1
|
Lan C, Fang G, Li X, Chen X, Chen Y, Hu T, Wang X, Cai H, Hao J, Li H, Zhang Y, Peng K, Xu Z, Yang D, Kang X, Xin Q, Yang Y. SerpinB1 targeting safeguards against pathological cardiac hypertrophy and remodelling by suppressing cardiomyocyte pyroptosis and inflammation initiation. Cardiovasc Res 2024:cvae241. [PMID: 39688818 DOI: 10.1093/cvr/cvae241] [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: 01/09/2024] [Revised: 07/06/2024] [Accepted: 09/15/2024] [Indexed: 12/18/2024] Open
Abstract
AIMS While the pivotal role of inflammation in pathological cardiac hypertrophy and remodelling is widely acknowledged, the mechanisms triggering inflammation initiation remain largely obscure. This study aims to elucidate the role and mechanism of serpin family B member 1 (SerpinB1) in pro-inflammatory cardiomyocyte pyroptosis, heart inflammation, and cardiac remodelling. METHODS AND RESULTS C57BL/6J wild-type, inducible cardiac-specific SerpinB1 overexpression or knockout mice underwent transverse aortic constriction (TAC) surgery. Cardiac hypertrophy and remodelling were assessed through echocardiography and histology. Cardiomyocyte pyroptosis and heart inflammation were monitored. Adeno-associated virus 9 -mediated gene manipulations and molecular assays were employed to explore the mechanisms through which SerpinB1 regulates cardiomyocyte pyroptosis and heart inflammation. Finally, recombinant mouse SerpinB1 protein (rSerpinB1) was administrated both in vivo through osmotic minipump delivery and in vitro to investigate the therapeutic potential of SerpinB1 in cardiac remodelling. Myocardial SerpinB1 overexpression was up-regulated shortly upon TAC or phenylephrine challenge, with no further elevation during prolonged hypertrophic stimuli. It is important to note that cardiac-specific overexpression of SerpinB1 markedly attenuated TAC-induced cardiac remodelling, while deletion of SerpinB1 exacerbated it. At the mechanistic level, SerpinB1 gain-of-function inhibited cardiomyocyte pyroptosis and inflammation in hypertrophic hearts; the protective effect was nullified by overexpression of either cleaved N-terminal gasdermin D or cleaved caspase-1. Co-immunoprecipitation and confocal assays confirmed that SerpinB1 directly interacts with caspase-1 in cardiomyocytes. Remarkably, rSerpinB1 replicated the cardioprotective effect against cardiac hypertrophy and remodelling. CONCLUSION SerpinB1 safeguards against pathological cardiac hypertrophy and remodelling by impeding cardiomyocyte pyroptosis to suppress inflammation initiation, achieved through interaction with caspase-1 to inhibit its activation. Targeting SerpinB1 could represent a novel therapeutic strategy for treating pathological cardiac hypertrophy and remodelling.
Collapse
Affiliation(s)
- Cong Lan
- Department of Cardiology, The General Hospital of Western Theater Command, No. 270, Tianhui Road, Jinniu District, Chengdu , Sichuan 610083, P.R. China
- Pancreatic Injury and Repair Key Laboratory of Sichuan Province, The General Hospital of Western Theater Command, No. 270, Tianhui Road, Jinniu District, Chengdu, Sichuan 610083, P.R. China
- College of Medicine, Southwest Jiaotong University, No. 144, Jiaoda Road, Jinniu District, Chengdu, Sichuan 610031, P.R. China
| | - Gangyao Fang
- Department of Cardiology, The General Hospital of Western Theater Command, No. 270, Tianhui Road, Jinniu District, Chengdu , Sichuan 610083, P.R. China
| | - Xiuchuan Li
- Department of Cardiology, The General Hospital of Western Theater Command, No. 270, Tianhui Road, Jinniu District, Chengdu , Sichuan 610083, P.R. China
| | - Xiao Chen
- Department of General Practice, The General Hospital of Western Theater Command, No. 270, Tianhui Road, Jinniu District, Chengdu, Sichuan 610083, P.R. China
| | - Yingmei Chen
- Department of Cardiology, The General Hospital of Western Theater Command, No. 270, Tianhui Road, Jinniu District, Chengdu , Sichuan 610083, P.R. China
| | - Tao Hu
- Department of Cardiology, The General Hospital of Western Theater Command, No. 270, Tianhui Road, Jinniu District, Chengdu , Sichuan 610083, P.R. China
| | - Xuenan Wang
- School of Clinical Medicine, Southwest Medical University, No. 319, Zhongshan Road, Jiangyang District, Luzhou, Sichuan 646000, P.R. China
| | - Huiling Cai
- School of Clinical Medicine, Southwest Medical University, No. 319, Zhongshan Road, Jiangyang District, Luzhou, Sichuan 646000, P.R. China
| | - Jiajin Hao
- Department of Cardiology, The General Hospital of Western Theater Command, No. 270, Tianhui Road, Jinniu District, Chengdu , Sichuan 610083, P.R. China
- College of Medicine, Southwest Jiaotong University, No. 144, Jiaoda Road, Jinniu District, Chengdu, Sichuan 610031, P.R. China
| | - Haoran Li
- Department of Cardiology, The General Hospital of Western Theater Command, No. 270, Tianhui Road, Jinniu District, Chengdu , Sichuan 610083, P.R. China
- College of Medicine, Southwest Jiaotong University, No. 144, Jiaoda Road, Jinniu District, Chengdu, Sichuan 610031, P.R. China
| | - Yan Zhang
- Department of Cardiology, The General Hospital of Western Theater Command, No. 270, Tianhui Road, Jinniu District, Chengdu , Sichuan 610083, P.R. China
| | - Ke Peng
- Department of Cardiology, The General Hospital of Western Theater Command, No. 270, Tianhui Road, Jinniu District, Chengdu , Sichuan 610083, P.R. China
| | - Zaicheng Xu
- Department of Cancer Center, Second Affiliated Hospital, Chongqing Medical University, No. 288, Tianwen Road, Nanan District, Chongqing 400072, P.R. China
| | - Dachun Yang
- Department of Cardiology, The General Hospital of Western Theater Command, No. 270, Tianhui Road, Jinniu District, Chengdu , Sichuan 610083, P.R. China
| | - Xia Kang
- Pancreatic Injury and Repair Key Laboratory of Sichuan Province, The General Hospital of Western Theater Command, No. 270, Tianhui Road, Jinniu District, Chengdu, Sichuan 610083, P.R. China
| | - Qian Xin
- Department of Cardiology, Sixth Medical Center of Chinese PLA General Hospital, No. 6, Fucheng Road, Haidian District, Beijing 100048, P.R. China
| | - Yongjian Yang
- Department of Cardiology, The General Hospital of Western Theater Command, No. 270, Tianhui Road, Jinniu District, Chengdu , Sichuan 610083, P.R. China
- College of Medicine, Southwest Jiaotong University, No. 144, Jiaoda Road, Jinniu District, Chengdu, Sichuan 610031, P.R. China
- School of Clinical Medicine, Southwest Medical University, No. 319, Zhongshan Road, Jiangyang District, Luzhou, Sichuan 646000, P.R. China
| |
Collapse
|
2
|
Prajapati AK, Shah G. Exploring in vivo and in vitro models for heart failure with biomarker insights: a review. Egypt Heart J 2024; 76:141. [PMID: 39432214 PMCID: PMC11493927 DOI: 10.1186/s43044-024-00568-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Accepted: 09/27/2024] [Indexed: 10/22/2024] Open
Abstract
BACKGROUND Heart failure (HF) is a condition characterized by the heart's inability to meet the body's demands, resulting in various complications. Two primary types of HF exist, namely HF with preserved left ventricular ejection fraction (LVEF) and HF reduced with LVEF. The progression of HF involves compensatory mechanisms such as cardiac hypertrophy, fibrosis, and alterations in gene expression. Pressure overload and volume overload are common etiologies of HF, with pressure overload often stemming from conditions like hypertension, leading to left ventricular hypertrophy and fibrosis. In contrast, volume overload can arise from chronic valvular regurgitant disease, also inducing left ventricular hypertrophy. MAIN BODY In vitro cell culture techniques serve as vital tools in studying HF pathophysiology, allowing researchers to investigate cellular responses and potential therapeutic targets. Additionally, biomarkers, measurable biological characteristics, play a crucial role in diagnosing and predicting HF. Some notable biomarkers include adrenomedullin, B-type natriuretic peptide, copeptin, galectin-3, interleukin-6, matrix metalloproteinases (MMPs), midregional pro-atrial natriuretic peptide, myostatin, procollagen type I C-terminal propeptide, procollagen type III N-terminal propeptide and tissue inhibitors of metalloproteinases (TIMPs). These biomarkers aid in HF diagnosis, assessing its severity, and monitoring treatment response, contributing to a deeper understanding of the disease and potentially leading to improved management strategies and outcomes. CONCLUSIONS This review provides comprehensive insights into various in vivo models of HF, commonly utilized cell lines in HF research, and pivotal biomarkers with diagnostic relevance for HF. By synthesizing this information, researchers gain valuable resources to further explore HF pathogenesis, identify novel therapeutic targets, and enhance diagnostic and prognostic approaches.
Collapse
Affiliation(s)
- Anil Kumar Prajapati
- Pharmacology Department, L. M. College of Pharmacy, Ahmedabad, Gujarat, 380009, India
- Research Scholar, Gujarat Technological University, Ahmedabad, Gujarat, 382424, India
| | - Gaurang Shah
- Pharmacology Department, L. M. College of Pharmacy, Ahmedabad, Gujarat, 380009, India.
| |
Collapse
|
3
|
Grzeczka A, Graczyk S, Kordowitzki P. Pleiotropic Effects of Resveratrol on Aging-Related Cardiovascular Diseases-What Can We Learn from Research in Dogs? Cells 2024; 13:1732. [PMID: 39451250 PMCID: PMC11505706 DOI: 10.3390/cells13201732] [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: 09/20/2024] [Revised: 10/17/2024] [Accepted: 10/18/2024] [Indexed: 10/26/2024] Open
Abstract
Resveratrol (RES) is a polyphenol with natural anti-inflammatory and antioxidant properties. It is found in abundance in plants, i.e., grapes and mulberry fruit. In addition, synthetic forms of RES exist. Since the discovery of its specific biological properties, RES has emerged as a candidate substance not only with modeling effects on the immune response but also as an important factor in preventing the onset and progression of cardiovascular disease (CVD). Previous research provided strong evidence of the effects of RES on platelets, mitochondria, cardiomyocytes, and vascular endothelial function. In addition, RES positively affects the coagulation system and vasodilatory function and improves blood flow. Not only in humans but also in veterinary medicine, cardiovascular diseases have one of the highest incidence rates. Canine and human species co-evolved and share recent evolutionary selection processes, and interestingly, numerous pathologies of companion dogs have a human counterpart. Knowledge of the impact of RES on the cardiovascular system of dogs is becoming clearer in the literature. Dogs have long been recognized as valuable animal models for the study of various human diseases as they share many physiological and genetic similarities with humans. In this review, we aim to shed light on the pleiotropic effects of resveratrol on cardiovascular health in dogs as a translational model for human cardiovascular diseases.
Collapse
Affiliation(s)
| | | | - Pawel Kordowitzki
- Department for Basic and Preclinical Sciences, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, 87-100 Toruń, Poland; (A.G.)
| |
Collapse
|
4
|
Napiórkowska-Baran K, Doligalska A, Drozd M, Czarnowska M, Łaszczych D, Dolina M, Szymczak B, Schmidt O, Bartuzi Z. Management of a Patient with Cardiovascular Disease Should Include Assessment of Primary and Secondary Immunodeficiencies: Part 2-Secondary Immunodeficiencies. Healthcare (Basel) 2024; 12:1977. [PMID: 39408157 PMCID: PMC11477378 DOI: 10.3390/healthcare12191977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 09/19/2024] [Accepted: 09/24/2024] [Indexed: 10/20/2024] Open
Abstract
BACKGROUND Cardiovascular diseases are among the most common chronic diseases, generating high social and economic costs. Secondary immunodeficiencies occur more often than primary ones and may result from the co-occurrence of specific diseases, treatment, nutrient deficiencies and non-nutritive bio-active compounds that result from the industrial nutrient practices. OBJECTIVES The aim of this article is to present selected secondary immunodeficiencies and their impact on the cardiovascular system. RESULTS The treatment of a patient with cardiovascular disease should include an assess-ment for immunodeficiencies, because the immune and cardiovascular systems are closely linked. CONCLUSIONS Immune system dysfunctions can significantly affect the course of cardiovascular diseases and their treatment. For this reason, comprehensive care for a patient with cardiovascular disease requires taking into account potential immunodeficiencies, which can have a significant impact on the patient's health.
Collapse
Affiliation(s)
- Katarzyna Napiórkowska-Baran
- Department of Allergology, Clinical Immunology and Internal Diseases, Collegium Medicum Bydgoszcz, Nicolaus Copernicus University Torun, 85-067 Bydgoszcz, Poland;
| | - Agata Doligalska
- Student Research Club of Clinical Immunology, Department of Allergology, Clinical Immunology and Internal Diseases, Collegium Medicum Bydgoszcz, Nicolaus Copernicus University Torun, 85-067 Bydgoszcz, Poland; (A.D.); (M.D.); (M.C.); (D.Ł.); (M.D.); (B.S.); (O.S.)
| | - Magdalena Drozd
- Student Research Club of Clinical Immunology, Department of Allergology, Clinical Immunology and Internal Diseases, Collegium Medicum Bydgoszcz, Nicolaus Copernicus University Torun, 85-067 Bydgoszcz, Poland; (A.D.); (M.D.); (M.C.); (D.Ł.); (M.D.); (B.S.); (O.S.)
| | - Marta Czarnowska
- Student Research Club of Clinical Immunology, Department of Allergology, Clinical Immunology and Internal Diseases, Collegium Medicum Bydgoszcz, Nicolaus Copernicus University Torun, 85-067 Bydgoszcz, Poland; (A.D.); (M.D.); (M.C.); (D.Ł.); (M.D.); (B.S.); (O.S.)
| | - Dariusz Łaszczych
- Student Research Club of Clinical Immunology, Department of Allergology, Clinical Immunology and Internal Diseases, Collegium Medicum Bydgoszcz, Nicolaus Copernicus University Torun, 85-067 Bydgoszcz, Poland; (A.D.); (M.D.); (M.C.); (D.Ł.); (M.D.); (B.S.); (O.S.)
| | - Marcin Dolina
- Student Research Club of Clinical Immunology, Department of Allergology, Clinical Immunology and Internal Diseases, Collegium Medicum Bydgoszcz, Nicolaus Copernicus University Torun, 85-067 Bydgoszcz, Poland; (A.D.); (M.D.); (M.C.); (D.Ł.); (M.D.); (B.S.); (O.S.)
| | - Bartłomiej Szymczak
- Student Research Club of Clinical Immunology, Department of Allergology, Clinical Immunology and Internal Diseases, Collegium Medicum Bydgoszcz, Nicolaus Copernicus University Torun, 85-067 Bydgoszcz, Poland; (A.D.); (M.D.); (M.C.); (D.Ł.); (M.D.); (B.S.); (O.S.)
| | - Oskar Schmidt
- Student Research Club of Clinical Immunology, Department of Allergology, Clinical Immunology and Internal Diseases, Collegium Medicum Bydgoszcz, Nicolaus Copernicus University Torun, 85-067 Bydgoszcz, Poland; (A.D.); (M.D.); (M.C.); (D.Ł.); (M.D.); (B.S.); (O.S.)
| | - Zbigniew Bartuzi
- Department of Allergology, Clinical Immunology and Internal Diseases, Collegium Medicum Bydgoszcz, Nicolaus Copernicus University Torun, 85-067 Bydgoszcz, Poland;
| |
Collapse
|
5
|
Ding B, Jiang L, Zhang N, Zhou L, Luo H, Wang H, Chen X, Gao Y, Zhao Z, Wang C, Wang Z, Guo Z, Wang Y. Santalum album L. alleviates cardiac function injury in heart failure by synergistically inhibiting inflammation, oxidative stress and apoptosis through multiple components. Chin Med 2024; 19:98. [PMID: 39010069 PMCID: PMC11251102 DOI: 10.1186/s13020-024-00968-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 06/30/2024] [Indexed: 07/17/2024] Open
Abstract
BACKGROUND Heart failure (HF) is a complex cardiovascular syndrome with high mortality. Santalum album L. (SAL) is a traditional Chinese medicine broadly applied for various diseases treatment including HF. However, the potential active compounds and molecular mechanisms of SAL in HF treatment are not well understood. METHODS The active compounds and possible mechanisms of action of SAL were analyzed and validated by a systems pharmacology framework and an ISO-induced mouse HF model. RESULTS We initially confirmed that SAL alleviates heart damage in ISO-induced HF model. A total of 17 potentially active components in SAL were identified, with Luteolin (Lut) and Syringaldehyde (SYD) in SAL been identified as the most effective combination through probabilistic ensemble aggregation (PEA) analysis. These compounds, individually and in their combination (COMB), showed significant therapeutic effects on HF by targeting multiple pathways involved in anti-oxidation, anti-inflammation, and anti-apoptosis. The active ingredients in SAL effectively suppressed inflammatory mediators and pro-apoptotic proteins while enhancing the expression of anti-apoptotic factors and antioxidant markers. Furthermore, the synergistic effects of SAL on YAP and PI3K-AKT signaling pathways were further elucidated. CONCLUSIONS Mechanistically, the anti-HF effect of SAL is responsible for the synergistic effect of anti-inflammation, antioxidation and anti-apoptosis, delineating a multi-targeted therapeutic strategy for HF.
