1
|
Hou X, Hu G, Wang H, Yang Y, Sun Q, Bai X. Inhibition of Egr2 Protects against TAC-induced Heart Failure in Mice by Suppressing Inflammation and Apoptosis Via Targeting Acot1 in Cardiomyocytes. J Cardiovasc Transl Res 2025:10.1007/s12265-025-10602-5. [PMID: 40095198 DOI: 10.1007/s12265-025-10602-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Accepted: 02/26/2025] [Indexed: 03/19/2025]
Abstract
Heart failure (HF) is a clinical syndrome caused by structural or functional abnormalities in heart. Egr2 has been reported to be protective for multiple diseases, but its effect on HF remains unknown. The present study intended to investigate the potential role of Egr2 in HF and its possible downstream effectors. High Egr2 expression in heart was observed in HF mice. Egr2 knockdown alleviated cardiac damage and function in HF mice. Egr2 knockdown inhibited myocardial inflammation and apoptosis both in vivo and in vitro. Egr2 inhibited Acot1 transcription expression via directly binding to its promoter. Acot1 overexpression reduced Lipopolysaccharide (LPS)-induced cardiomyocyte inflammation and apoptosis. Functional rescue experiments revealed that Acot1 reversed the effects of Egr2 on LPS-induced cell apoptosis and inflammation. Overall, Egr2 knockdown might ameliorate HF by inhibiting inflammation and apoptosis in cardiomyocytes by targeting Acot1. This study might provide evidence to better understand the molecular mechanisms of HF pathogenesis.
Collapse
Affiliation(s)
- Xiaolu Hou
- Department of Cardiology, The Fourth Hospital of Harbin Medical University, 37 Yiyuan Street, Nangang District, Harbin, 150001, China
| | - Guoling Hu
- Department of Geratology, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Heling Wang
- Department of Cardiology, Langfang Changzheng Hospital, Langfang, China
| | - Ying Yang
- Department of Cardiology, Harbin 242 Hospital, Harbin, China
| | - Qi Sun
- Department of Cardiology, Beidahuang Group General Hospital, Harbin, China
| | - Xiuping Bai
- Department of Cardiology, The Fourth Hospital of Harbin Medical University, 37 Yiyuan Street, Nangang District, Harbin, 150001, China.
| |
Collapse
|
2
|
Fonseka O, Gare SR, Chen X, Zhang J, Alatawi NH, Ross C, Liu W. Molecular Mechanisms Underlying Heart Failure and Their Therapeutic Potential. Cells 2025; 14:324. [PMID: 40072053 PMCID: PMC11899429 DOI: 10.3390/cells14050324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 02/07/2025] [Accepted: 02/17/2025] [Indexed: 03/15/2025] Open
Abstract
Heart failure (HF) is a prominent fatal cardiovascular disorder afflicting 3.4% of the adult population despite the advancement of treatment options. Therefore, a better understanding of the pathogenesis of HF is essential for exploring novel therapeutic strategies. Hypertrophy and fibrosis are significant characteristics of pathological cardiac remodeling, contributing to HF. The mechanisms involved in the development of cardiac remodeling and consequent HF are multifactorial, and in this review, the key underlying mechanisms are discussed. These have been divided into the following categories thusly: (i) mitochondrial dysfunction, including defective dynamics, energy production, and oxidative stress; (ii) cardiac lipotoxicity; (iii) maladaptive endoplasmic reticulum (ER) stress; (iv) impaired autophagy; (v) cardiac inflammatory responses; (vi) programmed cell death, including apoptosis, pyroptosis, and ferroptosis; (vii) endothelial dysfunction; and (viii) defective cardiac contractility. Preclinical data suggest that there is merit in targeting the identified pathways; however, their clinical implications and outcomes regarding treating HF need further investigation in the future. Herein, we introduce the molecular mechanisms pivotal in the onset and progression of HF, as well as compounds targeting the related mechanisms and their therapeutic potential in preventing or rescuing HF. This, therefore, offers an avenue for the design and discovery of novel therapies for the treatment of HF.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Wei Liu
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK; (O.F.); (S.R.G.); (X.C.); (J.Z.); (N.H.A.)
| |
Collapse
|
3
|
Jia Y, Liu Y, Zuo Y, Zhang J, Li Y, Liu X, Lv S. The Potential Therapeutic Prospect of PANoptosis in Heart Failure. J Inflamm Res 2024; 17:9147-9168. [PMID: 39583864 PMCID: PMC11585275 DOI: 10.2147/jir.s485901] [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: 08/24/2024] [Accepted: 11/15/2024] [Indexed: 11/26/2024] Open
Abstract
Heart failure (HF) represents a serious manifestation or advanced stage of various cardiac diseases. HF continues to impose a significant global disease burden, characterized by high rates of hospitalization and fatality. Furthermore, the pathogenesis and pathophysiological processes underlying HF remain incompletely understood, complicating its prevention and treatment strategies. One significant pathophysiological mechanism associated with HF is the systemic inflammatory response. PANoptosis, a novel mode of inflammatory cell death, has been extensively studied in the context of infectious diseases, neurodegenerative disorders, cancers, and other inflammatory conditions. Recent investigations have revealed that PANoptosis-related genes are markedly dysregulated in HF specimens. Consequently, the PANoptosis-mediated inflammatory response may represent a potential mechanism and therapeutic target for HF. This paper conducts a comprehensive analysis of the molecular pathways that drive PANoptosis. We discuss its role and potential therapeutic targets in HF, thereby providing valuable insights for clinical treatment and the development of novel therapies.
Collapse
Affiliation(s)
- Yunfeng Jia
- Department of Geriatrics, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine (National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion), Tianjin, 300381, People’s Republic of China
| | - Yayi Liu
- Department of Geriatrics, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine (National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion), Tianjin, 300381, People’s Republic of China
| | - Yiming Zuo
- Department of Geriatrics, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine (National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion), Tianjin, 300381, People’s Republic of China
| | - Junping Zhang
- Department of Geriatrics, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine (National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion), Tianjin, 300381, People’s Republic of China
| | - Yanyang Li
- Department of Integrated Traditional and Western Medicine, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, People’s Republic of China
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, People’s Republic of China
| | - Xuezheng Liu
- Department of Geriatrics, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine (National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion), Tianjin, 300381, People’s Republic of China
| | - Shichao Lv
- Department of Geriatrics, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine (National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion), Tianjin, 300381, People’s Republic of China
| |
Collapse
|
4
|
Yerra VG, Connelly KA. Extrarenal Benefits of SGLT2 Inhibitors in the Treatment of Cardiomyopathies. Physiology (Bethesda) 2024; 39:0. [PMID: 38888433 DOI: 10.1152/physiol.00008.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 06/05/2024] [Accepted: 06/12/2024] [Indexed: 06/20/2024] Open
Abstract
Sodium-glucose cotransporter 2 (SGLT2) inhibitors have emerged as pivotal medications for heart failure, demonstrating remarkable cardiovascular benefits extending beyond their glucose-lowering effects. The unexpected cardiovascular advantages have intrigued and prompted the scientific community to delve into the mechanistic underpinnings of these novel actions. Preclinical studies have generated many mechanistic theories, ranging from their renal and extrarenal effects to potential direct actions on cardiac muscle cells, to elucidate the mechanisms linking these drugs to clinical cardiovascular outcomes. Despite the strengths and limitations of each theory, many await validation in human studies. Furthermore, whether SGLT2 inhibitors confer therapeutic benefits in specific subsets of cardiomyopathies akin to their efficacy in other heart failure populations remains unclear. By examining the shared pathological features between heart failure resulting from vascular diseases and other causes of cardiomyopathy, certain specific molecular actions of SGLT2 inhibitors (particularly those targeting cardiomyocytes) would support the concept that these medications will yield therapeutic benefits across a broad range of cardiomyopathies. This article aims to discuss the important mechanisms of SGLT2 inhibitors and their implications in hypertrophic and dilated cardiomyopathies. Furthermore, we offer insights into future research directions for SGLT2 inhibitor studies, which hold the potential to further elucidate the proposed biological mechanisms in greater detail.
Collapse
Affiliation(s)
- Veera Ganesh Yerra
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Canada
| | - Kim A Connelly
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Canada
| |
Collapse
|
5
|
Zhen K, Wei X, Zhi Z, Shang S, Zhang S, Xu Y, Fu X, Cheng L, Yao J, Li Y, Chen X, Liu P, Zhang H. Circulating Extracellular Vesicles from Heart Failure Patients Inhibit Human Cardiomyocyte Activities. J Cardiovasc Transl Res 2024:10.1007/s12265-024-10571-1. [PMID: 39384702 DOI: 10.1007/s12265-024-10571-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 10/02/2024] [Indexed: 10/11/2024]
Abstract
Extracellular vesicles (EVs) have been implicated in cardiac remodeling during heart failure (HF). However, the role of circulating EVs (CEVs) in the process of HF is poorly understood. To elucidate the molecular mechanism associated with CEVs in the context of HF, the proteome of 4D label-free EVs from plasma samples was identified. Among the identified proteins, 6 exhibited upregulation while 9 demonstrated downregulation in CEVs derived from HF patients (HCEVs) compared to healthy controls (NCEVs). Our results showed that up-regulated proteins mainly participate in the primary metabolic, glycerolipid metabolic processes, oxidation-reduction process, and inflammatory amplification. In contrast, the down-regulated proteins influenced cell development, differentiation, and proliferation. Compared to NCEVs, HCEVs significantly induced inflammation and triacylglycerol (TAG) accumulation in human cardiomyocytes (HCMs) in vitro. They also compromised their regenerative capacities, triggered endoplasmic reticulum (ER) stress and increased autophagy in HCMs. Further, HCEVs induced differentiation of human cardiac fibroblasts (HCFs), amplifying pro-inflammatory, and pro-fibrotic factors, and enhancing extracellular matrix deposition. Notably, HCEVs are also associated with an increase in the HF biomarker MMP9 within HCFs and demonstrate a negative correlation with autophagic flux. In conclusion, HCEVs appear pivotal in advancing HF via pathological cardiac remodeling.
Collapse
Affiliation(s)
- Ke Zhen
- Beijing Anzhen Hospital, Capital Medical University, Beijing, 100011, China
| | - Xiaojuan Wei
- Department of Cardiovascular Surgery, Air Force Medical Center, PLA, Beijing, 100048, China
| | - Zelun Zhi
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Shiyu Shang
- The First Clinical Medical College, Hebei North University, Zhangjiakou, 075132, China
| | - Shuyan Zhang
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yilu Xu
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, China
| | - Xiaochuan Fu
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Linjia Cheng
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, 650500, China
| | - Jing Yao
- Department of Cardiovascular Surgery, Air Force Medical Center, PLA, Beijing, 100048, China
| | - Yue Li
- Department of Cardiovascular Surgery, Air Force Medical Center, PLA, Beijing, 100048, China
| | - Xia Chen
- Department of Cardiovascular Surgery, Air Force Medical Center, PLA, Beijing, 100048, China
| | - Pingsheng Liu
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Hongchao Zhang
- Department of Cardiovascular Surgery, Air Force Medical Center, PLA, Beijing, 100048, China.
| |
Collapse
|
6
|
Kelling M, Dimza M, Bartlett A, Traktuev DO, Duarte JD, Keeley EC. Omega-3 fatty acids in the treatment of heart failure. Curr Probl Cardiol 2024; 49:102730. [PMID: 38950721 DOI: 10.1016/j.cpcardiol.2024.102730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Accepted: 06/26/2024] [Indexed: 07/03/2024]
Abstract
Omega-3 polyunsaturated fatty acids (Ω-3 PUFAs) have garnered increased attention as a therapeutic option in cardiovascular disease. Most of the research to date has focused on their lipid altering effects and clinical benefits in patients with coronary artery disease, however, there are data supporting their use in the treatment of heart failure. We review the mechanisms through which Ω-3 PUFAs exert their positive effects on the cardiovascular system and highlight the observational and treatment studies that assessed their effects in patients with heart failure.
Collapse
Affiliation(s)
- Matthew Kelling
- Department of Medicine, University of Florida, Gainesville, Florida; Division of Cardiovascular Medicine, University of Florida, Gainesville, Florida
| | - Michelle Dimza
- Department of Medicine, University of Florida, Gainesville, Florida; Division of Cardiovascular Medicine, University of Florida, Gainesville, Florida
| | - Alec Bartlett
- Department of Medicine, University of Arizona, Phoenix, Arizona, United States
| | - Dmitry O Traktuev
- Department of Medicine, University of Florida, Gainesville, Florida; Division of Pulmonary, Critical care and Sleep Medicine, University of Florida
| | - Julio D Duarte
- Department of Pharmacotherapy and Translational Research, University of Florida, Gainesville, Florida
| | - Ellen C Keeley
- Department of Medicine, University of Florida, Gainesville, Florida; Division of Cardiovascular Medicine, University of Florida, Gainesville, Florida.
| |
Collapse
|
7
|
Wang D, Yu X, Gao K, Li F, Li X, Pu H, Zhang P, Guo S, Wang W. Sweroside alleviates pressure overload-induced heart failure through targeting CaMKⅡδ to inhibit ROS-mediated NF-κB/NLRP3 in cardiomyocytes. Redox Biol 2024; 74:103223. [PMID: 38851078 PMCID: PMC11219961 DOI: 10.1016/j.redox.2024.103223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 06/02/2024] [Accepted: 06/03/2024] [Indexed: 06/10/2024] Open
Abstract
Ongoing inflammation in the heart is positively correlated with adverse remodeling, characterized by elevated levels of cytokines that stimulate activation of cardiac fibroblasts. It was found that CaMKIIδ response to Ang II or TAC triggers the accumulation of ROS in cardiomyocytes, which subsequently stimulates NF-κB/NLRP3 and leads to an increase in IL-6, IL-1β, and IL-18. This is an important causative factor in the occurrence of adverse remodeling in heart failure. Sweroside is a biologically active natural iridoids extracted from Lonicerae Japonicae Flos. It shows potent anti-inflammatory and antioxidant activity in various cardiovascular diseases. In this study, we found that sweroside inhibited ROS-mediated NF-κB/NLRP3 in Ang II-treated cardiomyocytes by directly binding to CaMKIIδ. Knockdown of CaMKⅡδ abrogated the effect of sweroside regulation on NF-κB/NLRP3 in cardiomyocytes. AAV-CaMKⅡδ induced high expression of CaMKⅡδ in the myocardium of TAC/Ang II-mice, and the inhibitory effect of sweroside on TAC/Ang Ⅱ-induced elevation of NF-κB/NLRP3 was impeded. Sweroside showed significant inhibitory effects on CaMKIIδ/NF-κB/NLRP3 in cardiomyocytes from TAC/Ang Ⅱ-induced mice. This would be able to mitigate the adverse events of myocardial remodeling and contractile dysfunction at 8 weeks after the onset of the inflammatory response. Taken together, our findings have revealed the direct protein targets and molecular mechanisms by which sweroside improves heart failure, thereby supporting the further development of sweroside as a therapeutic agent for heart failure.
