studies have found that microRNAs (miRNAs) play a potential role in sepsis as markers, but little is known about their functional effects on sepsis-induced cardiomyopathy (SIC). OBJECTIVE This study is designed to explore the possible role and underlying mechanisms of miR-702-3p in septic cardiomyopathy. METHODS As expected, H9c2 cells were induced with lipopolysaccharide (LPS) to construct the model of septic cardiomyopathy. The expression of miR-702-3p was detected by qRT-PCR assay and those of IL-1β, IL-6 and TNF-α by ELISA assay. The viability, proliferation and apoptosis of LPS-treated H9c2 cells were determined by CCK-8, EdU, flow cytometry and western blot assays. Moreover, Nucleotide-binding oligomerization domain-containing protein 1 (NOD1) was predicted and confirmed as a direct target of miR-702-3p by TargetScan, miRwalk and miRDB prediction and dual-luciferase reporter gene assays. RESULTS While LPS can weaken the viability of H9c2 cells, miR-702-3p enhances that of LPS-treated H9c2 cells by inhibit the expressions of TNF-α, IL-6, IL-1β. We found NOD1 is a target gene of miR-702-3p, and over-expression of NOD1 restores the inhibitory effects of miR-702-3p on the LPS-treated H9c2 cells. CONCLUSION MiR-702-3p played an important role in the pathogenesis of sepsis cardiomyopathy via targeting NOD1, suggesting that miR-702-3p may be a potential new target for the treatment of SIC.
Collapse
|
63
|
Surviving Sepsis Campaign: International Guidelines for Management of Sepsis and Septic Shock 2021. Crit Care Med 2021; 49:e1063-e1143. [PMID: 34605781 DOI: 10.1097/ccm.0000000000005337] [Citation(s) in RCA: 1232] [Impact Index Per Article: 308.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
64
|
Chen L, Tian Q, Shi Z, Qiu Y, Lu Q, Liu C. Melatonin Alleviates Cardiac Function in Sepsis-Caused Myocarditis via Maintenance of Mitochondrial Function. Front Nutr 2021; 8:754235. [PMID: 34708067 PMCID: PMC8542660 DOI: 10.3389/fnut.2021.754235] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 09/10/2021] [Indexed: 12/13/2022] Open
Abstract
Melatonin (N-acetyl-5-methoxytryptamine) has been shown to have a cardioprotective effect against myocarditis. However, the mechanisms underlying the protective role of melatonin (MLT) in sepsis-induced myocarditis are yet to be revealed. In this study, MLT was administrated to mice, 14 days before cecal ligation puncture surgery. Echocardiography results showed that MLT alleviated cardiac dysfunction in sepsis-induced myocarditis. Furthermore, MLT reduced cardiac inflammation by inhibiting the expression of Il-1α, Il-1β, Il-6, and Mcp-1 messenger RNA (mRNA) levels. The RNA sequencing (RNA-seq) assays with heart tissues showed that MLT maintains the mitochondrial function in sepsis-caused myocarditis. Additionally, the production of reactive oxygen species (ROS) in heart tissues was suppressed by MLT. Taken together, in evaluating the therapeutic effect of MLT on sepsis-induced myocarditis, the results showed that MLT alleviated cardiac damage by regulating mitochondrial function and mitochondrial ROS.
Collapse
Affiliation(s)
- Liyang Chen
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Qing Tian
- Intensive Care Unit of Wuhan Asia Heart Hospital, Wuhan, China
| | - Zhiguang Shi
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Yu Qiu
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA, United States
| | - Qiulun Lu
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Chao Liu
- Hubei Key Laboratory of Diabetes and Angiopathy, Hubei University of Science and Technology, Xianning, China
| |
Collapse
|
65
|
Chen W, Gao G, Yan M, Yu M, Shi K, Yang P. Long noncoding RNA MAPKAPK5-AS1 promoted lipopolysaccharide-induced inflammatory damage in the myocardium by sponging microRNA-124-3p/E2F3. Mol Med 2021; 27:131. [PMID: 34666672 PMCID: PMC8524853 DOI: 10.1186/s10020-021-00385-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 09/22/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Myocardial dysfunction caused by sepsis (SIMD) leads to high mortality in critically ill patients. We investigated the function and mechanism of long non-coding RNA MAPKAPK5-AS1 (lncRNA MAPKAPK-AS1) on lipopolysaccharide (LPS)-induced inflammation response in vivo and in vitro. METHOD Male SD rats were utilized for in vivo experiments. Rat cardiomyocytes (H9C2) were employed for in vitro experiments. Western blotting was employed to measure protein expression, and RT-PCR was performed to measure mRNA expression of inflammation factors. TUNEL and flow cytometry were carried out to evulate cell apoptosis. RESULT The results showed that the expression of MAPKAPK5-AS1 was increased, while the expression of miR-124-3p was decreased in the inflammatory damage induced by LPS in vivo and in vitro. Knockdown of MAPKAPK5-AS1 reduced LPS-induced cell apoptosis and inflammation response, while overexpression of miR-124-3p weakened the effects of MAPKAPK5-AS1 knockdown on LPS-induced cell apoptosis and inflammation response. Moreover, miR-124-3p was identified as a downstream miRNA of MAPKAPK5-AS1, and E2F3 was a target of miR-214-3p. MAPKAPK5-AS1 knockdown increased the expression of miR-124-3p, while miR-124-3p overexpression reduced the expression of MAPKAPK5-AS1. In addition, miR-124-3p was found to downregulate E2F3 expression in H9C2 cells. CONCLUSION MAPKAPK5-AS1/miR-124-3p/E2F3 axis regulates LPS-related H9C2 cell apoptosis and inflammatory response.
Collapse
Affiliation(s)
- Weiwei Chen
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun City, 130033, Jilin Province, People's Republic of China
- Jilin Provincial Key Laboratory for Genetic Diagnosis of Cardiovascular Disease, Changchun City, 130033, Jilin Province, People's Republic of China
| | - Guangyuan Gao
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun City, 130033, Jilin Province, People's Republic of China
- Jilin Provincial Key Laboratory for Genetic Diagnosis of Cardiovascular Disease, Changchun City, 130033, Jilin Province, People's Republic of China
| | - Mengjie Yan
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun City, 130033, Jilin Province, People's Republic of China
- Jilin Provincial Key Laboratory for Genetic Diagnosis of Cardiovascular Disease, Changchun City, 130033, Jilin Province, People's Republic of China
| | - Ming Yu
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun City, 130033, Jilin Province, People's Republic of China
- Jilin Provincial Key Laboratory for Genetic Diagnosis of Cardiovascular Disease, Changchun City, 130033, Jilin Province, People's Republic of China
| | - Kaiyao Shi
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun City, 130033, Jilin Province, People's Republic of China
- Jilin Provincial Key Laboratory for Genetic Diagnosis of Cardiovascular Disease, Changchun City, 130033, Jilin Province, People's Republic of China
| | - Ping Yang
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun City, 130033, Jilin Province, People's Republic of China.
- Jilin Provincial Key Laboratory for Genetic Diagnosis of Cardiovascular Disease, Changchun City, 130033, Jilin Province, People's Republic of China.
| |
Collapse
|
66
|
Chen P, An Q, Huang Y, Zhang M, Mao S. Prevention of endotoxin-induced cardiomyopathy using sodium tanshinone IIA sulfonate: Involvement of augmented autophagy and NLRP3 inflammasome suppression. Eur J Pharmacol 2021; 909:174438. [PMID: 34437885 DOI: 10.1016/j.ejphar.2021.174438] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 08/16/2021] [Accepted: 08/17/2021] [Indexed: 11/17/2022]
Abstract
Increasing evidence indicates that patients or experimental animals exposure to endotoxin (lipopolysaccharides, LPS) exert deleterious cardiac functions that greatly contribute to morbidity and mortality. The pathophysiologic processes, including NLRP3 inflammasome overactivation and cardiac inflammatory injury, are complicated. Sodium tanshinone IIA sulfonate (STS), a water-soluble derivative of tanshinone IIA, is a naturally occurring compound extracted from Salvia miltiorrhiza and has anti-inflammatory and cardioprotective properties. In this study we examined the effect of STS on endotoxin-induced cardiomyopathy and investigated the underlying mechanisms. An endotoxemic mouse model was established by injecting LPS (10 mg/kg). Different doses of STS were administered intraperitoneally (5, 10, or 50 mg/kg) at different time points (2/12 h, 4/12 h, and 8/12 h) after LPS challenge to assess its effect on survival of mice with endotoxemia. In parallel, cardiac function, myocardial inflammatory cytokines, cardiomyocyte pyroptosis and autophagy were evaluated to determine the extent of myocardial damage due to sepsis in the presence and absence of STS at the optimal dose (10 mg/kg) and time-point (2/12 h). The results demonstrated that STS increased the survival rates, improved the compromised cardiac function and reduced myocardial inflammatory injury associated with enhanced autophagy and mitigated NLRP3 inflammasome activation. Moreover, inhibiting of autophagy or blocking the AMPK pathway reversed STS-elicited prevention of cardiomyopathy and activated the NLRP3 inflammasome in endotoxemic mice. Collectively, our study demonstrates that STS attenuates endotoxemia-induced mortality and cardiomyopathy, which may be associated with promotion of autophagy and inhibition of NLRP3 inflammasome overactivation.
Collapse
Affiliation(s)
- Peipei Chen
- Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; Department of Critical Care Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, 510120, China
| | - Qiyuan An
- Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; Department of Critical Care Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, 510120, China; Southern Medical University, Guangzhou, 510515, China
| | - Yuxin Huang
- Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Minzhou Zhang
- Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; Department of Critical Care Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, 510120, China
| | - Shuai Mao
- Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; Department of Critical Care Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, 510120, China; Guangdong Provincial Branch of National Clinical Research Centre for Chinese Medicine Cardiology, Guangzhou, 510120, China.
| |
Collapse
|
67
|
Yang C, Wen K. Predictive value and regulatory mechanism of serum miR-499a-5p on myocardial dysfunction in sepsis. J Cardiothorac Surg 2021; 16:301. [PMID: 34654440 PMCID: PMC8518260 DOI: 10.1186/s13019-021-01679-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 09/29/2021] [Indexed: 12/13/2022] Open
Abstract
Background This study sought to investigate the predictive value and regulatory mechanism of serum miR-499a-5p in sepsis-induced myocardial dysfunction (SIMD). Methods A total of 60 patients with sepsis and 60 healthy volunteers were enrolled in this study. The serum levels of miRNAs (miR-451, miR-378 and miR-499a-5p) were detected. Receiver operating characteristic curve and logistic regression analysis were used to evaluate the diagnostic and prognostic value of miR-499a-5p in SIMD patients. AC16 cells were used to establish SIMD model in vitro using lipopolysaccharide (LPS). An analysis was conducted for miR-499a-5p expression, cell viability, and the concentration of creatine kinase-MB isoform (CK-MB), brain natriuretic peptide (BNP), superoxide dismutase (SOD) and cytochrome C oxidase IV (COX IV). The downstream target of miR-499a-5p was verified. Results Our results revealed a poor expression of miR-499a-5p in the serum of SIMD patients, while no significant difference was evident for miR-451 and miR-378. The level of miR-499a-5p in the survival group was higher than the non-survival group. miR-499a-5p elicited good diagnostic and prognostic value for SIMD. Our findings revealed that miR-499a-5p was decreased significantly in LPS-treated cardiomyocytes. After overexpression of miR-499a-5p, the cell viability increased, and the concentrations of CK-MB and BNP were decreased, while the concentrations of SOD and COX IV were increased. EIF4E was validated as the target of miR-499a-5p. After overexpression of EIF4E, the cell viability was decreased and the concentrations of CK-MB and BNP were increased while the concentrations of SOD and COX IV were decreased. Conclusion The level of miR-499a-5p is weak in SIMD patients. miR-499a-5p has a good diagnostic and prognostic value for SIMD by inhibiting EIF4E transcription.
