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
|
Namazi M, Eftekhar SP, Mosaed R, Shiralizadeh Dini S, Hazrati E. Pulmonary Hypertension and Right Ventricle: A Pathophysiological Insight. CLINICAL MEDICINE INSIGHTS-CARDIOLOGY 2024; 18:11795468241274744. [PMID: 39257563 PMCID: PMC11384539 DOI: 10.1177/11795468241274744] [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] [Received: 05/18/2024] [Accepted: 07/21/2024] [Indexed: 09/12/2024]
Abstract
Background Pulmonary hypertension (PH) is a pulmonary vascular disease characterized by elevated pulmonary vascular pressure. Long-term PH, irrespective of its etiology, leads to increased right ventricular (RV) pressure, RV hypertrophy, and ultimately, RV failure. Main body Research indicates that RV failure secondary to hypertrophy remains the primary cause of mortality in pulmonary arterial hypertension (PAH). However, the impact of PH on RV structure and function under increased overload remains incompletely understood. Several mechanisms have been proposed, including extracellular remodeling, RV hypertrophy, metabolic disturbances, inflammation, apoptosis, autophagy, endothelial-to-mesenchymal transition, neurohormonal dysregulation, capillary rarefaction, and ischemia. Conclusions Studies have demonstrated the significant role of oxidative stress in the development of RV failure. Understanding the interplay among these mechanisms is crucial for the prevention and management of RV failure in patients with PH.
Collapse
Affiliation(s)
- Mehrshad Namazi
- Trauma and Surgery Research Center, AJA University of Medical Sciences, Tehran, Iran
- Clinical Biomechanics and Ergonomics Research Center, AJA University of Medical Sciences, Tehran, Iran
| | - Seyed Parsa Eftekhar
- Trauma and Surgery Research Center, AJA University of Medical Sciences, Tehran, Iran
| | - Reza Mosaed
- Trauma and Surgery Research Center, AJA University of Medical Sciences, Tehran, Iran
| | | | - Ebrahim Hazrati
- Trauma and Surgery Research Center, AJA University of Medical Sciences, Tehran, Iran
| |
Collapse
|
3
|
Belcher DJ, Kim N, Navarro‐Llinas B, Möller M, López‐Soriano FJ, Busquets S, Nader GA. Anabolic deficits and divergent unfolded protein response underlie skeletal and cardiac muscle growth impairments in the Yoshida hepatoma tumor model of cancer cachexia. Physiol Rep 2024; 12:e70044. [PMID: 39294861 PMCID: PMC11410559 DOI: 10.14814/phy2.70044] [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: 07/24/2024] [Revised: 08/30/2024] [Accepted: 08/30/2024] [Indexed: 09/21/2024] Open
Abstract
Cancer cachexia manifests as whole body wasting, however, the precise mechanisms governing the alterations in skeletal muscle and cardiac anabolism have yet to be fully elucidated. In this study, we explored changes in anabolic processes in both skeletal and cardiac muscles in the Yoshida AH-130 ascites hepatoma model of cancer cachexia. AH-130 tumor-bearing rats experienced significant losses in body weight, skeletal muscle, and heart mass. Skeletal and cardiac muscle loss was associated with decreased ribosomal (r)RNA, and hypophosphorylation of the eukaryotic factor 4E binding protein 1. Endoplasmic reticulum stress was evident by higher activating transcription factor mRNA in skeletal muscle and growth arrest and DNA damage-inducible protein (GADD)34 mRNA in both skeletal and cardiac muscles. Tumors provoked an increase in tissue expression of interferon-γ in the heart, while an increase in interleukin-1β mRNA was apparent in both skeletal and cardiac muscles. We conclude that compromised skeletal muscle and heart mass in the Yoshida AH-130 ascites hepatoma model involves a marked reduction translational capacity and efficiency. Furthermore, our observations suggest that endoplasmic reticulum stress and tissue production of pro-inflammatory factors may play a role in the development of skeletal and cardiac muscle wasting.
Collapse
Affiliation(s)
- Daniel J. Belcher
- Huck Institutes of the Life SciencesThe Pennsylvania State UniversityUniversity ParkPennsylvaniaUSA
| | - Nina Kim
- Department of KinesiologyThe Pennsylvania State UniversityUniversity ParkPennsylvaniaUSA
| | - Blanca Navarro‐Llinas
- Department of Biochemistry and Molecular MedicineUniversity of BarcelonaBarcelonaSpain
| | - Maria Möller
- Department of Biochemistry and Molecular MedicineUniversity of BarcelonaBarcelonaSpain
| | - Francisco J. López‐Soriano
- Department of Biochemistry and Molecular MedicineUniversity of BarcelonaBarcelonaSpain
- Institut de Biomedicina de la Universitat de BarcelonaBarcelonaSpain
| | - Silvia Busquets
- Department of Biochemistry and Molecular MedicineUniversity of BarcelonaBarcelonaSpain
- Institut de Biomedicina de la Universitat de BarcelonaBarcelonaSpain
| | - Gustavo A. Nader
- Huck Institutes of the Life SciencesThe Pennsylvania State UniversityUniversity ParkPennsylvaniaUSA
- Department of KinesiologyThe Pennsylvania State UniversityUniversity ParkPennsylvaniaUSA
- Penn State Cancer InstituteThe Pennsylvania State UniversityUniversity ParkPennsylvaniaUSA
| |
Collapse
|
4
|
Lian Z, Yu SR, Cui YX, Li SF, Su L, Song JX, Lee CY, Chen QX, Chen H. Rosuvastatin Enhances Lymphangiogenesis after Myocardial Infarction by Regulating the miRNAs/Vascular Endothelial Growth Factor Receptor 3 (miRNAs/VEGFR3) Pathway. ACS Pharmacol Transl Sci 2024; 7:335-347. [PMID: 38357274 PMCID: PMC10863446 DOI: 10.1021/acsptsci.3c00151] [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: 07/17/2023] [Revised: 12/24/2023] [Accepted: 01/15/2024] [Indexed: 02/16/2024]
Abstract
BACKGROUND Several clinical studies have suggested that the early administration of statins could reduce the risk of in-hospital mortality in acute myocardial infarction (AMI) patients. Recently, some studies have identified that stimulating lymphangiogenesis after AMI could improve cardiac function by reducing myocardial edema and inflammation. This study aimed to identify the effect of rosuvastatin on postinfarct lymphangiogenesis and to identify the underlying mechanism of this effect. METHOD Myocardial infarction (MI) was induced by ligation of the left anterior descending coronary artery in mice orally administered rosuvastatin for 7 days. The changes in cardiac function, pathology, and lymphangiogenesis following MI were measured by echocardiography and immunostaining. EdU, Matrigel tube formation, and scratch wound assays were used to evaluate the effect of rosuvastatin on the proliferation, tube formation, and migration of the lymphatic endothelial cell line SVEC4-10. The expression of miR-107-3p, miR-491-5p, and VEGFR3 was measured by polymerase chain reaction (PCR) and Western blotting. A gain-of-function study was performed using miR-107-3p and miR-491-5p mimics. RESULTS The rosuvastatin-treated mice had a significantly improved ejection fraction and increased lymphatic plexus density 7 days after MI. Rosuvastatin also reduced myocardial edema and inflammatory response after MI. We used a VEGFR3 inhibitor to partially reverse these effects. Rosuvastatin promoted the proliferation, migration, and tube formation of SVEC4-10 cells. PCR and Western blot analyses revealed that rosuvastatin intervention downregulated miR-107-3p and miR-491-5p and promoted VEGFR3 expression. The gain-of-function study showed that miR-107-3p and miR-491-5p could inhibit the proliferation, migration, and tube formation of SVEC4-10 cells. CONCLUSION Rosuvastatin could improve heart function by promoting lymphangiogenesis after MI by regulating the miRNAs/VEGFR3 pathway.