Collapse
Affiliation(s)
- Dong Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Xue Yu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Kuo Gao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Fanghe Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Xiang Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Haiyin Pu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Peng Zhang
- Wuhan Hospital of Traditional Chinese Medicine, Wuhan, 430014, China.
| | - Shuzhen Guo
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 102488, China.
| | - Wei Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 102488, China.
| |
Collapse
|
8
|
Zapolski T, Kornecki W, Jaroszyński A. The Influence of Balneotherapy Using Salty Sulfide-Hydrogen Sulfide Water on Selected Markers of the Cardiovascular System: A Prospective Study. J Clin Med 2024; 13:3526. [PMID: 38930055 PMCID: PMC11204439 DOI: 10.3390/jcm13123526] [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: 05/21/2024] [Revised: 06/10/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
Background: The sulfide-hydrogen sulfide brine balneotherapy (HSBB), including a combination of dissolved hydrogen sulfide (H2S) gas, inorganic sulfur ions (S2-), and hydrosulfide ions (HS-), is one of the most important and most effective forms of spa treatment in patients with osteoarticular disorders (OADs). Some cardiovascular diseases (CVDs) are often considered to be contraindications to HSBB since the presence of thiol groups may lead to an increased quantity of reactive oxygen species (ROS), which damage the vascular endothelium, and endothelial dysfunction is considered to be the main cause of atherosclerosis. However, there are a number of literature reports suggesting this theory to be false. H2S is a member of the endogenous gaseous transmitter family and, since it is a relatively recent addition, it has the least well-known biological properties. H2S-NO interactions play an important role in oxidative stress in CVDs. The general objective of this study was to assess the cardiovascular safety of HSBB and analyze the effect of HSBB on selected cardiovascular risk markers. Methods: A total of 100 patients at the age of 76.3 (±7.5) years from the Włókniarz Sanatorium in Busko-Zdrój were initially included in the study. The following parameters were assessed: age, sex, height, body weight, body surface area (BSA), body mass index (BMI), systolic (SBP) and diastolic blood pressure (DBP), heart rate, the diagnosis of OAD that was the indication for balneotherapy, creatinine (CREAT), glomerular filtration rate (GFR), lipid panel, C-reactive protein (CRP), uric acid (UA), and fibrinogen (FIBR) and cardiovascular markers: (cardiac troponin T (cTnT), N-terminal pro-B-type natriuretic peptide (NT-proBNP). Results: A significant decrease in DBP and a trend towards SBP reduction were observed over the course of the study. A significant decrease was observed in CRP levels decreasing from 2.7 (±3.6) mg/L to 2.06 (±1.91) mg/L, whereas FIBR rose significantly from 2.95 (±0.59) g/L to 3.23 (±1.23) g/L. LDL-C levels decreased slightly, statistically significant, from 129.36 (±40.67) mg/dL to 123.74 (±36.14) mg/dL. HSBB did not affect the levels of evaluated cardiovascular biomarkers, namely NT-proBNP (137.41 (±176.52) pg/mL vs. 142.89 (±182.82) pg/mL; p = 0.477) and cTnT (9.64 (±4.13) vs. 9.65 (±3.91) ng/L; p = 0.948). A multiple regression analysis of pre-balneotherapy and post-balneotherapy values showed cTnT levels to be independently correlated only with CREAT levels and GFR values. None of the assessed parameters independently correlated with the NT-proBNP level. Conclusions: HSBB resulted in a statistically significant improvement in a subclinical pro-inflammatory state. HSBB has a beneficial effect in modifying key cardiovascular risk factors by reducing LDL-C levels and DBP values. HSBB has a neutral effect on cardiovascular ischemia/injury. Despite slightly elevated baseline levels of the biochemical marker of HF (NT-proBNP), HSBB causes no further increase in this marker. The use of HSBB in patients with OAD has either a neutral effect or a potentially beneficial effect on the cardiovascular system, which may constitute grounds for further studies to verify the current cardiovascular contraindications for this form of therapy.
Collapse
Affiliation(s)
- Tomasz Zapolski
- Department of Cardiology, Medical University of Lublin, 20-093 Lublin, Poland
| | | | - Andrzej Jaroszyński
- Department of Internal Medicine and Family Medicine, Collegium Medicum, Jan Kochanowski University in Kielce, 25-369 Kielce, Poland;
| |
Collapse
|
9
|
Zhang Y, Feng L, Zhu Z, He Y, Li X. Association between blood inflammatory indices and heart failure: a cross-sectional study of NHANES 2009-2018. Acta Cardiol 2024; 79:473-485. [PMID: 38771356 DOI: 10.1080/00015385.2024.2356325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Accepted: 05/12/2024] [Indexed: 05/22/2024]
Abstract
BACKGROUND Inflammation plays a pivotal role in the pathogenesis of heart failure (HF). This study was aimed to the potential association between complete blood cell count (CBC)-derived inflammatory biomarkers and HF. METHODS Data from the National Health and Nutrition Examination Survey (NHANES) 2009-2018 were utilised. We evaluated the associations between HF and five systemic inflammation markers derived from CBC: systemic immune-inflammation index (SII), systemic inflammatory response index (SIRI), neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), and monocyte-to-lymphocyte ratio (MLR). Demographic characteristics, physical examinations, and laboratory data were systematically collected for comparative analysis between HF and non-HF individuals. Fitted smoothing curves and threshold effect analysis delineated the relationship. In addition, Spearman correlation and subgroup analyses were further conducted. RESULTS A total of 26,021 participants were categorised into HF (n = 858) and non-HF (n = 25,163) groups. After adjusting for confounding variables, SIRI, NLR, and MLR had significant positive correlations with the risk of HF. Participants in the highest quarter groups of SIRI, NLR, and MLR showed a increased risk of developing HF compared to those in the lowest quarter group. Furthermore, subgroup and sensitivity analyses indicated that SIRI, NLR, and MLR had a stronger correlation to HF (all p < 0.05). Smoothing curve fitting highlighted a nonlinear relationship between CBC-derived inflammatory biomarkers and HF. CONCLUSIONS Our results illustrated a significant association between elevated levels of SIRI, NLR, and MLR and an increased risk of HF. SIRI, NLR, and MLR could potentially serve as systemic inflammation hazard markers for HF.
Collapse
Affiliation(s)
- Yayun Zhang
- Department of Cardiovascular Medicine, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Lu Feng
- Department of Nursing, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
| | - Zixiong Zhu
- Department of Cardiovascular Medicine, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Yubin He
- Department of Cardiovascular Medicine, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Xuewen Li
- Department of Cardiovascular Medicine, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| |
Collapse
|
10
|
Asiwe JN, Ojetola AA, Ekene NE, Osirim E, Nnamudi AC, Oritsemuelebi B, Onuelu JE, Asiwe N, Eruotor HO, Inegbenehi S. Pleiotropic attenuating effect of Ginkgo biloba against isoprenaline-induced myocardial infarction via improving Bcl-2/mTOR/ERK1/2/Na +, K +-ATPase activities. CHINESE HERBAL MEDICINES 2024; 16:282-292. [PMID: 38706831 PMCID: PMC11064635 DOI: 10.1016/j.chmed.2023.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 08/10/2023] [Accepted: 11/23/2023] [Indexed: 05/07/2024] Open
Abstract
Objective Myocardial infarction (MI) is linked to an imbalance in the supply and demand of blood oxygen in the heart muscles. Beta-blockers and calcium antagonists are just two of the common medications used to treat MI. However, these have reportedly been shown to be either ineffective or to have undesirable side effects. Extract of Ginkgo biloba leaves (GBE), a Chinese herbal product offers special compatibility benefits in therapeutic settings relating to inflammatory diseases and oxidative stress. In order to better understand how GBE affects MI in rats insulted by isoprenaline (ISO), the current study was designed. Methods The heart weight index, serum lipid profile, cardiac marker enzymes, endogenous antioxidants [catalase (CAT), superoxide dismutase (SOD), glutathione (GSH), nitrites and malondialdehyde (MDA)], inflammatory mediators [tumour necrosis factor alpha (TNF-α) and interleukin-6 (IL-6)], immunohistochemical expressions of B-cell lymphoma factor-2 (Bcl-2), extracellular signal-regulated kinase (ERK1/2), and mammalian target of rapamycin (mTOR) and histopathological analysis were used to assess the cardioprotective properties of GBE. Results The findings showed that GBE effectively attenuated myocardial infarction by boosting the body's natural antioxidant defense system and reducing the release of inflammatory cytokines as well as heart injury marker enzymes. The expression of Bcl-2, ERK1/2 and mTOR was increased while the histomorphological alterations were reversed. Conclusion The cardioprotective effects of GBE may be due to a mechanism involving increased Bcl-2/mTOR/ERK1/2/Na+, K+-ATPase activity.
Collapse
Affiliation(s)
- Jerome Ndudi Asiwe
- Department of Physiology, Delta State University, Abraka 1, Nigeria
- Department of Physiology, University of Ibadan, Ibadan 3017, Nigeria
| | | | | | | | | | | | | | - Nicholas Asiwe
- Department of Anatomy, University of Port Harcourt, Choba 5323, Nigeria
| | | | - Saviour Inegbenehi
- Department of Biochemistry, PAMO University of Medical Sciences, Port Harcourt 500211, Nigeria
| |
Collapse
|
11
|
Cheng MD, Zheng YY, Zhang XY, Ruzeguli T, Sureya Y, Didaer Y, Ailiman M, Zhang JY. The Simplified Thrombo-Inflammatory Score as a Novel Predictor of All-Cause Mortality in Patients with Heart Failure: A Retrospective Cohort Study. J Inflamm Res 2024; 17:1845-1855. [PMID: 38523685 PMCID: PMC10961063 DOI: 10.2147/jir.s452544] [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: 11/30/2023] [Accepted: 03/08/2024] [Indexed: 03/26/2024] Open
Abstract
Background The simplified thrombo-inflammatory score (sTIPS) has recently emerged as a novel prognostic score. Hence, we investigated the prognostic value of sTIPS for predicting long-term mortality in patients with heart failure (HF). Methods A total of 3741 patients were analyzed in this study. The sTIPS was calculated based on the white blood cell count (WBC) and the mean platelet volume to platelet count (MPV/PC) ratio at admission. The mean follow-up time was 22.75 months. Multivariable Cox regression analyses were used to investigate the associations between the sTIPS and all-cause mortality (ACM). Results In the whole study population, multivariate Cox regression analysis showed that patients in both the sTIPS 2 and sTIPS 1 groups had significantly increased risk of ACM as compared with patients in the sTIPS 0 group (hazard ratio [HR]=1.706, 95% confidence interval [CI]: 1.405-2.072, P<0.001 and HR = 1.431, 95% CI 1.270-1.612, P<0.001). The same significant trend was observed in heart failure with preserved ejection fraction (HFpEF) patients (sTIPS1 vs sTIPS0: HR = 1.366, 95% CI 1.100-1.697, P = 0.005; sTIPS2 vs sTIPS0: HR = 1.995, 95% CI 1.460-2.725, P<0.001). However, only sTIPS 1 group had a significantly increased the risk of ACM compared to the sTIPS 0 group among patients with HFmrEF (sTIPS1 vs sTIPS0: HR = 1.648, 95% CI 1.238-2.194, P = 0.001) and HFrEF (sTIPS1 vs sTIPS0: HR = 1.322, 95% CI 1.021-1.712, P = 0.035). Conclusion sTIPS is useful in predicting risk for long-term mortality in patients with HF.
Collapse
Affiliation(s)
- Meng-Die Cheng
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People’s Republic of China
| | - Ying-Ying Zheng
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, People’s Republic of China
| | - Xing-Yan Zhang
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, People’s Republic of China
| | - Tuersun Ruzeguli
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, People’s Republic of China
| | - Yisimayili Sureya
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, People’s Republic of China
| | - Yisha Didaer
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, People’s Republic of China
| | - Mahemuti Ailiman
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, People’s Republic of China
| | - Jin-Ying Zhang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People’s Republic of China
| |
Collapse
|
12
|
Li X, Zeng L, Qu Z, Zhang F. Huoxin pill protects verapamil-induced zebrafish heart failure through inhibition of oxidative stress-triggered inflammation and apoptosis. Heliyon 2024; 10:e23402. [PMID: 38169776 PMCID: PMC10758798 DOI: 10.1016/j.heliyon.2023.e23402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 11/27/2023] [Accepted: 12/04/2023] [Indexed: 01/05/2024] Open
Abstract
Heart failure (HF) is a major and growing public health concern. Although advances in medical and surgical therapies have been achieved over the last decades, there is still no firmly evidence-based treatment with many traditional Chinese medicines (TCMs) for HF. Huoxin Pill (HXP), a TCM, has been widely used to treat patients with coronary heart disease and angina pectoris. However, the underlying molecular mechanism is poorly understood. In this study, using a verapamil-induced zebrafish HF model, we validated the efficacy and revealed the underlying mechanism of HXP in the treatment of HF. Zebrafish embryos were pretreated with different concentrations of HXP followed by verapamil administration, and we found that HXP significantly improved cardiac function in HF zebrafish, such as by effectively alleviating venous congestion and increasing heart rates. Mechanistically, HXP evidently inhibited verapamil-induced ROS and H2O2 production and upregulated CAT activity in HF zebrafish. Moreover, transgenic lines Tg(mpx:EGFP) and Tg(nfkb:EGFP) were administered for inflammation evaluation, and we found that neutrophil infiltration in HF zebrafish hearts and the activated NF-kB level could be reduced by HXP. Furthermore, HXP significantly downregulated the level of cell apoptosis in HF zebrafish hearts, as assessed by AO staining. Molecularly, RT‒qPCR results showed that pretreatment with HXP upregulated antioxidant-related genes such as gpx-1a and gss and downregulated the expression of the stress-related gene hsp70, proinflammatory genes such as tnf-α, il-6 and lck, and apoptosis-related indicators such as apaf1, puma and caspase9. In conclusion, HXP exerts a protective effect on verapamil-induced zebrafish HF through inhibition of oxidative stress-triggered inflammation and apoptosis.