Collapse
Affiliation(s)
- Chuang Yang
- Department of Critical Care Medicine, The Second Hospital of Shandong University, No. 247 Beiyuan Dajie Street, Jinan City, 250012, Shandong Province, China
| | - Kun Wen
- Department of Critical Care Medicine, The Second Hospital of Shandong University, No. 247 Beiyuan Dajie Street, Jinan City, 250012, Shandong Province, China.
| |
Collapse
|
68
|
Evans L, Rhodes A, Alhazzani W, Antonelli M, Coopersmith CM, French C, Machado FR, Mcintyre L, Ostermann M, Prescott HC, Schorr C, Simpson S, Wiersinga WJ, Alshamsi F, Angus DC, Arabi Y, Azevedo L, Beale R, Beilman G, Belley-Cote E, Burry L, Cecconi M, Centofanti J, Coz Yataco A, De Waele J, Dellinger RP, Doi K, Du B, Estenssoro E, Ferrer R, Gomersall C, Hodgson C, Møller MH, Iwashyna T, Jacob S, Kleinpell R, Klompas M, Koh Y, Kumar A, Kwizera A, Lobo S, Masur H, McGloughlin S, Mehta S, Mehta Y, Mer M, Nunnally M, Oczkowski S, Osborn T, Papathanassoglou E, Perner A, Puskarich M, Roberts J, Schweickert W, Seckel M, Sevransky J, Sprung CL, Welte T, Zimmerman J, Levy M. Surviving sepsis campaign: international guidelines for management of sepsis and septic shock 2021. Intensive Care Med 2021; 47:1181-1247. [PMID: 34599691 PMCID: PMC8486643 DOI: 10.1007/s00134-021-06506-y] [Citation(s) in RCA: 2075] [Impact Index Per Article: 518.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Accepted: 08/05/2021] [Indexed: 02/07/2023]
Affiliation(s)
- Laura Evans
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Washington, Seattle, WA, USA.
| | - Andrew Rhodes
- Adult Critical Care, St George's University Hospitals NHS Foundation Trust & St George's University of London, London, UK
| | - Waleed Alhazzani
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON, Canada
| | - Massimo Antonelli
- Dipartimento di Scienze dell'Emergenza, Anestesiologiche e della Rianimazione, Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | | | | | - Flávia R Machado
- Anesthesiology, Pain and Intensive Care Department, Federal University of São Paulo, Hospital of São Paulo, São Paulo, Brazil
| | | | | | - Hallie C Prescott
- University of Michigan and VA Center for Clinical Management Research, Ann Arbor, MI, USA
| | | | - Steven Simpson
- University of Kansas Medical Center, Kansas City, KS, USA
| | - W Joost Wiersinga
- ESCMID Study Group for Bloodstream Infections, Endocarditis and Sepsis, Division of Infectious Diseases, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Fayez Alshamsi
- Department of Internal Medicine, College of Medicine and Health Sciences, Emirates University, Al Ain, United Arab Emirates
| | - Derek C Angus
- University of Pittsburgh Critical Care Medicine CRISMA Laboratory, Pittsburgh, PA, USA
| | - Yaseen Arabi
- Intensive Care Department, Ministry of National Guard Health Affairs, King Abdullah International Medical Research Center, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Kingdom of Saudi Arabia
| | - Luciano Azevedo
- School of Medicine, University of Sao Paulo, São Paulo, Brazil
| | | | | | | | - Lisa Burry
- Mount Sinai Hospital & University of Toronto (Leslie Dan Faculty of Pharmacy), Toronto, ON, Canada
| | - Maurizio Cecconi
- Department of Biomedical Sciences, Humanitas University Pieve Emanuele, Milan, Italy.,Department of Anaesthesia and Intensive Care, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - John Centofanti
- Department of Anesthesia, McMaster University, Hamilton, ON, Canada
| | - Angel Coz Yataco
- Lexington Veterans Affairs Medical Center/University of Kentucky College of Medicine, Lexington, KY, USA
| | | | | | - Kent Doi
- The University of Tokyo, Tokyo, Japan
| | - Bin Du
- Medical ICU, Peking Union Medical College Hospital, Beijing, China
| | - Elisa Estenssoro
- Hospital Interzonal de Agudos San Martin de La Plata, Buenos Aires, Argentina
| | - Ricard Ferrer
- Intensive Care Department, Vall d'Hebron University Hospital, Vall d'Hebron Institut de Recerca, Barcelona, Spain
| | | | - Carol Hodgson
- Australian and New Zealand Intensive Care Research Centre, Monash University, Melbourne, VIC, Australia
| | - Morten Hylander Møller
- Department of Intensive Care 4131, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | | | - Shevin Jacob
- Liverpool School of Tropical Medicine, Liverpool, UK
| | | | - Michael Klompas
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA.,Department of Population Medicine, Harvard Medical School, and Harvard Pilgrim Health Care Institute, Boston, MA, USA
| | - Younsuck Koh
- ASAN Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Anand Kumar
- University of Manitoba, Winnipeg, MB, Canada
| | - Arthur Kwizera
- Makerere University College of Health Sciences, Kampala, Uganda
| | - Suzana Lobo
- Intensive Care Division, Faculdade de Medicina de São José do Rio Preto, São Paulo, Brazil
| | - Henry Masur
- Critical Care Medicine Department, NIH Clinical Center, Bethesda, MD, USA
| | | | | | - Yatin Mehta
- Medanta the Medicity, Gurugram, Haryana, India
| | - Mervyn Mer
- Charlotte Maxeke Johannesburg Academic Hospital and Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Mark Nunnally
- New York University School of Medicine, New York, NY, USA
| | - Simon Oczkowski
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON, Canada
| | - Tiffany Osborn
- Washington University School of Medicine, St. Louis, MO, USA
| | | | | | - Michael Puskarich
- University of Minnesota/Hennepin County Medical Center, Minneapolis, MN, USA
| | - Jason Roberts
- Faculty of Medicine, University of Queensland Centre for Clinical Research, The University of Queensland, Brisbane, Australia.,Department of Pharmacy, Royal Brisbane and Women's Hospital, Brisbane, Australia.,Department of Intensive Care Medicine, Royal Brisbane and Women's Hospital, Brisbane, Australia.,Division of Anaesthesiology Critical Care Emergency and Pain Medicine, Nîmes University Hospital, University of Montpellier, Nîmes, France
| | | | | | | | - Charles L Sprung
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel.,Department of Anesthesiology, Critical Care and Pain Medicine, Hadassah Medical Center, Jerusalem, Israel
| | - Tobias Welte
- Medizinische Hochschule Hannover and German Center of Lung Research (DZL), Hannover, Germany
| | - Janice Zimmerman
- World Federation of Intensive and Critical Care, Brussels, Belgium
| | - Mitchell Levy
- Warren Alpert School of Medicine at Brown University, Providence, Rhode Island & Rhode Island Hospital, Providence, RI, USA
| |
Collapse
|
69
|
Tang X, Xu Y, Dai X, Xing Y, Yang D, Huang Q, Li H, Lv X, Wang Y, Lu D, Wang H. The Long-term Effect of Dobutamine on Intrinsic Myocardial Function and Myocardial Injury in Septic Rats with Myocardial Dysfunction. Shock 2021; 56:582-592. [PMID: 34524268 DOI: 10.1097/shk.0000000000001718] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
ABSTRACT Dobutamine (DOB) is recommended as an inotrope for septic patients with low cardiac output, but its long-term impact on sepsis-induced cardiomyopathy remains unclear. This study investigated the long-term effect of DOB on septic myocardial dysfunction and injury. Rats were exposed to cecal ligation and puncture (CLP), the intrinsic myocardial function, other organ functions, hemodynamics, inflammatory response, serum myocardial injury biomarkers, myocardial apoptosis, and vascular permeability were determined. At 6 h after CLP, the left ventricular ±dP/dt were significantly depressed, cardiac tumor necrosis factor-α and vascular cell adhesion molecule-1 expression were increased, but not serum cardiac troponin I (cTnI), N-terminal pro-brain natriuretic peptide (NT-proBNP), heart-type fatty acid-binding protein (H-FABP), creatinine, and urea nitrogen concentrations in CLP group compared with controls. At 9 h after CLP, hepatic dysfunction was present in CLP rats compared with controls. At 6 h after CLP, DOB treatment did not affect hemodynamics, the left ventricular ±dP/dt, cytokine levels in serum and myocardium, as well as cardiomyocyte apoptosis and cardiac vascular hyperpermeability at 20 h after CLP. However, DOB (10.0 μg/kg) increased serum IL-10 level and improved survival in septic rats. These results indicate that the intrinsic myocardial depression occurs earlier than hepatic and renal dysfunction in sepsis and serum cTnI, NT-proBNP, and H-FABP are not suitable as early biomarkers for sepsis-induced myocardial dysfunction. Although DOB treatment (10.0 μg/kg) in the presence of myocardial dysfunction improves survival in septic rats, it neither improves myocardial function and hemodynamics nor attenuates myocardial injury at the later stage of sepsis.
Collapse
Affiliation(s)
- Xiangxu Tang
- Department of Pathophysiology, Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
70
|
Tang W, Luo L, Hu B, Zheng M. Butorphanol alleviates lipopolysaccharide-induced inflammation and apoptosis of cardiomyocytes via activation of the κ-opioid receptor. Exp Ther Med 2021; 22:1248. [PMID: 34539844 PMCID: PMC8438658 DOI: 10.3892/etm.2021.10683] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 04/16/2021] [Indexed: 12/25/2022] Open
Abstract
Sepsis-induced myocardial dysfunction is a leading cause of the high mortality rates associated with sepsis. The aim of the present study was to investigate the effect of butorphanol on sepsis-induced cardiomyocyte dysfunction. Lipopolysaccharide (LPS) was used to induce H9C2 cardiomyocytes to establish an in vitro sepsis model. The effect of butorphanol on the viability of LPS-induced H9C2 cells was analyzed using a Cell Counting Kit-8 assay. The levels of tumor necrosis factor-α, interleukin (IL)-1β and IL-6 were detected using ELISA. Western blotting was used to analyze the expression levels of inflammation-and apoptosis-related proteins. Cell apoptosis was measured using a TUNEL assay. The expression levels of κ-opioid receptor (KOR) were analyzed using reverse transcription-quantitative PCR analysis and western blotting. Following LPS induction, the levels of inflammatory cytokines and proapoptotic proteins were found to be upregulated in H9C2 cells, while butorphanol treatment downregulated these levels. The expression levels of KOR were also upregulated following butorphanol treatment in LPS-induced H9C2 cells. Addition of the KOR inhibitor, nor-binaltorphimine, alleviated the inhibitory effects of butorphanol on inflammation and apoptosis in LPS-induced H9C2 cells. In conclusion, the findings of the present study provided evidence indicating that butorphanol may alleviate LPS-induced inflammation and apoptosis in cardiomyocytes by upregulating KOR expression, which may provide a novel insight into the potential therapeutic effects of butorphanol and its underlying mechanism of action.
Collapse
Affiliation(s)
- Weiqing Tang
- Department of Anesthesiology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, P.R. China
| | - Liu Luo
- Department of Anesthesiology, The Affiliated Zhuzhou Hospital of Xiangya School of Medicine, Central South University, Zhuzhou Central Hospital, Zhuzhou, Hunan 412000, P.R. China
| | - Baoji Hu
- Department of Anesthesiology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, P.R. China
| | - Mingzhi Zheng
- Department of Anesthesiology, The Affiliated Zhuzhou Hospital of Xiangya School of Medicine, Central South University, Zhuzhou Central Hospital, Zhuzhou, Hunan 412000, P.R. China
| |
Collapse
|
71
|
Hao Y, Wang Z, Wang X, Zhan W, Wu D. OGDH is involved in sepsis induced acute lung injury through the MAPK pathway. J Thorac Dis 2021; 13:5042-5054. [PMID: 34527342 PMCID: PMC8411135 DOI: 10.21037/jtd-21-948] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 07/02/2021] [Indexed: 11/06/2022]
Abstract
Background Acute lung injury (ALI) induced by sepsis is a common cause of death in clinical practice, and there remains a lack of clinical effective treatment. Cecal ligation and puncture (CLP) is a classic animal model of sepsis, which can induce ALI. Studies have shown that in the lung injury cell model, OGDH (oxoglutarate dehydrogenase) transcription is up-regulated, which is a potential therapeutic target for acute pneumonia. The purpose of this study was to confirm the effects of OGDH on lung injury and inflammation in animal and cell models, and to explore its mechanism. Methods By analyzing the GSE16650 gene set, the upregulated OGDH gene was detected in the lung injury cell model. In a sepsis animal model established by CLP and a lung injury cell model, RT-PCR, immunohistochemistry, WB, and other techniques were used to verify the upregulation of OGDH expression, which was then was down-regulated with shRNA to confirm its relationship with ALI. Further, ELISA, RT-PCR, and WB were used to detect the effect of OGDH on the expression of pro-inflammatory factors including IL-1β, IL-6, IL-18, and TNF-α. The downstream pathway of OGDH was predicted using KEGG and GSEA tools and verified by WB and immunofluorescence. Results The results showed OGDH was highly expressed in a lung injury cell model and the lung tissue of ALI mice induced by CLP, and downregulation of OGDH alleviated sepsis induced ALI. In animal models and cell models, the expression of OGDH was positively correlated with the expression of pro-inflammatory factors. OGDH may act through the MAPK pathway. Conclusions Under the pathological condition of sepsis, OGDH amplifies the inflammatory response through the MAPK pathway, releases pro-inflammatory factors, and induces ALI.