Collapse
Affiliation(s)
- Zheng Lian
- Cardiovascular
Center, Beijing Tongren Hospital, Capital
Medical University, Xihuan South Road No. 2, Economic-Technological
Development Area, Beijing 100176, China
- Department
of Cardiology, Peking University People’s
Hospital, Xizhimen South Road No. 11, Xicheng District, Beijing 100044, China
- Beijing
Key Laboratory of Early Prediction and Intervention of Acute Myocardial
Infarction, Peking University People’s
Hospital, Xizhimen South
Road No. 11, Xicheng District, Beijing 100044, China
- Center
for Cardiovascular Translational Research, Peking University People’s Hospital, Xizhimen South Road No. 11, Xicheng
District, Beijing 100044, China
| | - Shi-Ran Yu
- Department
of Cardiology, Peking University People’s
Hospital, Xizhimen South Road No. 11, Xicheng District, Beijing 100044, China
- Beijing
Key Laboratory of Early Prediction and Intervention of Acute Myocardial
Infarction, Peking University People’s
Hospital, Xizhimen South
Road No. 11, Xicheng District, Beijing 100044, China
- Center
for Cardiovascular Translational Research, Peking University People’s Hospital, Xizhimen South Road No. 11, Xicheng
District, Beijing 100044, China
| | - Yu-Xia Cui
- Department
of Cardiology, Peking University People’s
Hospital, Xizhimen South Road No. 11, Xicheng District, Beijing 100044, China
- Beijing
Key Laboratory of Early Prediction and Intervention of Acute Myocardial
Infarction, Peking University People’s
Hospital, Xizhimen South
Road No. 11, Xicheng District, Beijing 100044, China
- Center
for Cardiovascular Translational Research, Peking University People’s Hospital, Xizhimen South Road No. 11, Xicheng
District, Beijing 100044, China
| | - Su-Fang Li
- Department
of Cardiology, Peking University People’s
Hospital, Xizhimen South Road No. 11, Xicheng District, Beijing 100044, China
- Beijing
Key Laboratory of Early Prediction and Intervention of Acute Myocardial
Infarction, Peking University People’s
Hospital, Xizhimen South
Road No. 11, Xicheng District, Beijing 100044, China
- Center
for Cardiovascular Translational Research, Peking University People’s Hospital, Xizhimen South Road No. 11, Xicheng
District, Beijing 100044, China
| | - Li−Na Su
- Department
of Cardiology, Peking University People’s
Hospital, Xizhimen South Road No. 11, Xicheng District, Beijing 100044, China
- Beijing
Key Laboratory of Early Prediction and Intervention of Acute Myocardial
Infarction, Peking University People’s
Hospital, Xizhimen South
Road No. 11, Xicheng District, Beijing 100044, China
- Center
for Cardiovascular Translational Research, Peking University People’s Hospital, Xizhimen South Road No. 11, Xicheng
District, Beijing 100044, China
| | - Jun-Xian Song
- Department
of Cardiology, Peking University People’s
Hospital, Xizhimen South Road No. 11, Xicheng District, Beijing 100044, China
- Beijing
Key Laboratory of Early Prediction and Intervention of Acute Myocardial
Infarction, Peking University People’s
Hospital, Xizhimen South
Road No. 11, Xicheng District, Beijing 100044, China
- Center
for Cardiovascular Translational Research, Peking University People’s Hospital, Xizhimen South Road No. 11, Xicheng
District, Beijing 100044, China
| | - Chong-Yoo Lee
- Department
of Cardiology, Peking University People’s
Hospital, Xizhimen South Road No. 11, Xicheng District, Beijing 100044, China
- Beijing
Key Laboratory of Early Prediction and Intervention of Acute Myocardial
Infarction, Peking University People’s
Hospital, Xizhimen South
Road No. 11, Xicheng District, Beijing 100044, China
- Center
for Cardiovascular Translational Research, Peking University People’s Hospital, Xizhimen South Road No. 11, Xicheng
District, Beijing 100044, China
| | - Qi-Xin Chen
- Department
of Cardiology, Peking University People’s
Hospital, Xizhimen South Road No. 11, Xicheng District, Beijing 100044, China
- Beijing
Key Laboratory of Early Prediction and Intervention of Acute Myocardial
Infarction, Peking University People’s
Hospital, Xizhimen South
Road No. 11, Xicheng District, Beijing 100044, China
- Center
for Cardiovascular Translational Research, Peking University People’s Hospital, Xizhimen South Road No. 11, Xicheng
District, Beijing 100044, China
| | - Hong Chen
- Department
of Cardiology, Peking University People’s
Hospital, Xizhimen South Road No. 11, Xicheng District, Beijing 100044, China
- Beijing
Key Laboratory of Early Prediction and Intervention of Acute Myocardial
Infarction, Peking University People’s
Hospital, Xizhimen South
Road No. 11, Xicheng District, Beijing 100044, China
- Center
for Cardiovascular Translational Research, Peking University People’s Hospital, Xizhimen South Road No. 11, Xicheng
District, Beijing 100044, China
| |
Collapse
|
5
|
Zhang H, Dhalla NS. The Role of Pro-Inflammatory Cytokines in the Pathogenesis of Cardiovascular Disease. Int J Mol Sci 2024; 25:1082. [PMID: 38256155 PMCID: PMC10817020 DOI: 10.3390/ijms25021082] [Citation(s) in RCA: 32] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 01/11/2024] [Accepted: 01/12/2024] [Indexed: 01/24/2024] Open
Abstract
With cardiovascular disease (CVD) being a primary source of global morbidity and mortality, it is crucial that we understand the molecular pathophysiological mechanisms at play. Recently, numerous pro-inflammatory cytokines have been linked to several different CVDs, which are now often considered an adversely pro-inflammatory state. These cytokines most notably include interleukin-6 (IL-6),tumor necrosis factor (TNF)α, and the interleukin-1 (IL-1) family, amongst others. Not only does inflammation have intricate and complex interactions with pathophysiological processes such as oxidative stress and calcium mishandling, but it also plays a role in the balance between tissue repair and destruction. In this regard, pre-clinical and clinical evidence has clearly demonstrated the involvement and dynamic nature of pro-inflammatory cytokines in many heart conditions; however, the clinical utility of the findings so far remains unclear. Whether these cytokines can serve as markers or risk predictors of disease states or act as potential therapeutic targets, further extensive research is needed to fully understand the complex network of interactions that these molecules encompass in the context of heart disease. This review will highlight the significant advances in our understanding of the contributions of pro-inflammatory cytokines in CVDs, including ischemic heart disease (atherosclerosis, thrombosis, acute myocardial infarction, and ischemia-reperfusion injury), cardiac remodeling (hypertension, cardiac hypertrophy, cardiac fibrosis, cardiac apoptosis, and heart failure), different cardiomyopathies as well as ventricular arrhythmias and atrial fibrillation. In addition, this article is focused on discussing the shortcomings in both pathological and therapeutic aspects of pro-inflammatory cytokines in CVD that still need to be addressed by future studies.
Collapse
Affiliation(s)
- Hannah Zhang
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, College of Medicine, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Naranjan S. Dhalla
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, College of Medicine, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| |
Collapse
|
6
|
Hu Y, Chen X, Mei X, Luo Z, Wu H, Zhang H, Zeng Q, Ren H, Xu D. Identification of diagnostic immune-related gene biomarkers for predicting heart failure after acute myocardial infarction. Open Med (Wars) 2023; 18:20230878. [PMID: 38152337 PMCID: PMC10751901 DOI: 10.1515/med-2023-0878] [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: 09/05/2023] [Revised: 11/02/2023] [Accepted: 11/24/2023] [Indexed: 12/29/2023] Open
Abstract
Post-myocardial infarction heart failure (HF) is a major public health concern. Previous studies have reported the critical role of immune response in HF pathogenesis. However, limited studies have reported predictive immune-associated biomarkers for HF. So we attempted to identify potential immune-related indicators for HF early diagnosis and therapy guidance. This study identified two potential immune-related hub genes (IRHGs), namely CXCR5 and FOS, using bioinformatic approaches. The expression levels of CXCR5 and FOS and their ability to predict long-term HF were analyzed. Functional enrichment analysis revealed that the hub genes were enriched in immune system processes, including the interleukin-17 and nuclear factor-kappa B signaling pathways, which are involved in the pathogenesis of HF. Quantitative real-time polymerase chain reaction revealed that the Fos mRNA levels, but not the Cxcr5 mRNA levels, were downregulated in the mice of the HF group. This study successfully identified two IRHGs that were significantly and differentially expressed in the HF group and could predict long-term HF, providing novel insights for future studies on HF and developing novel therapeutic targets for HF.