Collapse
Affiliation(s)
- Xianmei Li
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, PR China
| | - Laifeng Zeng
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, PR China
| | - Zhixin Qu
- Key Laboratory of Gastrointestinal Cancer (Ministry of Education), School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, PR China
| | - Fenghua Zhang
- Key Laboratory of Gastrointestinal Cancer (Ministry of Education), School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, PR China
| |
Collapse
|
13
|
Dasari TW, Chakraborty P, Mukli P, Akhtar K, Yabluchanskiy A, Cunningham MW, Csiszar A, Po SS. Noninvasive low-level tragus stimulation attenuates inflammation and oxidative stress in acute heart failure. Clin Auton Res 2023; 33:767-775. [PMID: 37943335 DOI: 10.1007/s10286-023-00997-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 10/24/2023] [Indexed: 11/10/2023]
Abstract
PURPOSE Acute decompensated heart failure (ADHF) is associated with inflammation, oxidative stress, and excess sympathetic drive. It is unknown whether neuromodulation would improve inflammation and oxidative stress in acute heart failure. We, therefore, performed this proof-of-concept study to evaluate the effects of neuromodulation using noninvasive low-level tragus stimulation on inflammation and oxidative stress in ADHF. METHODS Nineteen patients with ejection fraction < 40% were randomized to neuromodulation 4 h twice daily (6-10 a.m. and 6-10 p.m.) (n = 8) or sham stimulation (n = 11) during hospital admission. All patients received standard-of-care treatment. Blood samples were collected at admission and discharge. Serum cytokines were assayed using standard immunosorbent techniques. Reactive oxygen species inducibility from cultured coronary endothelial cells exposed to patient sera was determined using a dihydrodichlorofluorescein probe test (expressed as fluorescein units). RESULTS Compared to sham stimulation, neuromodulation was associated with a significant reduction of circulating serum interleukin-6 levels (-78% vs. -9%; p = 0.012). Similarly, neuromodulation led to a reduction of endothelial cell oxidative stress in the neuromodulation group (1363 units to 978 units, p = 0.003) compared to sham stimulation (1146 units to 1083 units, p = 0.094). No significant differences in heart rate, blood pressure, or renal function were noted between the two groups. CONCLUSION In this proof-of-concept pilot study, in acute decompensated heart failure, neuromodulation was feasible and safe and was associated with a reduction in systemic inflammation and attenuation of coronary endothelial cellular oxidative stress. CLINICAL TRIAL REGISTRATION NCT02898181.
Collapse
Affiliation(s)
- Tarun W Dasari
- Cardiovascular Section, Department of Internal Medicine, Heart Rhythm Institute, University of Oklahoma Health Sciences Center, 800 SL Young Blvd, COM 5400, Oklahoma City, OK, 73104, USA.
| | - Praloy Chakraborty
- Cardiovascular Section, Department of Internal Medicine, Heart Rhythm Institute, University of Oklahoma Health Sciences Center, 800 SL Young Blvd, COM 5400, Oklahoma City, OK, 73104, USA
| | - Peter Mukli
- Department of Neurosurgery, University of Oklahoma HSC, Oklahoma City, OK, USA
| | - Khawaja Akhtar
- Cardiovascular Section, Department of Internal Medicine, Heart Rhythm Institute, University of Oklahoma Health Sciences Center, 800 SL Young Blvd, COM 5400, Oklahoma City, OK, 73104, USA
| | | | - Madeleine W Cunningham
- Department of Microbiology and Immunology, University of Oklahoma HSC, Oklahoma City, OK, USA
| | - Anna Csiszar
- Department of Neurosurgery, University of Oklahoma HSC, Oklahoma City, OK, USA
| | - Sunny S Po
- Cardiovascular Section, Department of Internal Medicine, Heart Rhythm Institute, University of Oklahoma Health Sciences Center, 800 SL Young Blvd, COM 5400, Oklahoma City, OK, 73104, USA
| |
Collapse
|
14
|
Wu C, Zhang R, Wang J, Chen Y, Zhu W, Yi X, Wang Y, Wang L, Liu P, Li P. Dioscorea nipponica Makino: A comprehensive review of its chemical composition and pharmacology on chronic kidney disease. Biomed Pharmacother 2023; 167:115508. [PMID: 37716118 DOI: 10.1016/j.biopha.2023.115508] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 09/06/2023] [Accepted: 09/12/2023] [Indexed: 09/18/2023] Open
Abstract
Chronic kidney disease (CKD) is a widespread ailment that significantly impacts global health. It is characterized by high prevalence, poor prognosis, and substantial healthcare costs, making it a major public health concern. The current clinical treatments for CKD are not entirely satisfactory, leading to a high demand for alternative therapeutic options. Chinese herbal medicine, with its long history, diverse varieties, and proven efficacy, offers a promising avenue for exploration. One such Chinese herbal medicine, Dioscorea nipponica Makino (DNM), is frequently used to treat kidney diseases. In this review, we have compiled studies examining the mechanisms of action of DNM in the context of CKD, focusing on five primary areas: improvement of oxidative stress, inhibition of renal fibrosis, regulation of metabolism, reduction of inflammatory response, and regulation of autophagy.
Collapse
Affiliation(s)
- Chenguang Wu
- Renal Division, Department of Medicine, Heilongjiang Academy of Chinese Medicine Sciences, Harbin, China
| | - Rui Zhang
- Renal Division, Department of Medicine, Heilongjiang Academy of Chinese Medicine Sciences, Harbin, China
| | - Jingjing Wang
- Renal Division, Department of Medicine, Heilongjiang Academy of Chinese Medicine Sciences, Harbin, China
| | - Yao Chen
- Renal Division, Department of Medicine, Heilongjiang Academy of Chinese Medicine Sciences, Harbin, China
| | - Wenhui Zhu
- Renal Division, Department of Medicine, Heilongjiang Academy of Chinese Medicine Sciences, Harbin, China
| | - Xiang Yi
- Renal Division, Department of Medicine, Heilongjiang Academy of Chinese Medicine Sciences, Harbin, China
| | - Yan Wang
- Department of Nephrology, Peking University People's Hospital, Beijing, China
| | - Lifan Wang
- Renal Division, Department of Medicine, Heilongjiang Academy of Chinese Medicine Sciences, Harbin, China.
| | - Peng Liu
- Shunyi Hospital, Beijing Hospital of Traditional Chinese Medicine, Beijing, China.
| | - Ping Li
- Beijing Key Lab for Immune-Mediated Inflammatory Diseases, China-Japan Friendship Hospital, Beijing, China.
| |
Collapse
|
15
|
Dasari T, Chakraborty P, Mukli P, Akhtar K, Yabluchanskiy A, Cunningham MW, Csiszar A, Po SS. Noninvasive low-level tragus stimulation attenuates inflammation and oxidative stress in acute heart failure. RESEARCH SQUARE 2023:rs.3.rs-3323086. [PMID: 37790298 PMCID: PMC10543293 DOI: 10.21203/rs.3.rs-3323086/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
Purpose Acute decompensated heart failure is associated with inflammation, oxidative stress, and excess sympathetic drive. It is unknown if neuromodulation would improve inflammation and oxidative stress in acute heart failure. We, therefore, performed this proof-of-concept study to evaluate the effects of neuromodulation using noninvasive low-level Tragus stimulation on inflammation and oxidative stress in ADHF. Methods 19 patients with ejection fraction < 40% were randomized to neuromodulation- 4 hours twice daily (6 AM-10 AM and 6 PM-10 PM) (n = 8) or sham stimulation (n = 11) during hospital admission. All patients received standard-of-care treatment. Blood samples were collected at admission and discharge. Serum cytokines were assayed using standard immunosorbent techniques. Reactive oxygen species inducibility from cultured coronary endothelial cells exposed to patient sera was determined using dihydrodichlorofluorescein probe test (expressed as fluorescein units). Results Compared to sham stimulation, neuromodulation was associated with a significant reduction of circulating serum Interleukin-6 levels (-78% vs -9%; p = 0.012). Similarly, neuromodulation led to reduction of endothelial cell oxidative stress, in the neuromodulation group (1363 units to 978 units, p = 0.003) compared to sham stimulation (1146 units to 1083 units, p = 0.094). No significant difference in heart rate, blood pressure or renal function were noted between the two groups. Conclusion In this proof-of-concept pilot study, in acute systolic heart failure, neuromodulation was feasible and safe and was associated with a reduction in systemic inflammation and attenuation of cellular oxidative stress. Clinical trial NCT02898181.
Collapse
Affiliation(s)
- Tarun Dasari
- University of Oklahoma: The University of Oklahoma
| | | | - Peter Mukli
- University of Oklahoma: The University of Oklahoma
| | | | | | | | - Anna Csiszar
- University of Oklahoma: The University of Oklahoma
| | - Sunny S Po
- University of Oklahoma: The University of Oklahoma
| |
Collapse
|
16
|
Wołowiec A, Wołowiec Ł, Grześk G, Jaśniak A, Osiak J, Husejko J, Kozakiewicz M. The Role of Selected Epigenetic Pathways in Cardiovascular Diseases as a Potential Therapeutic Target. Int J Mol Sci 2023; 24:13723. [PMID: 37762023 PMCID: PMC10531432 DOI: 10.3390/ijms241813723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 09/03/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
Epigenetics is a rapidly developing science that has gained a lot of interest in recent years due to the correlation between characteristic epigenetic marks and cardiovascular diseases (CVDs). Epigenetic modifications contribute to a change in gene expression while maintaining the DNA sequence. The analysis of these modifications provides a thorough insight into the cardiovascular system from its development to its further functioning. Epigenetics is strongly influenced by environmental factors, including known cardiovascular risk factors such as smoking, obesity, and low physical activity. Similarly, conditions affecting the local microenvironment of cells, such as chronic inflammation, worsen the prognosis in cardiovascular diseases and additionally induce further epigenetic modifications leading to the consolidation of unfavorable cardiovascular changes. A deeper understanding of epigenetics may provide an answer to the continuing strong clinical impact of cardiovascular diseases by improving diagnostic capabilities, personalized medical approaches and the development of targeted therapeutic interventions. The aim of the study was to present selected epigenetic pathways, their significance in cardiovascular diseases, and their potential as a therapeutic target in specific medical conditions.
Collapse
Affiliation(s)
- Anna Wołowiec
- Department of Geriatrics, Division of Biochemistry and Biogerontology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, 87-100 Torun, Poland
| | - Łukasz Wołowiec
- Department of Cardiology and Clinical Pharmacology, Faculty of Health Sciences, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, 87-100 Torun, Poland
| | - Grzegorz Grześk
- Department of Cardiology and Clinical Pharmacology, Faculty of Health Sciences, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, 87-100 Torun, Poland
| | - Albert Jaśniak
- Department of Cardiology and Clinical Pharmacology, Faculty of Health Sciences, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, 87-100 Torun, Poland
| | - Joanna Osiak
- Department of Cardiology and Clinical Pharmacology, Faculty of Health Sciences, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, 87-100 Torun, Poland
| | - Jakub Husejko
- Department of Cardiology and Clinical Pharmacology, Faculty of Health Sciences, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, 87-100 Torun, Poland
| | - Mariusz Kozakiewicz
- Department of Geriatrics, Division of Biochemistry and Biogerontology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, 87-100 Torun, Poland
| |
Collapse
|
17
|
Yang X, Liu Z, Fang M, Zou T, Zhang Z, Meng X, Wang T, Meng H, Chen Y, Duan Y, Li Q. Novel pterostilbene derivatives ameliorate heart failure by reducing oxidative stress and inflammation through regulating Nrf2/NF-κB signaling pathway. Eur J Med Chem 2023; 258:115602. [PMID: 37406380 DOI: 10.1016/j.ejmech.2023.115602] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 06/14/2023] [Accepted: 06/25/2023] [Indexed: 07/07/2023]
Abstract
Pterostilbene is a demethylated resveratrol derivative with attractive anti-inflammatory, anti-tumor and anti-oxidative stress activities. However, the clinical use of pterostilbene is limited by its poor selectivity and druggability. Heart failure is a leading cause of morbidity and mortality worldwide, which is closely related to enhanced oxidative stress and inflammation. There is an urgent need for new effective therapeutic drugs that can reduce oxidative stress and inflammatory responses. Therefore, we designed and synthesized a series of novel pterostilbene chalcone and dihydropyrazole derivatives with antioxidant and anti-inflammatory activities by the molecular hybridization strategy. The preliminary anti-inflammatory activities and structure-activity relationships of these compounds were evaluated by nitric oxide (NO) inhibitory activity in lipopolysaccharide (LPS)-treated RAW264.7 cells, and compound E1 exhibited the most potent anti-inflammatory activities. Furthermore, pretreatment with compound E1 decreased reactive oxygen species (ROS) generation both in RAW264.7 and H9C2 cells by increasing the expression of nuclear factor erythroid 2-related factor 2 (Nrf2), as well as downstream antioxidant enzymes superoxide dismutase 1 (SOD1), catalase (CAT) and glutathione peroxidase 1 (GPX1). In addition, compound E1 also significantly inhibited LPS or doxorubicin (DOX)-induced inflammation in both RAW264.7 and H9C2 cells through reducing the expression of inflammatory cytokines by inhibiting nuclear factor-κB (NF-κB) signaling pathway. Moreover, we found that compound E1 improved DOX-induced heart failure by inhibiting inflammation and oxidative stress in mouse model, which is mediated by the potential of antioxidant and anti-inflammatory activities. In conclusion, this study demonstrated the novel pterostilbene dihydropyrazole derivative E1 was identified as a promising agent for heart failure treatment.
Collapse
Affiliation(s)
- Xiaoxiao Yang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China.
| | - Zhigang Liu
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Mengyuan Fang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Tingfeng Zou
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Zhen Zhang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Xianshe Meng
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Tianxiang Wang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Huawen Meng
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Yuanli Chen
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Yajun Duan
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Qingshan Li
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China.
| |
Collapse
|
18
|
Ucar A, Yeltekin AÇ, Köktürk M, Calimli MH, Nas MS, Parlak V, Alak G, Atamanalp M. Has PdCu@GO effect on oxidant/antioxidant balance? Using zebrafish embryos and larvae as a model. Chem Biol Interact 2023; 378:110484. [PMID: 37054932 DOI: 10.1016/j.cbi.2023.110484] [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: 03/06/2023] [Revised: 03/22/2023] [Accepted: 04/08/2023] [Indexed: 04/15/2023]
Abstract
Industrial products containing PdCu@GO can gain access to the aquaculture environment, causing dangerous effects on living biota. In this study, the developmental toxicity of zebrafish treated with different concentrations (50, 100, 250, 500 and 1000 μg/L) of PdCu@GO was investigated. The findings showed that PdCu@GO administration decreased the hatchability and survival rate, caused dose-dependent cardiac malformation. Reactive oxygen species (ROS) and apoptosis were also inhibited in a dose-dependent manner, with acetylcholinesterase (AChE) activity affected by nano-Pd exposure. As evidence for oxidative stress, malondialdehyde (MDA) level increased and superoxide dismutase (SOD), catalase (CAT) glutathione peroxidase (GPx) activities and glutathione (GSH) level decreased due to the increase in PdCu@GO concentration. Our research, it was determined that the oxidative stress stimulated by the increase in the concentration of PdCu@GO in zebrafish caused apoptosis (Caspase-3) and DNA damage (8-OHdG). Stimulation of ROS, inflammatory cytokines, tumor Necrosis Factor Alfa (TNF-α) and interleukin - 6 (IL-6), which act as signaling molecules to trigger proinflammatory cytokine production, induced zebrafish immunotoxicity. However, it was determined that the increase of ROS induced teratogenicity through the induction of nuclear factor erythroid 2 level (Nrf-2), NF-κB and apoptotic signaling pathways triggered by oxidative stress. Taken together with the research findings, the study contributed to a comprehensive assessment of the toxicological profile of PdCu@GO by investigating the effects on zebrafish embryonic development and potential molecular mechanisms.