Collapse
Affiliation(s)
- Yuewei Hao
- Department of Emergency, Shandong Second Provincial General Hospital, Jinan, China
| | - Zheng Wang
- Prehospital Emergency, Shandong Second Provincial General Hospital, Jinan, China
| | - Xinfang Wang
- Neonatal Intensive Care Unit, Shandong Second Provincial General Hospital, Jinan, China
| | - Wenming Zhan
- Department of Internal Medicine-Cardiovascular, Xishui People's Hospital, Xishui, China
| | - Dianshui Wu
- Department of Clinical Laboratory, Shandong Second Provincial General Hospital, Jinan, China
| |
Collapse
|
72
|
Huang X, Zhang MZ, Liu B, Ma SY, Yin X, Guo LH. Astragaloside IV Attenuates Polymicrobial Sepsis-Induced Cardiac Dysfunction in Rats via IKK/NF-κB Pathway. Chin J Integr Med 2021; 27:825-831. [PMID: 34432200 DOI: 10.1007/s11655-021-2869-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/20/2020] [Indexed: 01/17/2023]
Abstract
OBJECTIVE To evaluate the protective effects of Astragaloside IV (AST) in a rat model of myocardial injury induced by cecal ligation and puncture (CLP). METHODS The model of sepsis-induced cardiac dysfunction was induced by CLP. Using a random number table, 50 specific pathogen free grade of Sprague Dawley rats were randomized into 5 groups: the sham group (sham), the model group (CLP, 18 h/72 h) and AST group (18 h/72 h). Except the sham group, the rats in other groups received CLP surgery to induce sepsis. CLP groups received intragastric administration with normal saline after CLP. AST groups received intragastric administration with AST solution (40 mg/kg) once a day. The levels of inflammatory mediators and oxidative stress markers in the serum of the septic rats were determined via enzyme-linked immunosorbent assay (ELISA) at different time point, such as interleukin 6 (IL-6), IL-10, high mobility group box-1 protein B1 (HMGB-1), superoxide dismutase (SOD), and malondialdehyde (MDA). Cardiac function was determined by echocardiography. Moreover, changes in myocardial pathology were evaluated using hematoxylin and eosin staining. The levels of lactate dehydrogenase (LDH) and creatine kinase-MB (CK-MB) were analysed to determine the status of CLP-induced myocardium. In addition, the apotosis of myocardial cells was analysed by terminal-deoxynucleoitidyl transferase mediated nick end labeling (TUNEL). The protein levels of B-cell lymphoma-2 (Bcl-2), Bcl-2-associated X (Bax), IκB kinase α (IKKα), nuclear factor kappa B p65 (NF-κB p65) were detected by Western blot analysis. Moreover, survival rate was investigated. RESULTS AST improved the survival rate of CLP-induced rats by up to 33.3% (P<0.05). The cardioprotective effect of AST was observed by increased ejection fraction, fractional shortening and left ventricular internal diameter in diastole respectively (P<0.01 or P<0.05). Subsequently, AST attenuated CLP-induced myocardial apoptosis and the ratio of Bcl-2/Bax in the myocardium, as well as the histological alterations of myocardium (P<0.01 or P<0.05); the generation of inflammatory cytokines (IL-6, IL-10, HMGB-1) and oxidative stress markers (SOD, MDA) in the serum was significantly alleviated (P<0.01 or P<0.05). On the other hand, AST markedly suppressed CLP-induced accumulation of IKK-α and NF-κB p65 subunit phosphorylation (P<0.01 or P<0.05). CONCLUSIONS AST plays a significant protective role in sepsis-induced cardiac dysfunction and survival outcome. The possible mechanism of cardioprotection is dependent on the activation of the IKK/NF-κB pathway in cardiomyocytes.
Collapse
Affiliation(s)
- Xin Huang
- Intensive Care Research Team of Traditional Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, the Second Clinical Collage of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
| | - Min-Zhou Zhang
- Department of Critical Care Medicine, Guangdong Provincial Hospital of Chinese Medicine, the Second Clinical Collage of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
| | - Bo Liu
- Department of Radiology, Guangdong Provincial Hospital of Chinese Medicine, the Second Clinical Collage of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
| | - Shi-Yu Ma
- Intensive Care Research Team of Traditional Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, the Second Clinical Collage of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
| | - Xin Yin
- Department of Critical Care Medicine, Guangdong Provincial Hospital of Chinese Medicine, the Second Clinical Collage of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
| | - Li-Heng Guo
- Department of Critical Care Medicine, Guangdong Provincial Hospital of Chinese Medicine, the Second Clinical Collage of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China.
| |
Collapse
|
73
|
Cheng Z, Lv D, Luo M, Wang R, Guo Y, Yang X, Huang L, Li X, Li C, Shang FF, Huang B, Shen J, Luo S, Yan J. Tubeimoside I protects against sepsis-induced cardiac dysfunction via SIRT3. Eur J Pharmacol 2021; 905:174186. [PMID: 34033817 DOI: 10.1016/j.ejphar.2021.174186] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 05/06/2021] [Accepted: 05/12/2021] [Indexed: 12/31/2022]
Abstract
Sepsis-induced cardiac dysfunction (SICD) is one of the key complications in sepsis and it is associated with adverse outcomes and increased mortality. There is no effective drug to treat SICD. Previously, we reported that tubeimoside I (TBM) improved survival of septic mice. The aim of this study is to figure out whether TBM ameliorates SICD. Also, SIRT3 was reported to protects against SICD. Our second aim is to confirm whether SIRT3 plays essential roles in TBM's protective effects against SICD. Our results demonstrated that TBM could alleviate SICD and SICD's key pathological factor, inflammation, oxidative stress, and apoptosis were all reduced by TBM. Notably, SICD induced a significant decrease in cardiac SIRT3 expression, while TBM treatment could reverse SIRT3 expression. To clarify whether TBM provides protection via SIRT3, we injected a specific SIRT3 inhibitor 3-(1H-1,2,3-triazol-4-yl) pyridine (3-TYP) into mice before TBM treatment. Then the cardioprotective effects of TBM were largely abolished by 3-TYP. This suggests that SIRT3 plays an essential role in TBM's cardioprotective effects. In vitro, TBM also protected H9c2 cells against LPS-induced injury, and siSIRT3 diminished these protective effects. Taken together, our results demonstrate that TBM protects against SICD via SIRT3. TBM might be a potential drug candidate for SICD treatment.
Collapse
Affiliation(s)
- Zhe Cheng
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China; Institute of Life Sciences, Chongqing Medical University, Chongqing, 400010, China
| | - Dingyi Lv
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China; Institute of Life Sciences, Chongqing Medical University, Chongqing, 400010, China
| | - Minghao Luo
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China; Institute of Life Sciences, Chongqing Medical University, Chongqing, 400010, China
| | - Ruiyu Wang
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China; Institute of Life Sciences, Chongqing Medical University, Chongqing, 400010, China
| | - Yongzheng Guo
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China; Institute of Life Sciences, Chongqing Medical University, Chongqing, 400010, China
| | - Xiyang Yang
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China; Institute of Life Sciences, Chongqing Medical University, Chongqing, 400010, China
| | - Longxiang Huang
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China; Institute of Life Sciences, Chongqing Medical University, Chongqing, 400010, China
| | - Xingbing Li
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China; Institute of Life Sciences, Chongqing Medical University, Chongqing, 400010, China
| | - Chang Li
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China; Institute of Life Sciences, Chongqing Medical University, Chongqing, 400010, China
| | - Fei-Fei Shang
- Institute of Life Sciences, Chongqing Medical University, Chongqing, 400010, China
| | - Bi Huang
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Jian Shen
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China; Institute of Life Sciences, Chongqing Medical University, Chongqing, 400010, China
| | - Suxin Luo
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China; Institute of Life Sciences, Chongqing Medical University, Chongqing, 400010, China
| | - Jianghong Yan
- Institute of Life Sciences, Chongqing Medical University, Chongqing, 400010, China.
| |
Collapse
|
74
|
Use of Organ Dysfunction as a Primary Outcome Variable Following Cecal Ligation and Puncture: Recommendations for Future Studies. Shock 2021; 54:168-182. [PMID: 31764625 DOI: 10.1097/shk.0000000000001485] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Outcomes variables for research on sepsis have centered on mortality and changes in the host immune response. However, a recent task force (Sepsis-3) revised the definition of sepsis to "life-threatening organ dysfunction caused by a dysregulated host response to infection." This new definition suggests that human studies should focus on organ dysfunction. The appropriate criteria for organ dysfunction in either human sepsis or animal models are, however, poorly delineated, limiting the potential for translation. Further, in many systems, the difference between "dysfunction" and "injury" may not be clear. In this review, we identify criteria for organ dysfunction and/or injury in human sepsis and in rodents subjected to cecal ligation and puncture (CLP), the most commonly used animal model of sepsis. We further examine instances where overlap between human sepsis and CLP is sufficient to identify translational endpoints. Additional verification may demonstrate that these endpoints are applicable to other animals and to other sepsis models, for example, pneumonia. We believe that the use of these proposed measures of organ dysfunction will facilitate mechanistic studies on the pathobiology of sepsis and enhance our ability to develop animal model platforms to evaluate therapeutic approaches to human sepsis.
Collapse
|
75
|
Li D, Wang M, Ye J, Zhang J, Xu Y, Wang Z, Zhao M, Ye D, Wan J. Maresin 1 alleviates the inflammatory response, reduces oxidative stress and protects against cardiac injury in LPS-induced mice. Life Sci 2021; 277:119467. [PMID: 33811894 DOI: 10.1016/j.lfs.2021.119467] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 03/17/2021] [Accepted: 03/28/2021] [Indexed: 02/08/2023]
Abstract
BACKGROUND Maresin 1 (MaR1) is a pro-resolving lipid mediator that has been reported to have strong regulatory effects on oxidative stress and inflammation. This study aimed to determine the effect of MaR1 on lipopolysaccharide (LPS)-induced sepsis-related cardiac injury and explore its possible mechanisms. METHODS Mice were administered MaR1 or PBS and then treated with LPS or saline for 6 h. Then, cardiac function, cardiac injury markers, cardiac macrophage differentiation, oxidative stress and myocardial cell apoptosis in each group were measured. RESULTS MaR1 treatment significantly decreased the serum levels of lactate dehydrogenase (LDH) and kinase isoenzyme (CK-MB) and improved cardiac function in LPS-induced mice. Treatment with MaR1 also inhibited LPS-induced M1 macrophage differentiation and reduced M1 macrophage-related cytokine secretion while promoting M2 macrophage differentiation and increasing M2 macrophage-related inflammatory mediator expression. In addition, MaR1 decreased serum malondialdehyde (MDA) levels and increased serum levels of superoxide dismutase (SOD) and glutathione (GSH), as well as cardiac expression of nuclear factor erythroid-2 related factor 2 (Nrf-2) and heme oxygenase 1 (HO-1), in LPS-induced mice. Furthermore, fewer TUNEL-positive cells were observed in the LPS + MaR1 group than in the LPS group. CONCLUSIONS Our experimental results show that MaR1 alleviates cardiac injury and protects against cardiac dysfunction and may be beneficial in reducing sepsis-induced cardiac injury.