Collapse
Affiliation(s)
- Bojiao Ding
- Key Laboratory of Resource Biology and Modern Biotechnology in Western China, Ministry of Education, Northwest University, No. 229 TaiBai North Road, Xi'an, 710069, Shaanxi, China
- Jiuwei Institute of Life Sciences, Yangling, 712100, Shaanxi, China
| | - Li Jiang
- Key Laboratory of Phytomedicinal Resources Utilization, Ministry of Education, Shihezi University, Shihezi, 832000, Xinjiang, China
| | - Na Zhang
- Key Laboratory of Resource Biology and Modern Biotechnology in Western China, Ministry of Education, Northwest University, No. 229 TaiBai North Road, Xi'an, 710069, Shaanxi, China
- Jiuwei Institute of Life Sciences, Yangling, 712100, Shaanxi, China
| | - Li Zhou
- Key Laboratory of Resource Biology and Modern Biotechnology in Western China, Ministry of Education, Northwest University, No. 229 TaiBai North Road, Xi'an, 710069, Shaanxi, China
- Jiuwei Institute of Life Sciences, Yangling, 712100, Shaanxi, China
| | - Huiying Luo
- Key Laboratory of Resource Biology and Modern Biotechnology in Western China, Ministry of Education, Northwest University, No. 229 TaiBai North Road, Xi'an, 710069, Shaanxi, China
- Jiuwei Institute of Life Sciences, Yangling, 712100, Shaanxi, China
| | - Haiqing Wang
- Jiuwei Institute of Life Sciences, Yangling, 712100, Shaanxi, China
- Shaanxi Qinling Qiyao Collaborative Innovation Center Co. Ltd., Xianyang, 712100, Shaanxi, China
| | - Xuetong Chen
- Key Laboratory of Resource Biology and Modern Biotechnology in Western China, Ministry of Education, Northwest University, No. 229 TaiBai North Road, Xi'an, 710069, Shaanxi, China
- Jiuwei Institute of Life Sciences, Yangling, 712100, Shaanxi, China
- Shaanxi Qinling Qiyao Collaborative Innovation Center Co. Ltd., Xianyang, 712100, Shaanxi, China
| | - Yuxin Gao
- Key Laboratory of Resource Biology and Modern Biotechnology in Western China, Ministry of Education, Northwest University, No. 229 TaiBai North Road, Xi'an, 710069, Shaanxi, China
- Jiuwei Institute of Life Sciences, Yangling, 712100, Shaanxi, China
| | - Zezhou Zhao
- Jiuwei Institute of Life Sciences, Yangling, 712100, Shaanxi, China
- Key Laboratory of Phytomedicinal Resources Utilization, Ministry of Education, Shihezi University, Shihezi, 832000, Xinjiang, China
| | - Chao Wang
- National Key Laboratory On Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Jiangsu Kanion Pharmaceutical Co. Ltd., Lianyungang, 222002, Jiangsu, China
| | - Zhenzhong Wang
- National Key Laboratory On Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Jiangsu Kanion Pharmaceutical Co. Ltd., Lianyungang, 222002, Jiangsu, China
| | - Zihu Guo
- Key Laboratory of Resource Biology and Modern Biotechnology in Western China, Ministry of Education, Northwest University, No. 229 TaiBai North Road, Xi'an, 710069, Shaanxi, China.
- Jiuwei Institute of Life Sciences, Yangling, 712100, Shaanxi, China.
- Shaanxi Qinling Qiyao Collaborative Innovation Center Co. Ltd., Xianyang, 712100, Shaanxi, China.
| | - Yonghua Wang
- Key Laboratory of Resource Biology and Modern Biotechnology in Western China, Ministry of Education, Northwest University, No. 229 TaiBai North Road, Xi'an, 710069, Shaanxi, China.
- Jiuwei Institute of Life Sciences, Yangling, 712100, Shaanxi, China.
- Shaanxi Qinling Qiyao Collaborative Innovation Center Co. Ltd., Xianyang, 712100, Shaanxi, China.
- College of Pharmacy, Heze University, Heze, 274015, Shandong, China.
| |
Collapse
|
6
|
Krüger DN, Bosman M, Van Assche CXL, Wesley CD, Cillero-Pastor B, Delrue L, Heggermont W, Bartunek J, De Meyer GRY, Van Craenenbroeck EM, Guns PJ, Franssen C. Characterization of systolic and diastolic function, alongside proteomic profiling, in doxorubicin-induced cardiovascular toxicity in mice. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2024; 10:40. [PMID: 38909263 PMCID: PMC11193203 DOI: 10.1186/s40959-024-00241-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 06/10/2024] [Indexed: 06/24/2024]
Abstract
BACKGROUND The anthracycline doxorubicin (DOX) is a highly effective anticancer agent, especially in breast cancer and lymphoma. However, DOX can cause cancer therapy-related cardiovascular toxicity (CTR-CVT) in patients during treatment and in survivors. Current diagnostic criteria for CTR-CVT focus mainly on left ventricular systolic dysfunction, but a certain level of damage is required before it can be detected. As diastolic dysfunction often precedes systolic dysfunction, the current study aimed to identify functional and molecular markers of DOX-induced CTR-CVT with a focus on diastolic dysfunction. METHODS Male C57BL/6J mice were treated with saline or DOX (4 mg/kg, weekly i.p. injection) for 2 and 6 weeks (respectively cumulative dose of 8 and 24 mg/kg) (n = 8 per group at each time point). Cardiovascular function was longitudinally investigated using echocardiography and invasive left ventricular pressure measurements. Subsequently, at both timepoints, myocardial tissue was obtained for proteomics (liquid-chromatography with mass-spectrometry). A cohort of patients with CTR-CVT was used to complement the pre-clinical findings. RESULTS DOX-induced a reduction in left ventricular ejection fraction from 72 ± 2% to 55 ± 1% after 2 weeks (cumulative 8 mg/kg DOX). Diastolic dysfunction was demonstrated as prolonged relaxation (increased tau) and heart failure was evident from pulmonary edema after 6 weeks (cumulative 24 mg/kg DOX). Myocardial proteomic analysis revealed an increased expression of 12 proteins at week 6, with notable upregulation of SERPINA3N in the DOX-treated animals. The human ortholog SERPINA3 has previously been suggested as a marker in CTR-CVT. Upregulation of SERPINA3N was confirmed by western blot, immunohistochemistry, and qPCR in murine hearts. Thereby, SERPINA3N was most abundant in the endothelial cells. In patients, circulating SERPINA3 was increased in plasma of CTR-CVT patients but not in cardiac biopsies. CONCLUSION We showed that mice develop heart failure with impaired systolic and diastolic function as result of DOX treatment. Additionally, we could identify increased SERPINA3 levels in the mice as well as patients with DOX-induced CVT and demonstrated expression of SERPINA3 in the heart itself, suggesting that SERPINA3 could serve as a novel biomarker.
Collapse
Affiliation(s)
- Dustin N Krüger
- Laboratory of Psychopharmacology, Faculty of Medicine and Health Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, Campus Drie Eiken, University of Antwerp, Universiteitsplein 1, Antwerp, B-2610, Belgium.
| | - Matthias Bosman
- Laboratory of Psychopharmacology, Faculty of Medicine and Health Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, Campus Drie Eiken, University of Antwerp, Universiteitsplein 1, Antwerp, B-2610, Belgium
| | - Charles X L Van Assche
- Division M4I - Imaging Mass Spectrometry (IMS), Faculty of Health, Medicine and Life Sciences, Maastricht MultiModal Molecular Imaging Institute, Maastricht University, Universiteitssingel 50, Maastricht, 6229 ER, The Netherlands
| | - Callan D Wesley
- Laboratory of Psychopharmacology, Faculty of Medicine and Health Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, Campus Drie Eiken, University of Antwerp, Universiteitsplein 1, Antwerp, B-2610, Belgium
| | - Berta Cillero-Pastor
- Division M4I - Imaging Mass Spectrometry (IMS), Faculty of Health, Medicine and Life Sciences, Maastricht MultiModal Molecular Imaging Institute, Maastricht University, Universiteitssingel 50, Maastricht, 6229 ER, The Netherlands
- Department of Cell Biology-Inspired Tissue Engineering, Institute for Technology-Inspired Regenerative Medicine, Universiteitssingel 40, Maastricht, 6229 ER, The Netherlands
| | - Leen Delrue
- Cardiovascular Centre, OLV Hospital, Moorselbaan 164, Aalst, B-9300, Belgium
| | - Ward Heggermont
- Cardiovascular Centre, OLV Hospital, Moorselbaan 164, Aalst, B-9300, Belgium
| | - Jozef Bartunek
- Cardiovascular Centre, OLV Hospital, Moorselbaan 164, Aalst, B-9300, Belgium
| | - Guido R Y De Meyer
- Laboratory of Psychopharmacology, Faculty of Medicine and Health Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, Campus Drie Eiken, University of Antwerp, Universiteitsplein 1, Antwerp, B-2610, Belgium
| | - Emeline M Van Craenenbroeck
- Research Group Cardiovascular Diseases, University of Antwerp, Wilrijkstraat 10, Edegem, B-2650, Belgium
- Department of Cardiology, Antwerp University Hospital (UZA), Drie Eikenstraat 655, Edegem, B-2650, Belgium
| | - Pieter-Jan Guns
- Laboratory of Psychopharmacology, Faculty of Medicine and Health Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, Campus Drie Eiken, University of Antwerp, Universiteitsplein 1, Antwerp, B-2610, Belgium
| | - Constantijn Franssen
- Research Group Cardiovascular Diseases, University of Antwerp, Wilrijkstraat 10, Edegem, B-2650, Belgium
- Department of Cardiology, Antwerp University Hospital (UZA), Drie Eikenstraat 655, Edegem, B-2650, Belgium
| |
Collapse
|
7
|
Montecillo J, Pirker T, Pemberton C, Chew-Harris J. suPAR in cardiovascular disease. Adv Clin Chem 2024; 121:89-131. [PMID: 38797545 DOI: 10.1016/bs.acc.2024.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Soluble urokinase plasminogen activator receptor (suPAR), the soluble counterpart of urokinase plasminogen activator receptor, is found in the circulation at various levels. suPAR and its parent molecule, cell surface uPAR, exhibit similar structure and extracellular functional roles facilitating fibrinolysis, cellular adhesion, and migration. Studies have assessed the correlation between suPAR in cardiovascular disease (CVD). It is postulated that suPAR may serve as an indicator of inflammatory activation and burden during CVD progression. Increased suPAR independently predicts poorer outcomes in acute coronary syndromes, in heart failure, as well as in coronary artery disease and atherosclerosis. To guide translation into clinical utization, suPAR has been assessed in numerous CVD settings for improved risk discrimination independently or in association with established traditional risk factors. Whilst the involvement of suPAR has been explored in other diseases such as kidney diseases and cancer, there is only emerging evidence of suPAR's mechanistic involvement in cardiovascular disease. In this review, we provide a background into suPAR and its potential role as a biomarker in CVD.
Collapse
Affiliation(s)
- Jaya Montecillo
- Christchurch Heart Institute, University of Otago, Christchurch, New Zealand
| | - Thomas Pirker
- Christchurch Heart Institute, University of Otago, Christchurch, New Zealand
| | | | - Janice Chew-Harris
- Christchurch Heart Institute, University of Otago, Christchurch, New Zealand.
| |
Collapse
|
8
|
Athari SS, Mehrabi Nasab E, Jing K, Wang J. Interaction between cardiac resynchronization therapy and cytokines in heart failure patients. Cytokine 2024; 175:156479. [PMID: 38199086 DOI: 10.1016/j.cyto.2023.156479] [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: 04/20/2023] [Revised: 12/09/2023] [Accepted: 12/12/2023] [Indexed: 01/12/2024]
Abstract
Congestive heart failure (CHF) is a complex multistage syndrome that has a great financial burden on human societies. It was known that the damaged myocardium sends a signal to stimulate the immune system and proliferation of leukocytes. In continuous, cytokine storm can be initiated and causes the probability of CHF. Persistent inflammation by increasing the levels of pro-inflammatory cytokines, plays an important role in the pathogenesis of CHF and causes remodeling, which is a progressive processs. Although treatment by drugs can reduce mortality and partially control the symptoms of heart failure patients, but complications and mortality are still high. Therefore, other treatment options such as Cardiac Resynchronization Therapy (CRT) are necessary. Today, it is known that CRT can be an effective treatment for many patients with heart failure. CRT is novel, non-pharmacological, and device-based therapy that would be beneficial to know more about its performance in the management of heart failure. In this study, we have reviewed the immunological processes involved in heart failure and the effect of CRT in controlling of the cytokine storm.
Collapse
Affiliation(s)
- Seyyed Shamsadin Athari
- Department of Immunology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Entezar Mehrabi Nasab
- Department of Cardiology, School of Medicine, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Cardiology, School of Medicine, Valiasr Hospital, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Kai Jing
- Department of Proctology, The People's Hospital of Huaiyin Jinan, 250021 Shandong, China
| | - Jin Wang
- Department of Cardiology, The Fifth People's Hospital of Jinan, 250022 Shandong, China.
| |
Collapse
|
9
|
Farooq MU, Latib A, Jorde UP. Tricuspid Regurgitation in Congestive "End-Organ" Failure: Outline of an Opportunity. Cardiol Rev 2024; 32:18-23. [PMID: 35452428 DOI: 10.1097/crd.0000000000000455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Tricuspid regurgitation (TR) is a progressive disease that can be addressed only partially by medical therapy. Progression of TR is associated with worsening end-organ function and worse survival, yet tricuspid valve interventions are usually only performed in advanced stages. Recent evidence suggests a pivotal role for TR and pulsatile venous congestion in the pathophysiology of renal and hepatic dysfunction. This critical knowledge has provided the opportunity to optimally define the appropriate timing of transcatheter tricuspid valve interventions, integrating concurrent or impending functional consequences with severity of TR.
Collapse
Affiliation(s)
- Muhammed U Farooq
- From the Division of Cardiology, Montefiore Medical Center and Albert Einstein College of Medicine, Bronx, NY
| | | | | |
Collapse
|
10
|
Arvunescu AM, Ionescu RF, Cretoiu SM, Dumitrescu SI, Zaharia O, Nanea IT. Inflammation in Heart Failure-Future Perspectives. J Clin Med 2023; 12:7738. [PMID: 38137807 PMCID: PMC10743797 DOI: 10.3390/jcm12247738] [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: 11/05/2023] [Revised: 12/01/2023] [Accepted: 12/07/2023] [Indexed: 12/24/2023] Open
Abstract
Chronic heart failure is a terminal point of a vast majority of cardiac or extracardiac causes affecting around 1-2% of the global population and more than 10% of the people above the age of 65. Inflammation is persistently associated with chronic diseases, contributing in many cases to the progression of disease. Even in a low inflammatory state, past studies raised the question of whether inflammation is a constant condition, or if it is, rather, triggered in different amounts, according to the phenotype of heart failure. By evaluating the results of clinical studies which focused on proinflammatory cytokines, this review aims to identify the ones that are independent risk factors for heart failure decompensation or cardiovascular death. This review assessed the current evidence concerning the inflammatory activation cascade, but also future possible targets for inflammatory response modulation, which can further impact the course of heart failure.
Collapse
Affiliation(s)
- Alexandru Mircea Arvunescu
- Department of Internal Medicine and Cardiology, “Prof. Dr. Th. Burghele” Clinical Hospital, 061344 Bucharest, Romania; (O.Z.); (I.T.N.)
- Department of Cardio-Thoracic Pathology, Cardio-Thoracic Pathology, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 050471 Bucharest, Romania
| | - Ruxandra Florentina Ionescu
- Department of Cardiology I, Central Military Emergency Hospital “Dr Carol Davila”, 030167 Bucharest, Romania (S.I.D.)
- Department of Morphological Sciences, Cell and Molecular Biology and Histology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| | - Sanda Maria Cretoiu
- Department of Morphological Sciences, Cell and Molecular Biology and Histology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| | - Silviu Ionel Dumitrescu
- Department of Cardiology I, Central Military Emergency Hospital “Dr Carol Davila”, 030167 Bucharest, Romania (S.I.D.)