Collapse
Affiliation(s)
- Arzu Ucar
- Department of Aquaculture, Faculty of Fisheries, Ataturk University, Erzurum, Turkey.
| | | | - Mine Köktürk
- Department of Organic Agriculture Management, Faculty of Applied Sciences, Igdir University, TR-76000, Igdir, Turkey; Research Laboratory Application and Research Center (ALUM), Iğdır University, TR-76000, Iğdır, Turkey
| | - Mehmet Harbi Calimli
- Department of Medical Services and Techniques, Tuzluca Vocational School, Igdir University, TR-76000, Igdir, Turkey; Research Laboratory Application and Research Center (ALUM), Iğdır University, TR-76000, Iğdır, Turkey
| | - Mehmet Salih Nas
- Department of Environmental Engineering, Faculty of Engineering, Igdir University, TR-76000, Igdir, Turkey; Research Laboratory Application and Research Center (ALUM), Iğdır University, TR-76000, Iğdır, Turkey
| | - Veysel Parlak
- Department of Basic Sciences, Faculty of Fisheries, Ataturk University, Erzurum, Turkey
| | - Gonca Alak
- Department of Seafood Processing Technology, Faculty of Fisheries, Ataturk University, Erzurum, Turkey
| | - Muhammed Atamanalp
- Department of Aquaculture, Faculty of Fisheries, Ataturk University, Erzurum, Turkey.
| |
Collapse
|
19
|
Discovery of novel dihydropyrazole-stilbene derivatives for ameliorating heart failure through modulation of p38/NF-κB signaling pathway. Bioorg Chem 2022; 129:106206. [DOI: 10.1016/j.bioorg.2022.106206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 09/28/2022] [Accepted: 10/10/2022] [Indexed: 11/17/2022]
|
20
|
Vilella-Figuerola A, Padró T, Roig E, Mirabet S, Badimon L. New factors in heart failure pathophysiology: Immunity cells release of extracellular vesicles. Front Cardiovasc Med 2022; 9:939625. [PMID: 36407432 PMCID: PMC9669903 DOI: 10.3389/fcvm.2022.939625] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 09/12/2022] [Indexed: 07/30/2023] Open
Abstract
Leukocyte-shed extracellular vesicles (EVs) can play effector roles in the pathophysiological mechanisms of different diseases. These EVs released by membrane budding of leukocytes have been found in high amounts locally in inflamed tissues and in the circulation, indicating immunity cell activation. These EVs secreted by immune cell subsets have been minimally explored and deserve further investigation in many areas of disease. In this study we have investigated whether in heart failure there is innate and adaptive immune cell release of EVs. Patients with chronic heart failure (cHF) (n = 119) and in sex- and age-matched controls without this chronic condition (n = 60). Specifically, EVs were quantified and phenotypically characterized by flow cytometry and cell-specific monoclonal antibodies. We observed that even in well medically controlled cHF patients (with guideline-directed medical therapy) there are higher number of blood annexin-V+ (phosphatidylserine+)-EVs carrying activated immunity cell-epitopes in the circulation than in controls (p < 0.04 for all cell types). Particularly, EVs shed by monocytes and neutrophils (innate immunity) and by T-lymphocytes and natural-killer cells (adaptive immunity) are significantly higher in cHF patients. Additionally, EVs-shed by activated leukocytes/neutrophils (CD11b+, p = 0.006; CD29+/CD15+, p = 0.048), and T-lymphocytes (CD3+/CD45+, p < 0.02) were positively correlated with cHF disease severity (NYHA classification). Interestingly, cHF patients with ischemic etiology had the highest levels of EVs shed by lymphocytes and neutrophils (p < 0.045, all). In summary, in cHF patients there is a significant immune cell activation shown by high-release of EVs that is accentuated by clinical severity of cHF. These activated innate and adaptive immunity cell messengers may contribute by intercellular communication to the progression of the disease and to the common affectation of distant organs in heart failure (paracrine regulation) that contribute to the clinical deterioration of cHF patients.
Collapse
Affiliation(s)
- Alba Vilella-Figuerola
- Cardiovascular-Program ICCC, IR-Hospital Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain
- Department of Biochemical and Molecular Biology, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Teresa Padró
- Cardiovascular-Program ICCC, IR-Hospital Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain
- Centro de Investigación Biomédica en Red Cardiovascular, Instituto de Salud Carlos III, Madrid, Spain
| | - Eulàlia Roig
- Heart Failure Group, Department of Cardiology, Hospital Santa Creu i Sant Pau, Barcelona, Spain
| | - Sònia Mirabet
- Centro de Investigación Biomédica en Red Cardiovascular, Instituto de Salud Carlos III, Madrid, Spain
- Heart Failure Group, Department of Cardiology, Hospital Santa Creu i Sant Pau, Barcelona, Spain
| | - Lina Badimon
- Cardiovascular-Program ICCC, IR-Hospital Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain
- Centro de Investigación Biomédica en Red Cardiovascular, Instituto de Salud Carlos III, Madrid, Spain
- UAB-Chair Cardiovascular Research, Barcelona, Spain
| |
Collapse
|
21
|
Vural A, Aydın E. The Predictive Value of Eosinophil Indices for Major Cardiovascular Events in Patients with Acute Decompensated HFrEF. MEDICINA (KAUNAS, LITHUANIA) 2022; 58:1455. [PMID: 36295615 PMCID: PMC9611138 DOI: 10.3390/medicina58101455] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 10/04/2022] [Accepted: 10/12/2022] [Indexed: 11/09/2022]
Abstract
Background and Objectives: Heart failure is a chronic disease with a high risk of mortality and morbidity. In these patients, inflammatory markers have been shown to be associated with cardiovascular adverse outcomes and disease progression. To investigate the relationships between eosinophil indices and major cardiovascular events (MACE) in patients with acute decompensated heart failure (ADHF) with reduced ejection fraction. Materials and Methods: A total of 395 consecutive patients admitted to the intensive care unit (ICU) with ADHF and reduced ejection fraction between January 2017 and December 2021 were enrolled in this retrospective study. MACE was defined as the composite of death and re-hospitalization for ADHF within 6 months of index hospitalization. All-cause mortality and MACE were assessed with respect to relationships with eosinophil indices, including neutrophil-to-eosinophil ratio (NER), leukocyte-to-eosinophil ratio (LER), eosinophil-to-lymphocyte ratio (ELR), and eosinophil-to-monocyte ratio (EMR). Results: NER and LER were significantly higher in subjects with MACE. Absolute eosinophil, lymphocyte and basophil count, hemoglobin, serum Na+, albumin, and CRP, and EMR and ELR were significantly lower in subjects with MACE compared to those without. NT-proBNP (OR: 1.682, 95% CI: 1.106−2.312, p = 0.001), Na+ (OR: 0.932, 95% CI: 0.897−0.969, p < 0.001), NER (OR: 2.740, 95 % CI: 1.797−4.177, p < 0.001), LER (OR: 2.705, 95% CI: 1.752−4.176, p < 0.001), EMR (OR:1.654, 95% CI 1.123−2.436, p = 0.011), ELR (OR: 2.112, 95% CI 1.424−3.134, p < 0.001), and eosinophil count (OR: 1.833, 95% CI 1.276−2.635) were independent predictors for development of MACE. Conclusions: Patients with ADHF and reduced ejection fraction who developed MACE within the first six months of index hospitalization had lower levels of absolute eosinophil and lymphocyte counts, and EMR and ELR values, whereas NER and LER were higher compared to those without MACE. The eosinophil indices were independently associated with mortality and MACE development. The eosinophil indices may be used to estimate MACE likelihood with acceptable sensitivity and specificity.
Collapse
Affiliation(s)
- Aslı Vural
- Clinic of Cardiology, Faculty of Medicine, Giresun University, Giresun 28200, Turkey
| | | |
Collapse
|
22
|
Fang J, Zhang Y, Chen D, Zheng Y, Jiang J. Exosomes and Exosomal Cargos: A Promising World for Ventricular Remodeling Following Myocardial Infarction. Int J Nanomedicine 2022; 17:4699-4719. [PMID: 36217495 PMCID: PMC9547598 DOI: 10.2147/ijn.s377479] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 09/21/2022] [Indexed: 11/23/2022] Open
Abstract
Exosomes are a pluripotent group of extracellular nanovesicles secreted by all cells that mediate intercellular communications. The effective information within exosomes is primarily reflected in exosomal cargos, including proteins, lipids, DNAs, and non-coding RNAs (ncRNAs), the most intensively studied molecules. Cardiac resident cells (cardiomyocytes, fibroblasts, and endothelial cells) and foreign cells (infiltrated immune cells, cardiac progenitor cells, cardiosphere-derived cells, and mesenchymal stem cells) are involved in the progress of ventricular remodeling (VR) following myocardial infarction (MI) via transferring exosomes into target cells. Here, we summarize the pathological mechanisms of VR following MI, including cardiac myocyte hypertrophy, cardiac fibrosis, inflammation, pyroptosis, apoptosis, autophagy, angiogenesis, and metabolic disorders, and the roles of exosomal cargos in these processes, with a focus on proteins and ncRNAs. Continued research in this field reveals a novel diagnostic and therapeutic strategy for VR.
Collapse
Affiliation(s)
- Jiacheng Fang
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, People’s Republic of China
| | - Yuxuan Zhang
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, People’s Republic of China
| | - Delong Chen
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, People’s Republic of China
| | - Yiyue Zheng
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, People’s Republic of China
| | - Jun Jiang
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, People’s Republic of China,Correspondence: Jun Jiang, Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, No. 88 Jiefang Road, Hangzhou, Zhejiang, 310009, People’s Republic of China, Tel/Fax +86 135 8870 6891, Email
| |
Collapse
|
23
|
Cediel G, Teis A, Codina P, Julve J, Domingo M, Santiago-Vacas E, Castelblanco E, Amigó N, Lupón J, Mauricio D, Alonso N, Bayés-Genís A. GlycA and GlycB as Inflammatory Markers in Chronic Heart Failure. Am J Cardiol 2022; 181:79-86. [PMID: 36008162 DOI: 10.1016/j.amjcard.2022.07.019] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 06/27/2022] [Accepted: 07/05/2022] [Indexed: 11/01/2022]
Abstract
The role of inflammation in heart failure (HF) has been extensively described, but it is uncertain whether inflammation exerts a different prognostic influence according to etiology. We aimed to examine the inflammatory state in chronic HF by measuring N-acetylglucosamine/galactosamine (GlycA) and sialic acid (GlycB), evolving proton nuclear magnetic resonance biomarkers of systemic inflammation, and explore their prognostic value in patients with chronic HF. The primary end point was a composite of all-cause death and HF readmission. A total of 429 patients were included. GlycB correlated with interleukin-1 receptor-like 1 in the whole cohort (r2 = 0.14, p = 0.011) and the subgroup of nonischemic etiology (r2 = 0.31, p <0.001). No association was found with New York Heart Association functional class or left ventricular ejection fraction. In patients with nonischemic HF (52.2%, n = 224), GlycA and GlycB exhibited significant association with the composite end point (hazard ratio [HR] 1.19, 95% confidence interval [CI] 1.06 to 1.33, p = 0.004 and HR 2.13, 95% CI 1.43 to 3.13, p <0.001; respectively) and GlycB with HF readmission after multivariable adjustment (HR 2.25, 95% CI 1.54 to 3.30, p <0.001). GlycB levels were also associated with a greater risk of HF-related recurrent admissions (adjusted incidence rate ratio 1.33, 95% CI = 1.07 to 1.65, p = 0.009). None of the markers were associated with the clinical end points in patients with ischemic HF. In conclusion, GlycA and GlycB represent an evolving approach to inflammation status with prognostic value in long-term outcomes in patients with nonischemic HF.
Collapse
Affiliation(s)
- German Cediel
- Heart Failure Unit and Cardiology Department, Hospital Universitari Germans Trias I Pujol, Badalona, Spain; Center for Biomedical Research on Cardiovascular Diseases (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Albert Teis
- Heart Failure Unit and Cardiology Department, Hospital Universitari Germans Trias I Pujol, Badalona, Spain; Center for Biomedical Research on Cardiovascular Diseases (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain; Department of Medicine, Universitat Autonoma de Barcelona, Barcelona, Spain
| | - Pau Codina
- Heart Failure Unit and Cardiology Department, Hospital Universitari Germans Trias I Pujol, Badalona, Spain
| | - Josep Julve
- Center for Biomedical Research on Diabetes and Associated Metabolic Diseases (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain; Sant Pau Biomedical Research Institute (IIB Sant Pau), Barcelona, Spain
| | - Mar Domingo
- Heart Failure Unit and Cardiology Department, Hospital Universitari Germans Trias I Pujol, Badalona, Spain
| | - Evelyn Santiago-Vacas
- Heart Failure Unit and Cardiology Department, Hospital Universitari Germans Trias I Pujol, Badalona, Spain; Center for Biomedical Research on Cardiovascular Diseases (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Esmeralda Castelblanco
- Department of Internal Medicine, Endocrinology, Metabolism and Lipid Research Division, Washington University School of Medicine, St Louis, Missouri; Unitat de Suport a la Recerca Barcelona, Institut Universitari d'Investigació en Atenció Primària Jordi Gol i Gurina (IDIAP Jordi Gol), Barcelona, Spain
| | - Nuria Amigó
- Center for Biomedical Research on Diabetes and Associated Metabolic Diseases (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain; Departamento de Ciencias Médicas Básicas, Universidad Rovira i Virgili, Tarragona, Spain; Biosfer Teslab - Metabolomic Platform, Universidad Rovira i Virgili, Tarragona, Spain
| | - Josep Lupón
- Heart Failure Unit and Cardiology Department, Hospital Universitari Germans Trias I Pujol, Badalona, Spain; Center for Biomedical Research on Cardiovascular Diseases (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain; Department of Medicine, Universitat Autonoma de Barcelona, Barcelona, Spain
| | - Didac Mauricio
- Center for Biomedical Research on Diabetes and Associated Metabolic Diseases (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain; Unitat de Suport a la Recerca Barcelona, Institut Universitari d'Investigació en Atenció Primària Jordi Gol i Gurina (IDIAP Jordi Gol), Barcelona, Spain; Department of Endocrinology & Nutrition, Hospital de la Santa Creu i Sant Pau & Sant Pau Biomedical Research Institute (IIB Sant Pau), Barcelona, Spain; Faculty of Medicine, University of Vic (UVIC), Vic, Spain
| | - Nuria Alonso
- Center for Biomedical Research on Diabetes and Associated Metabolic Diseases (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain; Department of Endocrinology & Nutrition, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Antoni Bayés-Genís
- Heart Failure Unit and Cardiology Department, Hospital Universitari Germans Trias I Pujol, Badalona, Spain; Center for Biomedical Research on Cardiovascular Diseases (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain; Department of Medicine, Universitat Autonoma de Barcelona, Barcelona, Spain.
| |
Collapse
|
24
|
Helleberg S, Engel A, Ahmed S, Ahmed A, Rådegran G. Higher plasma IL-6 and PTX3 are associated with worse survival in left heart failure with pulmonary hypertension. AMERICAN HEART JOURNAL PLUS : CARDIOLOGY RESEARCH AND PRACTICE 2022; 20:100190. [PMID: 38560419 PMCID: PMC10978361 DOI: 10.1016/j.ahjo.2022.100190] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 08/06/2022] [Indexed: 04/04/2024]
Abstract
Introduction Left heart failure (LHF) is commonly complicated by pulmonary hypertension (PH), increasing morbidity and mortality. The present study aimed to evaluate the prognostic value of inflammatory proteins in LHF with PH (LHF-PH). Materials and methods The levels of 65 plasma proteins, analysed with proximity extension assay, were compared between healthy controls (n = 20), patients with LHF-PH (n = 67) comprising both HFpEF-PH (n = 31) and HFrEF-PH (n = 36), and in a LHF subpopulation before and after heart transplantation (HT, n = 19). Haemodynamic parameters were measured using right heart catheterization. Results Plasma levels of Interleukin 6 (IL-6) and Pentraxin related protein PTX3 (PTX3) were elevated in LHF-PH vs. controls (p < 0.001), and these decreased after HT compared to before HT (p < 0.001). Plasma IL-6 and PTX3 correlated to elevated NT-proBNP (r = 0.44, p = 0.0002 and r = 0.4, p = 0.0009, respectively). Additionally, IL-6 correlated with mean pulmonary arterial pressure (r = 0.4, p = 0.0009) and mean right atrial pressure (r = 0.51, p < 0.0001). Higher levels of IL-6 and PTX3 were associated with worse survival rates in patients with LHF-PH (Log rank p < 0.01). Discussion In patients with LHF-PH, higher plasma levels of IL-6 and PTX3 were associated with worse survival rates. Future larger studies to validate and investigate the direct clinical applicability of IL-6 and PTX3 as potential prognostic biomarkers are encouraged.