Collapse
Affiliation(s)
- Dan Li
- Department of Pediatric, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Menglong Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan 430060, China.
| | - Jing Ye
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Jishou Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Yao Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Zhen Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Mengmeng Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Di Ye
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Jun Wan
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan 430060, China.
| |
Collapse
|
76
|
Laghlam D, Jozwiak M, Nguyen LS. Renin-Angiotensin-Aldosterone System and Immunomodulation: A State-of-the-Art Review. Cells 2021; 10:cells10071767. [PMID: 34359936 PMCID: PMC8303450 DOI: 10.3390/cells10071767] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 06/30/2021] [Accepted: 07/09/2021] [Indexed: 12/11/2022] Open
Abstract
The renin–angiotensin system (RAS) has long been described in the field of cardiovascular physiology as the main player in blood pressure homeostasis. However, other effects have since been described, and include proliferation, fibrosis, and inflammation. To illustrate the immunomodulatory properties of the RAS, we chose three distinct fields in which RAS may play a critical role and be the subject of specific treatments. In oncology, RAS hyperactivation has been associated with tumor migration, survival, cell proliferation, and angiogenesis; preliminary data showed promise of the benefit of RAS blockers in patients treated for certain types of cancer. In intensive care medicine, vasoplegic shock has been associated with severe macro- and microcirculatory imbalance. A relative insufficiency in angiotensin II (AngII) was associated to lethal outcomes and synthetic AngII has been suggested as a specific treatment in these cases. Finally, in solid organ transplantation, both AngI and AngII have been associated with increased rejection events, with a regional specificity in the RAS activity. These elements emphasize the complexity of the direct and indirect interactions of RAS with immunomodulatory pathways and warrant further research in the field.
Collapse
|
77
|
Xie L, Wu Y, Zhou C, Tan Z, Xu H, Chen G, Chen H, Huang G, Fan H, Gao L, Liu B, Zhou Y. Piceatannol protects against sepsis-induced myocardial dysfunction via direct inhibition of JAK2. Int Immunopharmacol 2021; 96:107639. [PMID: 34162128 DOI: 10.1016/j.intimp.2021.107639] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 03/24/2021] [Accepted: 03/31/2021] [Indexed: 11/26/2022]
Abstract
Sepsis-induced myocardial dysfunction (SIMD) represents one of the serious complications secondary to sepsis, which is a leading cause of the high mortality rate among septic cases. Subsequent cardiomyocyte apoptosis, together with the uncontrolled inflammatory response, has been suggested to be closely related to SIMD. Piceatannol (PIC) is verified with potent anti-apoptotic and anti-inflammatory effects, but its function and molecular mechanism in SIMD remain unknown so far. This study aimed to explore the potential role and mechanism of action of PIC in resisting SIMD. The interaction of PIC with JAK2 proteins was evaluated by molecular docking, molecular dynamics (MD) simulation and surface plasmon resonance imaging (SPRi). The cecal ligation and puncture-induced septicemia mice and the LPS-stimulated H9C2 cardiomyocytes were prepared as the models in vivo and in vitro, separately. Molecular docking showed that JAK2-PIC complex had the -8.279 kcal/mol binding energy. MD simulations showed that JAK2-PIC binding was stable. SPRi analysis also showed that PIC has a strong binding affinity to JAK2. PIC treatment significantly ameliorated the cardiac function, attenuated the sepsis-induced myocardial loss, and suppressed the myocardial inflammatory responses both in vivo and in vitro. Further detection revealed that PIC inhibited the activation of the JAK2/STAT3 signaling, which was tightly associated with apoptosis and inflammation. Importantly, pre-incubation with a JAK2 inhibitor (AG490) partially blocked the cardioprotective effects of PIC. Collectively, the findings demonstrated that PIC restored the impaired cardiac function by attenuating the sepsis-induced apoptosis and inflammation via suppressing the JAK2/STAT3 pathway both in septic mice and H9C2 cardiomyocytes.
Collapse
Affiliation(s)
- Lingpeng Xie
- School of Traditional Chinese Medicine, Department of Traditional Chinese Medicine, Nanfang Hospital (ZengCheng Branch), Southern Medical University, Guangzhou 510515, China
| | - Yuting Wu
- School of Traditional Chinese Medicine, Department of Traditional Chinese Medicine, Nanfang Hospital (ZengCheng Branch), Southern Medical University, Guangzhou 510515, China; Department of Traditional Chinese Medicine, Binzhou Medical University Hospital, Binzhou 256603, China
| | - Chuying Zhou
- School of Traditional Chinese Medicine, Department of Traditional Chinese Medicine, Nanfang Hospital (ZengCheng Branch), Southern Medical University, Guangzhou 510515, China
| | - Zhangbin Tan
- Department of Traditional Chinese Medicine (Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, State Key Laboratory of Respiratory Disease), the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 510260, China
| | - Honglin Xu
- School of Traditional Chinese Medicine, Department of Traditional Chinese Medicine, Nanfang Hospital (ZengCheng Branch), Southern Medical University, Guangzhou 510515, China
| | - Guanghong Chen
- School of Traditional Chinese Medicine, Department of Traditional Chinese Medicine, Nanfang Hospital (ZengCheng Branch), Southern Medical University, Guangzhou 510515, China
| | - Hongmei Chen
- School of Traditional Chinese Medicine, Department of Traditional Chinese Medicine, Nanfang Hospital (ZengCheng Branch), Southern Medical University, Guangzhou 510515, China
| | - Guiqiong Huang
- Department of Internal Medicine, Huizhou Hospital of Guangzhou University of Traditional Chinese Medicine, Huizhou 516000, China
| | - Huijie Fan
- TCM Health Construction Department of Yangjiang People's Hospital, Guangdong Province, Yangjiang 529500, China
| | - Lei Gao
- School of Traditional Chinese Medicine, Department of Traditional Chinese Medicine, Nanfang Hospital (ZengCheng Branch), Southern Medical University, Guangzhou 510515, China.
| | - Bin Liu
- Department of Traditional Chinese Medicine (Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, State Key Laboratory of Respiratory Disease), the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 510260, China.
| | - Yingchun Zhou
- School of Traditional Chinese Medicine, Department of Traditional Chinese Medicine, Nanfang Hospital (ZengCheng Branch), Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
78
|
Silencing Cardiac Troponin I-Interacting Kinase Reduces Lipopolysaccharide-Induced Sepsis-Induced Myocardial Dysfunction in Rat by Regulating Apoptosis-Related Proteins. BIOMED RESEARCH INTERNATIONAL 2021; 2021:5520051. [PMID: 34136567 PMCID: PMC8175134 DOI: 10.1155/2021/5520051] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 05/13/2021] [Indexed: 11/18/2022]
Abstract
The aim of this study was to investigate the effect of cardiac troponin I-interacting kinase (TNNI3K) on sepsis-induced myocardial dysfunction (SIMD) and further explore the underlying molecular mechanisms. In this study, a lipopolysaccharide- (LPS-) induced myocardial injury model was used. qRT-PCR was performed to detect the mRNA expression of TNNI3K. Western blot was conducted to quantitatively detect the expression of TNNI3K and apoptosis-related proteins (Bcl-2, Bax, and caspase-3). ELISA was performed to detect the content of lactate dehydrogenase (LDH) and creatine kinase (CK). TUNEL assay was used to detect the apoptosis of H9C2 cells. In LPS-induced H9C2 cells, TNNI3K was up regulated. Besides, the CK activity, the content of LDH, and the apoptosis of H9C2 cells were significantly increased after treatment with LPS. Silencing TNNI3K decreased the LDH release activity and CK activity and inhibited apoptosis of H9C2 cell. Further research illustrated that si-TNNI3K promoted the protein expression of Bcl-2 and decreased the protein expression of Bax and cleaved caspase-3. The study concluded that TNNI3K was upregulated in LPS-induced H9C2 cells. Importantly, functional research findings indicated that silencing TNNI3K alleviated LPS-induced H9C2 cell injury by regulating apoptosis-related proteins.
Collapse
|
79
|
Early myocardial damage (EMD) and valvular dysfunction after femur fracture in pigs. Sci Rep 2021; 11:8503. [PMID: 33875675 PMCID: PMC8055677 DOI: 10.1038/s41598-021-86151-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 10/01/2020] [Indexed: 12/27/2022] Open
Abstract
Musculoskeletal injuries are the most common reason for surgery in severely injured patients. In addition to direct cardiac damage after physical trauma, there is rising evidence that trauma induces secondary cardiac structural and functional damage. Previous research associates hip fractures with the appearance of coronary heart disease: As 25% of elderly patients developed a major adverse cardiac event after hip fracture. 20 male pigs underwent femur fracture with operative stabilization via nailing (unreamed, reamed, RIA I and a new RIA II; each group n = 5). Blood samples were collected 6 h after trauma and the concentration of troponin I and heart-type fatty acid binding protein (HFABP) as biomarkers for EMD were measured. At baseline and 6 h after trauma, transesophageal ECHO (TOE) was performed; and invasive arterial and left ventricular blood pressure were measured to evaluate the cardiac function after femur fracture. A systemic elevation of troponin I and HFABP indicate an early myocardial damage after femur fracture in pigs. Furthermore, various changes in systolic (ejection fraction and cardiac output) and diastolic (left ventricular end-diastolic pressure, mitral valve deceleration time and E/A ratio) parameters illustrate the functional impairment of the heart. These findings were accompanied by the development of valvular dysfunction (pulmonary and tricuspid valve). To the best of our knowledge, we described for the first time the development of functional impairment of the heart in the context of EMD after long bone fracture in pigs. Next to troponin and HFABP elevation, alterations in the systolic and diastolic function occurred and were accompanied by pulmonary and tricuspid valvular insufficiency. Regarding EMD, none of the fracture stabilization techniques (unreamed nailing, reaming, RIA I and RIA II) was superior.
Collapse
|
80
|
Wang XT, Peng Z, An YY, Shang T, Xiao G, He S, Chen X, Zhang H, Wang Y, Wang T, Zhang JH, Gao X, Zhu Y, Feng Y. Paeoniflorin and Hydroxysafflor Yellow A in Xuebijing Injection Attenuate Sepsis-Induced Cardiac Dysfunction and Inhibit Proinflammatory Cytokine Production. Front Pharmacol 2021; 11:614024. [PMID: 33986658 PMCID: PMC8112230 DOI: 10.3389/fphar.2020.614024] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 12/07/2020] [Indexed: 12/15/2022] Open
Abstract
Sepsis-induced myocardial dysfunction is a major contributor to the poor outcomes of septic shock. As an add-on with conventional sepsis management for over 15 years, the effect of Xuebijing injection (XBJ) on the sepsis-induced myocardial dysfunction was not well understood. The material basis of Xuebijing injection (XBJ) in managing infections and infection-related complications remains to be defined. A murine cecal ligation and puncture (CLP) model and cardiomyocytes in vitro culture were adopted to study the influence of XBJ on infection-induced cardiac dysfunction. XBJ significantly improved the survival of septic-mice and rescued cardiac dysfunction in vivo. RNA-seq revealed XBJ attenuated the expression of proinflammatory cytokines and related signalings in the heart which was further confirmed on the mRNA and protein levels. Xuebijing also protected cardiomyocytes from LPS-induced mitochondrial calcium ion overload and reduced the LPS-induced ROS production in cardiomyocytes. The therapeutic effect of XBJ was mediated by the combination of paeoniflorin and hydroxysafflor yellow A (HSYA) (C0127-2). C0127-2 improved the survival of septic mice, protected their cardiac function and cardiomyocytes while balancing gene expression in cytokine-storm-related signalings, such as TNF-α and NF-κB. In summary, Paeoniflorin and HSYA are key active compounds in XBJ for managing sepsis, protecting cardiac function, and controlling inflammation in the cardiac tissue partially by limiting the production of IL-6, IL-1β, and CXCL2.