Collapse
Affiliation(s)
- Yingchun Hu
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Xiaoyu Chen
- Department of Nephrology, Rheumatism and Immunology, Chongqing Jiulongpo People’s Hospital, Chongqing, 400050, China
| | - Xiyuan Mei
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Zhen Luo
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Hongguang Wu
- Department of Arrhythmic, Cardiovascular Medical Center, Shenzhen Hospital of University of Hong Kong, Shenzhen, 518040, Guangdong, China
| | - Hao Zhang
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Qingchun Zeng
- Department of Cardiology, Nanfang Hospital, Southern Medical University,
Guangzhou, 510515, Guangdong, China
| | - Hao Ren
- Department of Rheumatology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Dingli Xu
- Department of Cardiology, Nanfang Hospital, Southern Medical University,
Guangzhou, 510515, Guangdong, China
| |
Collapse
|
7
|
Li Z, Lin C, Cai X, Hu S, Lv F, Yang W, Zhu X, Ji L. Anti-inflammatory therapies were associated with reduced risk of myocardial infarction in patients with established cardiovascular disease or high cardiovascular risks: A systematic review and meta-analysis of randomized controlled trials. Atherosclerosis 2023; 379:117181. [PMID: 37527612 DOI: 10.1016/j.atherosclerosis.2023.06.972] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 06/27/2023] [Accepted: 06/28/2023] [Indexed: 08/03/2023]
Abstract
BACKGROUND AND AIMS We aimed to evaluate the association between anti-inflammatory therapies and the incidence of cardiovascular events in patients with established cardiovascular disease (CVD) or high cardiovascular risks. METHODS Literature retrieval was conducted in PubMed, Medline, Embase, the Cochrane Central Register of Controlled Trials and Clinicaltrial.gov website from the inception to December 2022. Randomized controlled trials comparing anti-inflammatory therapies with placebo in patients with established CVD or high cardiovascular risks were included. The results of the meta-analysis were computed as the risk ratio (RR) with 95% confidence interval (CI). RESULTS Compared with placebo, anti-inflammatory therapies were associated with decreased incidence of myocardial infarction (MI) (RR = 0.93, 95% CI, 0.88 to 0.98), which was mainly driven by therapies targeting central IL-6 signaling pathway (RR = 0.83, 95% CI, 0.74 to 0.93). IL-1 inhibitors treatment was associated with reduced risks of heart failure (RR = 0.38, 95% CI, 0.18 to 0.80) while lower incidence of stroke was observed in patients with colchicine treatment (RR = 0.47, 95% CI, 0.28 to 0.77). MI events were less frequent in patients over 65 years of age (RR = 0.90, 95% CI, 0.83 to 0.98) or with follow-up duration over 1 year (RR = 0.90, 95% CI, 0.85 to 0.96) when comparing anti-inflammatory therapies with placebo. CONCLUSIONS Anti-inflammatory therapies, especially those targeting the central IL-6 signaling pathway, may serve as promising treating strategies to ameliorate the risk of MI. IL-1 inhibitor and colchicine were associated with decreased risks of heart failure and stroke, respectively. MI risk reduction by anti-inflammatory therapies seemed to be more prominent in older patients with long follow-up duration.
Collapse
Affiliation(s)
- Zonglin Li
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Beijing, China
| | - Chu Lin
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Beijing, China
| | - Xiaoling Cai
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Beijing, China.
| | - Suiyuan Hu
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Beijing, China
| | - Fang Lv
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Beijing, China
| | - Wenjia Yang
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Beijing, China
| | - Xingyun Zhu
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Beijing, China
| | - Linong Ji
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Beijing, China.
| |
Collapse
|
8
|
Francisco J, Guan J, Zhang Y, Nakada Y, Mareedu S, Sung EA, Hu CM, Oka S, Zhai P, Sadoshima J, Del Re DP. Suppression of myeloid YAP antagonizes adverse cardiac remodeling during pressure overload stress. J Mol Cell Cardiol 2023; 181:1-14. [PMID: 37235928 PMCID: PMC10524516 DOI: 10.1016/j.yjmcc.2023.05.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 05/05/2023] [Accepted: 05/16/2023] [Indexed: 05/28/2023]
Abstract
Inflammation is an integral component of cardiovascular disease and is thought to contribute to cardiac dysfunction and heart failure. While ischemia-induced inflammation has been extensively studied in the heart, relatively less is known regarding cardiac inflammation during non-ischemic stress. Recent work has implicated a role for Yes-associated protein (YAP) in modulating inflammation in response to ischemic injury; however, whether YAP influences inflammation in the heart during non-ischemic stress is not described. We hypothesized that YAP mediates a pro-inflammatory response during pressure overload (PO)-induced non-ischemic injury, and that targeted YAP inhibition in the myeloid compartment is cardioprotective. In mice, PO elicited myeloid YAP activation, and myeloid-specific YAP knockout mice (YAPF/F;LysMCre) subjected to PO stress had better systolic function, and attenuated pathological remodeling compared to control mice. Inflammatory indicators were also significantly attenuated, while pro-resolving genes including Vegfa were enhanced, in the myocardium, and in isolated macrophages, of myeloid YAP KO mice after PO. Experiments using bone marrow-derived macrophages (BMDMs) from YAP KO and control mice demonstrated that YAP suppression shifted polarization toward a resolving phenotype. We also observed attenuated NLRP3 inflammasome priming and function in YAP deficient BMDMs, as well as in myeloid YAP KO hearts following PO, indicating disruption of inflammasome induction. Finally, we leveraged nanoparticle-mediated delivery of the YAP inhibitor verteporfin and observed attenuated PO-induced pathological remodeling compared to DMSO nanoparticle control treatment. These data implicate myeloid YAP as an important molecular nodal point that facilitates cardiac inflammation and fibrosis during PO stress and suggest that selective inhibition of YAP may prove a novel therapeutic target in non-ischemic heart disease.
Collapse
Affiliation(s)
- Jamie Francisco
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Jin Guan
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Yu Zhang
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Yasuki Nakada
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Satvik Mareedu
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Eun-Ah Sung
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Che-Ming Hu
- Institute of Biomedical Sciences, Academia Sinica, Taiwan
| | - Shinichi Oka
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Peiyong Zhai
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Junichi Sadoshima
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Dominic P Del Re
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, NJ, USA.
| |
Collapse
|
9
|
d'Aiello A, Bonanni A, Vinci R, Pedicino D, Severino A, De Vita A, Filomia S, Brecciaroli M, Liuzzo G. Meta-Inflammation and New Anti-Diabetic Drugs: A New Chance to Knock Down Residual Cardiovascular Risk. Int J Mol Sci 2023; 24:ijms24108643. [PMID: 37239990 DOI: 10.3390/ijms24108643] [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: 04/03/2023] [Revised: 04/28/2023] [Accepted: 05/08/2023] [Indexed: 05/28/2023] Open
Abstract
Type 2 diabetes mellitus (DM) represents, with its macro and microvascular complications, one of the most critical healthcare issues for the next decades. Remarkably, in the context of regulatory approval trials, sodium-glucose cotransporter 2 inhibitors (SGLT2i) and glucagon-like peptide 1 receptor agonists (GLP-1 RAs) proved a reduced incidence of major adverse cardiovascular events (MACEs), i.e., cardiovascular death and heart failure (HF) hospitalizations. The cardioprotective abilities of these new anti-diabetic drugs seem to run beyond mere glycemic control, and a growing body of evidence disclosed a wide range of pleiotropic effects. The connection between diabetes and meta-inflammation seems to be the key to understanding how to knock down residual cardiovascular risk, especially in this high-risk population. The aim of this review is to explore the link between meta-inflammation and diabetes, the role of newer glucose-lowering medications in this field, and the possible connection with their unexpected cardiovascular benefits.