Collapse
Affiliation(s)
- Sara Helleberg
- Department of Clinical Sciences Lund, Cardiology, Lund University, Sweden
- The Haemodynamic Lab, Section for Heart Failure and Valvular Disease, VO. Heart and Lung Medicine, Skåne University Hospital, Lund, Sweden
| | - Adam Engel
- Department of Clinical Sciences Lund, Cardiology, Lund University, Sweden
- The Haemodynamic Lab, Section for Heart Failure and Valvular Disease, VO. Heart and Lung Medicine, Skåne University Hospital, Lund, Sweden
| | - Salaheldin Ahmed
- Department of Clinical Sciences Lund, Cardiology, Lund University, Sweden
- The Haemodynamic Lab, Section for Heart Failure and Valvular Disease, VO. Heart and Lung Medicine, Skåne University Hospital, Lund, Sweden
| | - Abdulla Ahmed
- Department of Clinical Sciences Lund, Cardiology, Lund University, Sweden
- The Haemodynamic Lab, Section for Heart Failure and Valvular Disease, VO. Heart and Lung Medicine, Skåne University Hospital, Lund, Sweden
| | - Göran Rådegran
- Department of Clinical Sciences Lund, Cardiology, Lund University, Sweden
- The Haemodynamic Lab, Section for Heart Failure and Valvular Disease, VO. Heart and Lung Medicine, Skåne University Hospital, Lund, Sweden
| |
Collapse
|
25
|
Investigation of the Potential Key Genes and the Multitarget Mechanisms of Polygonum cuspidatum against Heart Failure Based on Network Pharmacology and Experimental Validation. DISEASE MARKERS 2022; 2022:7784021. [PMID: 35669500 PMCID: PMC9167087 DOI: 10.1155/2022/7784021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 03/19/2022] [Accepted: 04/12/2022] [Indexed: 11/17/2022]
Abstract
In this study, systematic pharmacology and bioinformatic approaches were employed to identify the potential targets of Polygonum cuspidatum (PC) for treating heart failure (HF). The active ingredients of PC were screened by using the TCMSP database, and HF-related genes were identified in the GEO database. Then, the herb-HF targeted-gene networks were constructed using Cytoscape software. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) functional analyses were performed to obtain the enriched molecular pathways associated with the pathogenesis of HF. Finally, in vitro experiment was performed to evidence network pharmacology analysis. 170 intersection genes were obtained, and key genes (FOXO3, NFKB1, and TNF) were identified. Besides, GO and KEGG findings indicated that PC treatment of HF was achieved via regulating apoptosis, IL-17 signaling pathway, TNF signaling pathway, response to oxidative stress, and response to reactive oxygen species. And cell experiment revealed that PC could decrease the expression of NFKB1 and TNF and increase the expression of FOXO3, SOD1, and GPX1 in H9C2 cells. These findings showed that the therapeutic mechanism of PC in the treatment of HF may be associated with the regulation of inflammation-related and oxidative stress-related genes.
Collapse
|
26
|
Wang A, Zhao W, Yan K, Huang P, Zhang H, Zhang Z, Zhang D, Ma X. Mechanisms and Efficacy of Traditional Chinese Medicine in Heart Failure. Front Pharmacol 2022; 13:810587. [PMID: 35281941 PMCID: PMC8908244 DOI: 10.3389/fphar.2022.810587] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Accepted: 01/24/2022] [Indexed: 12/11/2022] Open
Abstract
Heart failure (HF) is one of the main public health problems at present. Although some breakthroughs have been made in the treatment of HF, the mortality rate remains very high. However, we should also pay attention to improving the quality of life of patients with HF. Traditional Chinese medicine (TCM) has a long history of being used to treat HF. To demonstrate the clinical effects and mechanisms of TCM, we searched published clinical trial studies and basic studies. The search results showed that adjuvant therapy with TCM might benefit patients with HF, and its mechanism may be related to microvascular circulation, myocardial energy metabolism, oxidative stress, and inflammation.
Collapse
Affiliation(s)
- Anzhu Wang
- Graduate School, China Academy of Chinese Medical Sciences, Beijing, China.,Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Wei Zhao
- Yidu Central Hospital of Weifang, Weifang, China
| | - Kaituo Yan
- Yidu Central Hospital of Weifang, Weifang, China
| | - Pingping Huang
- Graduate School, China Academy of Chinese Medical Sciences, Beijing, China.,Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Hongwei Zhang
- Graduate School, China Academy of Chinese Medical Sciences, Beijing, China.,Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhibo Zhang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.,Xiyuan Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Dawu Zhang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.,National Clinical Research Center for Chinese Medicine Cardiology, Beijing, China
| | - Xiaochang Ma
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.,National Clinical Research Center for Chinese Medicine Cardiology, Beijing, China
| |
Collapse
|
27
|
Yin T, Shi S, Zhu X, Cheang I, Lu X, Gao R, Zhang H, Yao W, Zhou Y, Li X. A Survival Prediction for Acute Heart Failure Patients via Web-Based Dynamic Nomogram with Internal Validation: A Prospective Cohort Study. J Inflamm Res 2022; 15:1953-1967. [PMID: 35342297 PMCID: PMC8947803 DOI: 10.2147/jir.s348139] [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: 11/06/2021] [Accepted: 03/09/2022] [Indexed: 12/12/2022] Open
Abstract
Purpose The current study aimed to develop a convenient and accurate prognostic dynamic nomogram model for the risk of all-cause death in acute heart failure (AHF) patients that incorporates clinical characteristics including N-terminal pro-brain natriuretic peptide (NT-pro BNP) and growth stimulation expresses gene 2 protein (ST2). Patients and Methods We prospectively studied 537 consecutive AHF patients and derived a clinical prediction model. The least absolute shrinkage and selection operator regression model combined with clinical characteristics were used for dimensional reduction and feature selection. Multivariate Cox proportional hazard analysis and “Dynnom” package were used to build the dynamic nomogram for prediction of 1-,2-,and 5-year overall survival for AHF. With bootstrap validation, the time-dependent concordance index (C-index) and calibration curves were used to assess predictive discrimination and accuracy. The contributions of NT-pro BNP and ST2 to the nomogram were evaluated using integrated discrimination improvement (IDI) and net reclassification improvement (NRI), while decision curve analysis (DCA) was used to assess clinical value. Results Patients were randomly divided into derivation (74.9%, n=402) and validation (25.1%, n=135) cohorts. Optimal independent prognostic factors for 1-,2-, and 5-year all-cause mortality were BS-ACMR (B: NT-pro BNP; S: ST2; A: age; C: complete right bundle branch block; M: mean arterial pressure; and R: red cell distribution width >14.5%); these were incorporated into the dynamic nomogram (https://bs-acmr-nom.shinyapps.io/dynnomapp/) with bootstrap validation. The C-indexes in the derivation (0.793) and validation (0.782) cohorts were consistent with comparable performance parameters. The calibration curve showed good agreement between the nomogram-predicted and actual survival. Adding NT-pro BNP and ST2 provided a significant net benefit and improved performance over other less adequate schemes in terms of DCA of survival probability compared to those neglecting either of these two factors. Conclusion The study constructed a dynamic BS-ACMR nomogram, which is a convenient, practical and effective clinical decision-making tool for providing accurate prognosis in AHF patients.
Collapse
Affiliation(s)
- Ting Yin
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, People’s Republic of China
| | - Shi Shi
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, People’s Republic of China
| | - Xu Zhu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, People’s Republic of China
| | - Iokfai Cheang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, People’s Republic of China
| | - Xinyi Lu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, People’s Republic of China
| | - Rongrong Gao
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, People’s Republic of China
| | - Haifeng Zhang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, People’s Republic of China
- Department of Cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, 215002, People’s Republic of China
| | - Wenming Yao
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, People’s Republic of China
| | - Yanli Zhou
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, People’s Republic of China
| | - Xinli Li
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, People’s Republic of China
- Correspondence: Xinli Li; Yanli Zhou, Tel +86 136 1157 3111; +86 137 7787 9077, Email ;
| |
Collapse
|
28
|
Kuna J, Żuber Z, Chmielewski G, Gromadziński L, Krajewska-Włodarczyk M. Role of Distinct Macrophage Populations in the Development of Heart Failure in Macrophage Activation Syndrome. Int J Mol Sci 2022; 23:2433. [PMID: 35269577 PMCID: PMC8910409 DOI: 10.3390/ijms23052433] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 02/18/2022] [Accepted: 02/21/2022] [Indexed: 12/12/2022] Open
Abstract
Macrophage activation syndrome (MAS) is one of the few entities in rheumatology with the potential to quickly cause multiple organ failure and loss of life, and as such, requires urgent clinical intervention. It has a broad symptomatology, depending on the organs it affects. One especially dangerous aspect of MAS's course of illness is myocarditis leading to acute heart failure and possibly death. Research in recent years has proved that macrophages settled in different organs are not a homogenous group, with particular populations differing in both structure and function. Within the heart, we can determine two major groups, based on the presence of the C-C 2 chemokine receptor (CCR2): CCR2+ and CCR2-. There are a number of studies describing their function and the changes in the population makeup between normal conditions and different illnesses; however, to our knowledge, there has not been one touching on the matter of changes occurring in the populations of heart macrophages during MAS and their possible consequences. This review summarizes the most recent knowledge on heart macrophages, the influence of select cytokines (those particularly significant in the development of MAS) on their activity, and both the immediate and long-term consequences of changes in the makeup of specific macrophage populations-especially the loss of CCR2- cells that are responsible for regenerative processes, as well as the substitution of tissue macrophages by the highly proinflammatory CCR2+ macrophages originating from circulating monocytes. Understanding the significance of these processes may lead to new discoveries that could improve the therapeutic methods in the treatment of MAS.
Collapse
Affiliation(s)
- Jakub Kuna
- Department of Rheumatology, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-900 Olsztyn, Poland;
| | - Zbigniew Żuber
- Department of Pediatrics, Faculty of Medicine and Health Sciences, Andrzej Frycz Modrzewski Kraków University, 30-705 Kraków, Poland;
| | - Grzegorz Chmielewski
- Department of Rheumatology, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-900 Olsztyn, Poland;
| | - Leszek Gromadziński
- Department of Cardiology and Internal Medicine, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-900 Olsztyn, Poland;
| | - Magdalena Krajewska-Włodarczyk
- Department of Rheumatology, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-900 Olsztyn, Poland;
| |
Collapse
|
29
|
Salzer U, Müller A, Zhou Q, Nieters A, Grundmann S, Wehr C. Susceptibility to infections and adaptive immunity in adults with heart failure. ESC Heart Fail 2022; 9:1195-1205. [PMID: 35032103 PMCID: PMC8934962 DOI: 10.1002/ehf2.13793] [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: 09/14/2021] [Revised: 11/25/2021] [Accepted: 12/14/2021] [Indexed: 12/11/2022] Open
Abstract
Aims Heart failure (HF) is a systemic inflammatory disorder with infections being an important cause of morbidity and mortality. We asked if HF patients have a higher susceptibility to infections compared with the general population and if a subtle secondary immunodeficiency facilitates infectious complications. Methods and results In a cohort of 92 patients with HF with reduced ejection fraction, we analysed recirculating lymphocyte subpopulations, serum immunoglobulin levels, and specific antibody titres against pneumococcal antigens. We quantified susceptibility to infections of the respiratory tract with a validated questionnaire and compared it to the general population. Susceptibility to infections of the respiratory tract was comparable in HF patients and the general population. Hypogammaglobulinaemia was present in 16% of HF patients, but anti‐pneumococcal titres showed no evidence of specific secondary antibody deficiency. Relative lymphopaenia in our HF cohort was due to B lymphocytopenia with a relative reduction in naive B‐cells and expansion of memory B‐cells while CD4+ and CD8+ T‐lymphocytes as well as NK‐cell counts were comparable between HF and healthy donors. The intake of the angiotensin receptor neprilysin (CD10) inhibitor (ARNI) sacubitril/valsartan was associated with increased B‐lymphocyte counts, possibly by an increased output of CD10+ transitional B lymphocytes from the bone marrow. Conclusion Despite a reduction of B lymphocytes in HF and mild hypogammaglobulinaemia, patients showed no evidence of secondary immunodeficiency or increased susceptibility to infections. The relevance of B‐cell lymphopenia in HF patients and modulation of B‐cell counts under ARNI treatment remains to be investigated.