Collapse
Affiliation(s)
- Xin-Tong Wang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology and Medicine, Tianjin, China
| | - Zhen Peng
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology and Medicine, Tianjin, China
| | - Ying-Ying An
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology and Medicine, Tianjin, China
| | - Ting Shang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology and Medicine, Tianjin, China
| | - Guangxu Xiao
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology and Medicine, Tianjin, China
| | - Shuang He
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology and Medicine, Tianjin, China
| | - Xi Chen
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Han Zhang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuefei Wang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Tao Wang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jun-Hua Zhang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiumei Gao
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yan Zhu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology and Medicine, Tianjin, China
| | - Yuxin Feng
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology and Medicine, Tianjin, China
| |
Collapse
|
81
|
Wang Y, Zhai X, Zhu M, Pan Y, Yang M, Yu K, He B. Risk factors for postoperative sepsis-induced cardiomyopathy in patients undergoing general thoracic surgery: a single center experience. J Thorac Dis 2021; 13:2486-2494. [PMID: 34012595 PMCID: PMC8107539 DOI: 10.21037/jtd-21-492] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Background The current study aimed to investigate the incidence of sepsis-induced cardiomyopathy (SICM) in patients who received general thoracic surgery, along with the risk factors and management strategies for this complication. Methods The clinical records of 163 patients with postoperative sepsis were retrospectively reviewed. After propensity score matching, 144 patients were divided into 2 groups by stroke volume: the SICM group (n=72) and the non-SICM group (n=72). Results The overall incidence of postoperative SICM was 53.99%. Multiple logistic regression analysis showed that stroke volume and C-reactive protein were independent predictors of mortality in patients with postoperative sepsis. Statistical analysis by t-test and χ2 test indicated that mortality (P=0.000), B-type natriuretic peptide (P=0.001), left ventricular ejection fraction (P=0.000), the mitral peak velocity of early filling/early diastolic mitral annular velocity (E/e’) (P=0.049), C-reactive protein (P=0.016), procalcitonin (P=0.013), serum creatinine (P=0.016), platelets (P=0.028), and lactic acid (P=0.002) were significantly associated with the occurrence of postoperative SICM. Among these parameters, B-type natriuretic peptide was identified as the best biomarker for predicting SICM by receiver operating characteristic (ROC) curve analysis. Conclusions It is vital to improve the diagnosis and standard management of SICM. A combined strategy comprising early detection of suspected infection, adequate use of antibiotics, close monitoring, effective drainage, and supportive care may improve the outcomes of patients with postoperative SICM.
Collapse
Affiliation(s)
- Yinghua Wang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China.,Department of Intensive Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xinming Zhai
- Department of Intensive Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Minfang Zhu
- Department of Intensive Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yan Pan
- Department of Pharmacy, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Min Yang
- Department of Intensive Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Kaiyan Yu
- Department of Intensive Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Ben He
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
82
|
Adams JA, Lopez JR, Uryash A, Sackner MA. Whole body periodic acceleration (pGz) improves endotoxin induced cardiomyocyte contractile dysfunction and attenuates the inflammatory response in mice. Heliyon 2021; 7:e06444. [PMID: 33748496 PMCID: PMC7970274 DOI: 10.1016/j.heliyon.2021.e06444] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 11/10/2020] [Accepted: 03/04/2021] [Indexed: 11/01/2022] Open
Abstract
Sepsis-induces myocardial contractile dysfunction. We previously showed that whole body periodic acceleration (pGz), the sinusoidal motion of the supine body head-foot ward direction significantly improves survival and decreases microvascular permeability in a lethal model of sepsis. We tested the hypothesis that pGz improves LPS induced cardiomyocyte contractile dysfunction and decreases LPS pro-inflammatory cytokine response when applied pre- or post-treatment. Isolated cardiomyocytes were obtained from mice that received LPS who had been pre-treated with pGz for three days (pGz-LPS) or control. Peak shortening (PS), maximal velocity of shortening (+dL/dt), and relengthening (-dL/dt) as well as diastolic intracellular calcium concentration ([Ca+2]d), sodium ([Na+]d), reactive oxygen species (ROS), and cardiac troponin (cTnT) production were measured. LPS decreased PS, +dL/dt, and -dL/dt, by 37%, 41% and 35% change respectively (p < 0.01), increased [Ca+2]d, [Na+]d, ROS, and cTnT by 343%, 122%, 298%, and 610% change respectively (p < 0.01) compared to control. pGz pre-treatment attenuated the parameters mentioned above. In a separate cohort, the effects of a lethal dose of LPS on protein expression of nitric oxide synthases (iNOS, eNOS, nNOS), pro- and anti-inflammatory cytokines in hearts of mice was studied in pre-treated with pGz for three days prior to LPS (pGz-LPS) and post-treated with pGz 30 min after LPS (LPS-pGz) were determined. LPS increased expression of early and late iNOS and decreased expression of eNOS, phosphorylated eNOS (p-eNOS), and nNOS. Both pre- and post-treatment with pGz markedly reduced early and late pro-inflammatory surge. Therefore, pre- and post-treatment with pGz improves LPS-induced cardiomyocyte dysfunction, decreases iNOS expression, and increases cytoprotective eNOS and nNOS, with decreased pro-inflammatory response. Such results have potential for translation to benefit outcomes in human sepsis.
Collapse
Affiliation(s)
- Jose A Adams
- Division of Neonatology, Mount Sinai Medical Center, Miami Beach, FL, USA
| | - Jose R Lopez
- Department of Research, Mount Sinai Medical Center, Miami Beach, FL, USA
| | - Arkady Uryash
- Division of Neonatology, Mount Sinai Medical Center, Miami Beach, FL, USA
| | - Marvin A Sackner
- Department of Medicine, Mount Sinai Medical Center, Miami Beach, FL, USA
| |
Collapse
|
83
|
Sun F, Chen G, Yang Y, Lei M. Fatty acid-binding protein 4 silencing protects against lipopolysaccharide-induced cardiomyocyte hypertrophy and apoptosis by inhibiting the Toll-like receptor 4-nuclear factor-κB pathway. J Int Med Res 2021; 49:300060521998233. [PMID: 33719658 PMCID: PMC7952852 DOI: 10.1177/0300060521998233] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Objective To explore the effects and potential mechanisms of fatty acid-binding protein 4 (FABP4) in a lipopolysaccharide (LPS)-induced in vitro septic cardiomyopathy model. Methods Rat cardiomyocyte H9c2 cells were transfected with small interfering RNA (siRNA) against FABP4 (siFABP4), then induced with LPS. The following parameters were measured: cell viability, lactate dehydrogenase release, cardiac hypertrophy and related marker expression, apoptosis, inflammatory cytokine release and expression, and the activation of Toll-like receptor 4 (TLR4) and nuclear factor-κB (NF-κB) pathways. Results LPS increased the mRNA and protein expression of FABP4 in H9c2 cells. FABP4 silencing by siFABP4 significantly inhibited LPS-induced cardiac hypertrophy and reduced the mRNA expression of the myocardial hypertrophy markers atrial natriuretic peptide and brain natriuretic peptide. siFABP4 also attenuated LPS-induced increase in TUNEL-positive apoptotic cells, caspase-3 and caspase-9 activities, and the release and expression of proinflammatory cytokines. Mechanistically, we found that FABP4 silencing inhibited the mRNA and protein expression of TLR4 and suppressed the NF-kappa B signaling pathway, as evidenced by reduced nuclear NF-κB p65 and increased cytoplasmic I-κBα expression in LPS-stimulated H9c2 cells. Conclusion FABP4 silencing reduces LPS-induced cardiomyocyte hypertrophy and apoptosis by down-regulating the TLR4/NF-κB axis.
Collapse
Affiliation(s)
- Fangyuan Sun
- Department of Intensive Care Medicine, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, P.R. China
| | - Gang Chen
- Department of Intensive Care Medicine, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, P.R. China
| | - Yingyao Yang
- Yueyang Hospital of Integrated Chinese and Western Medicine, Shanghai University of TCM, Shanghai, P.R. China
| | - Ming Lei
- Department of Intensive Care Medicine, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, P.R. China
| |
Collapse
|
84
|
Montealegre F, Lyons BM. Fluid Therapy in Dogs and Cats With Sepsis. Front Vet Sci 2021; 8:622127. [PMID: 33718468 PMCID: PMC7947228 DOI: 10.3389/fvets.2021.622127] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 02/03/2021] [Indexed: 01/20/2023] Open
Abstract
Sepsis is currently defined as life-threatening organ dysfunction caused by a dysregulated host response to infection. Sepsis may occur secondary to infection anywhere in the body, and its pathogenesis is complex and not yet fully understood. Variations in the host immune response result in diverse clinical manifestations, which complicates clinical recognition and fluid therapy both in humans and veterinary species. Septic shock is a subset of sepsis in which particularly profound circulatory, cellular, and metabolic abnormalities are associated with a greater risk of mortality than with sepsis alone. Although septic shock is a form of distributive shock, septic patients frequently present with hypovolemic and cardiogenic shock as well, further complicating fluid therapy decisions. The goals of this review are to discuss the clinical recognition of sepsis in dogs and cats, the basic mechanisms of its pathogenesis as it affects hemodynamic function, and considerations for fluid therapy. Important pathophysiologic changes, such as cellular interaction, microvascular alterations, damage to the endothelial glycocalyx, hypoalbuminemia, and immune paralysis will be also reviewed. The advantages and disadvantages of treatment with crystalloids, natural and synthetic colloids, and blood products will be discussed. Current recommendations for evaluating fluid responsiveness and the timing of vasopressor therapy will also be considered. Where available, the veterinary literature will be used to guide recommendations.
Collapse
Affiliation(s)
- Federico Montealegre
- Department of Medical and Scientific Affairs, Nova Biomedical, Waltham, MA, United States
| | - Bridget M Lyons
- Department of Veterinary Medicine and Surgery, University of Missouri, Columbia, MO, United States
| |
Collapse
|
85
|
Liu Y, Liu L, Zhang J. Protective role of matrine in sepsis-associated cardiac dysfunction through regulating the lncRNA PTENP1/miR-106b-5p axis. Biomed Pharmacother 2021; 134:111112. [PMID: 33341669 DOI: 10.1016/j.biopha.2020.111112] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 11/16/2020] [Accepted: 12/04/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Matrine has attractive cardioprotective effects in some diseases. This study aimed to evaluate the therapeutic potential of matrine against cardiac dysfunction induced by sepsis in vivo and in vitro, and further explore the related mechanisms. METHODS Cecal ligation and puncture (CLP) was used to induce a sepsis mice model, and H9C2 cells treated with lipopolysaccharide (LPS) were used as a cardiac myoblast injury model. The evaluation of cardiac function of mice was performed by measuring cardiac function biomarker levels and hemodynamic indicators. An ELISA method was used to examine inflammatory cytokine levels. H9C2 cell viability was measured using MTT assay. The expression of non-coding RNAs that might be involved in matrine function was analyzed using real-time quantitative PCR. RESULTS Matrine could significantly improve the cardiac function and attenuate the inflammatory response of the mice model, and could increase H9C2 viability and inhibit inflammation in the cell model. By matrine administration, the expression of PTENP1 was downregulated, but miR-106b-5p expression was upregulated both in vivo and in vitro. The cardioprotective effects of matrine in mice and cell models could be reversed by the overexpression of PTENP1 or the knockdown of miR-106b-5p, and the overexpression of miR-106b-5p could significantly abolish the effects of PTENP1 on cardiac function and inflammation. CONCLUSION All the data revealed that matrine can alleviate sepsis-related cardiac dysfunction by enhancing cardiac myoblast viability and attenuating inflammatory responses through the PTENP1/miR-106b-5p axis.