Collapse
Affiliation(s)
- Alessia d'Aiello
- Department of Cardiovascular Sciences, Fondazione Policlinico A. Gemelli, IRCCS, 00168 Rome, Italy
- Department of Cardiovascular and Pneumological Sciences, Catholic University of Sacred Heart, 00168 Rome, Italy
| | - Alice Bonanni
- Department of Cardiovascular Sciences, Fondazione Policlinico A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Ramona Vinci
- Department of Cardiovascular Sciences, Fondazione Policlinico A. Gemelli, IRCCS, 00168 Rome, Italy
- Department of Cardiovascular and Pneumological Sciences, Catholic University of Sacred Heart, 00168 Rome, Italy
| | - Daniela Pedicino
- Department of Cardiovascular Sciences, Fondazione Policlinico A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Anna Severino
- Department of Cardiovascular Sciences, Fondazione Policlinico A. Gemelli, IRCCS, 00168 Rome, Italy
- Department of Cardiovascular and Pneumological Sciences, Catholic University of Sacred Heart, 00168 Rome, Italy
| | - Antonio De Vita
- Department of Cardiovascular Sciences, Fondazione Policlinico A. Gemelli, IRCCS, 00168 Rome, Italy
- Department of Cardiovascular and Pneumological Sciences, Catholic University of Sacred Heart, 00168 Rome, Italy
| | - Simone Filomia
- Department of Cardiovascular and Pneumological Sciences, Catholic University of Sacred Heart, 00168 Rome, Italy
| | - Mattia Brecciaroli
- Department of Cardiovascular and Pneumological Sciences, Catholic University of Sacred Heart, 00168 Rome, Italy
| | - Giovanna Liuzzo
- Department of Cardiovascular Sciences, Fondazione Policlinico A. Gemelli, IRCCS, 00168 Rome, Italy
- Department of Cardiovascular and Pneumological Sciences, Catholic University of Sacred Heart, 00168 Rome, Italy
| |
Collapse
|
10
|
Screever EM, Yousif LIE, Moslehi JJ, Salem JE, Voors AA, Silljé HHW, de Boer RA, Meijers WC. Circulating immune checkpoints predict heart failure outcomes. ESC Heart Fail 2023. [PMID: 37186066 PMCID: PMC10375122 DOI: 10.1002/ehf2.14304] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 01/16/2023] [Indexed: 05/17/2023] Open
Abstract
AIMS There are limited data examining the role of immune checkpoint (IC) ligands in the pathophysiology of heart failure (HF). Therefore, we explore this in three HF animal models and in three different human cohorts (healthy, stable, and worsening HF). METHODS AND RESULTS Transcriptomic analyses of cardiac tissue of three different HF mouse models revealed differentially expressed IC receptors and their ligands compared with control mice. Based on this observation, serum levels of three well-known IC ligands (i.e. sPD-L1, sPD-L2 and galectin-9) were measured in stable HF patients from the Vitamin D Chronic Heart Failure (VitD-CHF) study (n = 101), as well as healthy individuals from the Prevention of Renal and Vascular End-stage Disease (PREVEND) study (n = 58). sPD-L1, sPD-L2, and galectin-9 were all associated with New York Heart Association classification. In multivariate linear regression analyses, all three IC ligands were associated with galectin-3 (β = 0.230, β = 0.283, and β = 0.304, respectively). sPD-L1 and galectin-9 were also associated with hs-troponin-T (β = 0.386 and β = 0.314). Regarding prognosis, higher serum levels of sPD-L1 and galectin-9 were significantly associated with increased risk for HF hospitalization and all-cause mortality [hazard ratio 1.69 (1.09-2.59) and hazard ratio 1.50 (1.06-2.12)]. Furthermore, the importance of IC ligands was tested in another stage of HF, namely worsening HF patients. In the worsening HF cohort (The BIOlogy Study to Tailored Treatment in Chronic Heart Failure) (n = 2032), sPD-L2 and galectin-9 were associated with New York Heart Association classification and significantly predicted outcome with an increased relative risk of 15% and 20%, after multivariable adjustment, respectively. CONCLUSIONS IC ligands are expressed in cardiac disease models, and serum levels of IC ligands are elevated in HF patients, are associated with disease severity, and significantly predict prognosis. These data indicate a potential role for IC ligands in HF pathogenesis.
Collapse
Affiliation(s)
- Elles M Screever
- Department of Cardiology, University Medical Center Groningen, Groningen, The Netherlands
- Division of Experimental Cardiology, Department of Cardiology, Thorax Center, Erasmus University Medical Center, PO Box 2040, 3000CA, Rotterdam, The Netherlands
| | - Laura I E Yousif
- Division of Experimental Cardiology, Department of Cardiology, Thorax Center, Erasmus University Medical Center, PO Box 2040, 3000CA, Rotterdam, The Netherlands
| | - Javid J Moslehi
- Section of Cardio-Oncology and Immunology, Division of Cardiology and the Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, USA
| | - Joe-Elie Salem
- Department of Pharmacology, Assistance Publique-Hôpitaux de Paris (AP-HP), Sorbonne Université, INSERM, CIC-1901, UNICO-GRECO Cardio-oncology Program, Paris, France
| | - Adriaan A Voors
- Department of Cardiology, University Medical Center Groningen, Groningen, The Netherlands
| | - Herman H W Silljé
- Department of Cardiology, University Medical Center Groningen, Groningen, The Netherlands
| | - Rudolf A de Boer
- Department of Cardiology, University Medical Center Groningen, Groningen, The Netherlands
- Division of Experimental Cardiology, Department of Cardiology, Thorax Center, Erasmus University Medical Center, PO Box 2040, 3000CA, Rotterdam, The Netherlands
| | - Wouter C Meijers
- Department of Cardiology, University Medical Center Groningen, Groningen, The Netherlands
- Division of Experimental Cardiology, Department of Cardiology, Thorax Center, Erasmus University Medical Center, PO Box 2040, 3000CA, Rotterdam, The Netherlands
| |
Collapse
|
11
|
Hung MJ, Yeh CT, Kounis NG, Koniari I, Hu P, Hung MY. Coronary Artery Spasm-Related Heart Failure Syndrome: Literature Review. Int J Mol Sci 2023; 24:ijms24087530. [PMID: 37108691 PMCID: PMC10145866 DOI: 10.3390/ijms24087530] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 04/04/2023] [Accepted: 04/11/2023] [Indexed: 04/29/2023] Open
Abstract
Although heart failure (HF) is a clinical syndrome that becomes worse over time, certain cases can be reversed with appropriate treatments. While coronary artery spasm (CAS) is still underappreciated and may be misdiagnosed, ischemia due to coronary artery disease and CAS is becoming the single most frequent cause of HF worldwide. CAS could lead to syncope, HF, arrhythmias, and myocardial ischemic syndromes such as asymptomatic ischemia, rest and/or effort angina, myocardial infarction, and sudden death. Albeit the clinical significance of asymptomatic CAS has been undervalued, affected individuals compared with those with classic Heberden's angina pectoris are at higher risk of syncope, life-threatening arrhythmias, and sudden death. As a result, a prompt diagnosis implements appropriate treatment strategies, which have significant life-changing consequences to prevent CAS-related complications, such as HF. Although an accurate diagnosis depends mainly on coronary angiography and provocative testing, clinical characteristics may help decision-making. Because the majority of CAS-related HF (CASHF) patients present with less severe phenotypes than overt HF, it underscores the importance of understanding risk factors correlated with CAS to prevent the future burden of HF. This narrative literature review summarises and discusses separately the epidemiology, clinical features, pathophysiology, and management of patients with CASHF.
Collapse
Affiliation(s)
- Ming-Jui Hung
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital Keelung, Chang Gung University College of Medicine, Keelung City 24201, Taiwan
| | - Chi-Tai Yeh
- Department of Medical Research and Education, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan
- Continuing Education Program of Food Biotechnology Applications, College of Science and Engineering, National Taitung University, Taitung 95092, Taiwan
| | - Nicholas G Kounis
- Department of Cardiology, University of Patras Medical School, 26221 Patras, Greece
| | - Ioanna Koniari
- Cardiology Department, Liverpool Heart and Chest Hospital, Liverpool L14 3PE, UK
| | - Patrick Hu
- Department of Internal Medicine, School of Medicine, University of California, Riverside, Riverside, CA 92521, USA
- Department of Cardiology, Riverside Medical Clinic, Riverside, CA 92506, USA
| | - Ming-Yow Hung
- Division of Cardiology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, No.291, Zhongzheng Rd., Zhonghe District, New Taipei City 23561, Taiwan
- Taipei Heart Institute, Taipei Medical University, Taipei City 110301, Taiwan
- Division of Cardiology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, New Taipei City 23561, Taiwan
| |
Collapse
|
12
|
Bekedam FT, Goumans MJ, Bogaard HJ, de Man FS, Llucià-Valldeperas A. Molecular mechanisms and targets of right ventricular fibrosis in pulmonary hypertension. Pharmacol Ther 2023; 244:108389. [PMID: 36940790 DOI: 10.1016/j.pharmthera.2023.108389] [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: 11/29/2022] [Revised: 02/19/2023] [Accepted: 03/16/2023] [Indexed: 03/23/2023]
Abstract
Right ventricular fibrosis is a stress response, predominantly mediated by cardiac fibroblasts. This cell population is sensitive to increased levels of pro-inflammatory cytokines, pro-fibrotic growth factors and mechanical stimulation. Activation of fibroblasts results in the induction of various molecular signaling pathways, most notably the mitogen-activated protein kinase cassettes, leading to increased synthesis and remodeling of the extracellular matrix. While fibrosis confers structural protection in response to damage induced by ischemia or (pressure and volume) overload, it simultaneously contributes to increased myocardial stiffness and right ventricular dysfunction. Here, we review state-of-the-art knowledge of the development of right ventricular fibrosis in response to pressure overload and provide an overview of all published preclinical and clinical studies in which right ventricular fibrosis was targeted to improve cardiac function.