- Department of Cardiology, Faculty of Medicine, Titu Maiorescu University, 040441 Bucharest, Romania
| | - Ondin Zaharia
- Department of Internal Medicine and Cardiology, “Prof. Dr. Th. Burghele” Clinical Hospital, 061344 Bucharest, Romania; (O.Z.); (I.T.N.)
- Department of Cardio-Thoracic Pathology, Cardio-Thoracic Pathology, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 050471 Bucharest, Romania
| | - Ioan Tiberiu Nanea
- Department of Internal Medicine and Cardiology, “Prof. Dr. Th. Burghele” Clinical Hospital, 061344 Bucharest, Romania; (O.Z.); (I.T.N.)
- Department of Cardio-Thoracic Pathology, Cardio-Thoracic Pathology, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 050471 Bucharest, Romania
| |
Collapse
|
11
|
Cheeseman J, Badia C, Elgood-Hunt G, Gardner RA, Trinh DN, Monopoli MP, Kuhnle G, Spencer DIR, Osborn HMI. Elevated concentrations of Neu5Ac and Neu5,9Ac 2 in human plasma: potential biomarkers of cardiovascular disease. Glycoconj J 2023; 40:645-654. [PMID: 37991561 PMCID: PMC10788320 DOI: 10.1007/s10719-023-10138-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 10/25/2023] [Accepted: 10/30/2023] [Indexed: 11/23/2023]
Abstract
Cardiovascular disease (CVD) is a group of health conditions affecting the heart and vascular system with very high prevalence and mortality rates. The presence of CVD is characterised by high levels of inflammation which have previously been associated with increased plasma concentrations of N-acetyl neuraminic acid (Neu5Ac). While Neu5Ac has been studied in the context of CVD, Neu5,9Ac2 has not, despite being the second most abundant sialic acid in human plasma. A small-scale pilot study of thirty plasma samples from patients with diagnosed CVD, and thirty age and sex-matched healthy controls, was designed to gain insight into sialic acids as biomarkers for CVD and potential future areas of study. Each sample was assayed for Neu5Ac and Neu5,9Ac2 concentrations. Mean Neu5Ac and Neu5,9Ac2 concentrations were significantly elevated in patients with CVD compared to healthy controls (Neu5Ac: P < 0.001; Neu5,9Ac2: P < 0.04). Receiver operator curve (ROC) analysis indicated that both Neu5Ac and Neu5,9Ac2 have reasonable predictive power for the presence of CVD (Neu5Ac AUC: 0.86; Neu5,9Ac2 AUC: 0.71). However, while Neu5Ac had both good sensitivity (0.82) and specificity (0.81), Neu5,9Ac2 had equivalent specificity (0.81) but very poor sensitivity (0.44). A combination marker of Neu5Ac + Neu5,9Ac2 showed improvement over Neu5Ac alone in terms of predictive power (AUC: 0.93), sensitivity (0.87), and specificity (0.90). Comparison to a known inflammatory marker, high sensitivity c-reactive protein (hs-CRP: P-value: NS, ROC:0.50) was carried out, showing that both Neu5Ac and Neu5,9Ac2 outperformed this marker. Further to this, hs-CRP values were combined with the three different sialic acid markers to determine any effect on the AUC values. A slight improvement in AUC was noted for each of the combinations, with Neu5Ac + Neu5,9Ac2 + hs-CRP giving the best AUC of 0.97 overall. Thus, Neu5Ac would appear to offer good potential as a predictive marker for the presence of CVD, which the addition of Neu5,9Ac2 predictive power improves, with further improvement seen by the addition of hs-CRP.
Collapse
Affiliation(s)
- Jack Cheeseman
- School of Pharmacy, University of Reading, Whiteknights, Reading, RG6 6AD, UK
- Ludger Ltd, Culham Science Centre, Abingdon, OX14 3EB, UK
| | | | | | | | - Duong N Trinh
- Department of Chemistry, Royal College of Surgeons in Ireland (RCSI), Dublin 2, Dublin, D02 YN77, Ireland
- Department of Pharmaceutics and Pharmaceutical Technology, University of Medicine and Pharmacy, Vietnam National University, Hanoi, Vietnam
| | - Marco P Monopoli
- Department of Chemistry, Royal College of Surgeons in Ireland (RCSI), Dublin 2, Dublin, D02 YN77, Ireland
| | - Gunter Kuhnle
- Department of Food and Nutritional Sciences, University of Reading, Whiteknights, Reading, RG6 6AH, UK
| | | | - Helen M I Osborn
- School of Pharmacy, University of Reading, Whiteknights, Reading, RG6 6AD, UK.
| |
Collapse
|
12
|
Bai J, Chen L, Xu L, Zhang Q, Liu J, Zheng K. The value of serum Sema4D level in predicting the prognosis of patients with acute ST-segment elevation myocardial infarction and with high thrombus burden. BMC Cardiovasc Disord 2023; 23:230. [PMID: 37138227 PMCID: PMC10157983 DOI: 10.1186/s12872-023-03244-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 04/15/2023] [Indexed: 05/05/2023] Open
Abstract
BACKGROUND Acute ST-segment elevation myocardial infarction (STEMI) is a serious cardiovascular disease. High thrombus burden is an independent risk factor for poor prognosis of acute myocardial infarction. However, there is no study on the correlation between soluble semaphorin 4D (sSema4D) level and high thrombus burden in patients with STEMI. PURPOSE This study aimed to investigate the relationship between sSema4D level and the thrombus burden of STEMI and further explore its effect on the main predictive value of the occurrence of major adverse cardiovascular events (MACE). METHODS From October 2020 to June 2021, 100 patients with STEMI diagnosed in our hospital's cardiology department were selected. According to the thrombolysis in myocardial infarction(TIMI)score, STEMI patients were divided into high thrombus burden groups (55 cases) and non-high thrombus burden groups (45 cases) 0.74 patients with stable coronary heart disease (CHD) were selected as stable CHD group, and 75 patients with negative coronary angiography (CAG) were selected as control group. Serum sSema4D levels were measured in 4 groups. The correlation between serum sSema4D and high-sensitivity C-reactive protein (hs-CRP) in patients with STEMI was analyzed. The relationship of serum sSema4D levels between the high and non-high thrombus burden group was evaluated. The effect of sSema4D levels on the occurrence of MACE was explored in one year after percutaneous coronary intervention. RESULTS Serum sSema4D level was positively correlated with hs-CRP level in STEMI patients (P < 0.05) with a correlation coefficient of 0.493. The sSema4D level was significantly higher in the high versus non-high thrombus burden group (22.54(20.82,24.17), P < 0.05). Moreover, MACE occurred in 19 cases in high thrombus burden group and 3 cases in non-high thrombus burden group. The results of Cox regression analysis showed that sSema4D was an independent predictor of MACE (OR = 1.497,95% CI: 1.213-1.847, P < 0.001). CONCLUSION The sSema4D level is associated with coronary thrombus burden and is an independent risk factor for MACE.
Collapse
Affiliation(s)
- Jie Bai
- Department of Cardiology, Affiliated Hospital 2 of Nantong University, Nantong First People's Hospital , Nantong, 226001, Jiangsu, China
| | - Liang Chen
- Department of Cardiology, Affiliated Hospital 2 of Nantong University, Nantong First People's Hospital , Nantong, 226001, Jiangsu, China
| | - Louyuan Xu
- Department of Cardiology, Affiliated Hospital 2 of Nantong University, Nantong First People's Hospital , Nantong, 226001, Jiangsu, China
| | - Qingquan Zhang
- Department of Cardiology, Affiliated Hospital 2 of Nantong University, Nantong First People's Hospital , Nantong, 226001, Jiangsu, China
| | - Jun Liu
- Department of Cardiology, Affiliated Hospital 2 of Nantong University, Nantong First People's Hospital , Nantong, 226001, Jiangsu, China
| | - Koulong Zheng
- Department of Cardiology, Affiliated Hospital 2 of Nantong University, Nantong First People's Hospital , Nantong, 226001, Jiangsu, China.
| |
Collapse
|
13
|
Humes HD, Aaronson KD, Buffington DA, Sabbah HN, Westover AJ, Yessayan LT, Szamosfalvi B, Pagani FD. Translation of immunomodulatory therapy to treat chronic heart failure: Preclinical studies to first in human. PLoS One 2023; 18:e0273138. [PMID: 37023139 PMCID: PMC10079025 DOI: 10.1371/journal.pone.0273138] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 03/22/2023] [Indexed: 04/07/2023] Open
Abstract
BACKGROUND Inflammation has been associated with progression and complications of chronic heart failure (HF) but no effective therapy has yet been identified to treat this dysregulated immunologic state. The selective cytopheretic device (SCD) provides extracorporeal autologous cell processing to lessen the burden of inflammatory activity of circulating leukocytes of the innate immunologic system. AIM The objective of this study was to evaluate the effects of the SCD as an extracorporeal immunomodulatory device on the immune dysregulated state of HF. HF. METHODS AND RESULTS SCD treatment in a canine model of systolic HF or HF with reduced ejection fraction (HFrEF) diminished leukocyte inflammatory activity and enhanced cardiac performance as measured by left ventricular (LV) ejection fraction and stroke volume (SV) up to 4 weeks after treatment initiation. Translation of these observations in first in human, proof of concept clinical study was evaluated in a patient with severe HFrEFHFrEF ineligible for cardiac transplantation or LV LV assist device (LVAD) due to renal insufficiency and right ventricular dysfunction. Six hour SCD treatments over 6 consecutive days resulted in selective removal of inflammatory neutrophils and monocytes and reduction in key plasma cytokines, including tumor necrosis factor-alpha (TNF-α),), interleukin (IL)-6, IL-8, and monocyte chemoattractant protein (MCP)-1. These immunologic changes were associated with significant improvements in cardiac power output, right ventricular stroke work index, cardiac index and LVSV index…. Stabilization of renal function with progressive volume removal permitted successful LVAD implantation. CONCLUSION This translational research study demonstrates a promising immunomodulatory approach to improve cardiac performance in HFrEFHFrEF and supports the important role of inflammation in the progression of HFHF.
Collapse
Affiliation(s)
- H. David Humes
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
- Innovative Biotherapies, Ann Arbor, Michigan, United States of America
| | - Keith D. Aaronson
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Deborah A. Buffington
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Hani N. Sabbah
- Department of Medicine, Henry Ford Hospital, Detroit, Michigan, United States of America
| | - Angela J. Westover
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
- Innovative Biotherapies, Ann Arbor, Michigan, United States of America
| | - Lenar T. Yessayan
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Balazs Szamosfalvi
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Francis D. Pagani
- Department of Cardiovascular Surgery, University of Michigan, Ann Arbor, Michigan, United States of America
| |
Collapse
|
14
|
Matsiras D, Bezati S, Ventoulis I, Verras C, Parissis J, Polyzogopoulou E. Gut Failure: A Review of the Pathophysiology and Therapeutic Potentials in the Gut-Heart Axis. J Clin Med 2023; 12:2567. [PMID: 37048650 PMCID: PMC10095379 DOI: 10.3390/jcm12072567] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 03/26/2023] [Accepted: 03/27/2023] [Indexed: 03/31/2023] Open
Abstract
Despite considerable advances in the field, heart failure (HF) still poses a significant disease burden among affected individuals since it continues to cause high morbidity and mortality rates. Inflammation is considered to play a key role in disease progression, but the exact underlying pathophysiological mechanisms involved have not yet been fully elucidated. The gut, as a potential source of inflammation, could feasibly explain the state of low-grade inflammation seen in patients with chronic HF. Several derangements in the composition of the microbiota population, coupled with an imbalance between favorable and harmful metabolites and followed by gut barrier disruption and eventually bacterial translocation, could contribute to cardiac dysfunction and aggravate HF. On the other hand, HF-associated congestion and hypoperfusion alters intestinal function, thereby creating a vicious cycle. Based on this evidence, novel pharmaceutical agents have been developed and their potential therapeutic use has been tested in both animal and human subjects. The ultimate goal in these efforts is to reverse the aforementioned intestinal derangements and block the inflammation cascade. This review summarizes the gut-related causative pathways implicated in HF pathophysiology, as well as the associated therapeutic interventions described in the literature.
Collapse
Affiliation(s)
- Dionysis Matsiras
- Emergency Medicine Department, Attikon University Hospital, Rimini 1, 12462 Athens, Greece
| | - Sofia Bezati
- Emergency Medicine Department, Attikon University Hospital, Rimini 1, 12462 Athens, Greece
| | - Ioannis Ventoulis
- Department of Occupational Therapy, University of Western Macedonia, 50200 Ptolemaida, Greece
| | - Christos Verras
- Emergency Medicine Department, Attikon University Hospital, Rimini 1, 12462 Athens, Greece
| | - John Parissis
- Emergency Medicine Department, Attikon University Hospital, Rimini 1, 12462 Athens, Greece
- Emergency Medicine Department, National and Kapodistrian University of Athens, 15772 Athens, Greece
| | - Effie Polyzogopoulou
- Emergency Medicine Department, Attikon University Hospital, Rimini 1, 12462 Athens, Greece
- Emergency Medicine Department, National and Kapodistrian University of Athens, 15772 Athens, Greece
| |
Collapse
|
15
|
Torp MK, Vaage J, Stensløkken KO. Mitochondria-derived damage-associated molecular patterns and inflammation in the ischemic-reperfused heart. Acta Physiol (Oxf) 2023; 237:e13920. [PMID: 36617670 DOI: 10.1111/apha.13920] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 10/01/2022] [Accepted: 01/02/2023] [Indexed: 01/10/2023]
Abstract
Cardiac cell death after myocardial infarction release endogenous structures termed damage-associated molecular patterns (DAMPs) that trigger the innate immune system and initiate a sterile inflammation in the myocardium. Cardiomyocytes are energy demanding cells and 30% of their volume are mitochondria. Mitochondria are evolutionary endosymbionts originating from bacteria containing molecular patterns similar to bacteria, termed mitochondrial DAMPs (mDAMPs). Consequently, mitochondrial debris may be particularly immunogenic and damaging. However, the role of mDAMPs in myocardial infarction is not clarified. Identifying the most harmful mDAMPs and inhibiting their early inflammatory signaling may reduce infarct size and the risk of developing post-infarct heart failure. The focus of this review is the role of mDAMPs in the immediate pro-inflammatory phase after myocardial infarction before arrival of immune cells in the myocardium. We discuss different mDAMPs, their role in physiology and present knowledge regarding their role in the inflammatory response of acute myocardial infarction.
Collapse
Affiliation(s)
- May-Kristin Torp
- Division of Physiology, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Jarle Vaage
- Division of Physiology, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Research and Development, Division of Emergencies and Critical Care, Oslo University Hospital, Oslo, Norway
| | - Kåre-Olav Stensløkken
- Division of Physiology, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| |
Collapse
|
16
|
The AP-1 transcription factor Fosl-2 drives cardiac fibrosis and arrhythmias under immunofibrotic conditions. Commun Biol 2023; 6:161. [PMID: 36759717 PMCID: PMC9911788 DOI: 10.1038/s42003-023-04534-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 01/26/2023] [Indexed: 02/11/2023] Open
Abstract
Fibrotic changes in the myocardium and cardiac arrhythmias represent fatal complications in systemic sclerosis (SSc), however the underlying mechanisms remain elusive. Mice overexpressing transcription factor Fosl-2 (Fosl-2tg) represent animal model of SSc. Fosl-2tg mice showed interstitial cardiac fibrosis, disorganized connexin-43/40 in intercalated discs and deregulated expression of genes controlling conduction system, and developed higher heart rate (HR), prolonged QT intervals, arrhythmias with prevalence of premature ventricular contractions, ventricular tachycardias, II-degree atrio-ventricular blocks and reduced HR variability. Following stimulation with isoproterenol Fosl-2tg mice showed impaired HR response. In contrast to Fosl-2tg, immunodeficient Rag2-/-Fosl-2tg mice were protected from enhanced myocardial fibrosis and ECG abnormalities. Transcriptomics analysis demonstrated that Fosl-2-overexpression was responsible for profibrotic signature of cardiac fibroblasts, whereas inflammatory component in Fosl-2tg mice activated their fibrotic and arrhythmogenic phenotype. In human cardiac fibroblasts FOSL-2-overexpression enhanced myofibroblast signature under proinflammatory or profibrotic stimuli. These results demonstrate that under immunofibrotic conditions transcription factor Fosl-2 exaggerates myocardial fibrosis, arrhythmias and aberrant response to stress.