Collapse
Affiliation(s)
- Yujuan Liu
- Central Supply Room, Weifang Maternal and Child Health Hospital, Weifang, Shandong, 261011, China
| | - Lijun Liu
- Department of Gynecology, Weifang Maternal and Child Health Hospital, Weifang, Shandong, 261011, China
| | - Jun Zhang
- Department of Pharmacy, Weifang Maternal and Child Health Hospital, Weifang, Shandong, 261011, China.
| |
Collapse
|
86
|
Sun Y, Cai Y, Qian S, Chiou H, Zang QS. Beclin-1 improves mitochondria-associated membranes in the heart during endotoxemia. FASEB Bioadv 2021; 3:123-135. [PMID: 33733054 PMCID: PMC7944875 DOI: 10.1096/fba.2020-00039] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 08/13/2020] [Indexed: 01/24/2023] Open
Abstract
Mitochondria‐associated membranes (MAMs) are essential to mitochondria. This study was to determine whether endotoxemia rearranges MAMs in the heart, and whether Beclin‐1 regulates this process. Wild‐type mice and mice with a cardiac‐specific overexpression of Beclin‐1 (Becn1‐Tg), or a heterozygous knockout of Beclin‐1 (Becn1+/−) were given lipopolysaccharide (LPS) challenge. In the heart, the ultrastructure of MAMs was examined by electron microscopy and the histology evaluated by immunostaining. Additionally, MAMs were isolated by ultracentrifugation, and their content and function were quantified. The effects of Beclin‐1‐activating peptide (TB‐peptide) on MAMs were also examined. Data showed that endotoxemia decreased both the total mass and the function of MAMs, and these deficiencies became worse in Becn1+/− mice but were alleviated in Becn1‐Tg and TB‐peptide‐treated mice. Responses of myocardial MAMs to LPS and to TB‐peptide were additionally examined in AC16 human cardiomyocytes. In vitro findings recaptured the effects of LPS and TB‐peptide in cardiomyocytes; the challenge of LPS reduced the level and activity of MAMs, and TB‐peptide attenuated this defect. Together, the results suggest a new function of Beclin‐1 in improving cardiac MAMs during endotoxemia, providing a mechanism for the previously identified role of Beclin‐1 in protection of mitochondria and cardiac function.
Collapse
Affiliation(s)
- Yuxiao Sun
- Department of Surgery University of Texas Southwestern Medical Center Dallas TX USA
| | - Ying Cai
- Department of Surgery University of Texas Southwestern Medical Center Dallas TX USA.,Department of Developmental Cell Biology Key Laboratory of Cell Biology China Medical University Shenyang Liaoning Province P. R. China
| | - Suhong Qian
- Department of Surgery University of Texas Southwestern Medical Center Dallas TX USA
| | - Hellen Chiou
- Department of Surgery University of Texas Southwestern Medical Center Dallas TX USA
| | - Qun S Zang
- Department of Surgery University of Texas Southwestern Medical Center Dallas TX USA.,Department of Surgery, Burn and Shock Trauma Research Institute Stritch School of Medicine Loyola University Chicago Health Sciences Division Maywood IL USA
| |
Collapse
|
87
|
Xiao FP, Chen MY, Wang L, He H, Jia ZQ, Kuai L, Zhou HB, Liu M, Hong M. Outcomes of new-onset atrial fibrillation in patients with sepsis: A systematic review and meta-analysis of 225,841 patients. Am J Emerg Med 2021; 42:23-30. [PMID: 33429188 DOI: 10.1016/j.ajem.2020.12.062] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 12/15/2020] [Accepted: 12/20/2020] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND The outcomes of new-onset atrial fibrillation (AF) during sepsis are inconsistent and inconclusive. This meta-analysis aims to provide a comprehensive description of the impact of new-onset AF on the prognosis of sepsis. METHODS Three electronic databases (PubMed, Embase, and the Cochrane Library) were searched for relevant studies. Meta-analysis was performed using odds ratios (OR) and 95% confidence intervals (CI) as effect measures. RESULTS A total of 225,841 patients from 13 individual studies were incorporated to the meta-analysis. The summary results revealed that new-onset AF during sepsis was associated with increased odds of in-hospital mortality (pooled OR: 2.09; 95% CI: 1.53-2.86; p < 001), post-discharge mortality (pooled OR: 2.44; 95% CI: 1.81-3.29; p < .001), and stroke (pooled OR:1.88; 95% CI: 1.13-3.14; p < .05). Results also indicated that the incidence of new-onset AF varied from 1.9% for mild sepsis to 46.0% for septic shock. Furthermore, compared to those without AF, people with new-onset AF had longer ICU and hospital stays, as well as a higher recurrence of AF. CONCLUSIONS New-onset AF is frequently associated with adverse outcomes in patients with sepsis. This is a clinical issue that warrants more attention and should be managed appropriately to prevent poor prognosis.
Collapse
Affiliation(s)
- Fang-Ping Xiao
- Department of Cardiology, Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ming-Yue Chen
- Department of Geriatrics, Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Lei Wang
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Hao He
- Department of Cardiology, Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhi-Qiang Jia
- Department of Cardiology, Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Lin Kuai
- Department of Geriatrics, Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Hai-Bo Zhou
- Department of Cardiology, Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Meng Liu
- Department of Cardiology, Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Mei Hong
- Department of Cardiology, Second Affiliated Hospital of Nanjing Medical University, Nanjing, China; Department of Geriatrics, Second Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
88
|
Bilous T, Kretsu N, Koloskova O. Markers of myocardial dysfunction in neonates with sepsis of various gestational age. ACTA MEDICA INTERNATIONAL 2021. [DOI: 10.4103/amit.amit_53_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
89
|
Denning NL, Aziz M, Diao L, Prince JM, Wang P. Targeting the eCIRP/TREM-1 interaction with a small molecule inhibitor improves cardiac dysfunction in neonatal sepsis. Mol Med 2020; 26:121. [PMID: 33276725 PMCID: PMC7716442 DOI: 10.1186/s10020-020-00243-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 11/12/2020] [Indexed: 02/06/2023] Open
Abstract
Background Neonatal sepsis and the associated myocardial dysfunction remain a leading cause of infant mortality. Extracellular cold-inducible RNA-binding protein (eCIRP) acts as a ligand of triggering receptor expressed on myeloid cells-1 (TREM-1). M3 is a small CIRP-derived peptide that inhibits the eCIRP/TREM-1 interaction. We hypothesize that the eCIRP/TREM-1 interaction in cardiomyocytes contributes to sepsis-induced cardiac dysfunction in neonatal sepsis, while M3 is cardioprotective. Methods Serum was collected from neonates in the Neonatal Intensive Care Unit (NICU). 5–7-day old C57BL/6 mouse pups were used in this study. Primary murine neonatal cardiomyocytes were stimulated with recombinant murine (rm) CIRP with M3. TREM-1 mRNA and supernatant cytokine levels were assayed. Mitochondrial oxidative stress, ROS, and membrane potential were assayed. Neonatal mice were injected with rmCIRP and speckle-tracking echocardiography was conducted to measure cardiac strain. Sepsis was induced by i.p. cecal slurry. Mouse pups were treated with M3 or vehicle. After 16 h, echocardiography was performed followed by euthanasia for tissue analysis. A 7-day survival study was conducted. Results Serum eCIRP levels were elevated in septic human neonates. rmCIRP stimulation of cardiomyocytes increased TREM-1 gene expression. Stimulation of cardiomyocytes with rmCIRP upregulated TNF-α and IL-6 in the supernatants, while this upregulation was inhibited by M3. Stimulation of cardiomyocytes with rmCIRP resulted in a reduction in mitochondrial membrane potential (MMP) while M3 treatment returned MMP to near baseline. rmCIRP caused mitochondrial calcium overload; this was inhibited by M3. rmCIRP injection impaired longitudinal and radial cardiac strain. Sepsis resulted in cardiac dysfunction with a reduction in cardiac output and left ventricular end diastolic diameter. Both were improved by M3 treatment. Treatment with M3 attenuated serum, cardiac, and pulmonary levels of pro-inflammatory cytokines compared to vehicle-treated septic neonates. M3 dramatically increased sepsis survival. Conclusions Inhibition of eCIRP/TREM-1 interaction with M3 is cardioprotective, decreases inflammation, and improves survival in neonatal sepsis. Trial registration Retrospectively registered.
Collapse
Affiliation(s)
- Naomi-Liza Denning
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, 350 Community Dr, Manhasset, NY, 11030, USA.,Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, USA.,Department of Surgery, Donald and Barbara Zucker School of Medicine At Hofstra/Northwell, Hempstead, NY, USA
| | - Monowar Aziz
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, 350 Community Dr, Manhasset, NY, 11030, USA.,Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, USA
| | - Li Diao
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, 350 Community Dr, Manhasset, NY, 11030, USA
| | - Jose M Prince
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, 350 Community Dr, Manhasset, NY, 11030, USA.,Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, USA.,Department of Surgery, Donald and Barbara Zucker School of Medicine At Hofstra/Northwell, Hempstead, NY, USA.,Division of Pediatric Surgery, Cohen Children's Medical Center At Hofstra/Northwell, New Hyde Park, NY, USA
| | - Ping Wang
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, 350 Community Dr, Manhasset, NY, 11030, USA. .,Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, USA. .,Department of Surgery, Donald and Barbara Zucker School of Medicine At Hofstra/Northwell, Hempstead, NY, USA.
| |
Collapse
|
90
|
Mavraganis G, Aivalioti E, Chatzidou S, Patras R, Paraskevaidis I, Kanakakis I, Stamatelopoulos K, Dimopoulos MA. Cardiac arrest and drug-related cardiac toxicity in the Covid-19 era. Epidemiology, pathophysiology and management. Food Chem Toxicol 2020; 145:111742. [PMID: 32916218 PMCID: PMC7833119 DOI: 10.1016/j.fct.2020.111742] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Accepted: 08/17/2020] [Indexed: 02/07/2023]
Abstract
SARS-CoV-2 (Covid-19) infection has recently become a worldwide challenge with dramatic global economic and health consequences. As the pandemic is still spreading, new data concerning Covid-19 complications and related mechanisms become increasingly available. Accumulating data suggest that the incidence of cardiac arrest and its outcome are adversely affected during the Covid-19 period. This may be further exacerbated by drug-related cardiac toxicity of Covid-19 treatment regimens. Elucidating the underlying mechanisms that lead to Covid-19 associated cardiac arrest is imperative, not only in order to improve its effective management but also to maximize preventive measures. Herein we discuss available epidemiological data on cardiac arrest during the Covid-19 pandemic as well as possible associated causes and pathophysiological mechanisms and highlight gaps in evidence warranting further investigation. The risk of transmission during cardiopulmonary resuscitation (CPR) is also discussed in this review. Finally, we summarize currently recommended guidelines on CPR for Covid-19 patients including CPR in patients with cardiac arrest due to suspected drug-related cardiac toxicity in an effort to underscore the most important common points and discuss discrepancies proposed by established international societies.
Collapse
Affiliation(s)
- Georgios Mavraganis
- Department of Clinical Therapeutics, Alexandra Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Evmorfia Aivalioti
- Department of Clinical Therapeutics, Alexandra Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Sofia Chatzidou
- Department of Clinical Therapeutics, Alexandra Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Raphael Patras
- Department of Clinical Therapeutics, Alexandra Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Ioannis Paraskevaidis
- Department of Clinical Therapeutics, Alexandra Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Ioannis Kanakakis
- Department of Clinical Therapeutics, Alexandra Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Kimon Stamatelopoulos
- Department of Clinical Therapeutics, Alexandra Hospital, National and Kapodistrian University of Athens, Athens, Greece.
| | | |
Collapse
|
91
|
Lucci C, Cosentino N, Genovese S, Campodonico J, Milazzo V, De Metrio M, Rondinelli M, Riggio D, Biondi ML, Rubino M, Celentano K, Bonomi A, Capra N, Veglia F, Agostoni P, Bartorelli AL, Marenzi G. Prognostic impact of admission high-sensitivity C-reactive protein in acute myocardial infarction patients with and without diabetes mellitus. Cardiovasc Diabetol 2020; 19:183. [PMID: 33081810 PMCID: PMC7576820 DOI: 10.1186/s12933-020-01157-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 10/10/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND High-sensitivity C-reactive protein (hs-CRP) elevation frequently occurs in acute myocardial infarction (AMI) and is associated with adverse outcomes. Since diabetes mellitus (DM) is characterized by an underlying chronic inflammation, hs-CRP may have a different prognostic power in AMI patients with and without DM. METHODS We prospectively included 2064 AMI patients; hs-CRP was measured at hospital admission. Patients were grouped according to hs-CRP quartiles and DM status. The primary endpoint was a composite of in-hospital mortality, cardiogenic shock, and acute pulmonary edema. Two-year all-cause mortality was the secondary endpoint. RESULTS Twenty-six percent (n = 548) of patients had DM and they had higher hs-CRP levels than non-DM patients (5.32 vs. 3.24 mg/L; P < 0.0001). The primary endpoint incidence in the overall population (7%, 9%, 13%, 22%; P for trend < 0.0001), in DM (14%, 9%, 21%, 27%; P = 0.0001), and non-DM (5%, 8%, 10%, 19%; P < 0.0001) patients increased in parallel with hs-CRP quartiles. The adjusted risk of the primary endpoint increased in parallel with hs-CRP quartiles in DM and non-DM patients but this relationship was less evident in DM patients. In the overall population, the adjusted OR of the primary endpoint associated with an hs-CRP value ≥ 2 mg/L was 2.10 (95% CI 1.46-3.00). For the same risk, hs-CRP was 7 and 2 mg/L in patients with and without DM. A similar behavior was observed for the secondary endpoint when the HR associated with an hs-CRP value ≥ 2 mg/L found in the overall population was 2.25 (95% CI 1.57-3.22). For the same risk, hs-CRP was 8 and 1.5 mg/L in DM and non-DM patients. CONCLUSIONS This study shows that hs-CRP predicts in-hospital outcome and two-year mortality in AMI patients with and without DM. However, in DM patients, the same risk of developing events as in non-DM patients is associated to higher hs-CRP levels.