Collapse
Affiliation(s)
- F T Bekedam
- Amsterdam UMC location Vrije Universiteit Amsterdam, PHEniX laboratory, Department of Pulmonary Medicine, De Boelelaan 1117, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, Pulmonary Hypertension and Thrombosis, Amsterdam, the Netherlands
| | - M J Goumans
- Department of Cell and Chemical Biology, Leiden UMC, 2300 RC Leiden, the Netherlands
| | - H J Bogaard
- Amsterdam UMC location Vrije Universiteit Amsterdam, PHEniX laboratory, Department of Pulmonary Medicine, De Boelelaan 1117, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, Pulmonary Hypertension and Thrombosis, Amsterdam, the Netherlands
| | - F S de Man
- Amsterdam UMC location Vrije Universiteit Amsterdam, PHEniX laboratory, Department of Pulmonary Medicine, De Boelelaan 1117, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, Pulmonary Hypertension and Thrombosis, Amsterdam, the Netherlands.
| | - A Llucià-Valldeperas
- Amsterdam UMC location Vrije Universiteit Amsterdam, PHEniX laboratory, Department of Pulmonary Medicine, De Boelelaan 1117, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, Pulmonary Hypertension and Thrombosis, Amsterdam, the Netherlands.
| |
Collapse
|
13
|
Puukila S, Lawrence MD, De Pasquale CG, Bersten AD, Bihari S, McEvoy-May J, Nemec-Bakk A, Dixon DL. Monocyte chemotactic protein (MCP)-1 (CCL2) and its receptor (CCR2) are elevated in chronic heart failure facilitating lung monocyte infiltration and differentiation which may contribute to lung fibrosis. Cytokine 2023; 161:156060. [PMID: 36219898 DOI: 10.1016/j.cyto.2022.156060] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 09/25/2022] [Accepted: 09/29/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Dyspnea, the cardinal manifestation of chronic heart failure (CHF), may reflect both pulmonary oedema and pulmonary remodeling resulting in tissue stiffening. Emerging evidence suggests that predominance of distinct phenotypes of alveolar and recruited macrophages, designated M1 and M2, may regulate the course of inflammatory tissue repair and remodeling in the lung. METHODS In a CHF rat model, we found fibrotic reinforcement of the extracellular matrix with an increase in monocyte chemotactic protein (MCP)-1/CCL2 in bronchoalveolar lavage (BAL), corresponding to a 3-fold increase in recruited macrophages. In this clinical cross sectional study, we aimed to examine potential mediators of leukocyte activation and lung infiltration in parallel BAL and blood from CHF patients compared to non-CHF controls. RESULTS Mini-BAL and peripheral blood samples were obtained from hospitalized CHF, acute decompensated CHF and non-CHF patients. CHF patients and decompensated CHF patients demonstrated increases from non-CHF patients in BAL MCP-1, as well as the M2 macrophage cytokines interleukin-10 and transforming growth factor-β. BAL and plasma MCP-1 were significantly correlated; however, MCP-1 was 20-fold higher in epithelial lining fluid in BAL, indicative of an alveolar chemotactic gradient. An increase in transglutaminase 2 positive M2 macrophages in parallel with a decrease in the MCP-1 receptor, CC chemokine receptor 2 (CCR2), was apparent in BAL cells of CHF patients compared to non-CHF. CONCLUSION These data suggest a pathway of MCP-1 mediated M2 macrophage prevalence in the lungs of CHF patients which may contribute to pulmonary fibrotic remodeling and consequent increased severity of dyspnea.
Collapse
Affiliation(s)
- Stephanie Puukila
- College of Medicine and Public Health, Flinders University, Adelaide, Australia; Northern Ontario School of Medicine, Thunder Bay, Canada
| | - Mark D Lawrence
- College of Medicine and Public Health, Flinders University, Adelaide, Australia
| | - Carmine G De Pasquale
- College of Medicine and Public Health, Flinders University, Adelaide, Australia; Cardiac Services, Flinders Medical Centre, Adelaide, Australia
| | - Andrew D Bersten
- College of Medicine and Public Health, Flinders University, Adelaide, Australia; Intensive and Critical Care Unit, Flinders Medical Centre, Adelaide, Australia
| | - Shailesh Bihari
- College of Medicine and Public Health, Flinders University, Adelaide, Australia; Intensive and Critical Care Unit, Flinders Medical Centre, Adelaide, Australia
| | - James McEvoy-May
- College of Medicine and Public Health, Flinders University, Adelaide, Australia
| | - Ashley Nemec-Bakk
- Department of Biotechnology, Lakehead University, Thunder Bay, Canada
| | - Dani-Louise Dixon
- College of Medicine and Public Health, Flinders University, Adelaide, Australia; Northern Ontario School of Medicine, Thunder Bay, Canada; Intensive and Critical Care Unit, Flinders Medical Centre, Adelaide, Australia.
| |
Collapse
|
14
|
Siddiqui SW, Ashok T, Patni N, Fatima M, Lamis A, Anne KK. Anemia and Heart Failure: A Narrative Review. Cureus 2022; 14:e27167. [PMID: 36017290 PMCID: PMC9393312 DOI: 10.7759/cureus.27167] [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] [Accepted: 07/22/2022] [Indexed: 11/06/2022] Open
Abstract
Anemia in heart failure patients is a relatively common finding and has been linked with an increased risk of hospital admissions, morbidities, and significant mortality making its correction a significant factor in improving the quality of life and clinical outcomes in those suffering from it. This review article has discussed the multifactorial pathophysiology, including iron deficiency, longstanding inflammation, abnormal levels of human erythropoietin (Epo), and the abnormal activation of the renin-angiotensin-aldosterone system (RAAS) being the most significant. The diagnostic guidelines as well as research-based management modalities specifically with iron supplements and erythropoietin stimulating agents have also been discussed, although research done in this area has been limited and shown conflicting results.
Collapse
|
15
|
Hasselbach L, Weidner J, Elsässer A, Theilmeier G. Heart Failure Relapses in Response to Acute Stresses - Role of Immunological and Inflammatory Pathways. Front Cardiovasc Med 2022; 9:809935. [PMID: 35548445 PMCID: PMC9081344 DOI: 10.3389/fcvm.2022.809935] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 03/23/2022] [Indexed: 12/15/2022] Open
Abstract
Cardiovascular diseases continue to be the most imminent health care problems in the western world, accounting for numerous deaths per year. Heart failure (HF), namely the reduction of left ventricular function, is one of the major cardiovascular disease entities. It is chronically progressing with relapsing acute decompensations and an overall grave prognosis that is little different if not worse than most malignant diseases. Interestingly acute metabolically and/or immunologically challenging events like infections or major surgical procedures will cause relapses in the course of preexisting chronic heart failure, decrease the patients wellbeing and worsen myocardial function. HF itself and or its progression has been demonstrated to be driven at least in part by inflammatory pathways that are similarly turned on by infectious or non-infectious stress responses. These thus add to HF progression or relapse. TNF-α plasma levels are associated with disease severity and progression in HF. In addition, several cytokines (e.g., IL-1β, IL-6) are involved in deteriorating left ventricular function. Those observations are based on clinical studies using inhibitors of cytokines or their receptors or they stem from animal studies examining the effect of cytokine mediated inflammation on myocardial remodeling in models of heart failure. This short review summarizes the known underlying immunological processes that are shared by and drive all: chronic heart failure, select infectious diseases, and inflammatory stress responses. In conclusion the text provides a brief summary of the current development in immunomodulatory therapies for HF and their overlap with treatments of other disease entities.
Collapse
Affiliation(s)
- Lisa Hasselbach
- Division of Cardiology and Division of Perioperative Inflammation and Infection, Department Human Medicine, University of Oldenburg, Oldenburg, Germany
| | - Johannes Weidner
- Division of Perioperative Inflammation and Infection, Department Human Medicine, University of Oldenburg, Oldenburg, Germany
| | - Albrecht Elsässer
- Division of Cardiology, Department Human Medicine, University of Oldenburg, Oldenburg, Germany
| | - Gregor Theilmeier
- Division of Perioperative Inflammation and Infection, Department Human Medicine, University of Oldenburg, Oldenburg, Germany
| |
Collapse
|
16
|
Fries RC, Kadotani S, Stack JP, Kruckman L, Wallace G. Prognostic Value of Neutrophil-to-Lymphocyte Ratio in Cats With Hypertrophic Cardiomyopathy. Front Vet Sci 2022; 9:813524. [PMID: 35359679 PMCID: PMC8964083 DOI: 10.3389/fvets.2022.813524] [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: 11/19/2021] [Accepted: 02/09/2022] [Indexed: 11/13/2022] Open
Abstract
ObjectiveTo assess the prognostic value of neutrophil-to-lymphocyte ratio (NLR) for cardiac death in cats with hypertrophic cardiomyopathy.Study DesignProspective observation study.AnimalsNinety-six client-owned cats.MethodsComplete blood count samples were collected from 38 healthy and 58 cats with hypertrophic cardiomyopathy (HCM), and the NLR ratios were analyzed. All cats had echocardiographic measurements performed on the same day as blood collection. Spearman rank correlation was used to assess the relationship between echocardiographic measurements and NLR. Long-term outcome data were obtained, and time to cardiac death and variables associated with cardiac death were analyzed using Kaplan–Meier survival curves and Cox proportional hazards models, respectively.ResultsThe NLR was significantly higher in cats with confirmed congestive heart failure. When evaluating HCM patients, cats in the third NLR tertile had a significantly higher risk of cardiac death with a hazard ratio of 10.26 (95% CI: 1.84–57.14; p = 0.0001) when compared with that of patients in the first tertile. NLR was significantly associated with echocardiographic measures of left atrial size, left auricular function, the presence of left atrial spontaneous echo contrast (SEC), and thrombus formation.Conclusions and Clinical RelevanceIncreased NLR is a negative prognostic indicator in cats with HCM.