Collapse
|
17
|
Cardiovascular Disease as a Consequence or a Cause of Cancer: Potential Role of Extracellular Vesicles. Biomolecules 2023; 13:biom13020321. [PMID: 36830690 PMCID: PMC9953640 DOI: 10.3390/biom13020321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 02/02/2023] [Accepted: 02/06/2023] [Indexed: 02/11/2023] Open
Abstract
Both cardiovascular disease and cancer continue to be causes of morbidity and mortality all over the world. Preventing and treating heart disease in patients undergoing cancer treatment remain an important and ongoing challenge for improving the lives of cancer patients, but also for their survival. Despite ongoing efforts to improve patient survival, minimal advances have been made in the early detection of cardiovascular disease in patients suffering from cancer. Understanding the communication between cancer and cardiovascular disease can be based on a deeper knowledge of the molecular mechanisms that define the profile of the bilateral network and establish disease-specific biomarkers and therapeutic targets. The role of exosomes, microvesicles, and apoptotic bodies, together defined as extracellular vesicles (EVs), in cross talk between cardiovascular disease and cancer is in an incipient form of research. Here, we will discuss the preclinical evidence on the bilateral connection between cancer and cardiovascular disease (especially early cardiac changes) through some specific mediators such as EVs. Investigating EV-based biomarkers and therapies may uncover the responsible mechanisms, detect the early stages of cardiovascular damage and elucidate novel therapeutic approaches. The ultimate goal is to reduce the burden of cardiovascular diseases by improving the standard of care in oncological patients treated with anticancer drugs or radiotherapy.
Collapse
|
18
|
Extracellular Vesicles in Aging: An Emerging Hallmark? Cells 2023; 12:cells12040527. [PMID: 36831194 PMCID: PMC9954704 DOI: 10.3390/cells12040527] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 02/01/2023] [Accepted: 02/01/2023] [Indexed: 02/08/2023] Open
Abstract
Extracellular vesicles (EVs) are membrane-enclosed particles secreted by cells and circulating in body fluids. Initially considered as a tool to dispose of unnecessary material, they are now considered an additional method to transmit cell signals. Aging is characterized by a progressive impairment of the physiological functions of tissues and organs. The causes of aging are complex and interconnected, but there is consensus that genomic instability, telomere erosion, epigenetic alteration, and defective proteostasis are primary hallmarks of the aging process. Recent studies have provided evidence that many of these primary stresses are associated with an increased release of EVs in cell models, able to spread senescence signals in the recipient cell. Additional investigations on the role of EVs during aging also demonstrated the great potential of EVs for the modulation of age-related phenotypes and for pro-rejuvenation therapies, potentially beneficial for many diseases associated with aging. Here we reviewed the current literature on EV secretion in senescent cell models and in old vs. young individual body fluids, as well as recent studies addressing the potential of EVs from different sources as an anti-aging tool. Although this is a recent field, the robust consensus on the altered EV release in aging suggests that altered EV secretion could be considered an emerging hallmark of aging.
Collapse
|
19
|
Iwai-Saito K, Sato K, Kondo K. Associations of influenza and pneumococcal vaccinations with burdens of older family caregivers: The Japan Gerontological Evaluation study (JAGES) cross-sectional study. Vaccine 2023; 41:444-451. [PMID: 36470685 DOI: 10.1016/j.vaccine.2022.11.047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 10/28/2022] [Accepted: 11/20/2022] [Indexed: 12/03/2022]
Abstract
BACKGROUND Influenza and pneumonia tend to be severe in older adults; thus, vaccination is necessary to prevent these illnesses. Vaccination is especially important for older family caregivers (OFCs) not only to prevent them from becoming ill, but also to prevent secondary infections in the family care receivers (FCRs), who are mostly frail older adults and have a higher risk of severe illness. Thus, we investigated whether caregiving burdens were associated with the vaccinations among older adults. METHODS We used cross-sectional data from the Japan Gerontological Evaluation Study (JAGES), which was conducted in 64 Japanese municipalities from November 2019 to January 2020. The target population consisted of 26,177 individuals aged 65 years or older who were independent and did not need public long-term care. The primary outcome was the uptakes of either or both influenza and pneumococcal vaccinations. Multinomial logistic regressions were performed, setting those who underwent neither vaccinations as the reference group. RESULTS Among the participants, 23.3 %, 25.8 %, 9.4 %, or 41.5 % underwent neither, only influenza, only pneumococcal, or the both vaccinations, respectively. The caregiving frequency, time length in a day, or dementia of FCR were negatively associated with influenza vaccination (caregiving almost every day: relative risk ratio {RRR}: 0.39, 95 % confident interval {95 % CI} [0.24-0.63]; caregiving almost all day: 0.44, 95 % CI: 0.23-0.85; caregiving for FCR: RRR:0.55, 95 % CI: 0.34-0.91). On the other hand, those caregiving burdens were not associated with pneumococcal only or the both vaccinations. Having a family physician mitigated all the negative effect of the caregiving burdens on the vaccinations. CONCLUSION Our results suggest that the caregiving burden is a barrier to influenza vaccination but not to pneumococcal vaccination and that having a physician mitigates the negative effect regardless of the burden kind.
Collapse
Affiliation(s)
- Kousuke Iwai-Saito
- Division of International Health, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan.
| | - Koryu Sato
- Department of Social Epidemiology, Graduate School of Medicine and School of Public Health, Kyoto University, Kyoto, Japan.
| | - Katsunori Kondo
- Department of Social Preventive Medical Sciences, Center for Preventive Medical Sciences, Chiba University, 1-8-1, Inohana, Chuo-ku, Chiba 360-0856, Japan; Department of Gerontological Evaluation, Center for Gerontology and Social Science, Research Institution, National Center for Geriatrics and Gerontology, Aichi, Japan.
| |
Collapse
|
20
|
Liu T, Wang B, Xiao S, Sun L, Zhu Z, Wang S, Li B, Yao J, Huang C, Ge W, Qian L, Lu Z, Pan Y. Correlation analysis between the static and the changed neutrophil-to-lymphocyte ratio and in-hospital mortality in critical patients with acute heart failure. Postgrad Med 2023; 135:50-57. [PMID: 36154549 DOI: 10.1080/00325481.2022.2129177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
OBJECTIVE Association between neutrophil-to-lymphocyte ratio (NLR) on admission and poor prognosis in patients with acute heart failure (AHF) has been well established. However, the relationship between dynamic changes in NLR and in-hospital mortality in AHF patients has not been studied. Our purpose was to determine if an early change in NLR within the first week after AHF patients was admitted to intensive care unit (ICU) was associated with in-hospital mortality. METHODS Data from the medical information mart for intensive care IV (the MIMIC-IV) database was analyzed. The effect of baseline NLR on in-hospital mortality in critical patients with AHF was evaluated utilizing smooth curve fitting and multivariable logistic regression analysis. Moreover, comparison of the dynamic change in NLR among survivors and non-survivors was performed using the generalized additive mixed model (GAMM). RESULTS There were 1169 participants who took part in the present study, 986 of whom were in-hospital survivors and 183 of whom were in-hospital non-survivors. The smooth curve fitting revealed a positive relationship between baseline NLR and in-hospital mortality, and multivariable logistic regression analysis indicated that baseline NLR was an independent risk factor for in-hospital mortality (OR 1.04, 95% CI 1.02,1.07, P-value = 0.001). After adjusting for confounders, GAMM showed that the difference in NLR between survivors and non-survivors grew gradually during the first week after ICU admission, and the difference grew by an average of 0.51 per day (β = 0.51, 95% CI 0.45-0.56, P-value <0.001). CONCLUSIONS Baseline NLR was associated with poor prognosis in critical patients with AHF. Early rises in NLR were linked to higher in-hospital mortality, which suggests that keeping track of how NLR early changes might help identify short-term prognosis of critical patients with AHF.
Collapse
Affiliation(s)
- Tao Liu
- Department of Cardiology, Jinshan Branch of Shanghai Sixth People's Hospital, Shanghai, China
| | - Bing Wang
- Department of Cardiology, Jinshan Branch of Shanghai Sixth People's Hospital, Shanghai, China
| | - Shengjue Xiao
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Lifang Sun
- Department of Cardiology, Jinshan Branch of Shanghai Sixth People's Hospital, Shanghai, China
| | - Zhijian Zhu
- Department of Cardiology, Jinshan Branch of Shanghai Sixth People's Hospital, Shanghai, China
| | - Shasha Wang
- Department of Cardiology, Jinshan Branch of Shanghai Sixth People's Hospital, Shanghai, China
| | - Baoyin Li
- Department of Cardiology, Jinshan Branch of Shanghai Sixth People's Hospital, Shanghai, China
| | - Jianhui Yao
- Department of Cardiology, Jinshan Branch of Shanghai Sixth People's Hospital, Shanghai, China
| | - Conggang Huang
- Department of Cardiology, Jinshan Branch of Shanghai Sixth People's Hospital, Shanghai, China
| | - Wei Ge
- Department of Cardiology, Jinshan Branch of Shanghai Sixth People's Hospital, Shanghai, China
| | - Lei Qian
- Department of Cardiology, Jinshan Branch of Shanghai Sixth People's Hospital, Shanghai, China
| | - Zhigang Lu
- Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yesheng Pan
- Department of Cardiology, Jinshan Branch of Shanghai Sixth People's Hospital, Shanghai, China
| |
Collapse
|
21
|
Abdallah YEH, Chahal S, Jamali F, Mahmoud SH. Drug-disease interaction: Clinical consequences of inflammation on drugs action and disposition. JOURNAL OF PHARMACY & PHARMACEUTICAL SCIENCES : A PUBLICATION OF THE CANADIAN SOCIETY FOR PHARMACEUTICAL SCIENCES, SOCIETE CANADIENNE DES SCIENCES PHARMACEUTIQUES 2023; 26:11137. [PMID: 36942294 PMCID: PMC9990632 DOI: 10.3389/jpps.2023.11137] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 01/23/2023] [Indexed: 02/07/2023]
Abstract
Inflammation is a culprit in many conditions affecting millions of people worldwide. A plethora of studies has revealed that inflammation and inflammatory mediators such as cytokines and chemokines are associated with altered expression and activity of various proteins such as those involved in drug metabolism, specifically cytochrome P450 enzymes (CYPs). Emphasis of most available reports is on the inflammation-induced downregulation of CYPs, subsequently an increase in their substrate concentrations, and the link between the condition and the inflammatory mediators such as interleukin-6 and tumor necrosis factor alpha. However, reports also suggest that inflammation influences expression and/or activity of other proteins such as those involved in the drug-receptor interaction. These multifaced involvements render the clinical consequence of the inflammation unexpected. Such changes are shown in many inflammatory conditions including rheumatoid arthritis, Crohn's disease, acute respiratory illnesses as well as natural processes such as aging, among others. For example, some commonly used cardiovascular drugs lose their efficacy when patients get afflicted with inflammatory conditions such as rheumatoid arthritis and Crohn's disease. Interestingly, this is despite increased concentration subsequent to reduced clearance. The observation is attributed to a simultaneous reduction in the expression of target receptor proteins such as the calcium and potassium channel and β-adrenergic receptor as well as the metabolic enzymes. This narrative review summarizes the current understanding and clinical implications of the inflammatory effects on both CYPs and drug-receptor target proteins.
Collapse
|
22
|
MacDonnell S, Megna J, Ruan Q, Zhu O, Halasz G, Jasewicz D, Powers K, E H, del Pilar Molina-Portela M, Jin X, Zhang D, Torello J, Feric NT, Graziano MP, Shekhar A, Dunn ME, Glass D, Morton L. Activin A directly impairs human cardiomyocyte contractile function indicating a potential role in heart failure development. Front Cardiovasc Med 2022; 9:1038114. [PMID: 36440002 PMCID: PMC9685658 DOI: 10.3389/fcvm.2022.1038114] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 10/10/2022] [Indexed: 09/27/2023] Open
Abstract
Activin A has been linked to cardiac dysfunction in aging and disease, with elevated circulating levels found in patients with hypertension, atherosclerosis, and heart failure. Here, we investigated whether Activin A directly impairs cardiomyocyte (CM) contractile function and kinetics utilizing cell, tissue, and animal models. Hydrodynamic gene delivery-mediated overexpression of Activin A in wild-type mice was sufficient to impair cardiac function, and resulted in increased cardiac stress markers (N-terminal pro-atrial natriuretic peptide) and cardiac atrophy. In human-induced pluripotent stem cell-derived (hiPSC) CMs, Activin A caused increased phosphorylation of SMAD2/3 and significantly upregulated SERPINE1 and FSTL3 (markers of SMAD2/3 activation and activin signaling, respectively). Activin A signaling in hiPSC-CMs resulted in impaired contractility, prolonged relaxation kinetics, and spontaneous beating in a dose-dependent manner. To identify the cardiac cellular source of Activin A, inflammatory cytokines were applied to human cardiac fibroblasts. Interleukin -1β induced a strong upregulation of Activin A. Mechanistically, we observed that Activin A-treated hiPSC-CMs exhibited impaired diastolic calcium handling with reduced expression of calcium regulatory genes (SERCA2, RYR2, CACNB2). Importantly, when Activin A was inhibited with an anti-Activin A antibody, maladaptive calcium handling and CM contractile dysfunction were abrogated. Therefore, inflammatory cytokines may play a key role by acting on cardiac fibroblasts, causing local upregulation of Activin A that directly acts on CMs to impair contractility. These findings demonstrate that Activin A acts directly on CMs, which may contribute to the cardiac dysfunction seen in aging populations and in patients with heart failure.
Collapse
Affiliation(s)
| | - Jake Megna
- Regeneron Pharmaceuticals, Tarrytown, NY, United States
| | - Qin Ruan
- Regeneron Pharmaceuticals, Tarrytown, NY, United States
| | - Olivia Zhu
- Regeneron Pharmaceuticals, Tarrytown, NY, United States
| | - Gabor Halasz
- Regeneron Pharmaceuticals, Tarrytown, NY, United States
| | - Dan Jasewicz
- Regeneron Pharmaceuticals, Tarrytown, NY, United States
| | - Kristi Powers
- Regeneron Pharmaceuticals, Tarrytown, NY, United States
| | - Hock E
- Regeneron Pharmaceuticals, Tarrytown, NY, United States
| | | | - Ximei Jin
- Regeneron Pharmaceuticals, Tarrytown, NY, United States
| | - Dongqin Zhang
- Regeneron Pharmaceuticals, Tarrytown, NY, United States
| | | | - Nicole T. Feric
- TARA Biosystems Inc., Alexandria Center for Life Sciences, New York, NY, United States
| | - Michael P. Graziano
- TARA Biosystems Inc., Alexandria Center for Life Sciences, New York, NY, United States
| | | | | | - David Glass
- Regeneron Pharmaceuticals, Tarrytown, NY, United States
| | - Lori Morton
- Regeneron Pharmaceuticals, Tarrytown, NY, United States
| |
Collapse
|
23
|
Wang R, Wu J, Ye H, Zhang X, Wang L. Application Value of Systemic Inflammatory Indexes in the Clinical Evaluation of Patients with Heart Failure with Preserved Ejection Fraction (HFpEF). MEDICINA (KAUNAS, LITHUANIA) 2022; 58:medicina58101473. [PMID: 36295633 PMCID: PMC9611882 DOI: 10.3390/medicina58101473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/17/2022] [Accepted: 10/12/2022] [Indexed: 11/07/2022]
Abstract
Background: In areas where medical resources are scarce, an economical and convenient way to assess patients’ condition so that treatment plans can be adjusted in a timely manner makes sense. The clinical value of systemic inflammatory indexes (SII) such as neutrophil-to-lymphocyte ratio (NLR), lymphocyte-to-monocyte ratio (LMR), albumin-to-gamma-glutamyl-transferase ratio (AGR), white-blood-cell-count-to-mean-platelet-volume ratio (WMR), high-density-lipoprotein-cholesterol-to-C-reactive-protein ratio (HCR), etc. were explored in heart failure (HF) with preserved ejection fraction (HFpEF) because of their easy availability and clinical value in the diagnosis, therapy and prognosis of cardiovascular diseases. Methods: 189 inpatients (including 48 patients with New York Heart Association (NYHA) I in the control group, and 141 patients with NYHA II-IV in the study group) from The First Affiliated Hospital of Jinan University, during the period July 2018 to March 2022, were included by retrieving electronic medical records. Logistic regression analysis, Spearman’s correlation coefficient, operating characteristic curve, etc. were used to analyze the data. Results: In patients with HFpEF, LMR (OR = 0.463, 95% CI 0.348−0.617, p = 0.000), NLR and N-terminal pro-B-type natriuretic peptide (NT-proBNP) were independent predictors for the presence of HF, and LMR (OR = 2.630, 95% CI 2.016−3.435, p = 0.000), NLR, FAG, MHR, AGR and NT-proBNP were independent predictors for increased NYHA functional classification. There were good correlations (r > 0.4) between LMR (r = −0.667, p = 0.000), NLR, WMR, HCR, NT-proBNP (r = −0.681, p = 0.000) and NYHA functional classification, and LMR (AUC = 0.803, 95% CI 0.729−0.849, p = 0.0001), NLR and NT-proBNP (AUC = 0.805, 95% CI 0.738−0.861, p = 0.0001) had good diagnostic values (AUC > 0.7) for HF in patients with HFpEF. In addition, there were certain correlations between LMR, NT-proBNP and echocardiography indicators of cardiac structural. Conclusions: SII have a potential application value in the clinical evaluation of patients with HFpEF in the follow-up, especially in areas with limited medical resources, as they are more convenient and cost effective. Among different SII, LMR is probably the most promising metric. However, large-scale clinical trials are needed in the future to confirm these findings.