Collapse
Affiliation(s)
- Claudia Lucci
- Centro Cardiologico Monzino IRCCS, Via Parea 4, Milan, 20138, Italy
| | - Nicola Cosentino
- Centro Cardiologico Monzino IRCCS, Via Parea 4, Milan, 20138, Italy
| | - Stefano Genovese
- Centro Cardiologico Monzino IRCCS, Via Parea 4, Milan, 20138, Italy
| | | | | | - Monica De Metrio
- Centro Cardiologico Monzino IRCCS, Via Parea 4, Milan, 20138, Italy
| | | | - Daniela Riggio
- Centro Cardiologico Monzino IRCCS, Via Parea 4, Milan, 20138, Italy
| | | | - Mara Rubino
- Centro Cardiologico Monzino IRCCS, Via Parea 4, Milan, 20138, Italy
| | - Katia Celentano
- Centro Cardiologico Monzino IRCCS, Via Parea 4, Milan, 20138, Italy
| | - Alice Bonomi
- Centro Cardiologico Monzino IRCCS, Via Parea 4, Milan, 20138, Italy
| | - Nicolò Capra
- Centro Cardiologico Monzino IRCCS, Via Parea 4, Milan, 20138, Italy
| | - Fabrizio Veglia
- Centro Cardiologico Monzino IRCCS, Via Parea 4, Milan, 20138, Italy
| | - Piergiuseppe Agostoni
- Centro Cardiologico Monzino IRCCS, Via Parea 4, Milan, 20138, Italy
- Department of Clinical Sciences and Community Health - Cardiovascular Section, University of Milan, Milan, Italy
| | - Antonio L Bartorelli
- Centro Cardiologico Monzino IRCCS, Via Parea 4, Milan, 20138, Italy
- Department of Biomedical and Clinical Sciences, "Luigi Sacco", University of Milan, Milan, Italy
| | - Giancarlo Marenzi
- Centro Cardiologico Monzino IRCCS, Via Parea 4, Milan, 20138, Italy.
| |
Collapse
|
92
|
Liu JJ, Li Y, Yang MS, Chen R, Cen CQ. SP1-induced ZFAS1 aggravates sepsis-induced cardiac dysfunction via miR-590-3p/NLRP3-mediated autophagy and pyroptosis. Arch Biochem Biophys 2020; 695:108611. [PMID: 33002446 DOI: 10.1016/j.abb.2020.108611] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 09/24/2020] [Accepted: 09/27/2020] [Indexed: 12/21/2022]
Abstract
BACKGROUND Sepsis-induced cardiac dysfunction is one of the leading complications of sepsis, contributing to the high morbidity and mortality of septic patients. Several lines of evidence have demonstrated that autophagy and pyroptosis may be involved in septic cardiac dysfunction. In this study, we examined the impact of zinc finger antisense 1 (ZFAS1) on sepsis-induced myocardial dysfunction via regulating pyroptosis and autophagy. METHOD Mice with cecal ligation and puncture (CLP)-induced sepsis was constructed in vivo. Myocardial injury was assessed by H&E staining, immunohistochemistry (IHC) for NLRP3, caspase 1, and interleukin (IL)-1β, as well as ELISA assay for serum levels of creatine kinase (CK), CK-MB, tumor necrosis factor α (TNF-α), and IL-1β. Primary cardiomyocytes exposed to lipopolysaccharide (LPS) were established to simulate sepsis-induced cardiac dysfunction in vitro. Cell viability was examined by MTT assay and concentration of TNF-α and IL-1β was measured by ELISA. Flow cytometry, immunofluorescent staining and western blotting were performed to assess pyroptosis and autophagy. The transcriptional regulation of SP1 on ZFAS1 was determined using ChIP assay. Luciferase reporter assay was performed to verify the ZFAS1/miR-590-3p interaction. Besides, activation of AMPK/mTOR signaling was detected using western blotting. RESULTS Highly expressed ZFAS1 was observed in sepsis-induced cardiac dysfunction in the in vivo and in vitro model. Knockdown of ZFAS1 robustly abolished LPS-induced pyroptosis and attenuated the inhibition of autophagy. SP1 was identified to be an essential transcription factor to positively regulate ZFAS1 expression. Moreover, miR-590-3p functioned as a downstream effector to reverse ZFAS1-mediated sepsis-induced cardiac dysfunction. AMPK/mTOR signaling was involved in miR-590-3p-regulated autophagy and pyroptosis of cardiomyocytes. Furthermore, the regulatory network of ZFAS1/miR-590-3p on AMPK/mTOR signaling was verified in vivo. CONCLUSION ZFAS1, activated by SP1, aggravates the progression of sepsis-induced cardiac dysfunction via targeting miR-590-3p/AMPK/mTOR signaling-mediated autophagy and pyroptosis of cardiomyocytes.
Collapse
Affiliation(s)
- Jing-Jing Liu
- Department of Intensive Care Medicine, The Third Xiangya Hospital of Central South University, Changsha, 410013, Hunan Province, PR China
| | - Yong Li
- Department of Emergency Medicine, The Third Xiangya Hospital of Central South University, Changsha, 410013, Hunan Province, PR China
| | - Ming-Shi Yang
- Department of Intensive Care Medicine, The Third Xiangya Hospital of Central South University, Changsha, 410013, Hunan Province, PR China
| | - Rui Chen
- Department of Orthopedics, The First Naval Hospital Southern Theater Command, Zhanjiang, 524000, Guangdong Province, PR China
| | - Chao-Qun Cen
- Department of Emergency Medicine, The Third Xiangya Hospital of Central South University, Changsha, 410013, Hunan Province, PR China.
| |
Collapse
|
93
|
Miocarditis por gram negativos. REVISTA COLOMBIANA DE CARDIOLOGÍA 2020. [DOI: 10.1016/j.rccar.2020.05.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
94
|
Overexpression of miR-150-5p Alleviates Apoptosis in Sepsis-Induced Myocardial Depression. BIOMED RESEARCH INTERNATIONAL 2020; 2020:3023186. [PMID: 32908879 PMCID: PMC7477614 DOI: 10.1155/2020/3023186] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 08/03/2020] [Accepted: 08/08/2020] [Indexed: 12/13/2022]
Abstract
Sepsis-induced myocardial depression has high mortality and is very common in intensive care units. Previous studies have found that microRNAs play an important role in regulating sepsis-induced myocardial depression. miR-150-5p is involved in many biological processes; however, the mechanism underlying its role in sepsis-induced myocardial depression is still unclear. In this study, we generated rat models of septic shock induced by lipopolysaccharide. Whole genomic RNA sequencing was performed on 12 left ventricles collected after LPS treatment to identify miRNAs. Most of the target genes of the differently expressed microRNAs were involved in apoptosis, according to Gene Ontology. We also observed apoptosis in the heart tissue and in H9C2 cardiomyocytes stimulated with lipopolysaccharide, indicating that cell apoptosis may be an important mechanism in sepsis-induced myocardial depression. Furthermore, the expression of miR-150-5p was reduced, and overexpression of miR-150-5p with mimics resulted in a decrease in apoptosis, decreased expression of cleaved caspase3 and Bax, and increased expression of Bcl-2. Additionally, after H9C2 cells were transfected with miR-150-5p mimics or an inhibitor, the expression of Akt2 decreased or increased, respectively. These findings suggest that miR-150-5p can alleviate apoptosis and may be a novel therapeutic target for sepsis-induced myocardial depression.
Collapse
|
95
|
MG53 Protects against Sepsis-Induced Myocardial Dysfunction by Upregulating Peroxisome Proliferator-Activated Receptor- α. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:7413693. [PMID: 32908637 PMCID: PMC7474382 DOI: 10.1155/2020/7413693] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 07/02/2020] [Accepted: 07/10/2020] [Indexed: 12/20/2022]
Abstract
Background The heart is one of the most commonly affected organs during sepsis. Mitsugumin-53 (MG53) has attracted attention in research due to its cardioprotective function. However, the role of MG53 in sepsis-induced myocardial dysfunction (SIMD) remains unknown. The purpose of this study was to explore the underlying mechanism of MG53 in SIMD and investigate its potential relationship with peroxisome proliferator-activated receptor-α (PPARα). Methods The cecal ligation and puncture (CLP) model was created to induce SIMD in rats. Protein levels of MG53 and PPARα, cardiac function, cardiomyocyte injury, myocardial oxidative stress and inflammatory indicators, and cardiomyocyte apoptosis were measured at 18 h after CLP. The effects of MG53 on PPARα in SIMD were investigated via preconditioning recombinant human MG53 (rhMG53) and PPARα antagonist GW6471. Results The expression of MG53 and PPARα sharply decreased in the myocardium at 18 h after CLP. Compared with the sham group, cardiac function was significantly depressed, which was associated with the destructed myocardium, upregulated oxidative stress indicators and proinflammatory cytokines, and excessive cardiomyocyte apoptosis in the CLP group. Supplementation with rhMG53 enhanced myocardial MG53, increased the survival rate with improved cardiac function, and reduced oxidative stress, inflammation, and myocardial apoptosis, which were associated with PPARα upregulation. Pretreatment with GW6471 abolished the abovementioned protective effects induced by MG53. Conclusions Both MG53 and PPARα were downregulated after sepsis shock. MG53 supplement protects the heart against SIMD by upregulating PPARα expression. Our results provide a new treatment strategy for SIMD.
Collapse
|
96
|
Jin Y, Wang H, Li J, Dang M, Zhang W, Lei Y, Zhao H. Exploring the beneficial role of telmisartan in sepsis-induced myocardial injury through inhibition of high-mobility group box 1 and glycogen synthase kinase-3β/nuclear factor-κB pathway. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2020; 24:311-317. [PMID: 32587125 PMCID: PMC7317178 DOI: 10.4196/kjpp.2020.24.4.311] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 01/31/2020] [Indexed: 02/02/2023]
Abstract
In the present experimental study, cecal ligation and puncture significantly increased the myocardial injury assessed in terms of excess release of creative kinase-MB (CK-MB), cardiac troponin I (cTnI), interleukin (IL)-6 and decrease of IL-10 in the blood following 12 h of laparotomy procedure as compared to normal control. Also, a significant increase in protein expression levels of high-mobility group box 1 (HMGB1) and decreased phosphorylation of glycogen synthase kinase-3β (GSK-3β) was observed in the myocardial tissue as compared to normal control. A single independent administration of telmisartan (2 and 4 mg/kg) and AR-A014418 (1 and 2 mg/kg) substantially reduced sepsis-induced myocardial injury in terms of decrease levels of CK-MB, cTnI and IL-6, HMGB1, GSK-3β and increase in IL-10 and p-GSK-3β in the blood in sepsis- subjected rats. The effects of telmisartan at dose 4 mg/kg and AR-A014418 at a dose of 2 mg/kg were significantly higher than the telmisartan at a dose of 2 mg/kg and AR-A014418 1 mg/kg respectively. Further, no significant effects on different parameters were observed in the sham control group in comparison to normal. Therefore it is plausible to suggest that sepsis may increase the levels of angiotensin II to trigger GSK-3β-dependent signaling to activate the HMGB1/receptors for advanced glycation end products, which may promote inflammation and myocardial injury in sepsis-subjected rats.