Collapse
Affiliation(s)
- Ryan C. Fries
- Department of Veterinary Clinical Medicine, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, United States
- *Correspondence: Ryan C. Fries
| | - Saki Kadotani
- Department of Veterinary Clinical Medicine, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Jonathan P. Stack
- Department of Veterinary Clinical Medicine, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, United States
- VCA Loomis Basin Veterinary Clinic, Loomis, CA, United States
| | - Leah Kruckman
- Department of Veterinary Clinical Medicine, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Gabrielle Wallace
- Department of Veterinary Clinical Medicine, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, United States
- Pacific Northwest Pet ER and Specialty Center, Vancouver, WA, United States
| |
Collapse
|
17
|
Targeting Inflammation in the Diagnosis, Management, and Prevention of Cardiovascular Diseases. Glob Heart 2022; 17:80. [PMID: 36382160 PMCID: PMC9635324 DOI: 10.5334/gh.1156] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 09/01/2022] [Indexed: 01/25/2023] Open
Abstract
Inflammation plays an important role in the development and progression of cardiovascular diseases (CVDs). Hypertension and hyperlipidemia are the key risk factors of CVDs and induce inflammation in the heart and vessels. Unhealthy diet, infection, and smoking coupled with genetic factors lead to the development of CVDs as well as induce inflammation. Risk factors of CVDs such as hypertension and hyperlipidemia along with diabetes activate nuclear factor kappa B, which regulates the transcription of immunoglobulin free light chain (FLC) κ in B cells and the production of multiple inflammatory molecules, leading to inflammation. FLCs are novel biomarkers of inflammation, and their levels have been associated with overall mortality in a general population and with cardiovascular outcomes. In this review, the role of inflammation in the pathogenesis of CVDs, new biomarkers of inflammation, and dietary options counterbalancing inflammatory processes, such as the polyphenol-rich French maritime pine bark extract Pycnogenol, are discussed.
Collapse
|
18
|
Koniari I, Artopoulou E, Velissaris D, Ainslie M, Mplani V, Karavasili G, Kounis N, Tsigkas G. Biomarkers in the clinical management of patients with atrial fibrillation and heart failure. J Geriatr Cardiol 2021; 18:908-951. [PMID: 34908928 PMCID: PMC8648548 DOI: 10.11909/j.issn.1671-5411.2021.11.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Atrial fibrillation (AF) and heart failure (HF) are two cardiovascular diseases with an increasing prevalence worldwide. These conditions share common pathophysiologiesand frequently co-exit. In fact, the occurrence of either condition can 'cause' the development of the other, creating a new patient group that demands different management strategies to that if they occur in isolation. Regardless of the temproral association of the two conditions, their presence is linked with adverse cardiovascular outcomes, increased rate of hospitalizations, and increased economic burden on healthcare systems. The use of low-cost, easily accessible and applicable biomarkers may hasten the correct diagnosis and the effective treatment of AF and HF. Both AF and HF effect multiple physiological pathways and thus a great number of biomarkers can be measured that potentially give the clinician important diagnostic and prognostic information. These will then guide patient centred therapeutic management. The current biomarkers that offer potential for guiding therapy, focus on the physiological pathways of miRNA, myocardial stretch and injury, oxidative stress, inflammation, fibrosis, coagulation and renal impairment. Each of these has different utility in current clinincal practice.
Collapse
Affiliation(s)
- Ioanna Koniari
- Manchester Heart Institute, Manchester University Foundation Trust, Manchester, United Kingdom
| | - Eleni Artopoulou
- Department of Internal Medicine, University Hospital of Patras, Patras, Greece
| | | | - Mark Ainslie
- Manchester Heart Institute, Manchester University Foundation Trust, Manchester, United Kingdom
- Division of Cardiovascular Sciences, University of Manchester
| | - Virginia Mplani
- Department of Cardiology, University Hospital of Patras, Patras, Greece
| | - Georgia Karavasili
- Manchester Heart Institute, Manchester University Foundation Trust, Manchester, United Kingdom
| | - Nicholas Kounis
- Department of Cardiology, University Hospital of Patras, Patras, Greece
| | - Grigorios Tsigkas
- Department of Cardiology, University Hospital of Patras, Patras, Greece
| |
Collapse
|
19
|
Zelko IN, Dassanayaka S, Malovichko MV, Howard CM, Garrett LF, Uchida S, Brittian KR, Conklin DJ, Jones SP, Srivastava S. Chronic Benzene Exposure Aggravates Pressure Overload-Induced Cardiac Dysfunction. Toxicol Sci 2021; 185:64-76. [PMID: 34718823 DOI: 10.1093/toxsci/kfab125] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Benzene is a ubiquitous environmental pollutant abundant in household products, petrochemicals and cigarette smoke. Benzene is a well-known carcinogen in humans and experimental animals; however, little is known about the cardiovascular toxicity of benzene. Recent population-based studies indicate that benzene exposure is associated with an increased risk for heart failure. Nonetheless, it is unclear whether benzene exposure is sufficient to induce and/or exacerbate heart failure. We examined the effects of benzene (50 ppm, 6 h/day, 5 days/week, 6 weeks) or HEPA-filtered air exposure on transverse aortic constriction (TAC)-induced pressure overload in male C57BL/6J mice. Our data show that benzene exposure had no effect on cardiac function in the Sham group; however, it significantly compromised cardiac function as depicted by a significant decrease in fractional shortening and ejection fraction, as compared with TAC/Air-exposed mice. RNA-seq analysis of the cardiac tissue from the TAC/benzene-exposed mice showed a significant increase in several genes associated with adhesion molecules, cell-cell adhesion, inflammation, and stress response. In particular, neutrophils were implicated in our unbiased analyses. Indeed, immunofluorescence studies showed that TAC/benzene exposure promotes infiltration of CD11b+/S100A8+/myeloperoxidase+-positive neutrophils in the hearts by 3-fold. In vitro, the benzene metabolites, hydroquinone and catechol, induced the expression of P-selectin in cardiac microvascular endothelial cells by 5-fold and increased the adhesion of neutrophils to these endothelial cells by 1.5-2.0-fold. Benzene metabolite-induced adhesion of neutrophils to the endothelial cells was attenuated by anti-P-selectin antibody. Together, these data suggest that benzene exacerbates heart failure by promoting endothelial activation and neutrophil recruitment.