Collapse
Affiliation(s)
- Ulrich Salzer
- Department of Rheumatology and Clinical Immunology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Alisa Müller
- Department of General Paediatrics, Children's Hospital, University of Bonn, Bonn, Germany.,Center for Chronic Immunodeficiency, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Qian Zhou
- University Heart Center Freiburg-Bad Krozingen, Department of Cardiology and Angiology I, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Department of Medicine, Division of Cardiology, University Hospital Basel, Basel, Switzerland
| | - Alexandra Nieters
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Sebastian Grundmann
- University Heart Center Freiburg-Bad Krozingen, Department of Cardiology and Angiology I, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Claudia Wehr
- Center for Chronic Immunodeficiency, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Department of Medicine I, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
30
|
Diabetes, Heart Failure and Beyond: Elucidating the Cardioprotective Mechanisms of Sodium Glucose Cotransporter 2 (SGLT2) Inhibitors. Am J Cardiovasc Drugs 2022; 22:35-46. [PMID: 34189716 DOI: 10.1007/s40256-021-00486-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/27/2021] [Indexed: 02/07/2023]
Abstract
Approximately 5 million individuals in the US are living with congestive heart failure (CHF), with 650,000 new cases being diagnosed every year. CHF has a multifactorial etiology, ranging from coronary artery disease, hypertension, valvular abnormalities and diabetes mellitus. Currently, guidelines by the American College of Cardiology advocate the use of angiotensin-converting enzyme (ACE) inhibitors, angiotensin II receptor blockers, β-blockers, diuretics, aldosterone antagonists, and inotropes for the medical management of heart failure. The sodium glucose cotransporter 2 (SGLT2) inhibitors are a class of drug that have been widely used in the management of type 2 diabetes mellitus that work by inhibiting the reabsorption of glucose in the proximal convoluted tubule. Since the EMPA-REG OUTCOME trial, several studies have demonstrated the benefits of SGLT2 inhibitors in reducing cardiovascular risk related to heart failure. While the cardiovascular benefits could be explained by their ability to reduce weight, improve glycemic index and lower blood pressure, several recent trials have suggested that SGLT2 inhibitors exhibit pleiotropic effects that underlie their cardioprotective properties. These findings have led to an expansion in preclinical and clinical research aiming to understand the mechanisms by which SGLT2 inhibitors improve heart failure outcomes.
Collapse
|
31
|
Reina-Couto M, Pereira-Terra P, Quelhas-Santos J, Silva-Pereira C, Albino-Teixeira A, Sousa T. Inflammation in Human Heart Failure: Major Mediators and Therapeutic Targets. Front Physiol 2021; 12:746494. [PMID: 34707513 PMCID: PMC8543018 DOI: 10.3389/fphys.2021.746494] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 09/20/2021] [Indexed: 12/28/2022] Open
Abstract
Inflammation has been recognized as a major pathophysiological contributor to the entire spectrum of human heart failure (HF), including HF with reduced ejection fraction, HF with preserved ejection fraction, acute HF and cardiogenic shock. Nevertheless, the results of several trials attempting anti-inflammatory strategies in HF patients have not been consistent or motivating and the clinical implementation of anti-inflammatory treatments for HF still requires larger and longer trials, as well as novel and/or more specific drugs. The present work reviews the different inflammatory mechanisms contributing to each type of HF, the major inflammatory mediators involved, namely tumor necrosis factor alpha, the interleukins 1, 6, 8, 10, 18, and 33, C-reactive protein and the enzymes myeloperoxidase and inducible nitric oxide synthase, and their effects on heart function. Furthermore, several trials targeting these mediators or involving other anti-inflammatory treatments in human HF are also described and analyzed. Future therapeutic advances will likely involve tailored anti-inflammatory treatments according to the patient's inflammatory profile, as well as the development of resolution pharmacology aimed at stimulating resolution of inflammation pathways in HF.
Collapse
Affiliation(s)
- Marta Reina-Couto
- Departamento de Biomedicina – Unidade de Farmacologia e Terapêutica, Faculdade de Medicina, Universidade do Porto, Porto, Portugal
- Centro de Investigação Farmacológica e Inovação Medicamentosa, Universidade do Porto (MedInUP), Porto, Portugal
- Departamento de Medicina Intensiva, Centro Hospitalar e Universitário São João, Porto, Portugal
| | - Patrícia Pereira-Terra
- Departamento de Biomedicina – Unidade de Farmacologia e Terapêutica, Faculdade de Medicina, Universidade do Porto, Porto, Portugal
| | - Janete Quelhas-Santos
- Departamento de Biomedicina – Unidade de Farmacologia e Terapêutica, Faculdade de Medicina, Universidade do Porto, Porto, Portugal
| | - Carolina Silva-Pereira
- Departamento de Biomedicina – Unidade de Farmacologia e Terapêutica, Faculdade de Medicina, Universidade do Porto, Porto, Portugal
- Centro de Investigação Farmacológica e Inovação Medicamentosa, Universidade do Porto (MedInUP), Porto, Portugal
| | - António Albino-Teixeira
- Departamento de Biomedicina – Unidade de Farmacologia e Terapêutica, Faculdade de Medicina, Universidade do Porto, Porto, Portugal
- Centro de Investigação Farmacológica e Inovação Medicamentosa, Universidade do Porto (MedInUP), Porto, Portugal
| | - Teresa Sousa
- Departamento de Biomedicina – Unidade de Farmacologia e Terapêutica, Faculdade de Medicina, Universidade do Porto, Porto, Portugal
- Centro de Investigação Farmacológica e Inovação Medicamentosa, Universidade do Porto (MedInUP), Porto, Portugal
| |
Collapse
|
32
|
Tourki B, Halade GV. Heart Failure Syndrome With Preserved Ejection Fraction Is a Metabolic Cluster of Non-resolving Inflammation in Obesity. Front Cardiovasc Med 2021; 8:695952. [PMID: 34409075 PMCID: PMC8367012 DOI: 10.3389/fcvm.2021.695952] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 06/08/2021] [Indexed: 12/20/2022] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is an emerging disease with signs of nonresolving inflammation, endothelial dysfunction, and multiorgan defects. Moreover, based on the clinical signs and symptoms and the rise of the obesity epidemic, the number of patients developing HFpEF is increasing. From recent molecular and cellular studies, it becomes evident that HFpEF is not a single and homogenous disease but a cluster of heterogeneous pathophysiology with aging at the base of the pyramid. Obesity superimposed on aging drives the number of inflammatory pathways that intersect with metabolic dysfunction and suboptimal inflammation. Here, we compiled information on obesity-directed macrophage dysfunction that coincide with metabolic defects. Obesity-associated proinflammatory stimuli facilitates heart and interorgan inflammation in HFpEF. Furthermore, diversified mechanisms that drive heart failure urge the need of studying pervasive and unresolved inflammation in animal models to understand HFpEF. A broad and system-based approach will help to study major translational aspects of HFpEF, since no single animal model recapitulates all signs of differential HFpEF stages in the clinical setting. Here, we covered experimental models that target HFpEF and emphasized the advances observed with formyl peptide 2 (FPR2) receptor, a prime sensor that is important in inflammation-resolution signaling. Dysfunction of FPR2 led to the development of spontaneous obesity, impaired macrophage function, and triggered kidney fibrosis, providing evidence of multiorgan defects in HFpEF in an obesogenic aging experimental model.
Collapse
Affiliation(s)
- Bochra Tourki
- Division of Cardiovascular Sciences, Department of Medicine, The University of South Florida, Tampa, FL, United States
| | - Ganesh V Halade
- Division of Cardiovascular Sciences, Department of Medicine, The University of South Florida, Tampa, FL, United States
| |
Collapse
|
33
|
Xiang N, Liao H, Zhai Z, Gong J. Expression and significance of inflammatory reactions mediated by the IL-33/ST2 signaling pathway in the serum of heart failure patients. Am J Transl Res 2021; 13:8247-8252. [PMID: 34377313 PMCID: PMC8340166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 04/01/2021] [Indexed: 06/13/2023]
Abstract
OBJECTIVE This research aimed to explore the clinical significance of inflammatory reactions mediated by the IL-33/ST2 signaling pathway in heart failure (HF) patients. METHODS A total of 100 HF patients treated in the Department of Cardiology in our hospital were prospectively regarded as the observation group, and 100 healthy age and gender matched patients who were undergoing physical examination were considered as the control group. The levels of interleukin-33 (IL-33), ST2, tumor necrosis factor-α (TNF-α) and pro B-type natriuretic peptide (pro-BNP) in the peripheral blood of patients were detected. The potential correlation between IL-33 and ST2, TNF-α and pro-BNP was analyzed by Pearson. RESULTS The levels of IL-33, IL-10, ST2 and pro-BNP in the peripheral blood of patients in the observation group were higher than those in the control group; and they increased with the rise of cardiac function grade (all P<0.05). In addition, IL-33 was positively correlated with TNF-α, ST2 and pro-BNP (r=0.863, 0.879, 0.945; all P<0.05). Multivariate Logistic analysis revealed that the increase of IL-33 and ST2 were independent risk factors of HF. CONCLUSION The IL-33 and ST2 levels in the peripheral serum of HF patients are correlated with TNF-α and BNP, the finding of which can assist in clinical diagnosis and treatment.
Collapse
Affiliation(s)
- Ning Xiang
- Department of Geriatrics, Affiliated Hospital of Guizhou Medical UniversityGuiyang, Guizhou Province, China
| | - Haijun Liao
- Guizhou Medical UniversityGuiyang, Guizhou Province, China
| | - Zichen Zhai
- Guizhou Medical UniversityGuiyang, Guizhou Province, China
| | - Jingwen Gong
- Guizhou Medical UniversityGuiyang, Guizhou Province, China
| |
Collapse
|
34
|
Druzhaeva N, Nemec Svete A, Ihan A, Pohar K, Domanjko Petrič A. Peripheral blood lymphocyte subtypes in dogs with different stages of myxomatous mitral valve disease. J Vet Intern Med 2021; 35:2112-2122. [PMID: 34236111 PMCID: PMC8478039 DOI: 10.1111/jvim.16207] [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: 02/28/2021] [Revised: 06/14/2021] [Accepted: 06/18/2021] [Indexed: 12/19/2022] Open
Abstract
Background Data on alterations in peripheral blood lymphocyte (PBL) subtypes in dogs with myxomatous mitral valve disease (MMVD) is lacking. Objectives To investigate PBL subtypes and their correlation with parameters of inflammation and MMVD progression markers in dogs with different stages of MMVD. Animals Seventy‐eight client‐owned dogs: 65 with MMVD (American College of Veterinary Internal Medicine [ACVIM] classification stages B2, C, and D) and 13 healthy controls. Methods Prospective cross‐sectional study. Complete cardiac assessment, flow cytometry (T lymphocytes [CD3+], their subtypes [CD3+CD4+, CD3+CD8+, CD3+CD4+CD8+, CD3+CD4−CD8−], and B lymphocytes [CD45+CD21+]) and measurement of N‐terminal pro B‐type natriuretic peptide, cardiac troponin I, and C‐reactive protein concentrations were performed. Results The percentage of CD3+CD4+ lymphocytes was significantly lower in stable ACVIM C patients (P = .01) and unstable ACVIM C and D patients (P = .003), the percentage of CD3+CD8+ lymphocytes was significantly higher in stable ACVIM C patients (P = .01) and unstable ACVIM C and D patients (P = .01), CD3+CD8+ lymphocyte concentration was significantly higher in unstable ACVIM C and D patients (P = .05), and the CD3+CD4+/CD3+CD8+ ratio was significantly lower in stable ACVIM C patients (P = .01) and unstable ACVIM C and D patients (P = .01) compared with healthy controls. Conclusions and Clinical Importance The percentages of CD3+CD4+ and CD3+CD8+ PBL and CD4+/CD8+ ratio were altered in MMVD dogs with congestive heart failure (ACVIM C, D), but not in ACVIM B2, suggesting involvement of these PBL subtypes in the pathogenesis of congestive heart failure in dogs with MMVD.
Collapse
Affiliation(s)
- Natalia Druzhaeva
- Small Animal Clinic, Veterinary Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Alenka Nemec Svete
- Small Animal Clinic, Veterinary Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Alojz Ihan
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Katka Pohar
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | | |
Collapse
|
35
|
Lin CY, Chen HA, Hsu TC, Wu CH, Su YJ, Hsu CY. Time-Dependent Analysis of Risk of New-Onset Heart Failure Among Patients With Polymyositis and Dermatomyositis. Arthritis Rheumatol 2021; 74:140-149. [PMID: 34180158 DOI: 10.1002/art.41907] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 05/30/2021] [Accepted: 06/23/2021] [Indexed: 12/23/2022]
Abstract
OBJECTIVE To determine the risk and time trends of heart failure (HF) leading to hospitalization in individuals newly diagnosed as having polymyositis/dermatomyositis (PM/DM) relative to non-PM/DM controls at the general population level. METHODS A retrospective cohort study was conducted using data from a nationwide insurance database in Taiwan. Patients with incident PM/DM and without a history of HF were selected between 2000 and 2013. Unmatched and propensity score-matched cohorts were established separately. A multivariable Cox proportional hazards regression model was used to estimate the adjusted hazard ratio (HR) for the risk of HF in the unmatched cohort. In the propensity score-matched cohort, general population controls were selected and matched at a 1:1 ratio to the patients with PM/DM based on propensity scores, which accounted for the confounding factors of age, sex, index date (year) of first diagnosis, comorbidities, and medication usage. The cumulative incidence of HF was estimated using the Kaplan-Meier method. A stratified Cox proportional hazards model was used to calculate the HR for the risk of HF events at different follow-up time points among patients with PM/DM compared with non-PM/DM controls in the propensity score-matched cohort. RESULTS In the unmatched cohort, the study assessed 2,025 patients with PM/DM and 196,109 general population controls. Results of multivariable Cox regression analysis, adjusted for age, sex, comorbidities, and medication usage, revealed a greater risk of HF leading to hospitalization in the PM/DM group than in the control group (adjusted HR 3.29, 95% confidence interval [95% CI] 2.60-4.18). After matching based on propensity score, a total of 1,997 pairs of PM/DM patients and general population controls were identified. In this propensity score-matched cohort, the cumulative incidence of HF in patients with PM/DM at 3 years, 5 years, and 10 years was 3.3%, 4.4%, and 7.4%, respectively. The absolute difference in HF risk in the PM/DM group compared with the control group was 1.8% at 3 years, 2.1% at 5 years, and 3.0% at 10 years. Compared with general population controls, patients with PM/DM exhibited an augmented risk of HF (HR 2.06, 95% CI 1.36-3.12). Analyses stratified according to follow-up time point revealed that the increased risk of HF persisted for up to 10 years after the PM/DM diagnosis. CONCLUSION These results indicate that the risk of HF leading to hospitalization was increased in patients with PM/DM throughout the study period, supporting the need for greater vigilance in the monitoring of patients with PM/DM for the development of this potentially lethal complication.