Collapse
Affiliation(s)
- Yan Jin
- Emergency Department, Second Affiliated Hospital of Dalian Medical University Dalian, Jinan, Shandong 116027, P.R. China
| | - Hong Wang
- Emergency Department, Second Affiliated Hospital of Dalian Medical University Dalian, Jinan, Shandong 116027, P.R. China
| | - Jing Li
- Department of Cardiology, Hospital Affiliated to Shenyang Medical College, Shenyang, Liaoning 110021, P.R. China
| | - Minyan Dang
- Innoscience Research Sdn Bhd, Subang Jaya, Selangor 47650, Malaysia
| | - Wenzhi Zhang
- Innoscience Research Sdn Bhd, Subang Jaya, Selangor 47650, Malaysia
| | - Yan Lei
- Innoscience Research Sdn Bhd, Subang Jaya, Selangor 47650, Malaysia
| | - Hao Zhao
- mergency Department, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250014, P.R. China
| |
Collapse
|
97
|
Xu Y, Zhang S, Rong J, Lin Y, Du L, Wang Y, Zhang Z. Sirt3 is a novel target to treat sepsis induced myocardial dysfunction by acetylated modulation of critical enzymes within cardiac tricarboxylic acid cycle. Pharmacol Res 2020; 159:104887. [PMID: 32526680 DOI: 10.1016/j.phrs.2020.104887] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 04/29/2020] [Accepted: 05/01/2020] [Indexed: 12/16/2022]
Abstract
Sepsis induced myocardial dysfunction (SIMD) results in high morbidity and mortality. However, the effective therapeutic strategies for SIMD treatment remain limited. Sirt3 is the main mitochondrial Sirtuin member and is a key modulator of mitochondrial metabolism and function. In this study, we aimed to investigate the effect and mechanism of Sirt3 on SIMD. SIMD was induced by 20 mg/kg Lipopolysaccharides (LPS) injection for 6 h in mice. Sepsis could induce the reduction of cardiac Sirt3 expression and global deficiency of Sirt3 exacerbated cardiac function. Quantitative acetyl-proteomics and cardiac metabolomics analysis revealed that loss of Sirt3 led to hyper-acetylation of critical enzymes within cardiac tricarboxylic acid (TCA) cycle and generation of lactate and NADH, subsequently promotion of cardiac dysfunction after sepsis. Additionally, to evaluate whether Emodin could be utilized as a potential Sirt3 modulator to treat SIMD, male wild type mice (WT mice) or global Sirt3 deficient mice (Sirt3-/- mice) were intraperitoneally injected with 40 mg/kg Emodin for 5 days followed by 20 mg/kg LPS administration for another 6 h and observed that exogenous administration of Emodin could attenuate myocardial dysfunction in septic WT mice. However, septic Sirt3-/- mice can not gain benefit on cardiac performance from Emodin infusion. In conclusion, this study presented the protective role of Sirt3 targeting SIMD, which may provide a potential novel approach to maintain normal cardiac performance after sepsis.
Collapse
Affiliation(s)
- Yinchuan Xu
- Department of Cardiology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Shujing Zhang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jiabing Rong
- Department of Cardiology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Yao Lin
- Department of Intensive Care Unit, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Linlin Du
- Department of Intensive Care Unit, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Yi Wang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.
| | - Zhaocai Zhang
- Department of Intensive Care Unit, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China.
| |
Collapse
|
98
|
Song MJ, Lee SH, Leem AY, Kim SY, Chung KS, Kim EY, Jung JY, Kang YA, Kim YS, Chang J, Park MS. Predictors and outcomes of sepsis-induced cardiomyopathy in critically ill patients. Acute Crit Care 2020; 35:67-76. [PMID: 32407613 PMCID: PMC7280797 DOI: 10.4266/acc.2020.00024] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 04/07/2020] [Indexed: 11/30/2022] Open
Abstract
Background Sepsis-induced cardiomyopathy (SIC) occurs frequently in critically ill patients, but the clinical features and prognostic impact of SIC on sepsis outcome remain controversial. Here, we investigated the predictors and outcomes of SIC. Methods Patients admitted to a single medical intensive care unit from June 2016 to September 2017 were retrospectively reviewed. SIC was diagnosed by ejection fraction (EF) <50% and ≥10% decrease in baseline EF that recovered within 2 weeks. Results In total, 342 patients with sepsis met the inclusion criteria, and 49 patients (14.3%) were diagnosed with SIC; the latter were compared with 259 patients whose EF was not deteriorated by sepsis (non-SIC). Low systolic blood pressure and increased left ventricular end-diastolic diameter (LVEDD) were identified as predictors of SIC. SIC and non-SIC patients did not differ significantly in terms of 28-day all-cause mortality (24.5% vs. 26.3%, P=0.936). Acute Physiology and Chronic Health Evaluation II (APACHE II; hazard ratio [HR], 1.10; 95% confidential interval [CI], 1.02 to 1.18; P=0.009) and delta neutrophil index (DNI; HR, 1.02; 95% CI, 1.00 to 1.08; P=0.026) were independent risk factors for 28-day mortality with SIC. DNI, APACHE II, and lactate were identified as risk factors for 28-day mortality in sepsis patients as a whole. Conclusions SIC was not associated with increased mortality compared to non-SIC. Low systolic blood pressure and increased LVEDD were predictors of SIC. High APACHE II score and elevated DNI, which reflect sepsis severity, predict 28-day all-cause mortality.
Collapse
|
99
|
Baranwal AK, Deepthi G, Rohit MK, Jayashree M, Angurana SK, Kumar-M P. Longitudinal Study of CPK-MB and Echocardiographic Measures of Myocardial Dysfunction in Pediatric Sepsis: Are Patients with Shock Different from Those without? Indian J Crit Care Med 2020; 24:109-115. [PMID: 32205942 PMCID: PMC7075059 DOI: 10.5005/jp-journals-10071-23340] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background Sepsis-induced myocardial dysfunction has implications on outcome. For lack of echocardiography in resource-limited settings, myocardial biomarkers may be an alternative monitoring tool. Objective This study was planned to explore the longitudinal behavior of creatine phosphokinase-MB (CPK-MB) in children with sepsis with and without shock, and its correlation with clinical and echocardiographic parameters over the first 10 days. Design Prospective observational study. Setting Tertiary care hospital in a lower-middle-income economy of South Asia. Patients Children (3 months to 12 years) with nonshock sepsis (NSS) (n = 40) and septic shock survivors (SSSs) (n = 40) after optimal resuscitation. Patients with catecholamine refractory shock, preexisting heart disease, and cardiorespiratory event within the past 1 month were excluded from the study. Measurements and main results Pediatric logistic organ dysfunction (PeLOD) score, vasoactive inotrope score (VIS), CPK-MB, and echocardiographic measures of myocardial function were recorded on days 1, 3, 7, and 10. Echocardiography was repeated at 1 month. Both groups were similar at baseline. The SSSs had higher CPK-MB (180 vs 53 IU/L; p < 0.001) and PeLOD score (2 ± 0.4 vs 11.7 ± 5.1, p < 0.001) on day 1 compared to the NSS children. More than half of the SSS and none of the NSS patients had myocardial dysfunction. Reduction in CPK-MB over 10 days correlated well with improvement in PeLOD (p < 0.01), VIS (p = 0.04), and echocardiographic measures of myocardial dysfunction (p < 0.05) among SSSs. At 1 month follow-up, all had normal echocardiography. Conclusion The SSSs had markedly elevated CPK-MB, and its fall paralleled the improvement in clinical status and myocardial dysfunctions. The CPK-MB could be a potential monitoring tool for septic cardiomyopathy in resource-limited settings. How to cite this article Baranwal AK, Deepthi G, Rohit MK, Jayashree M, Angurana SK, Kumar-M P. Longitudinal Study of CPK-MB and Echocardiographic Measures of Myocardial Dysfunction in Pediatric Sepsis: Are Patients with Shock Different from Those without? Indian J Crit Care Med 2020;24(2):109–115.
Collapse
Affiliation(s)
- Arun K Baranwal
- Division of Pediatric Critical Care and Pediatric Cardiac Care Unit, Advanced Pediatrics Center, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | | | - Manoj K Rohit
- Department of Cardiology, Advanced Cardiac Center, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Muralidharan Jayashree
- Division of Pediatric Critical Care, Advanced Pediatrics Center, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Suresh K Angurana
- Division of Pediatric Critical Care, Advanced Pediatrics Center, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Praveen Kumar-M
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
100
|
Di S, Wang Z, Hu W, Yan X, Ma Z, Li X, Li W, Gao J. The Protective Effects of Melatonin Against LPS-Induced Septic Myocardial Injury: A Potential Role of AMPK-Mediated Autophagy. Front Endocrinol (Lausanne) 2020; 11:162. [PMID: 32373063 PMCID: PMC7176935 DOI: 10.3389/fendo.2020.00162] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 03/09/2020] [Indexed: 12/12/2022] Open
Abstract
Aim: Melatonin is an indolamine secreted by the pineal gland, as well as most of the organs and tissues. In addition to regulating circadian biology, studies have confirmed the multiple pharmacological effects of melatonin. Melatonin provides a strong defense against septic myocardial injury. However, the underlying mechanism has not been fully described. In this study, we investigated the protective effects of melatonin against lipopolysaccharide (LPS)-induced myocardial injury as well as the mechanisms involved. Methods: Mice were intraperitoneally injected with LPS to induce a septic myocardial injury model or an LPS shock model, depending on the dose of LPS. Melatonin was given (20 mg/kg/day, via intraperitoneal injection) for a week prior to LPS insult. 6 h after LPS injection, echocardiographic analysis, TUNEL staining, transmission electron microscopy (TEM), western blot, quantitative real-time PCR and ELISA were used to investigate the protective effects of melatonin against LPS induced myocardial injury. AMPK inhibitor, autophagy activator and inhibitor, siRNAs were used for further validation. Results: Survival test showed that melatonin significantly increased the survival rate after LPS-induced shock. In the sepsis model, melatonin markedly ameliorated myocardial dysfunction, decreased the release of inflammatory cytokines, activated AMP-activated protein kinase (AMPK), improved mitochondrial function, and activated autophagy. To confirm whether the protection of melatonin was mediated by AMPK and autophagy, Compound C, an AMPK inhibitor; 3-MA, an autophagy inhibitor; and Rapamycin (Rapa), an autophagy activator, were used in this study. AMPK inhibition down-regulated autophagy, abolished protection of melatonin, as indicated by significantly decreased cardiac function, increased inflammation and damaged mitochondrial function. Furthermore, autophagy inhibition by 3-MA significantly impaired the protective effects of melatonin, whereas autophagy activation by Rapa reversed LPS + Compound C induced myocardial injury. In addition, in vitro studies further confirmed the protection of melatonin against LPS-induced myocardial injury and the mechanisms involving AMPK-mediated autophagy signaling. Conclusions: In summary, our results demonstrated that melatonin protects against LPS-induced septic myocardial injury by activating AMPK mediated autophagy pathway.
Collapse
Affiliation(s)
- Shouyin Di
- Department of Geriatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
- Department of Thoracic Surgery, Sixth Medical Center of PLA General Hospital, Beijing, China
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Zheng Wang
- Department of Cardiothoracic Surgery, Central Theater Command General Hospital of Chinese People's Liberation Army, Wuhan, China
| | - Wei Hu
- Department of Immunology, The Fourth Military Medical University, Xi'an, China
| | - Xiaolong Yan
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Zhiqiang Ma
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Xiaofei Li
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Weimiao Li
- Department of Oncology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- *Correspondence: Weimiao Li
| | - Jianyuan Gao
- Department of Geriatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
- Jianyuan Gao
| |
Collapse
|