Collapse
Affiliation(s)
- Ruxin Wang
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Juan Wu
- Clinical Laboratory, Suqian First People’s Hospital Affiliated to Nanjing Medical University, Suqian 223812, China
| | - Haowen Ye
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Xiaofang Zhang
- Clinical Experimental Center, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
- Correspondence: (X.Z.); (L.W.)
| | - Lihong Wang
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
- Correspondence: (X.Z.); (L.W.)
| |
Collapse
|
24
|
Iwai-Saito K, Sato K, Kondo K. Association of functional competencies with vaccination among older adults: a JAGES cross-sectional study. Sci Rep 2022; 12:17247. [PMID: 36241910 PMCID: PMC9568573 DOI: 10.1038/s41598-022-22192-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 10/11/2022] [Indexed: 01/06/2023] Open
Abstract
It is unknown whether higher functions in sublevels of competence other than instrumental activities of daily living (IADL) are associated with vaccinations. This study examined whether higher functions, including intellectual activity (IA) and social role (SR), were associated with vaccinations among 26,177 older adults. Older adults with incapable activities in IA and SR had increased risks for non-receipt of influenza vaccinations (IA: for one incapable task/activity: incident rate ratio (IRR) = 1.05, 95% confidence interval (CI) = 1.02-1.09; SR: for two incapable tasks: IRR = 1.12, 95% CI = 1.08-1.16). Those with incapable activities in IADL and IA had increased risks for non-receipt of pneumococcal vaccination (IADL: for two incapable tasks: IRR = 1.13, 95% CI = 1.05-1.23; IA: for two incapable tasks: IRR = 1.10, 95% CI = 1.08-1.12). Those with incapable activities in IADL, IA, and SR had increased risks for non-receipt of both of the two vaccinations (IADL: for two incapable tasks: IRR = 1.17, 95% CI = 1.03-1.33; IA: for two incapable tasks: IRR = 1.18, 95% CI = 1.11-1.25; SR: for two incapable tasks: IRR = 1.13, 95% CI = 1.07-1.20). Having a family physician mitigated associations for non-receipt, regardless of competency. Our results suggest-maintaining the higher functions are important for older adults to undergo recommended vaccinations as scheduled; also, having a family physician to promote vaccinations is beneficial even for older adults with limited functions.
Collapse
Affiliation(s)
- Kousuke Iwai-Saito
- Division of International Health, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan.
| | - Koryu Sato
- Department of Social Epidemiology, Graduate School of Medicine and School of Public Health, Kyoto University, Kyoto, Japan
| | - Katsunori Kondo
- Department of Social Preventive Medical Sciences, Center for Preventive Medical Sciences, Chiba University, 1-8-1, Inohana, Chuo-Ku, Chiba, 360-0856, Japan
- Department of Gerontological Evaluation, Center for Gerontology and Social Science, Research Institution, National Center for Geriatrics and Gerontology, Aichi, Japan
| |
Collapse
|
25
|
Prameswari HS, Putra ICS, Raffaello WM, Nathaniel M, Suhendro AS, Khalid AF, Pranata R. Managing Covid-19 in patients with heart failure: current status and future prospects. Expert Rev Cardiovasc Ther 2022; 20:807-828. [PMID: 36185009 DOI: 10.1080/14779072.2022.2132230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
INTRODUCTION COVID-19 may contribute to decompensation of previously stable chronic HF or cause a de-novo heart failure, which may come from the hyperinflammatory response and subsequent increase in metabolic demand. AREAS COVERED Two independent investigators searched MEDLINE (via PubMed), Europe PMC, and ScienceDirect databases with the following search terms: COVID-19, heart failure, COVID-19 drugs, heart failure drugs, and device therapy. All of the included full-text articles were rigorously evaluated by both authors in case there was disagreement about whether research should be included or not. In total, 157 studies were included and underwent extensive reading by the authors. EXPERT OPINION The World Health Organization (WHO) and the National Institute of Health (NIH) have published COVID-19 drug recommendations, although recommendations for HF-specific drug choices in COVID-19 are still lacking. We hope that this review can answer the void of comprehensive research data regarding the management options of HF in the COVID-19 condition so that clinicians can at least choose a more beneficial therapy or avoid combination therapies that have a high burden of side effects on HF; thus, morbidity and mortality in COVID-19 patients with HF may be reduced.
Collapse
Affiliation(s)
- Hawani Sasmaya Prameswari
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| | - Iwan Cahyo Santosa Putra
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| | | | - Michael Nathaniel
- School of Medicine and Health Sciences Atma Jaya Catholic University of Indonesia, Jakarta, Indonesia
| | - Adrian Sebastian Suhendro
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| | - Achmad Fitrah Khalid
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| | - Raymond Pranata
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| |
Collapse
|
26
|
Qin L, Cui J, Li J. Sympathetic Nerve Activity and Blood Pressure Response to Exercise in Peripheral Artery Disease: From Molecular Mechanisms, Human Studies, to Intervention Strategy Development. Int J Mol Sci 2022; 23:ijms231810622. [PMID: 36142521 PMCID: PMC9505475 DOI: 10.3390/ijms231810622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/08/2022] [Accepted: 09/09/2022] [Indexed: 11/16/2022] Open
Abstract
Sympathetic nerve activity (SNA) regulates the contraction of vascular smooth muscle and leads to a change in arterial blood pressure (BP). It was observed that SNA, vascular contractility, and BP are heightened in patients with peripheral artery disease (PAD) during exercise. The exercise pressor reflex (EPR), a neural mechanism responsible for BP response to activation of muscle afferent nerve, is a determinant of the exaggerated exercise-induced BP rise in PAD. Based on recent results obtained from a series of studies in PAD patients and a rat model of PAD, this review will shed light on SNA-driven BP response and the underlying mechanisms by which receptors and molecular mediators in muscle afferent nerves mediate the abnormalities in autonomic activities of PAD. Intervention strategies, particularly non-pharmacological strategies, improving the deleterious exercise-induced SNA and BP in PAD, and enhancing tolerance and performance during exercise will also be discussed.
Collapse
|
27
|
Long K, Zhao Z, Chen J, Zhi L, Wang C, Liao D, Wang M, Gao P. Yang-xin-xue keli exerts therapeutic effects via regulating mitochondrial homeostasis and function in doxorubicin-induced rat heart failure. Front Pharmacol 2022; 13:931453. [PMID: 36110548 PMCID: PMC9468485 DOI: 10.3389/fphar.2022.931453] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 08/02/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Heart failure, especially chronic heart failure, is generally induced by the accumulation of reactive oxygen species (ROS), as well as the subsequent loss of mitochondrial permeability transition pore (mPTP) openings and pathological mitochondrial dysfunction. Herein, we explored the therapeutic effects of the Chinese medicine Yangxin Keli (YXXKL) on chronic heart failure and its underlying working mechanism. Methods: To mimic oxidative stress-induced chronic heart failure, a rat heart failure model was induced by the administration of DOX. Transthoracic echocardiography was performed to confirm the successful establishment of the heart failure model by observing significantly decreased cardiac function in the rats. Mitochondrial membrane potential, function, and ATP synthesis activity were measured after YXXKL was employed. Results The administration of YXXKL not only significantly improved cardiac function but also reversed the myocardium loss and fibrosis induced via DOX. Moreover, the administration of YXXKL also increased ATP synthesis and mitochondrial DNA mass in left ventricular tissues, which indicated that mitochondria may be a key target of YXXKL. Thus, we employed rat cardiomyocyte H9c2 and primary rat cardiac myocytes (RCMs) to induce oxidative stress-induced myocardial injury via DOX treatment. YXXKL-medicated serum promoted cell proliferation, which was inhibited by the addition of IC30 DOX, and the serum also inhibited cell apoptosis, which was promoted by the addition of IC50 DOX. YXKL-medicated serum was able to scavenge ROS and maintain the mitochondrial membrane potential as well as promote mitochondrial function, including the promotion of ATP synthesis, mitochondrial DNA mass, and transcriptional activity. Furthermore, we also observed that YXXKL-medicated serum inhibited DOX-induced autophagy/mitophagy by scavenging ROS. Conclusion: Taken together, we conclude that YXXKLI may exert therapeutic effects on oxidative stress-related heart failure via the regulation of mitochondria.
Collapse
Affiliation(s)
- Kunlan Long
- Intensive Care Unit, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ziyi Zhao
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jun Chen
- Intensive Care Unit, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lijia Zhi
- Intensive Care Unit, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chunxia Wang
- Intensive Care Unit, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Dan Liao
- Intensive Care Unit, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Meng Wang
- Intensive Care Unit, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Peiyang Gao
- Intensive Care Unit, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- *Correspondence: Peiyang Gao,
| |
Collapse
|
28
|
Maroofi A, Moro T, Agrimi J, Safari F. Cognitive decline in heart failure: Biomolecular mechanisms and benefits of exercise. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166511. [PMID: 35932891 DOI: 10.1016/j.bbadis.2022.166511] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 07/18/2022] [Accepted: 07/29/2022] [Indexed: 11/24/2022]
Abstract
By definition, heart failure (HF) is a human pathological condition affecting the structure and function of all organs in the body, and the brain is not an exception to that. Failure of the heart to pump enough blood centrally and peripherally is at the foundation of HF patients' inability to attend even the most ordinary daily activities and progressive deterioration of their cognitive capacity. What is more, between heart and brain exists a bidirectional relationship that goes well beyond hemodynamics and concerns bioelectric and endocrine signaling. This increasingly consolidated evidence makes the scenario even more complex. Studies have mainly chased how HF impairs cognition without focusing much on preventive measures, notably cardio-cerebral health proxies. Here, we aim to provide a brief account of known and hypothetical factors that may explain how exercise can help obviate cognitive dysfunction associated with HF in its different forms. As we shall see, there is a stringent need for a deeper grasp of such mechanisms. Indeed, gaining this new knowledge will automatically shed new light on the inner workings of HF itself, thus resulting in more effective prevention and treatment of this escalating syndrome.
Collapse
Affiliation(s)
- Abdulbaset Maroofi
- Department of Exercise Physiology, Faculty of Physical Education & Sport Sciences, University of Guilan, Rasht, Iran
| | - Tatiana Moro
- Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy
| | - Jacopo Agrimi
- Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy.
| | - Fatemeh Safari
- Department of Physiology, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| |
Collapse
|
29
|
Wang X, Ni Q, Wang J, Wu S, Chen P, Xing D. Systemic Inflammation Response Index Is a Promising Prognostic Marker in Elderly Patients With Heart Failure: A Retrospective Cohort Study. Front Cardiovasc Med 2022; 9:871031. [PMID: 35911534 PMCID: PMC9330028 DOI: 10.3389/fcvm.2022.871031] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Accepted: 06/23/2022] [Indexed: 01/03/2023] Open
Abstract
Background Heart failure (HF) is a clinical syndrome caused by ventricular dysfunction, which leads to the decline of activity tolerance and repeated hospitalization, which seriously affects the quality of life and is the main cause of death of the elderly. It has long been observed that the pathophysiological mechanism of HF is associated with systemic inflammation. This study aims to explore the association between the systemic inflammation response index (SIRI), a novel biomarker of inflammation, and outcomes in elderly patients with HF. Methods Data was extracted from the Medical Information Mart data for Intensive Care III (MIMIC-III) database and the Second Affiliated Hospital of Wenzhou Medical University. The primary outcome was 90-day all-cause mortality. The secondary outcomes included 1-year all-cause mortality, the length of hospital or intensive care unit (ICU) stay, and the need for renal replacement therapy (RRT). Cox proportional hazards regression, linear regression, and logistic regression models were used to assess the association between SIRI levels and all-cause mortality, the length of hospital or ICU stay, the need for RRT, respectively. Moreover, Pearson correlation analysis was conducted to evaluate the correlation between SIRI and C-reactive protein (CRP). Results This study cohort included 3,964 patients from the MIMIC-III database and 261 patients from the Second Affiliated Hospital of Wenzhou Medical University. The result suggested that SIRI was independently associated with the 90-day, and 1-year all-cause mortality in elderly patients with HF (tertile 3 vs. tertile 1: adjusted HR, 95% CI: 1.41 (1.18, 1.68), 1.19 (1.03, 1.37); p trend = 0.0013, 0.0260; respectively). Elevated SIRI was associated with increased the length of hospital or ICU stay after adjusting for multiple confounders (tertile 3 vs. tertile 1: β, 95% CI: 0.85 (0.16, 1.54); 0.62 (0.18, 1.06); p trend = 0.0095, 0.0046; respectively). Furthermore, we found that patients with higher SIRI levels were more likely to require RRT (tertile 3 vs. tertile 1: OR, 95% CI: 1.55 (1.06, 2.28); p trend = 0.0459). Moreover, we confirmed that SIRI was statistically positively correlated with CRP (correlation coefficient r = 0.343, p <0.001). Conclusions SIRI could be a novel promising inflammatory biomarker for predicting all-cause mortality in elderly patients with HF. And the patients with higher SIRI values had the longer length of hospital or ICU stay and were more likely to require for RRT. Of note, this study also verified a statistically significant positive correlation between SIRI and the inflammatory marker CRP, highlighting the importance of systemic inflammation as a determinant of outcome in patients with HF.
Collapse
Affiliation(s)
- Xue Wang
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Qingwei Ni
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Jie Wang
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Shujie Wu
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Peng Chen
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Dawei Xing
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
30
|
Effects of Coenzyme Q10 Supplementation on Oxidative Stress Markers, Inflammatory Markers, Lymphocyte Subpopulations, and Clinical Status in Dogs with Myxomatous Mitral Valve Disease. Antioxidants (Basel) 2022; 11:antiox11081427. [PMID: 35892628 PMCID: PMC9394267 DOI: 10.3390/antiox11081427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 07/18/2022] [Accepted: 07/19/2022] [Indexed: 11/28/2022] Open
Abstract
Scarce data exist on the effects of coenzyme Q10 (CoQ10) supplementation in dogs with myxomatous mitral valve disease (MMVD). The purpose of this study was to investigate the effect of CoQ10 supplementation on oxidative stress markers (glutathione peroxidase, F2-isoprostanes), markers of inflammation (tumor necrosis factor-α, TNF soluble receptor II, leucocytes, and their subtypes), lymphocyte subpopulations (T helper and cytotoxic T lymphocytes, including activated T lymphocytes, and B lymphocytes), and echocardiographic and clinical parameters in dogs with MMVD. In this randomized, controlled, double-blind, longitudinal study, 43 MMVD dogs in stages ACVIM (American College of Veterinary Internal Medicine classification) B2 and ACVIM C and D (congestive heart failure (CHF)) received water-soluble coenzyme Q10 (100 mg twice daily) or placebo for 3 months, and 12 non-supplemented healthy dogs served as controls. All parameters were measured before and after supplementation in MMVD dogs and once in healthy dogs. CoQ10 supplementation had a positive impact on neutrophil percentage, lymphocyte percentage, and lymphocyte concentration in our cohort of dogs with CHF (ACVIM C and D). Conclusion: CoQ10 as an oral supplement may have benefits in terms of decreasing inflammation in dogs with MMVD and CHF.
Collapse
|
31
|
Cancer- and cardiac-induced cachexia: same fate through different inflammatory mediators? Inflamm Res 2022; 71:771-783. [PMID: 35680678 DOI: 10.1007/s00011-022-01586-y] [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: 11/17/2021] [Revised: 05/04/2022] [Accepted: 05/09/2022] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND Inflammation is widely recognized as the driving force of cachexia induced by chronic diseases; however, therapies targeting inflammation do not always reverse cachexia. Thus, whether inflammation per se plays an important role in the clinical course of cachectic patients is still a matter of debate. AIMS To give new insights into cachexia's pathogenesis and diagnosis, we performed a comprehensive literature search on the contribution of inflammatory markers to this syndrome, focusing on the noncommunicable diseases cancer and cardiovascular diseases. METHODS A systematic review was performed in PubMed using the keywords ("cancer" OR "cardiac" cachexia AND "human" OR "patient" AND "plasma" or "serum"). A total of 744 studies were retrieved and, from these, 206 were selected for full-text screening. In the end, 98 papers focusing on circulating biomarkers of cachexia were identified, which resulted in a list of 113 different mediators. RESULTS Data collected from the literature highlight the contribution of interleukin-6 (IL-6) and C-reactive protein (CRP) to cachexia, independently of the underlying condition. Despite not being specific, once the diagnosis of cachexia is established, CRP might help to monitor the effectiveness of anti-cachexia therapies. In cardiac diseases, B-type natriuretic peptide (BNP), renin, and obestatin might be putative markers of body wasting, whereas in cancer, growth differentiation factor (GDF) 15, transforming growth factor (TGF)-β1 and vascular endothelial growth factor (VEGF) C seem to be better markers of this syndrome. Independently of the circulating mediators, NF-κB and JAK/STAT signaling pathways play a key role in bridging inflammation with muscle wasting; however, therapies targeting these pathways were not proven effective for all cachectic patients. CONCLUSION The critical and integrative analysis performed herein will certainly feed future research focused on the better comprehension of cachexia pathogenesis toward the improvement of its diagnosis and the development of personalized therapies targeting specific cachexia phenotypes.