Collapse
Affiliation(s)
- Chun-Yu Lin
- Division of Rheumatology, Allergy, and Immunology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Hung-An Chen
- Department of Internal Medicine, Chi Mei Medical Center, Tainan, Taiwan
| | | | - Chun-Hsin Wu
- Division of Rheumatology, Allergy, and Immunology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yu-Jih Su
- Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Chung-Yuan Hsu
- Division of Rheumatology, Allergy, and Immunology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, and School of Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
36
|
Li X, Liu S, Qu L, Chen Y, Yuan C, Qin A, Liang J, Huang Q, Jiang M, Zou W. Dioscin and diosgenin: Insights into their potential protective effects in cardiac diseases. JOURNAL OF ETHNOPHARMACOLOGY 2021; 274:114018. [PMID: 33716083 DOI: 10.1016/j.jep.2021.114018] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 02/07/2021] [Accepted: 03/07/2021] [Indexed: 06/12/2023]
Abstract
BACKGROUND AND ETHNOPHARMACOLOGICAL RELEVANCE Dioscin and diosgenin derived from plants of the genus Dioscoreaceae such as D. nipponica and D. panthaica Prain et Burk. Were utilized as the main active ingredients of traditional herbal medicinal products for coronary heart disease in the former Soviet Union and China since 1960s. A growing number of research showed that dioscin and diosgenin have a wide range of pharmacological activities in heart diseases. AIM OF THE STUDY To summarize the evidence of the effectiveness of dioscin and diosgenin in cardiac diseases, and to provide a basis and reference for future research into their clinical applications and drug development in the field of cardiac disease. METHODS Literatures in this review were searched in PubMed, ScienceDirect, Google Scholar, China National Knowledge Infrastructure (CNKI) and Web of Science. All eligible studies are analyzed and summarized in this review. RESULTS The pharmacological activities and therapeutic potentials of dioscin and diosgenin in cardiac diseases are similar, can effectively improve hypertrophic cardiomyopathy, arrhythmia, myocardial I/R injury and cardiotoxicity caused by doxorubicin. But the bioavailability of dioscin and diosgenin may be too low as a result of poor absorption and slow metabolism, which hinders their development and utilization. CONCLUSION Dioscin and diosgenin need further in-depth experimental research, clinical transformation and structural modification or research of new preparations before they can be expected to be developed into new therapeutic drugs in the field of cardiac disease.
Collapse
Affiliation(s)
- Xiaofen Li
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Sili Liu
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Liping Qu
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Yang Chen
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Chuqiao Yuan
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Anquan Qin
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Jiyi Liang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Qianqian Huang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Miao Jiang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Wenjun Zou
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| |
Collapse
|
37
|
Zayas-Arrabal J, Alquiza A, Tuncay E, Turan B, Gallego M, Casis O. Molecular and Electrophysiological Role of Diabetes-Associated Circulating Inflammatory Factors in Cardiac Arrhythmia Remodeling in a Metabolic-Induced Model of Type 2 Diabetic Rat. Int J Mol Sci 2021; 22:ijms22136827. [PMID: 34202017 PMCID: PMC8268936 DOI: 10.3390/ijms22136827] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/16/2021] [Accepted: 06/22/2021] [Indexed: 12/29/2022] Open
Abstract
Background: Diabetic patients have prolonged cardiac repolarization and higher risk of arrhythmia. Besides, diabetes activates the innate immune system, resulting in higher levels of plasmatic cytokines, which are described to prolong ventricular repolarization. Methods: We characterize a metabolic model of type 2 diabetes (T2D) with prolonged cardiac repolarization. Sprague-Dawley rats were fed on a high-fat diet (45% Kcal from fat) for 6 weeks, and a low dose of streptozotozin intraperitoneally injected at week 2. Body weight and fasting blood glucose were measured and electrocardiograms of conscious animals were recorded weekly. Plasmatic lipid profile, insulin, cytokines, and arrhythmia susceptibility were determined at the end of the experimental period. Outward K+ currents and action potentials were recorded in isolated ventricular myocytes by patch-clamp. Results: T2D animals showed insulin resistance, hyperglycemia, and elevated levels of plasma cholesterol, triglycerides, TNFα, and IL-1b. They also developed bradycardia and prolonged QTc-interval duration that resulted in increased susceptibility to severe ventricular tachycardia under cardiac challenge. Action potential duration (APD) was prolonged in control cardiomyocytes incubated 24 h with plasma isolated from diabetic rats. However, adding TNFα and IL-1b receptor blockers to the serum of diabetic animals prevented the increased APD. Conclusions: The elevation of the circulating levels of TNFα and IL-1b are responsible for impaired ventricular repolarization and higher susceptibility to cardiac arrhythmia in our metabolic model of T2D.
Collapse
Affiliation(s)
- Julian Zayas-Arrabal
- Departament of Physiology, Facultad de Farmacia, Universidad del País Vasco UPV/EHU, 01006 Vitoria-Gasteiz, Spain; (J.Z.-A.); (A.A.); (M.G.)
| | - Amaia Alquiza
- Departament of Physiology, Facultad de Farmacia, Universidad del País Vasco UPV/EHU, 01006 Vitoria-Gasteiz, Spain; (J.Z.-A.); (A.A.); (M.G.)
| | - Erkan Tuncay
- Department of Biophysics, Faculty of Medicine, Ankara University, 06100 Ankara, Turkey;
| | - Belma Turan
- Department of Biophysics, Faculty of Medicine, Lokman Hekim University, 06510 Ankara, Turkey;
| | - Monica Gallego
- Departament of Physiology, Facultad de Farmacia, Universidad del País Vasco UPV/EHU, 01006 Vitoria-Gasteiz, Spain; (J.Z.-A.); (A.A.); (M.G.)
| | - Oscar Casis
- Departament of Physiology, Facultad de Farmacia, Universidad del País Vasco UPV/EHU, 01006 Vitoria-Gasteiz, Spain; (J.Z.-A.); (A.A.); (M.G.)
- Correspondence: ; Tel.: +34-945013033
| |
Collapse
|
38
|
Li S, Liu H, Li Y, Qin X, Li M, Shang J, Xing W, Gong Y, Liu W, Zhou M. Shen-Yuan-Dan Capsule Attenuates Verapamil-Induced Zebrafish Heart Failure and Exerts Antiapoptotic and Anti-Inflammatory Effects via Reactive Oxygen Species-Induced NF-κB Pathway. Front Pharmacol 2021; 12:626515. [PMID: 33732158 PMCID: PMC7959770 DOI: 10.3389/fphar.2021.626515] [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: 11/06/2020] [Accepted: 01/15/2021] [Indexed: 01/08/2023] Open
Abstract
Background: Heart failure (HF) is the end stage of ischemic cardiovascular diseases; nonetheless, safe and effective therapeutic agents for HF are still lacking, and their discovery remains challenging. Our previous studies demonstrated that Shen-Yuan-Dan Capsule (SYDC), a hospital preparation of traditional Chinese herbal, effectively protected ischemic injury in cardiovascular diseases. However, its therapeutic effects and possible mechanisms on HF remain unclear. Methods: A zebrafish HF model treated with verapamil was developed to assess the therapeutic effect of SYDC on HF zebrafish. Zebrafish were administered with SYDC and digoxin (positive control) by direct soaking. After drug treatment, zebrafish were randomly assigned to the visual observation and image acquisition using a Zebralab Blood Flow System. The reactive oxygen species (ROS), MDA, and SOD levels were determined by fluorescence signal detection, TBA, and WST-8 methods. RT-PCR determined the mRNA expressions of Caspase-3, Caspase-1, Bcl-2, Bax, IL-1β, NF-κB, and TNF-α. Results: SYDC significantly inhibited the levels of heart dilatation and venous congestion and markedly increased the levels of cardiac output, blood flow dynamics, and heart rates in HF zebrafish (p < 0.05, p < 0.01, and p < 0.001). Moreover, SYDC also significantly decreased the levels of MDA and ROS and increased the level of SOD in HF zebrafish. The RT-PCR results revealed that SYDC decreased the expression of Caspase-1, Caspase-3, Bax, IL-1β, NF-κB, and TNF-α but increased the expression of Bcl-2 in HF zebrafish (p < 0.05, p < 0.01, and p < 0.001). Conclusions: SYDC improved the heart function in verapamil-induced HF zebrafish and alleviated inflammation and apoptosis by inhibiting the ROS-mediated NF-κB pathway.
Collapse
Affiliation(s)
- Sinai Li
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, Beijing, China
| | - Hongxu Liu
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Yue Li
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, Beijing, China
| | - Xiaomei Qin
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, Beijing, China
| | - Mengjie Li
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, Beijing, China
| | - Juju Shang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Wenlong Xing
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Yanbing Gong
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Weihong Liu
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, Beijing, China
| | - Mingxue Zhou
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, Beijing, China
| |
Collapse
|
39
|
Purwowiyoto SL, Prawara AS. Metabolic syndrome and heart failure: mechanism and management. Med Pharm Rep 2021; 94:15-21. [PMID: 33629043 PMCID: PMC7880077 DOI: 10.15386/mpr-1884] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 10/15/2020] [Accepted: 11/06/2020] [Indexed: 12/14/2022] Open
Abstract
Heart failure (HF) and metabolic syndrome (MetS) are syndromes that affect a large proportion of the world population. MetS is known to be one of the risk factors of HF, and it can also act as comorbidity in HF. This review aims to further discuss the mechanism of MetS in causing HF, the management of MetS in order to prevent HF, and the management of MetS in HF patients. Visceral adiposity is the primary trigger of MetS which is followed by chronic inflammation, insulin resistance, and neurohormonal activation. All the mechanisms causing MetS play also an important role in the progression of HF. The MetS approach can be achieved by managing its components according to the current guidelines and careful management of MetS should be done in patients with HF. MetS is closely related to the progression of HF so that comprehensive management which involves a multidisciplinary team is necessary for managing patients with metabolic syndrome and heart failure.
Collapse
Affiliation(s)
- Sidhi Laksono Purwowiyoto
- Cardiac Catheterization Laboratory, Department of Cardiology and Vascular Medicine, RSUD Pasar Rebo, East Jakarta, Indonesia.,Faculty of Medicine, Universitas Muhammadiyah Prof. DR. Hamka, Tangerang, Indonesia
| | | |
Collapse
|
40
|
Gupta AK, Tomasoni D, Sidhu K, Metra M, Ezekowitz JA. Evidence-Based Management of Acute Heart Failure. Can J Cardiol 2021; 37:621-631. [PMID: 33440229 DOI: 10.1016/j.cjca.2021.01.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 12/21/2020] [Accepted: 01/04/2021] [Indexed: 10/22/2022] Open
Abstract
Acute heart failure (AHF) is a complex, heterogeneous, clinical syndrome with high morbidity and mortality, incurring significant health care costs. Patients transition from home to the emergency department, the hospital, and home again and require decisions surrounding diagnosis, treatment, and prognosis at each step of the way. The purpose of this review is to examine the epidemiology, etiology, and classifications of AHF and specifically focus on practical information relevant to the clinician. We examine the mechanisms of decompensation relevant to clinical presentations-including precipitating factors, neuroendocrine interactions, and inflammation-along with how consideration of these factors may help select therapies for an individual patient. The prevalence and significance of end-organ manifestations such as renal, gastrointestinal, respiratory, and neurologic manifestations are discussed. We also highlight how the development of renal dysfunction relates to the choice of a variety of diuretics that may be useful in specific circumstances and review guideline-directed medical therapy. We discuss the practical use (and pitfalls) of a variety of evidence-based clinical scoring criteria available to risk stratify patients with AHF. Finally, evidence-based management of AHF is discussed, including both pharmacologic and nonpharmacologic therapies, including the lack of evidence for using old and new vasodilators and the recent evidence regarding initiation of newer therapies in hospital. Overall, we suggest that clinicians consider implementing the newer data in AHF and subject existing practice patterns and treatments to the same rigour as new therapies.
Collapse
Affiliation(s)
- Arjun K Gupta
- Division of Cardiology, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Daniela Tomasoni
- Institute of Cardiology, ASST Spedali Civili di Brescia and Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia, Brescia, Italy
| | - Kiran Sidhu
- Section of Cardiology, Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Marco Metra
- Institute of Cardiology, ASST Spedali Civili di Brescia and Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia, Brescia, Italy
| | - Justin A Ezekowitz
- Division of Cardiology, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada; Canadian VIGOUR Centre, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
41
|
Khoshkhouy F, Farshbaf A, Mahmoudabady M, Gholamnezhad Z. Effects of moderate exercise on lipopolysaccharide-induced inflammatory responses in rat's cardiac tissue. Cytokine 2020; 138:155409. [PMID: 33360764 DOI: 10.1016/j.cyto.2020.155409] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 12/13/2020] [Accepted: 12/14/2020] [Indexed: 01/03/2023]
Abstract
The effects of moderate exercise on cardiac tissue inflammation, oxidative stress markers and apoptosis in lipopolysaccharide (LPS)-administered rats were evaluated. Wistar rats were divided into three groups (N = 8): (1) control; (2) LPS (1 mg/kg); and (3) LPS + moderate training (LPS + EX: 15 m/min, 30 min/day, for 9 weeks (week 1-9)). LPS was injected intraperitoneally for 5 days during week 9. Finally, the rats' heart were removed for biochemical and expression assessments. LPS increased the levels of tumor necrosis factor α (TNF-α), interleukin (IL)- 1β, C-reactive protein (CRP), malondialdehyde (MDA) and nitric oxide (NO) metabolites in cardiac tissue, but decreased thiol contents and catalase (CAT) and superoxide dismutase (SOD) activity in cardiac tissue compared to the control group (p < 0.05-p < 0.001). In LPS + EX group, the level of NO metabolites was increased (p < 0.05) and thiol contents were decreased (p < 0.001) compared to the control group. Moderate training decreased the levels of TNF-α, IL-1β, CRP and NO metabolites while increased CAT activity in the LPS + EX group compared to the LPS group (p < 0.05-p < 0.001). The mRNA level of BAX in the LPS group and the BCL2/BAX ratio in both LPS and LPS + EX groups increased compared to the control group (p < 0.05-p < 0.01). These results indicated that moderate training improved LPS-induced deleterious effects on cardiac tissue by attenuating proinflammatory cytokine levels, apoptosis and oxidative damage.
Collapse
Affiliation(s)
- Fatemeh Khoshkhouy
- Department of Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alieh Farshbaf
- Dental Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maryam Mahmoudabady
- Department of Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zahra Gholamnezhad
- Department of Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
42
|
Topf A, Mirna M, Ohnewein B, Jirak P, Kopp K, Fejzic D, Haslinger M, Motloch LJ, Hoppe UC, Berezin A, Lichtenauer M. The Diagnostic and Therapeutic Value of Multimarker Analysis in Heart Failure. An Approach to Biomarker-Targeted Therapy. Front Cardiovasc Med 2020; 7:579567. [PMID: 33344515 PMCID: PMC7746655 DOI: 10.3389/fcvm.2020.579567] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 11/02/2020] [Indexed: 12/12/2022] Open
Abstract
Background: Heart failure is a pathophysiological state, which is still associated with high morbidity and mortality despite established therapies. Diverse well-known biomarkers fail to assess the variety of individual pathophysiology in the context of heart failure. Methods: An analysis of prospective, multimarker-specific therapeutic approaches to heart failure based on studies in current literature was performed. A total of 159 screened publications in the field of biomarkers in heart failure were hand-selected and found to be eligible for this study by a team of experts. Results: Established biomarkers of the inflammatory axis, matrix remodeling, fibrosis and oxidative stress axis, as well as potential therapeutic interventions were investigated. Interaction with end organs, such as cardio-hepatic, cardio-renal and cardio-gastrointestinal interactions show the complexity of the syndrome and could be of further therapeutic value. MicroRNAs are involved in a wide variety of physiologic and pathophysiologic processes in heart failure and could be useful in diagnostic as well as therapeutic setting. Conclusion: Based on our analysis by a biomarker-driven approach in heart failure therapy, patients could be treated more specifically in long term with a consideration of different aspects of heart failure. New studies evaluating a multimarker – based therapeutic approach could lead in a decrease in the morbidity and mortality of heart failure patients.