Collapse
Affiliation(s)
- Igor N Zelko
- University of Louisville Superfund Research Center.,Diabetes and Obesity Center.,Envirome Institute.,Department of Medicine, Division of Environmental Medicine, University of Louisville, Louisville, KY 40202
| | - Sujith Dassanayaka
- Diabetes and Obesity Center.,Department of Medicine, Division of Environmental Medicine, University of Louisville, Louisville, KY 40202
| | - Marina V Malovichko
- University of Louisville Superfund Research Center.,Diabetes and Obesity Center.,Envirome Institute.,Department of Medicine, Division of Environmental Medicine, University of Louisville, Louisville, KY 40202
| | - Caitlin M Howard
- Diabetes and Obesity Center.,Envirome Institute.,Department of Medicine, Division of Environmental Medicine, University of Louisville, Louisville, KY 40202
| | - Lauren F Garrett
- Diabetes and Obesity Center.,Envirome Institute.,Department of Medicine, Division of Environmental Medicine, University of Louisville, Louisville, KY 40202
| | - Shizuka Uchida
- Center for RNA Medicine, Department of Clinical Medicine, Aalborg University, Copenhagen SV, Denmark
| | - Kenneth R Brittian
- Diabetes and Obesity Center.,Envirome Institute.,Department of Medicine, Division of Environmental Medicine, University of Louisville, Louisville, KY 40202
| | - Daniel J Conklin
- University of Louisville Superfund Research Center.,Diabetes and Obesity Center.,Envirome Institute.,Department of Medicine, Division of Environmental Medicine, University of Louisville, Louisville, KY 40202
| | - Steven P Jones
- Diabetes and Obesity Center.,Envirome Institute.,Department of Medicine, Division of Environmental Medicine, University of Louisville, Louisville, KY 40202
| | - Sanjay Srivastava
- University of Louisville Superfund Research Center.,Diabetes and Obesity Center.,Envirome Institute.,Department of Medicine, Division of Environmental Medicine, University of Louisville, Louisville, KY 40202
| |
Collapse
|
20
|
Abstract
Neurohormones and inflammatory mediators have effects in both the heart and the peripheral vasculature. In patients with heart failure (HF), neurohormonal activation and increased levels of inflammatory mediators promote ventricular remodeling and development of HF, as well as vascular dysfunction and arterial stiffness. These processes may lead to a vicious cycle, whereby arterial stiffness perpetuates further ventricular remodeling leading to exacerbation of symptoms. Although significant advances have been made in the treatment of HF, currently available treatment strategies slow, but do not halt, this cycle. The current treatment for HF patients involves the inhibition of neurohormonal activation, which can reduce morbidity and mortality related to this condition. Beyond benefits associated with neurohormonal blockade, other strategies have focused on inhibition of inflammatory pathways implicated in the pathogenesis of HF. Unfortunately, attempts to target inflammation have not yet been successful to improve prognosis of HF. Further work is required to interrupt key maladaptive mechanisms involved in disease progression.
Collapse
|
21
|
Sansonetti M, De Windt LJ. Non-coding RNAs in cardiac inflammation: key drivers in the pathophysiology of heart failure. Cardiovasc Res 2021; 118:2058-2073. [PMID: 34097013 DOI: 10.1093/cvr/cvab192] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 06/04/2021] [Indexed: 12/15/2022] Open
Abstract
Heart failure is among the most progressive diseases and a leading cause of morbidity. Despite several advances in cardiovascular therapies, pharmacological treatments are limited to relieve symptoms without curing cardiac injury. Multiple observations point to the involvement of immune cells as key drivers in the pathophysiology of heart failure. In particular, there is a growing recognition that heart failure is related to a prolonged and insufficiently repressed inflammatory response leading to molecular, cellular, and functional cardiac alterations. Over the last decades, non-coding RNAs are recognized as prominent mediators of the cardiac inflammation, affecting the function of several immune cells. In the current review, we explore the contribution of the diverse immune cells in the progression of heart failure, revealing mechanistic functions for non-coding RNAs in cardiac immune cells as a new and exciting field of investigation.
Collapse
Affiliation(s)
- Marida Sansonetti
- Department of Molecular Genetics, Faculty of Science and Engineering; Faculty of Health, Medicine and Life Sciences; Maastricht University, Maastricht, The Netherlands
| | - Leon J De Windt
- Department of Molecular Genetics, Faculty of Science and Engineering; Faculty of Health, Medicine and Life Sciences; Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
22
|
Tripathi H, Peng H, Donahue R, Chelvarajan L, Gottipati A, Levitan B, Al-Darraji A, Gao E, Abdel-Latif A, Berron BJ. Isolation Methods for Human CD34 Subsets Using Fluorescent and Magnetic Activated Cell Sorting: an In Vivo Comparative Study. Stem Cell Rev Rep 2021; 16:413-423. [PMID: 31953639 DOI: 10.1007/s12015-019-09939-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
INTRODUCTION Acute myocardial infarction (AMI) and resulting cardiac damage and heart failure are leading causes of morbidity and mortality worldwide. Multiple studies have examined the utility of CD34+ cells for the treatment of acute and ischemic heart disease. However, the optimal strategy to enrich CD34 cells from clinical sources is not known. We examined the efficacy of fluorescence activated cell sorting (FACS) and magnetic beads cell sorting (MACS) methods for CD34 cell isolation from mobilized human mononuclear peripheral blood cells (mhPBMNCs). METHODS mhPBCs were processed following acquisition using FACS or MACS according to clinically established protocols. Cell viability, CD34 cell purity and characterization of surface marker expression were assessed using a flow cytometer. For in vivo characterization of cardiac repair, we conducted LAD ligation surgery on 8-10 weeks female NOD/SCID mice followed by intramyocardial transplantation of unselected mhPBMNCs, FACS or MACS enriched CD34+ cells. RESULTS Both MACS and FACS isolation methods achieved high purity rates, viability, and enrichment of CD34+ cells. In vivo studies following myocardial infarction demonstrated retention of CD34+ in the peri-infarct region for up to 30 days after transplantation. Retained CD34+ cells were associated with enhanced angiogenesis and reduced inflammation compared to unselected mhPBMNCs or PBS treatment arms. Cardiac scar and fibrosis as assessed by immunohistochemistry were reduced in FACS and MACS CD34+ treatment groups. Finally, reduced scar and augmented angiogenesis resulted in improved cardiac functional recovery, both on the global and regional function and remodeling assessments by echocardiography. CONCLUSION Cell based therapy using enriched CD34+ cells sorted by FACS or MACS result in better cardiac recovery after ischemic injury compared to unselected mhPBMNCs. Both enrichment techniques offer excellent recovery and purity and can be equally used for clinical applications.
Collapse
Affiliation(s)
- Himi Tripathi
- Gill Heart and Vascular Institute and Division of Cardiovascular Medicine, University of Kentucky and the Lexington VA Medical Center, Lexington, KY, USA
| | - Hsuan Peng
- Gill Heart and Vascular Institute and Division of Cardiovascular Medicine, University of Kentucky and the Lexington VA Medical Center, Lexington, KY, USA
| | - Renee Donahue
- Gill Heart and Vascular Institute and Division of Cardiovascular Medicine, University of Kentucky and the Lexington VA Medical Center, Lexington, KY, USA
| | - Lakshman Chelvarajan
- Gill Heart and Vascular Institute and Division of Cardiovascular Medicine, University of Kentucky and the Lexington VA Medical Center, Lexington, KY, USA
| | - Anuhya Gottipati
- Department of Chemical and Materials Engineering, University of Kentucky, Lexington, KY, USA
| | - Bryana Levitan
- Gill Heart and Vascular Institute and Division of Cardiovascular Medicine, University of Kentucky and the Lexington VA Medical Center, Lexington, KY, USA
| | - Ahmed Al-Darraji
- Gill Heart and Vascular Institute and Division of Cardiovascular Medicine, University of Kentucky and the Lexington VA Medical Center, Lexington, KY, USA
| | - Erhe Gao
- The Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Ahmed Abdel-Latif
- Gill Heart and Vascular Institute and Division of Cardiovascular Medicine, University of Kentucky and the Lexington VA Medical Center, Lexington, KY, USA
| | - Bradley J Berron
- Department of Chemical and Materials Engineering, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
23
|
Chen J, Norling LV, Cooper D. Cardiac Dysfunction in Rheumatoid Arthritis: The Role of Inflammation. Cells 2021; 10:881. [PMID: 33924323 PMCID: PMC8070480 DOI: 10.3390/cells10040881] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 04/08/2021] [Accepted: 04/10/2021] [Indexed: 12/25/2022] Open
Abstract
Rheumatoid arthritis is a chronic, systemic inflammatory disease that carries an increased risk of mortality due to cardiovascular disease. The link between inflammation and atherosclerotic disease is clear; however, recent evidence suggests that inflammation may also play a role in the development of nonischemic heart disease in rheumatoid arthritis (RA) patients. We consider here the link between inflammation and cardiovascular disease in the RA community with a focus on heart failure with preserved ejection fraction. The effect of current anti-inflammatory therapeutics, used to treat RA patients, on cardiovascular disease are discussed as well as whether targeting resolution of inflammation might offer an alternative strategy for tempering inflammation and subsequent inflammation-driven comorbidities in RA.
Collapse
Affiliation(s)
- Jianmin Chen
- Centre for Biochemical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK; (J.C.); (L.V.N.)
| | - Lucy V. Norling
- Centre for Biochemical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK; (J.C.); (L.V.N.)
- Centre for Inflammation and Therapeutic Innovation, Queen Mary University of London, London EC1M 6BQ, UK
| | - Dianne Cooper
- Centre for Biochemical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK; (J.C.); (L.V.N.)