Collapse
|
32
|
Mongirdienė A, Liobikas J. Phenotypic and Functional Heterogeneity of Monocyte Subsets in Chronic Heart Failure Patients. BIOLOGY 2022; 11:195. [PMID: 35205062 PMCID: PMC8869357 DOI: 10.3390/biology11020195] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 01/19/2022] [Accepted: 01/21/2022] [Indexed: 11/16/2022]
Abstract
Chronic heart failure (CHF) results when the heart cannot consistently supply the body's tissues with oxygen and required nutrients. CHF can be categorized as heart failure (HF) with preserved ejection fraction (HFpEF) or HF with reduced ejection fraction (HFrEF). There are different causes and mechanisms underlying HF pathogenesis; however, inflammation can be regarded as one of the factors that promotes both HFrEF and HFpEF. Monocytes, a subgroup of leukocytes, are known to be cellular mediators in response to cardiovascular injury and are closely related to inflammatory reactions. These cells are a vital component of the immune system and are the source of macrophages, which participate in cardiac tissue repair after injury. However, these monocytes are not as homogenous as thought and can present different functions under different cardiovascular disease conditions. In addition, there is still an open question regarding whether the functions of monocytes and macrophages should be regarded as causes or consequences in CHF development. Therefore, the aim of this work was to summarize current studies on the functions of various monocyte subsets in CHF with a focus on the role of a certain monocyte subset in HFpEF and HFrEF patients, as well as the subsets' relationship to inflammatory markers.
Collapse
Affiliation(s)
- Aušra Mongirdienė
- Department of Biochemistry, Medical Academy, Lithuanian University of Health Sciences, LT50161 Kaunas, Lithuania
| | - Julius Liobikas
- Laboratory of Biochemistry, Neuroscience Institute, Lithuanian University of Health Sciences, LT50162 Kaunas, Lithuania
| |
Collapse
|
33
|
Zhai G, Zhang B, Wang J, Liu Y, Zhou Y. Prognostic Value of Pericardial Effusion Size in Patients with Acute Heart Failure. Curr Vasc Pharmacol 2022; 20:508-516. [PMID: 35899953 PMCID: PMC10009891 DOI: 10.2174/1570161120666220721094739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/26/2022] [Accepted: 05/31/2022] [Indexed: 01/25/2023]
Abstract
BACKGROUND Pericardial Effusion (PEf) can occur with Acute Heart Failure (AHF). OBJECTIVE To evaluate the effect of PEf size on the prognosis of patients with AHF. METHODS According to the maximum size of PEf, all patients were divided into five groups. The primary outcome was in-hospital mortality. The independent effect of PEf size was determined by binary logistic regression analysis. The curve in line with the overall trend was drawn by local weighted regression (Lowess). RESULTS We included 192 patients with AHF complicated by PEf. As PEf size increased, in-hospital mortality increased significantly (Group 5 vs. Group 1: 34.8 vs. 8.9% p=0.042). After adjusting for confounders, there was no significant association between PEf groups and in-hospital mortality (Group 5 vs. Group 1: odd ratio (OR), 95% confidence interval (CI): 2.72, 0.41-18.22, p=0.298). However, when PEf size was analysed as a continuous variable, an independent association between increased risk of inhospital mortality and PEf size was observed (OR, 95% CI: 1.08, 1.00-1.16, p=0.037). The Lowess curve showed a positive relationship between PEf size and in-hospital mortality. Furthermore, as PEf groups increased, the length of hospital stay (Group 5 vs. Group 1 median and interquartile range: 16, 14-21 vs. 13, 8-17 days, p<0.001) was significantly prolonged. An association between PEf size with acute kidney injury (AKI) was not observed. CONCLUSION The PEf size was independently associated with the increased risk of in-hospital mortality in patients with AHF.
Collapse
Affiliation(s)
- Guangyao Zhai
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University Affiliated Anzhen Hospital, Beijing 100089, China
| | - Biyang Zhang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University Affiliated Anzhen Hospital, Beijing 100089, China
| | - Jianlong Wang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University Affiliated Anzhen Hospital, Beijing 100089, China
| | - Yuyang Liu
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University Affiliated Anzhen Hospital, Beijing 100089, China
| | - Yujie Zhou
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University Affiliated Anzhen Hospital, Beijing 100089, China
| |
Collapse
|
34
|
Sun G, Yafasova A, Baslund B, Faurschou M, Schou M, Shams-Eldin A, Kristensen SL, Weeke PE, Torp-Pedersen C, Fosbøl EL, Køber L, Butt JH. Long-term Risk of Heart Failure and Other Adverse Cardiovascular Outcomes in Granulomatosis With Polyangiitis: a Nationwide Cohort Study. J Rheumatol 2021; 49:291-298. [PMID: 34782450 DOI: 10.3899/jrheum.210677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/03/2021] [Indexed: 11/22/2022]
Abstract
OBJECTIVE To examine the long-term rates of heart failure and other adverse cardiovascular outcomes in a nationwide cohort of patients diagnosed with granulomatosis with polyangiitis (GPA) compared with the background population. METHODS Using Danish nationwide registries, patients with first-time diagnosed GPA were identified and matched 1:4 by age, sex, and comorbidities with subjects from background population. Outcomes were compared using Cox regression. Due to violation of the proportional hazard assumption, landmark analyses for the first year and from one year were performed. RESULTS Of the 1,923 patients with GPA, 1,781 patients (median age 59 years, 47.9% men) were matched with 7,124 subjects from the background population. The median follow-up was 6.4 years. The absolute 10-year risk of HF was 6.8% (95%CI, 5.5-8.2%) for GPA patients and 5.9% (5.3-6.6%) for the background population. During the first year after diagnosis, GPA was associated with a significantly higher rate of HF (HR 3.60 [95%CI, 2.28-5.67]) and other adverse outcomes, including atrial fibrillation/flutter (HR 6.50 [4.43-9.55]) and ischemic stroke (HR 3.24 [1.92-5.48]), compared with the background population. After the first year, GPA was not associated with higher rates of HF or other cardiovascular outcomes than the background population, except atrial fibrillation/flutter (HR 1.38 [1.12-1.70]). CONCLUSION During the first year after diagnosis, the rates of HF and other cardiovascular outcomes were higher in patients with GPA compared with the background population. However, after the first year, the rates of HF and other cardiovascular outcomes, except atrial fibrillation/flutter, were similar to those in the background population.
Collapse
Affiliation(s)
- Guoli Sun
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark; Department of Rheumatology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark; Department of Cardiology, Herlev-Gentofte Hospital, Copenhagen University Hospital, Copenhagen, Denmark; Department of Cardiology, Nordsjællands Hospital, Hillerød, Denmark. The Capital Region of Denmark approved this study (approval number: P-2019-348) in accordance with the General Data Protection Regulation. In Denmark, registry-based studies in which individuals cannot be identified do not require ethical approval or informed consent. Address for Correspondence: Jawad Haider Butt, MD, Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Blegdamsvej 9, 2100 Copenhagen, Denmark. E-mail:
| | - Adelina Yafasova
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark; Department of Rheumatology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark; Department of Cardiology, Herlev-Gentofte Hospital, Copenhagen University Hospital, Copenhagen, Denmark; Department of Cardiology, Nordsjællands Hospital, Hillerød, Denmark. The Capital Region of Denmark approved this study (approval number: P-2019-348) in accordance with the General Data Protection Regulation. In Denmark, registry-based studies in which individuals cannot be identified do not require ethical approval or informed consent. Address for Correspondence: Jawad Haider Butt, MD, Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Blegdamsvej 9, 2100 Copenhagen, Denmark. E-mail:
| | - Bo Baslund
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark; Department of Rheumatology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark; Department of Cardiology, Herlev-Gentofte Hospital, Copenhagen University Hospital, Copenhagen, Denmark; Department of Cardiology, Nordsjællands Hospital, Hillerød, Denmark. The Capital Region of Denmark approved this study (approval number: P-2019-348) in accordance with the General Data Protection Regulation. In Denmark, registry-based studies in which individuals cannot be identified do not require ethical approval or informed consent. Address for Correspondence: Jawad Haider Butt, MD, Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Blegdamsvej 9, 2100 Copenhagen, Denmark. E-mail:
| | - Mikkel Faurschou
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark; Department of Rheumatology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark; Department of Cardiology, Herlev-Gentofte Hospital, Copenhagen University Hospital, Copenhagen, Denmark; Department of Cardiology, Nordsjællands Hospital, Hillerød, Denmark. The Capital Region of Denmark approved this study (approval number: P-2019-348) in accordance with the General Data Protection Regulation. In Denmark, registry-based studies in which individuals cannot be identified do not require ethical approval or informed consent. Address for Correspondence: Jawad Haider Butt, MD, Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Blegdamsvej 9, 2100 Copenhagen, Denmark. E-mail:
| | - Morten Schou
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark; Department of Rheumatology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark; Department of Cardiology, Herlev-Gentofte Hospital, Copenhagen University Hospital, Copenhagen, Denmark; Department of Cardiology, Nordsjællands Hospital, Hillerød, Denmark. The Capital Region of Denmark approved this study (approval number: P-2019-348) in accordance with the General Data Protection Regulation. In Denmark, registry-based studies in which individuals cannot be identified do not require ethical approval or informed consent. Address for Correspondence: Jawad Haider Butt, MD, Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Blegdamsvej 9, 2100 Copenhagen, Denmark. E-mail:
| | - Abdulrahman Shams-Eldin
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark; Department of Rheumatology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark; Department of Cardiology, Herlev-Gentofte Hospital, Copenhagen University Hospital, Copenhagen, Denmark; Department of Cardiology, Nordsjællands Hospital, Hillerød, Denmark. The Capital Region of Denmark approved this study (approval number: P-2019-348) in accordance with the General Data Protection Regulation. In Denmark, registry-based studies in which individuals cannot be identified do not require ethical approval or informed consent. Address for Correspondence: Jawad Haider Butt, MD, Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Blegdamsvej 9, 2100 Copenhagen, Denmark. E-mail:
| | - Søren Lund Kristensen
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark; Department of Rheumatology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark; Department of Cardiology, Herlev-Gentofte Hospital, Copenhagen University Hospital, Copenhagen, Denmark; Department of Cardiology, Nordsjællands Hospital, Hillerød, Denmark. The Capital Region of Denmark approved this study (approval number: P-2019-348) in accordance with the General Data Protection Regulation. In Denmark, registry-based studies in which individuals cannot be identified do not require ethical approval or informed consent. Address for Correspondence: Jawad Haider Butt, MD, Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Blegdamsvej 9, 2100 Copenhagen, Denmark. E-mail:
| | - Peter E Weeke
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark; Department of Rheumatology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark; Department of Cardiology, Herlev-Gentofte Hospital, Copenhagen University Hospital, Copenhagen, Denmark; Department of Cardiology, Nordsjællands Hospital, Hillerød, Denmark. The Capital Region of Denmark approved this study (approval number: P-2019-348) in accordance with the General Data Protection Regulation. In Denmark, registry-based studies in which individuals cannot be identified do not require ethical approval or informed consent. Address for Correspondence: Jawad Haider Butt, MD, Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Blegdamsvej 9, 2100 Copenhagen, Denmark. E-mail:
| | - Christian Torp-Pedersen
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark; Department of Rheumatology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark; Department of Cardiology, Herlev-Gentofte Hospital, Copenhagen University Hospital, Copenhagen, Denmark; Department of Cardiology, Nordsjællands Hospital, Hillerød, Denmark. The Capital Region of Denmark approved this study (approval number: P-2019-348) in accordance with the General Data Protection Regulation. In Denmark, registry-based studies in which individuals cannot be identified do not require ethical approval or informed consent. Address for Correspondence: Jawad Haider Butt, MD, Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Blegdamsvej 9, 2100 Copenhagen, Denmark. E-mail:
| | - Emil L Fosbøl
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark; Department of Rheumatology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark; Department of Cardiology, Herlev-Gentofte Hospital, Copenhagen University Hospital, Copenhagen, Denmark; Department of Cardiology, Nordsjællands Hospital, Hillerød, Denmark. The Capital Region of Denmark approved this study (approval number: P-2019-348) in accordance with the General Data Protection Regulation. In Denmark, registry-based studies in which individuals cannot be identified do not require ethical approval or informed consent. Address for Correspondence: Jawad Haider Butt, MD, Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Blegdamsvej 9, 2100 Copenhagen, Denmark. E-mail:
| | - Lars Køber
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark; Department of Rheumatology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark; Department of Cardiology, Herlev-Gentofte Hospital, Copenhagen University Hospital, Copenhagen, Denmark; Department of Cardiology, Nordsjællands Hospital, Hillerød, Denmark. The Capital Region of Denmark approved this study (approval number: P-2019-348) in accordance with the General Data Protection Regulation. In Denmark, registry-based studies in which individuals cannot be identified do not require ethical approval or informed consent. Address for Correspondence: Jawad Haider Butt, MD, Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Blegdamsvej 9, 2100 Copenhagen, Denmark. E-mail:
| | - Jawad H Butt
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark; Department of Rheumatology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark; Department of Cardiology, Herlev-Gentofte Hospital, Copenhagen University Hospital, Copenhagen, Denmark; Department of Cardiology, Nordsjællands Hospital, Hillerød, Denmark. The Capital Region of Denmark approved this study (approval number: P-2019-348) in accordance with the General Data Protection Regulation. In Denmark, registry-based studies in which individuals cannot be identified do not require ethical approval or informed consent. Address for Correspondence: Jawad Haider Butt, MD, Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Blegdamsvej 9, 2100 Copenhagen, Denmark. E-mail:
| |
Collapse
|
35
|
Hoyer-Kimura C, Konhilas JP, Mansour HM, Polt R, Doyle KP, Billheimer D, Hay M. Neurofilament light: a possible prognostic biomarker for treatment of vascular contributions to cognitive impairment and dementia. J Neuroinflammation 2021; 18:236. [PMID: 34654436 PMCID: PMC8520282 DOI: 10.1186/s12974-021-02281-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 09/20/2021] [Indexed: 12/29/2022] Open
Abstract
Background Decreased cerebral blood flow and systemic inflammation during heart failure (HF) increase the risk for vascular contributions to cognitive impairment and dementia (VCID) and Alzheimer disease-related dementias (ADRD). We previously demonstrated that PNA5, a novel glycosylated angiotensin 1–7 (Ang-(1–7)) Mas receptor (MasR) agonist peptide, is an effective therapy to rescue cognitive impairment in our preclinical model of VCID. Neurofilament light (NfL) protein concentration is correlated with cognitive impairment and elevated in neurodegenerative diseases, hypoxic brain injury, and cardiac disease. The goal of the present study was to determine (1) if treatment with Ang-(1–7)/MasR agonists can rescue cognitive impairment and decrease VCID-induced increases in NfL levels as compared to HF-saline treated mice and, (2) if NfL levels correlate with measures of cognitive function and brain cytokines in our VCID model. Methods VCID was induced in C57BL/6 male mice via myocardial infarction (MI). At 5 weeks post-MI, mice were treated with daily subcutaneous injections for 24 days, 5 weeks after MI, with PNA5 or angiotensin 1–7 (500 microg/kg/day or 50 microg/kg/day) or saline (n = 15/group). Following the 24-day treatment protocol, cognitive function was assessed using the Novel Object Recognition (NOR) test. Cardiac function was measured by echocardiography and plasma concentrations of NfL were quantified using a Quanterix Simoa assay. Brain and circulating cytokine levels were determined with a MILLIPLEX MAP Mouse High Sensitivity Multiplex Immunoassay. Treatment groups were compared via ANOVA, significance was set at p < 0.05. Results Treatment with Ang-(1–7)/MasR agonists reversed VCID-induced cognitive impairment and significantly decreased NfL levels in our mouse model of VCID as compared to HF-saline treated mice. Further, NfL levels were significantly negatively correlated with cognitive scores and the concentrations of multiple pleiotropic cytokines in the brain. Conclusions These data show that treatment with Ang-(1–7)/MasR agonists rescues cognitive impairment and decreases plasma NfL relative to HF-saline-treated animals in our VCID mouse model. Further, levels of NfL are significantly negatively correlated with cognitive function and with several brain cytokine concentrations. Based on these preclinical findings, we propose that circulating NfL might be a candidate for a prognostic biomarker for VCID and may also serve as a pharmacodynamic/response biomarker for therapeutic target engagement.