Collapse
Affiliation(s)
- Albert Topf
- Department of Cardiology, Clinic of Internal Medicine II, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Moritz Mirna
- Department of Cardiology, Clinic of Internal Medicine II, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Bernhard Ohnewein
- Department of Cardiology, Clinic of Internal Medicine II, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Peter Jirak
- Department of Cardiology, Clinic of Internal Medicine II, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Kristen Kopp
- Department of Cardiology, Clinic of Internal Medicine II, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Dzeneta Fejzic
- Department of Cardiology, Clinic of Internal Medicine II, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Michael Haslinger
- Department of Cardiology, Clinic of Internal Medicine II, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Lukas J Motloch
- Department of Cardiology, Clinic of Internal Medicine II, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Uta C Hoppe
- Department of Cardiology, Clinic of Internal Medicine II, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Alexander Berezin
- Internal Medicine Department, State Medical University, Zaporozhye, Ukraine
| | - Michael Lichtenauer
- Department of Cardiology, Clinic of Internal Medicine II, Paracelsus Medical University of Salzburg, Salzburg, Austria
| |
Collapse
|
43
|
Dutka M, Bobiński R, Ulman-Włodarz I, Hajduga M, Bujok J, Pająk C, Ćwiertnia M. Sodium glucose cotransporter 2 inhibitors: mechanisms of action in heart failure. Heart Fail Rev 2020; 26:603-622. [PMID: 33150520 PMCID: PMC8024236 DOI: 10.1007/s10741-020-10041-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/12/2020] [Indexed: 12/16/2022]
Abstract
Diabetes is a key independent risk factor in the development of heart failure (HF) and a strong, adverse prognostic factor in HF patients. HF remains the primary cause of hospitalisation for diabetics and, as previous studies have shown, when HF occurs in these patients, intensive glycaemic control does not directly improve the prognosis. Recent clinical studies assessing a new class of antidiabetic drugs, sodium-glucose cotransporter 2 inhibitors (SGLT2is) showed some unexpected beneficial results. Patients treated with SGLT2is had a significant decrease in both cardiovascular (CV) and all-cause mortality and less hospitalisations due to HF compared to those given a placebo. These significant clinical benefits occurred quickly after the drugs were administered and were not solely due to improved glycaemic control. These groundbreaking clinical trials’ results have already changed clinical practice in the management of patients with diabetes at high CV risk. These trials have triggered numerous experimental studies aimed at explaining the mechanisms of action of this unique group of drugs. This article presents the current state of knowledge about the mechanisms of action of SGLT2is developed for the treatment of diabetes and which, thanks to their cardioprotective effects, may, in the future, become a treatment for patients with HF.
Collapse
Affiliation(s)
- Mieczysław Dutka
- Faculty of Health Sciences, Department of Biochemistry and Molecular Biology, University of Bielsko-Biała, Willowa St. 2, 43-309, Bielsko-Biała, Poland.
| | - Rafał Bobiński
- Faculty of Health Sciences, Department of Biochemistry and Molecular Biology, University of Bielsko-Biała, Willowa St. 2, 43-309, Bielsko-Biała, Poland
| | - Izabela Ulman-Włodarz
- Faculty of Health Sciences, Department of Biochemistry and Molecular Biology, University of Bielsko-Biała, Willowa St. 2, 43-309, Bielsko-Biała, Poland
| | - Maciej Hajduga
- Faculty of Health Sciences, Department of Biochemistry and Molecular Biology, University of Bielsko-Biała, Willowa St. 2, 43-309, Bielsko-Biała, Poland
| | - Jan Bujok
- Faculty of Health Sciences, Department of Biochemistry and Molecular Biology, University of Bielsko-Biała, Willowa St. 2, 43-309, Bielsko-Biała, Poland
| | - Celina Pająk
- Faculty of Health Sciences, Department of Biochemistry and Molecular Biology, University of Bielsko-Biała, Willowa St. 2, 43-309, Bielsko-Biała, Poland
| | - Michał Ćwiertnia
- Faculty of Health Sciences, Department of Emergency Medicine, University of Bielsko-Biała, Willowa St. 2, 43-309, Bielsko-Biała, Poland
| |
Collapse
|
44
|
Mayr F, Möller G, Garscha U, Fischer J, Rodríguez Castaño P, Inderbinen SG, Temml V, Waltenberger B, Schwaiger S, Hartmann RW, Gege C, Martens S, Odermatt A, Pandey AV, Werz O, Adamski J, Stuppner H, Schuster D. Finding New Molecular Targets of Familiar Natural Products Using In Silico Target Prediction. Int J Mol Sci 2020; 21:E7102. [PMID: 32993084 PMCID: PMC7582679 DOI: 10.3390/ijms21197102] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 09/19/2020] [Accepted: 09/21/2020] [Indexed: 12/01/2022] Open
Abstract
Natural products comprise a rich reservoir for innovative drug leads and are a constant source of bioactive compounds. To find pharmacological targets for new or already known natural products using modern computer-aided methods is a current endeavor in drug discovery. Nature's treasures, however, could be used more effectively. Yet, reliable pipelines for the large-scale target prediction of natural products are still rare. We developed an in silico workflow consisting of four independent, stand-alone target prediction tools and evaluated its performance on dihydrochalcones (DHCs)-a well-known class of natural products. Thereby, we revealed four previously unreported protein targets for DHCs, namely 5-lipoxygenase, cyclooxygenase-1, 17β-hydroxysteroid dehydrogenase 3, and aldo-keto reductase 1C3. Moreover, we provide a thorough strategy on how to perform computational target predictions and guidance on using the respective tools.
Collapse
Affiliation(s)
- Fabian Mayr
- Institute of Pharmacy/Pharmacognosy, Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria; (F.M.); (V.T.); (B.W.); (S.S.); (H.S.)
| | - Gabriele Möller
- Research Unit Molecular Endocrinology and Metabolism, Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764 Neuherberg, Germany; (G.M.); (J.A.)
| | - Ulrike Garscha
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, University Greifswald, Friedrich-Ludwig-Jahn-Straße 17, 17489 Greifswald, Germany; (U.G.); (J.F.)
| | - Jana Fischer
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, University Greifswald, Friedrich-Ludwig-Jahn-Straße 17, 17489 Greifswald, Germany; (U.G.); (J.F.)
| | - Patricia Rodríguez Castaño
- Pediatric Endocrinology, Diabetology and Metabolism, University Children’s Hospital Bern, Freiburgstrasse 15, 3010 Bern, Switzerland; (P.R.C.); (A.V.P.)
- Department of Biomedical Research, University of Bern, Freiburgstrasse 15, 3010 Bern, Switzerland
| | - Silvia G. Inderbinen
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland; (S.G.I.); (A.O.)
| | - Veronika Temml
- Institute of Pharmacy/Pharmacognosy, Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria; (F.M.); (V.T.); (B.W.); (S.S.); (H.S.)
| | - Birgit Waltenberger
- Institute of Pharmacy/Pharmacognosy, Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria; (F.M.); (V.T.); (B.W.); (S.S.); (H.S.)
| | - Stefan Schwaiger
- Institute of Pharmacy/Pharmacognosy, Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria; (F.M.); (V.T.); (B.W.); (S.S.); (H.S.)
| | - Rolf W. Hartmann
- Helmholtz Institute of Pharmaceutical Research Saarland (HIPS), Department for Drug Design and Optimization, Campus E8.1, 66123 Saarbrücken, Germany;
- Saarland University, Pharmaceutical and Medicinal Chemistry, Campus E8.1, 66123 Saarbrücken, Germany
| | - Christian Gege
- University of Heidelberg, Institute of Pharmacy and Molecular Biotechnology (IPMB), Medicinal Chemistry, Im Neuenheimer Feld 364, 69120 Heidelberg, Germany;
| | - Stefan Martens
- Research and Innovation Centre, Fondazione Edmund Mach (FEM), Via Mach 1, 38010 San Michele all’Adige, Italy;
| | - Alex Odermatt
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland; (S.G.I.); (A.O.)
| | - Amit V. Pandey
- Pediatric Endocrinology, Diabetology and Metabolism, University Children’s Hospital Bern, Freiburgstrasse 15, 3010 Bern, Switzerland; (P.R.C.); (A.V.P.)
- Department of Biomedical Research, University of Bern, Freiburgstrasse 15, 3010 Bern, Switzerland
| | - Oliver Werz
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University Jena, Philosophenweg 14, 07743 Jena, Germany;
| | - Jerzy Adamski
- Research Unit Molecular Endocrinology and Metabolism, Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764 Neuherberg, Germany; (G.M.); (J.A.)
- Lehrstuhl für Experimentelle Genetik, Technische Universität München, Emil-Erlenmeyer-Forum 5, 85356 Freising-Weihenstephan, Germany
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, 8 Medical Drive, Singapore 117597, Singapore
| | - Hermann Stuppner
- Institute of Pharmacy/Pharmacognosy, Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria; (F.M.); (V.T.); (B.W.); (S.S.); (H.S.)
| | - Daniela Schuster
- Institute of Pharmacy, Department of Pharmaceutical and Medicinal Chemistry, Paracelsus Medical University Salzburg, Strubergasse 21, 5020 Salzburg, Austria
- Institute of Pharmacy/Pharmaceutical Chemistry, Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria
| |
Collapse
|
45
|
Wang J, Deng B, Liu Q, Huang Y, Chen W, Li J, Zhou Z, Zhang L, Liang B, He J, Chen Z, Yan C, Yang Z, Xian S, Wang L. Pyroptosis and ferroptosis induced by mixed lineage kinase 3 (MLK3) signaling in cardiomyocytes are essential for myocardial fibrosis in response to pressure overload. Cell Death Dis 2020; 11:574. [PMID: 32710001 PMCID: PMC7382480 DOI: 10.1038/s41419-020-02777-3] [Citation(s) in RCA: 138] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 07/08/2020] [Accepted: 07/13/2020] [Indexed: 12/17/2022]
Abstract
Chronic heart failure (CHF) is the final outcome of many cardiovascular diseases, and is a severe health issue faced by the elderly population. Mixed lineage kinase 3 (MLK3), a member of MAP3K family, is associated with aging, inflammation, oxidative stress, and related diseases, such as CHF. MLK3 has also been reported to play an important role in protecting against cardiomyocyte injury; however, its function in myocardial fibrosis is unknown. To investigate the role of MLK3 in myocardial fibrosis, we inhibited the expression of MLK3, and examined cardiac function and remodeling in TAC mice. In addition, we assessed the expression of MLK3 protein in ventricular cells and its downstream associated protein. We found that MLK3 mainly regulates NF-κB/NLRP3 signaling pathway-mediated inflammation and that pyroptosis causes myocardial fibrosis in the early stages of CHF. Similarly, MLK3 mainly regulates the JNK/p53 signaling pathway-mediated oxidative stress and that ferroptosis causes myocardial fibrosis in the advanced stages of CHF. We also found that promoting the expression of miR-351 can inhibit the expression of MLK3, and significantly improve cardiac function in mice subjected to TAC. These results suggest the pyroptosis and ferroptosis induced by MLK3 signaling in cardiomyocytes are essential for adverse myocardial fibrosis, in response to pressure overload. Furthermore, miR-351, which has a protective effect on ventricular remodeling in heart failure caused by pressure overload, may be a key target for the regulation of MLK3.
Collapse
Affiliation(s)
- Junyan Wang
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- The First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Bo Deng
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- The First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Qing Liu
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- The First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Yusheng Huang
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- The First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- Guangzhou Key Laboratory of Chinese Medicine for Prevention and Treatment of Chronic Heart Failure, Guangzhou, 510405, China
| | - Weitao Chen
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- The First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Jing Li
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- The First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Zheng Zhou
- The First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Lu Zhang
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- The First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Birong Liang
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- The First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Jiaqi He
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- The First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Zixin Chen
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- The First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- Guangzhou Key Laboratory of Chinese Medicine for Prevention and Treatment of Chronic Heart Failure, Guangzhou, 510405, China
| | - Cui Yan
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- The First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Zhongqi Yang
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- The First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- Guangzhou Key Laboratory of Chinese Medicine for Prevention and Treatment of Chronic Heart Failure, Guangzhou, 510405, China
- National Clinical Research Base of Traditional Chinese Medicine, Guangzhou, 510405, China
| | - Shaoxiang Xian
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
- The First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
- Guangzhou Key Laboratory of Chinese Medicine for Prevention and Treatment of Chronic Heart Failure, Guangzhou, 510405, China.
- National Clinical Research Base of Traditional Chinese Medicine, Guangzhou, 510405, China.
| | - Lingjun Wang
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
- The First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
- Guangzhou Key Laboratory of Chinese Medicine for Prevention and Treatment of Chronic Heart Failure, Guangzhou, 510405, China.
- National Clinical Research Base of Traditional Chinese Medicine, Guangzhou, 510405, China.
| |
Collapse
|
46
|
Abstract
PURPOSE Hyperlipidemia, characterized by an increase in circulating lipid levels, doubles the chance of developing cardiovascular diseases. It prompts inflammation, immune activation, and oxidative stress in the bloodstream and organs of rats. Thus, we theorized that the metabolism of purines, an immunomodulatory mechanism, is altered in cells involved in the development of cardiovascular diseases. METHODS Therefore, we induced acute hyperlipidemia in Wistar rats with Poloxamer-407 and euthanized the animals 36 h later. The leucocyte differential, the rate of purine metabolism on the surface of platelets and heart cells, and markers of oxidative stress in the heart tissue were evaluated. These parameters were also assessed in animals pretreated for 30 days with curcumin and/or rutin. RESULTS Hyperlipidemia increased the hydrolyses of adenosine triphosphate (ATP), adenosine diphosphate (ADP), and adenosine monophosphate (AMP) in platelets. In heart cells, the metabolism of ATP and adenosine (ADO) were increased, while ADP hydrolysis was reduced. Additionally, lipid damage and antioxidant defenses were increased in heart homogenates. Hyperlipidemic rats also exhibited a reduced percentage of eosinophils and lymphocytes. CONCLUSION Together, these findings are indicative of an increased risk of developing cardiovascular diseases in hyperlipidemic rats. The pretreatments with antioxidants reverted some of the changes prompted by hyperlipidemia preventing detrimental changes in the cells and tissues. Graphical Abstract.
Collapse
|