Collapse
Affiliation(s)
| | - John P Konhilas
- Department of Physiology, The University of Arizona, Tucson, AZ, USA.,Department of Nutritional Sciences, The University of Arizona, Tucson, AZ, USA.,Department of Biomedical Engineering, The University of Arizona, Tucson, AZ, USA.,Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, AZ, USA
| | - Heidi M Mansour
- Department of Pharmacy, Skaggs Pharmaceutical Sciences Center, The University of Arizona, Tucson, AZ, USA.,Department of Medicine, Division of Translational and Regenerative Medicine, The University of Arizona, Tucson, AZ, USA
| | - Robin Polt
- Department of Chemistry and Biochemistry, The University of Arizona, Tucson, AZ, USA
| | - Kristian P Doyle
- Department of Immunobiology, The University of Arizona, Tucson, AZ, USA
| | - Dean Billheimer
- Department of Epidemiology and Biostatistics, The University of Arizona, Tucson, AZ, USA
| | - Meredith Hay
- Department of Physiology, The University of Arizona, Tucson, AZ, USA.,Department of Neurology, The University of Arizona, Tucson, AZ, USA.,Evelyn F. McKnight Brain Institute, The University of Arizona, Tucson, AZ, USA.,ProNeurogen, Inc, The University of Arizona, Tucson, AZ, USA
| |
Collapse
|
36
|
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: 67] [Impact Index Per Article: 16.8] [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
|
37
|
Heidarpour M, Bashiri S, Vakhshoori M, Heshmat-Ghahdarijani K, Khanizadeh F, Ferdowsian S, Shafie D. The association between platelet-to-lymphocyte ratio with mortality among patients suffering from acute decompensated heart failure. BMC Cardiovasc Disord 2021; 21:454. [PMID: 34537010 PMCID: PMC8449504 DOI: 10.1186/s12872-021-02260-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 09/13/2021] [Indexed: 02/08/2023] Open
Abstract
Background Platelet-to-lymphocyte ratio (PLR) is an inflammation index suggested to have the prognostic capability in heart failure (HF). We sought to investigate the association of PLR with cardiovascular disease (CVD) mortality and creatinine (Cr) rise among Iranian individuals suffering from acute decompensated HF (ADHF). Methods This retrospective cohort study was in the context of the Persian Registry Of cardioVascular diseasE/Heart Failure (PROVE/HF) study. 405 individuals with ADHF admitted to the emergency department were recruited from April 2019 to March 2020. PLR was calculated by division of platelet to absolute lymphocyte counts and categorized based on quartiles. We utilized the Kaplan–Meier curve to show the difference in mortality based on PLR quartiles. Cr rise was defined as the increment of at least 0.3 mg/dl from baseline. Cox proportional hazard ratio (HR) was used to investigate the association of PLR with CVDs mortality. Results Mean age of participants was 65.9 ± 13.49 years (males: 67.7%). The mean follow-up duration was 4.26 ± 2.2 months. CVDs mortality or re-hospitalization was not significantly associated with PLR status. Multivariate analysis of PLR quartiles showed a minimally reduced likelihood of CVDs death in 2nd quartile versus the first one (HR 0.40, 95% confidence interval (CI) 0.16–1.01, P = 0.054). Cr rise had no remarkable relation with PLR status in neither model. Conclusion PLR could not be used as an independent prognostic factor among ADHF patients. Several studies are required clarifying the exact utility of this index.
Collapse
Affiliation(s)
- Maryam Heidarpour
- Isfahan Endocrine and Metabolism Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Sepideh Bashiri
- Cardiac Rehabilitation Research Center, Isfahan Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mehrbod Vakhshoori
- Heart Failure Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Kiyan Heshmat-Ghahdarijani
- Heart Failure Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | - Shaghayegh Ferdowsian
- Heart Failure Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Davood Shafie
- Heart Failure Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
38
|
Su JH, Luo MY, Liang N, Gong SX, Chen W, Huang WQ, Tian Y, Wang AP. Interleukin-6: A Novel Target for Cardio-Cerebrovascular Diseases. Front Pharmacol 2021; 12:745061. [PMID: 34504432 PMCID: PMC8421530 DOI: 10.3389/fphar.2021.745061] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 08/09/2021] [Indexed: 12/18/2022] Open
Abstract
Cardio-Cerebrovascular Disease is a collective term for cardiovascular disease and cerebrovascular disease, being a serious threat to human health. A growing number of studies have proved that the content of inflammatory factors or mediators determines the stability of vascular plaque and the incidence of cardio-cerebrovascular event, and involves in the process of Cardio-Cerebrovascular Diseases. Interleukin-6 is a widely used cytokine that causes inflammation and oxidative stress, which would further result in cardiac and cerebral injury. The increased expression of interleukin-6 is closely related to atherosclerosis, myocardial infarction, heart failure and ischemic stroke. It is a key risk factor for these diseases by triggering inflammatory reaction and inducing other molecules release. Therefore, interleukin-6 may become a potential target for Cardio-Cerebrovascular Diseases in the future. This paper is aimed to discuss the expression changes and pathological mechanisms of interleukin-6 in Cardio-Cerebrovascular Diseases, and to provide a novel strategy for the prevention and treatment of Cardio-Cerebrovascular Diseases.
Collapse
Affiliation(s)
- Jian-Hui Su
- Institute of Clinical Research, Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang, China
- Hengyang Key Laboratory of Neurodegeneration and Cognitive Impairment, Department of Physiology, Institute of Neuroscience Research, Hengyang Medical College, University of South China, Hengyang, China
| | - Meng-Yi Luo
- Institute of Clinical Research, Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang, China
- Hengyang Key Laboratory of Neurodegeneration and Cognitive Impairment, Department of Physiology, Institute of Neuroscience Research, Hengyang Medical College, University of South China, Hengyang, China
| | - Na- Liang
- Department of Anesthesiology, Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang, China
| | - Shao-Xin Gong
- Department of Pathology, First Affiliated Hospital, Hengyang Medical College, University of South China, Hengyang, China
| | - Wei Chen
- Institute of Clinical Research, Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang, China
- Hengyang Key Laboratory of Neurodegeneration and Cognitive Impairment, Department of Physiology, Institute of Neuroscience Research, Hengyang Medical College, University of South China, Hengyang, China
| | - Wen-Qian Huang
- Institute of Clinical Research, Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang, China
- Hengyang Key Laboratory of Neurodegeneration and Cognitive Impairment, Department of Physiology, Institute of Neuroscience Research, Hengyang Medical College, University of South China, Hengyang, China
| | - Ying Tian
- Institute of Clinical Research, Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang, China
| | - Ai-Ping Wang
- Institute of Clinical Research, Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang, China
- Hengyang Key Laboratory of Neurodegeneration and Cognitive Impairment, Department of Physiology, Institute of Neuroscience Research, Hengyang Medical College, University of South China, Hengyang, China
| |
Collapse
|
39
|
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: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 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
|
40
|
Analytical methods of antibody surface coverage and orientation on bio-functionalized magnetic beads: application to immunocapture of TNF-α. Anal Bioanal Chem 2021; 413:6425-6434. [PMID: 34401927 PMCID: PMC8367650 DOI: 10.1007/s00216-021-03608-w] [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: 05/04/2021] [Revised: 08/03/2021] [Accepted: 08/06/2021] [Indexed: 11/30/2022]
Abstract
The use of magnetic beads bio-functionalized by antibodies (Ab) is constantly increasing with a wide range of biomedical applications. However, despite an urgent need for current methods to monitor Ab’s grafting process and orientation, existing methods are still either cumbersome and/or limited. In this work, we propose a new simple and rapid analytical approach to evaluate antibody orientation and density on magnetic beads. This approach relies on the cleavage by IdeS, a highly specific protease for human immunoglobulin G (hIgG), of immobilized antibodies. The F(ab)2 and Fc fragments could be then accurately quantified by size exclusion chromatography (SEC)-coupled to fluorescent detection (FLD), and the ratio of these fragments was used to give insight on the IgG orientation at the bead surface. Four different commercially available magnetic beads, bearing carboxyl groups, tosyl groups, streptavidin, or protein G on their surface have been used in this study. Results obtained showed that this approach ensures reliable information on hIgG orientation and bead surface coverage. Protein G magnetic beads demonstrated an optimal orientation of antibodies for antigen capture (75% of accessible F(ab)2 fragment) compared to tosylactivated, carboxylated, and streptavidin ones. Capture efficiency of the different functionalized beads towards human TNF-α immunocapture, a biomarker of inflammation, has been also compared. Protein G beads provided a more efficient capture compared to other beads. In the future, this approach could be applied to any type of surface and beads to assess hIgG coverage and orientation after any type of immobilization.
Collapse
|
41
|
Balogh V, MacAskill MG, Hadoke PWF, Gray GA, Tavares AAS. Positron Emission Tomography Techniques to Measure Active Inflammation, Fibrosis and Angiogenesis: Potential for Non-invasive Imaging of Hypertensive Heart Failure. Front Cardiovasc Med 2021; 8:719031. [PMID: 34485416 PMCID: PMC8416043 DOI: 10.3389/fcvm.2021.719031] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 07/22/2021] [Indexed: 12/11/2022] Open
Abstract
Heart failure, which is responsible for a high number of deaths worldwide, can develop due to chronic hypertension. Heart failure can involve and progress through several different pathways, including: fibrosis, inflammation, and angiogenesis. Early and specific detection of changes in the myocardium during the transition to heart failure can be made via the use of molecular imaging techniques, including positron emission tomography (PET). Traditional cardiovascular PET techniques, such as myocardial perfusion imaging and sympathetic innervation imaging, have been established at the clinical level but are often lacking in pathway and target specificity that is important for assessment of heart failure. Therefore, there is a need to identify new PET imaging markers of inflammation, fibrosis and angiogenesis that could aid diagnosis, staging and treatment of hypertensive heart failure. This review will provide an overview of key mechanisms underlying hypertensive heart failure and will present the latest developments in PET probes for detection of cardiovascular inflammation, fibrosis and angiogenesis. Currently, selective PET probes for detection of angiogenesis remain elusive but promising PET probes for specific targeting of inflammation and fibrosis are rapidly progressing into clinical use.
Collapse
Affiliation(s)
- Viktoria Balogh
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
- Edinburgh Imaging, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Mark G. MacAskill
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
- Edinburgh Imaging, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Patrick W. F. Hadoke
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Gillian A. Gray
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Adriana A. S. Tavares
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
- Edinburgh Imaging, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
42
|
Maeda D, Sakane K, Kanzaki Y, Horai R, Akamatsu K, Tsuda K, Ito T, Sohmiya K, Hoshiga M. Splenic Volume Index Determined Using Computed Tomography upon Admission Is Associated with Readmission for Heart Failure Among Patients with Acute Decompensated Heart Failure. Int Heart J 2021; 62:584-591. [PMID: 33994504 DOI: 10.1536/ihj.20-564] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The spleen is associated with inflammation, and the size of the spleen is affected by hemodynamic congestion and sympathetic stimulation. However, the association between splenic size and prognosis in patients with heart failure remains unknown. Between January 2015 and March 2017, we analyzed 125 patients with acute decompensated heart failure who were assessed by computed tomography (CT) on the day of admission. The spleen was measured by 3-dimensional CT and then the patients were assigned to groups according to their median splenic volume indexes (SpVi; splenic volume/body surface area). We then compared their baseline characteristics and rates of readmission for heart failure after one year. The median SpVi was 63.7 (interquartile range: 44.7-95.3) cm3/m2. Age did not significantly differ between the groups. Patients with a high SpVi had more significantly enlarged left atria and left ventricles. Multiple regression analysis identified significant positive correlations between SpVi and posterior wall thickness as well as left ventricular mass index. Kaplan-Meier analysis revealed lower event-free rates in the patients with a high, than a low SpVi (P = 0.041, log-rank test). After adjustment for potential cofounding factors, SpVi was independently associated with readmission for heart failure (Hazard ratio, 2.25; 95% confidence interval, 1.01-5.02; P = 0.047). In conclusion, increased splenic volume is independently associated with readmission for heart failure among patients with acute decompensated heart failure.
Collapse
Affiliation(s)
- Daichi Maeda
- Department of Cardiology, Osaka Medical and Pharmaceutical University
| | - Kazushi Sakane
- Department of Cardiology, Osaka Medical and Pharmaceutical University
| | - Yumiko Kanzaki
- Department of Cardiology, Osaka Medical and Pharmaceutical University
| | - Ryoto Horai
- Department of Cardiology, Osaka Medical and Pharmaceutical University
| | - Kanako Akamatsu
- Department of Cardiology, Osaka Medical and Pharmaceutical University
| | - Kosuke Tsuda
- Department of Cardiology, Osaka Medical and Pharmaceutical University
| | - Takahide Ito
- Department of Cardiology, Osaka Medical and Pharmaceutical University
| | - Koichi Sohmiya
- Department of Cardiology, Osaka Medical and Pharmaceutical University
| | - Masaaki Hoshiga
- Department of Cardiology, Osaka Medical and Pharmaceutical University
| |
Collapse
|
43
|
Wu X, Luo Q, Su Z, Li Y, Wang H, Yuan S, Yan F. Prognostic Value of Preoperative Absolute Lymphocyte Count in Children With Tetralogy of Fallot. J Am Heart Assoc 2021; 10:e019098. [PMID: 33998242 PMCID: PMC8483512 DOI: 10.1161/jaha.120.019098] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Background Tetralogy of Fallot (TOF) is the most common cyanotic congenital heart disease. Absolute lymphocyte count (ALC) is a low‐cost and easy‐to‐obtain inflammatory indicator; however, its association with the prognosis of patients with TOF remains unknown. This study aimed to determine the prognostic value of preoperative ALC in children with TOF. Methods and Results This retrospective study included 707 patients aged <6 years who underwent corrective operations for TOF between January 2016 and December 2018 in Fuwai Hospital, China. The end points were mortality, extracorporeal membrane oxygenation placement, postoperative hospital stay >30 days, and severe postoperative complications; patients were grouped on the basis of prognosis: poor prognosis (n=76) and good prognosis (n=631). Univariable and multivariable logistic regression analyses were performed to identify the independent risk factors for poor prognosis, on which a risk scoring system was based. The receiver operating characteristic curve was used to assess model performance. Using another model without ALC, the effect of the addition of ALC was assessed. Results suggested that ALC was an independent factor with a cutoff point of 4.36×109/L. The addition of ALC improved the area under the curve from 0.771 to 0.781 (P<0.001). To avoid reverse causality and further control for confounding factors, the patients were further divided on the basis of ALC level, and a propensity score matching was performed; 117 paired patients were identified for further analysis. Low ALC levels had an odds ratio of 3.500 (95% CI, 1.413–8.672). Conclusions Low preoperative ALC represents an independent predictor of poor prognosis in children with TOF.
Collapse
Affiliation(s)
- Xie Wu
- Department of Anesthesiology Fuwai HospitalNational Center of Cardiovascular DiseasesChinese Academy of Medical Sciences, and Peking Union Medical College Beijing China
| | - Qipeng Luo
- Department of Anesthesiology Fuwai HospitalNational Center of Cardiovascular DiseasesChinese Academy of Medical Sciences, and Peking Union Medical College Beijing China
| | - Zhanhao Su
- Center for Pediatric Cardiac Surgery Fuwai HospitalNational Center of Cardiovascular DiseasesChinese Academy of Medical Sciences, and Peking Union Medical College Beijing China
| | - Yinan Li
- Department of Anesthesiology Fuwai HospitalNational Center of Cardiovascular DiseasesChinese Academy of Medical Sciences, and Peking Union Medical College Beijing China
| | - Hongbai Wang
- Department of Anesthesiology Fuwai HospitalNational Center of Cardiovascular DiseasesChinese Academy of Medical Sciences, and Peking Union Medical College Beijing China
| | - Su Yuan
- Department of Anesthesiology Fuwai HospitalNational Center of Cardiovascular DiseasesChinese Academy of Medical Sciences, and Peking Union Medical College Beijing China
| | - Fuxia Yan
- Department of Anesthesiology Fuwai HospitalNational Center of Cardiovascular DiseasesChinese Academy of Medical Sciences, and Peking Union Medical College Beijing China
| |
Collapse
|
44
|
Fragoulis GE, Soulaidopoulos S, Sfikakis PP, Dimitroulas T, D Kitas G. Effect of Biologics on Cardiovascular Inflammation: Mechanistic Insights and Risk Reduction. J Inflamm Res 2021; 14:1915-1931. [PMID: 34017189 PMCID: PMC8131071 DOI: 10.2147/jir.s282691] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 04/14/2021] [Indexed: 12/17/2022] Open
Abstract
It is increasingly recognized that atherosclerosis and consequently cardiovascular disease (CVD) are closely linked with inflammatory processes. The latter is in the center of the pathogenic mechanism underlying autoimmune rheumatic diseases (ARD). It follows then, that optimal control of inflammation in ARDs may lead to a decrease of the accompanied CVD risk. Major trials (eg, CANTOS, CIRT), aimed at examining the possible benefits of immunomodulatory treatments in CVD, demonstrated conflicting results. On the other hand, substantial evidence is accumulating about the possible beneficial effects of biologic disease modifying antirheumatic drugs (bDMARDs) in patients with ARDs, particularly those with rheumatoid arthritis (RA). It seems that bDMARDs (some more than others) alter the lipid profile in RA patients but do not adversely affect, in most cases, the TC/HDL ratio. Favorable effects are noted for arterial stiffness and endothelial function. This is reflected in the lower risk for CVD events, seen in observational studies of RA patients treated with bDMARDs. It should be stressed that more data exist for the TNF-inhibitors than for other bDMARDs, such as tocilizumab, abatacept and rituximab. As regards the spondyloarthropathies (SpA), data are less robust. For TNF-inhibitors, effects appear to be on par with those seen in RA but no conclusions can be drawn for newer biologic drugs used in SpA (eg, IL-17 blockers). Finally, there is accumulating evidence for a beneficial effect of immunosuppressive treatment in cardiac inflammation and function in several ARDs. Introduction of newer therapeutic options in clinical practice seem to have a positive impact on CVD in the setting of ARD. This is probably due to better control of inflammation, but direct improvement in vascular pathology is also a valid hypothesis. Most data are derived from observational studies and, therefore, randomized controlled trials are needed to assess the possible favorable effect of bDMARDs on CVD outcomes.
Collapse
Affiliation(s)
- George E Fragoulis
- Rheumatology Unit, Joint Rheumatology Program, Medical School, First Department of Propaedeutic Internal Medicine, National and Kapodistrian University of Athens, "Laiko" General Hospital, Athens, 115 27, Greece
| | - Stergios Soulaidopoulos
- First Department of Cardiology, National and Kapodistrian University of Athens, Hippokration General Hospital, Athens, 115 27, Greece
| | - Petros P Sfikakis
- Rheumatology Unit, Joint Rheumatology Program, Medical School, First Department of Propaedeutic Internal Medicine, National and Kapodistrian University of Athens, "Laiko" General Hospital, Athens, 115 27, Greece
| | - Theodoros Dimitroulas
- Fourth Department of Internal Medicine, Hippokration Hospital, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, 546 41, Greece
| | - George D Kitas
- Department of Rheumatology, Russells Hall Hospital, Dudley Group NHS FT, Dudley, DY1 2HQ, UK.,Arthritis Research UK Epidemiology Unit, University of Manchester, Manchester, M13 9PT, UK
| |
Collapse
|
45
|
Nemec Svete A, Verk B, Čebulj-Kadunc N, Salobir J, Rezar V, Domanjko Petrič A. Inflammation and its association with oxidative stress in dogs with heart failure. BMC Vet Res 2021; 17:176. [PMID: 33902566 PMCID: PMC8077822 DOI: 10.1186/s12917-021-02878-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Accepted: 04/13/2021] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Inflammation and oxidative stress can contribute to the development and progression of heart failure. This study aimed to investigate the association between inflammatory and oxidative stress markers in dogs with congestive heart failure (CHF). Associations between the disease severity marker N-terminal pro-B-type natriuretic peptide (NT-proBNP) and markers of inflammation and oxidative stress were also determined. RESULTS Thirty-seven dogs with cardiovascular diseases (dilated cardiomyopathy, DCM (16 dogs), myxomatous mitral valve disease, MMVD (21 dogs)) and ten healthy dogs were included in this prospective study. The patients were further divided into groups with (26) and without CHF (11). We found a significantly higher serum concentration of C-reactive protein (P = 0.012), white blood cell (P = 0.001), neutrophil (P = 0.001) and monocyte counts (P = 0.001) in patients with CHF compared to control dogs. The concentration of tumor necrosis factor-alpha (TNF-α) was significantly higher in patients with CHF compared to patients without CHF (P = 0.030). No significant difference was found in most of the measured parameters between MMVD and DCM patients, except for glutathione peroxidase (GPX) and NT-proBNP. In patients with CHF, TNF-α correlated positively with malondialdehyde (P = 0.014, r = 0.474) and negatively with GPX (P = 0.026, r = - 0.453), and interleukin-6 correlated negatively with GPX (P = 0.046, r = - 0.412). NT-proBNP correlated positively with malondialdehyde (P = 0.011, r = 0.493). In patients without CHF none of the inflammatory and oxidative stress markers correlated significantly. Furthermore, in the group of all cardiac patients, GPX activity significantly negatively correlated with NT-proBNP (P = 0.050, r = - 0.339) and several markers of inflammation, including TNF-α (P = 0.010, r = - 0.436), interleukin-6 (P = 0.026, r = - 0.382), white blood cell (P = 0.032, r = - 0.369), neutrophil (P = 0.027, r = - 0.379) and monocyte counts (P = 0.024, r = - 0.386). CONCLUSION Inflammatory and oxidative stress markers are linked in canine CHF patients, but not in patients without CHF. These results suggest complex cross communication between the two biological pathways in advanced stages of CHF.
Collapse
Affiliation(s)
- Alenka Nemec Svete
- Small Animal Clinic, Veterinary Faculty, University of Ljubljana, Gerbičeva 60, 1000, Ljubljana, Slovenia
| | - Barbara Verk
- Small Animal Clinic, Veterinary Faculty, University of Ljubljana, Gerbičeva 60, 1000, Ljubljana, Slovenia
| | - Nina Čebulj-Kadunc
- Institute of Preclinical Sciences, Veterinary Faculty, University of Ljubljana, Gerbičeva 60, 1000, Ljubljana, Slovenia
| | - Janez Salobir
- Institute of Nutrition, Biotechnical Faculty, University of Ljubljana, Groblje 3, 1230, Domžale, Slovenia
| | - Vida Rezar
- Institute of Nutrition, Biotechnical Faculty, University of Ljubljana, Groblje 3, 1230, Domžale, Slovenia
| | - Aleksandra Domanjko Petrič
- Small Animal Clinic, Veterinary Faculty, University of Ljubljana, Gerbičeva 60, 1000, Ljubljana, Slovenia.
| |
Collapse
|
46
|
Liu L, Zhao Q, Kong M, Mao L, Yang Y, Xu Y. Myocardin-related transcription factor A (MRTF-A) regulates integrin beta 2 transcription to promote macrophage infiltration and cardiac hypertrophy in mice. Cardiovasc Res 2021; 118:844-858. [PMID: 33752236 DOI: 10.1093/cvr/cvab110] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 03/19/2021] [Indexed: 01/01/2023] Open
Abstract
AIMS Macrophage-mediated inflammatory response represents a key pathophysiological process in a host of cardiovascular diseases including heart failure. Regardless of etiology, heart failure is invariably preceded by cardiac hypertrophy. In the present study we investigated the effect of macrophage-specific deletion of myocardin-related transcription factor A (MRTF-A) on cardiac hypertrophy and the underlying mechanism. METHODS AND RESULTS We report that when subjected to transverse aortic constriction (TAC), macrophage MRTF-A conditional knockout (CKO) mice developed a less severe phenotype of cardiac hypertrophy compared to wild type (WT) littermates and were partially protected from the loss of heart function. In addition, there was less extensive cardiac fibrosis in the CKO mice than WT mice following the TAC procedure. Further analysis revealed that cardiac inflammation, as assessed by levels of pro-inflammatory cytokines and chemokines, was dampened in CKO mice paralleling reduced infiltration of macrophages in the heart. Mechanistically, MRTF-A deficiency attenuated the expression of integrin beta 2 (ITGB2/CD18) in macrophage thereby disrupting adhesion of macrophages to vascular endothelial cells. MRTF-A was recruited by Sp1 to the ITGB2 promoter and cooperated with Sp1 to activate ITGB2 transcription in macrophages. Administration of a CD18 blocking antibody attenuated TAC induced cardiac hypertrophy in mice. Interaction between MRTF-A and the histone demethylase KDM3A likely contributed to IGTB2 transcription and consequently adhesion of macrophages to endothelial cells. CONCLUSIONS Our data suggest that MRTF-A may regulate macrophage trafficking and contribute to the pathogenesis of cardiac hypertrophy by activating ITGB2 transcription.
Collapse
Affiliation(s)
- Li Liu
- Jiangsu Key Laboratory of Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Qianwen Zhao
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Ming Kong
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Lei Mao
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Yuyu Yang
- Jiangsu Key Laboratory of Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China.,Institute of Biomedical Research, Liaocheng University, Liaocheng, China
| | - Yong Xu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China.,Institute of Biomedical Research, Liaocheng University, Liaocheng, China
| |
Collapse
|
47
|
Damkjær M, Håkansson K, Kallemose T, Ulrik CS, Godtfredsen N. Statins in High-Risk Chronic Obstructive Pulmonary Disease Outpatients: No Impact on Time to First Exacerbation and All-Cause Mortality - The STATUETTE Cohort Study. Int J Chron Obstruct Pulmon Dis 2021; 16:579-589. [PMID: 33707941 PMCID: PMC7943323 DOI: 10.2147/copd.s296472] [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: 12/09/2020] [Accepted: 02/07/2021] [Indexed: 11/23/2022] Open
Abstract
Background Statins have, due to their anti-inflammatory properties, been suggested to potentially improve chronic obstructive pulmonary disease (COPD) outcomes. We aimed to investigate the effect of statins on time to first exacerbation and all-cause mortality in high-risk COPD outpatients. Methods All outpatients with COPD seen at the Department of Respiratory Medicine, Copenhagen University Hospital Amager and Hvidovre, Denmark in 2016 were identified and followed for 3.5 years in this retrospective, registry-based cohort study of time to first acute exacerbation of COPD (AECOPD) or death. AECOPD was defined as a rescue course of oral corticosteroid and/or hospital admission. The association was estimated using time-varying crude and multivariable Cox proportional hazard regression. Results The cohort comprised 950 COPD outpatients, mean (SD) age 71 (11) years, and FEV1 44% predicted (IQR 33%; 57%). The annual exacerbation rate was 0.88 (1.68) and 211 patients (22%) had a history of hospital admission for AECOPD in the 12 months prior to index date. Three hundred and ninety-three patients (41.4%) were defined as statin users, with 131 (33.3%) having filled the first prescription for statin after index date. Statin use was not associated with reduced risk of AECOPD. When stratifying for moderate and severe exacerbations in a sub-analysis in the same model, statin use did not have an increased HR for exacerbation of either severity (HR = 1.02 (95% CI 0.85to 1.24; p = 0.811) and HR = 1.07 (95% CI 0.89 to 1.29; p = 0.492) respectively). Statin use was not associated with all-cause mortality (HR 1.05 (95% CI, 0.75 to 1.47, p = 0.777)). Conclusion We did not find any association between statin use and risk of AECOPD or all-cause mortality. The result adds to the evidence that an aggressive approach with statin treatment upfront is not beneficial in COPD, unless prescribed according to current guidelines for cardiovascular diseases.
Collapse
Affiliation(s)
- Mathias Damkjær
- Department of Respiratory Medicine, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
| | - Kjell Håkansson
- Department of Respiratory Medicine, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
| | - Thomas Kallemose
- Clinical Research Centre, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
| | - Charlotte Suppli Ulrik
- Department of Respiratory Medicine, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark.,Institute of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Nina Godtfredsen
- Department of Respiratory Medicine, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark.,Institute of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
48
|
Laborie E, Bayle F, Bouville D, Smadja C, Dufour-Gergam E, Ammar M. Surface Biochemical Modification of Poly(dimethylsiloxane) for Specific Immune Cytokine Response. ACS APPLIED BIO MATERIALS 2021; 4:1307-1318. [DOI: 10.1021/acsabm.0c01188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Etienne Laborie
- Center for Nanosciences and Nanotechnologies, CNRS, Université Paris-Sud, Université Paris-Saclay, 10 Boulevard Thomas Gobert, 91120 Palaiseau, France
- Institut Galien Paris Sud, UMR 8612, Protein and Nanotechnology in Analytical Science (PNAS), CNRS, Université Paris-Sud, Université Paris-Saclay, 5 rue Jean Baptiste Clément, 92290 Châtenay-Malabry, France
| | - Fabien Bayle
- Center for Nanosciences and Nanotechnologies, CNRS, Université Paris-Sud, Université Paris-Saclay, 10 Boulevard Thomas Gobert, 91120 Palaiseau, France
| | - David Bouville
- Center for Nanosciences and Nanotechnologies, CNRS, Université Paris-Sud, Université Paris-Saclay, 10 Boulevard Thomas Gobert, 91120 Palaiseau, France
| | - Claire Smadja
- Institut Galien Paris Sud, UMR 8612, Protein and Nanotechnology in Analytical Science (PNAS), CNRS, Université Paris-Sud, Université Paris-Saclay, 5 rue Jean Baptiste Clément, 92290 Châtenay-Malabry, France
| | - Elisabeth Dufour-Gergam
- Center for Nanosciences and Nanotechnologies, CNRS, Université Paris-Sud, Université Paris-Saclay, 10 Boulevard Thomas Gobert, 91120 Palaiseau, France
| | - Mehdi Ammar
- Center for Nanosciences and Nanotechnologies, CNRS, Université Paris-Sud, Université Paris-Saclay, 10 Boulevard Thomas Gobert, 91120 Palaiseau, France
| |
Collapse
|
49
|
The interplay between gastrointestinal and cardiovascular diseases: a narrative review focusing on the clinical perspective. Eur J Gastroenterol Hepatol 2021; 32:132-139. [PMID: 32516176 DOI: 10.1097/meg.0000000000001779] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Both cardiovascular and gastrointestinal disorders represent considerable health burden on community clinics and hospitals with overwhelming economic cost. An overlap in the occurrence of these disorders is encountered in daily practice. Both affect each other in bidirectional manner through several mechanisms including altered hemodynamics, systemic inflammation, bacterial overgrowth and interactions and adverse effects of medications. In addition, to the known overlap in the symptoms occurrence of upper gastrointestinal tract diseases and cardiovascular diseases (CVDs). Awareness of this interplay and its clinical manifestations optimizes patient management, and could prevent catastrophic consequences and even save lives. In this review, we highlighted the clinical aspects of this bidirectional association between gastrointestinal and CVDs aiming to shed light on this topic and improve patients' care.
Collapse
|
50
|
Cetin Sanlialp S, Nar G, Senol H. Clinical significance of chitotriosidase in outpatients with advanced heart failure. J Investig Med 2020; 69:736-741. [PMID: 33272933 DOI: 10.1136/jim-2020-001595] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/17/2020] [Indexed: 11/03/2022]
Abstract
The previous studies have shown that plasma chitotriosidase (CHIT) levels increase in many diseases with inflammation. However, there are no reported studies investigating the relationship between CHIT and chronic heart failure (CHF) which is an inflammatory process. Therefore, we aimed to investigate the role of CHIT in diagnosis and severity of CHF in this study. 36 patients (50% male, mean age 63.17±10.18 years) with left ventricular ejection fraction <40% and 27 controls (44% male, mean age 61.33±8.73 years) were included in this study. Patients with CHF were divided into two groups as ischemic heart failure (IHF) and non-ischemic heart failure (NIHF) according to the underlying etiology. Plasma CHIT and N-terminal pro brain natriuretic peptide (NT-proBNP) levels were measured by ELISA method. Plasma CHIT and NT-proBNP levels were higher in patients with CHF than in controls (CHIT 931.25±461.39 ng/mL, 232.79±61.28 ng/mL, p<0.001; NT-proBNP, 595.31±428.11 pg/mL vs 78.13±30.47 pg/L; p<0.001). Also, the levels of these parameters increased in IHF compared with NIHF (CHIT, 1139.28±495.22 ng/mL, 671.22±237.21 ng/mL, p=0.002; NT-proBNP, 792.87±461.26 pg/mL vs 348.36±202.61 pg/mL, p=0.001) and there was a strong correlation between NT-proBNP and CHIT (r=0.969, p<0.001). According to this study findings, plasma CHIT level increases in CHF and its increased levels are correlated with NT-proBNP which is used diagnosis and prognosis of HF.
Collapse
Affiliation(s)
| | - Gokay Nar
- The Department of Cardiology, Pamukkale University Medical Faculty, Denizli, Turkey
| | - Hande Senol
- The Department of Biostatistics, Pamukkale University Medical Faculty, Denizli, Turkey
| |
Collapse
|