- Centre for Inflammation and Therapeutic Innovation, Queen Mary University of London, London EC1M 6BQ, UK
| |
Collapse
|
24
|
Lian Z, Song JX, Yu SR, Su LN, Cui YX, Li SF, Lee CY, Liang HZ, Chen H. Therapeutic targets of rosuvastatin on heart failure and associated biological mechanisms: A study of network pharmacology and experimental validation. Eur J Pharmacol 2021; 895:173888. [PMID: 33493484 DOI: 10.1016/j.ejphar.2021.173888] [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: 10/19/2020] [Revised: 01/12/2021] [Accepted: 01/14/2021] [Indexed: 11/20/2022]
Abstract
To explore the potential targets underlying the effect of rosuvastatin on heart failure (HF) by utilizing a network pharmacology approach and experiments to identify the results. PharmMapper and other databases were mined for information relevant to the prediction of rosuvastatin targets and HF-related targets. Then, the rosuvastatin-HF target gene networks were created in Cytoscape software. Eventually, the targets and enriched pathways were examined by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis. Furthermore, we constructed an HF animal model and used rosuvastatin to treat them, identifying the changes in heart function and related protein expression. We further used different cells to explore the mechanisms of rosuvastatin. Thirty-five intersection targets indicated the therapeutic targets linked to HF. GO analysis showed that 481 biological processes, 4 cellular components and 23 molecular functions were identified. KEGG analysis showed 13 significant treatment pathways. In animal experiments, rosuvastatin significantly improved the cardiac function of post-myocardial infarction mice and prevented the development of HF after myocardial infarction by inhibiting IL-1Β expression. Cell experiments showed that rosuvastatin could reduce the expression of IL-1B in HUVEC and THP-1 cells. The therapeutic mechanism of rosuvastatin against HF may be closely related to the inhibition of the expression of apoptosis-related proteins, inflammatory factors, and fibrosis-related genes. However, IL-1Β is one of the most important target genes.
Collapse
Affiliation(s)
- Zheng Lian
- Department of Cardiology, Peking University People's Hospital, Beijing, China; Beijing Key Laboratory of Early Prediction and Intervention of Acute Myocardial Infarction, Peking University People's Hospital, Beijing, China; Center for Cardiovascular Translational Research, Peking University People's Hospital, Beijing, China
| | - Jun-Xian Song
- Department of Cardiology, Peking University People's Hospital, Beijing, China; Beijing Key Laboratory of Early Prediction and Intervention of Acute Myocardial Infarction, Peking University People's Hospital, Beijing, China; Center for Cardiovascular Translational Research, Peking University People's Hospital, Beijing, China
| | - Shi-Ran Yu
- Department of Cardiology, Peking University People's Hospital, Beijing, China; Beijing Key Laboratory of Early Prediction and Intervention of Acute Myocardial Infarction, Peking University People's Hospital, Beijing, China; Center for Cardiovascular Translational Research, Peking University People's Hospital, Beijing, China
| | - Li-Na Su
- Department of Cardiology, Peking University People's Hospital, Beijing, China; Beijing Key Laboratory of Early Prediction and Intervention of Acute Myocardial Infarction, Peking University People's Hospital, Beijing, China; Center for Cardiovascular Translational Research, Peking University People's Hospital, Beijing, China
| | - Yu-Xia Cui
- Department of Cardiology, Peking University People's Hospital, Beijing, China; Beijing Key Laboratory of Early Prediction and Intervention of Acute Myocardial Infarction, Peking University People's Hospital, Beijing, China; Center for Cardiovascular Translational Research, Peking University People's Hospital, Beijing, China
| | - Su-Fang Li
- Department of Cardiology, Peking University People's Hospital, Beijing, China; Beijing Key Laboratory of Early Prediction and Intervention of Acute Myocardial Infarction, Peking University People's Hospital, Beijing, China; Center for Cardiovascular Translational Research, Peking University People's Hospital, Beijing, China
| | - Chong-Yoo Lee
- Department of Cardiology, Peking University People's Hospital, Beijing, China; Beijing Key Laboratory of Early Prediction and Intervention of Acute Myocardial Infarction, Peking University People's Hospital, Beijing, China; Center for Cardiovascular Translational Research, Peking University People's Hospital, Beijing, China
| | - Hui-Zhu Liang
- Department of Cardiology, Peking University People's Hospital, Beijing, China; Beijing Key Laboratory of Early Prediction and Intervention of Acute Myocardial Infarction, Peking University People's Hospital, Beijing, China; Center for Cardiovascular Translational Research, Peking University People's Hospital, Beijing, China
| | - Hong Chen
- Department of Cardiology, Peking University People's Hospital, Beijing, China; Beijing Key Laboratory of Early Prediction and Intervention of Acute Myocardial Infarction, Peking University People's Hospital, Beijing, China; Center for Cardiovascular Translational Research, Peking University People's Hospital, Beijing, China.
| |
Collapse
|
25
|
Funamoto M, Sunagawa Y, Katanasaka Y, Shimizu K, Miyazaki Y, Sari N, Shimizu S, Mori K, Wada H, Hasegawa K, Morimoto T. Histone Acetylation Domains Are Differentially Induced during Development of Heart Failure in Dahl Salt-Sensitive Rats. Int J Mol Sci 2021; 22:1771. [PMID: 33578969 PMCID: PMC7916721 DOI: 10.3390/ijms22041771] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 02/06/2021] [Accepted: 02/07/2021] [Indexed: 12/21/2022] Open
Abstract
Histone acetylation by epigenetic regulators has been shown to activate the transcription of hypertrophic response genes, which subsequently leads to the development and progression of heart failure. However, nothing is known about the acetylation of the histone tail and globular domains in left ventricular hypertrophy or in heart failure. The acetylation of H3K9 on the promoter of the hypertrophic response gene was significantly increased in the left ventricular hypertrophy stage, whereas the acetylation of H3K122 did not increase in the left ventricular hypertrophy stage but did significantly increase in the heart failure stage. Interestingly, the interaction between the chromatin remodeling factor BRG1 and p300 was significantly increased in the heart failure stage, but not in the left ventricular hypertrophy stage. This study demonstrates that stage-specific acetylation of the histone tail and globular domains occurs during the development and progression of heart failure, providing novel insights into the epigenetic regulatory mechanism governing transcriptional activity in these processes.
Collapse
Affiliation(s)
- Masafumi Funamoto
- Division of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan; (M.F.); (Y.S.); (Y.K.); (K.S.); (Y.M.); (N.S.); (S.S.); (K.H.)
- Kyoto Medical Center, Clinical Research Institute, National Hospital Organization, 1-1 Fukakusa Mukaihatacho, Fushimi-ku, Kyoto 612-8555, Japan;
| | - Yoichi Sunagawa
- Division of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan; (M.F.); (Y.S.); (Y.K.); (K.S.); (Y.M.); (N.S.); (S.S.); (K.H.)
- Kyoto Medical Center, Clinical Research Institute, National Hospital Organization, 1-1 Fukakusa Mukaihatacho, Fushimi-ku, Kyoto 612-8555, Japan;
- Shizuoka General Hospital, 4-27-1 Kitaando, Aoi-ku, Shizuoka 420-8527, Japan;
| | - Yasufumi Katanasaka
- Division of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan; (M.F.); (Y.S.); (Y.K.); (K.S.); (Y.M.); (N.S.); (S.S.); (K.H.)
- Kyoto Medical Center, Clinical Research Institute, National Hospital Organization, 1-1 Fukakusa Mukaihatacho, Fushimi-ku, Kyoto 612-8555, Japan;
- Shizuoka General Hospital, 4-27-1 Kitaando, Aoi-ku, Shizuoka 420-8527, Japan;
| | - Kana Shimizu
- Division of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan; (M.F.); (Y.S.); (Y.K.); (K.S.); (Y.M.); (N.S.); (S.S.); (K.H.)
- Kyoto Medical Center, Clinical Research Institute, National Hospital Organization, 1-1 Fukakusa Mukaihatacho, Fushimi-ku, Kyoto 612-8555, Japan;
| | - Yusuke Miyazaki
- Division of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan; (M.F.); (Y.S.); (Y.K.); (K.S.); (Y.M.); (N.S.); (S.S.); (K.H.)
- Kyoto Medical Center, Clinical Research Institute, National Hospital Organization, 1-1 Fukakusa Mukaihatacho, Fushimi-ku, Kyoto 612-8555, Japan;
- Shizuoka General Hospital, 4-27-1 Kitaando, Aoi-ku, Shizuoka 420-8527, Japan;
| | - Nurmila Sari
- Division of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan; (M.F.); (Y.S.); (Y.K.); (K.S.); (Y.M.); (N.S.); (S.S.); (K.H.)
| | - Satoshi Shimizu
- Division of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan; (M.F.); (Y.S.); (Y.K.); (K.S.); (Y.M.); (N.S.); (S.S.); (K.H.)
- Kyoto Medical Center, Clinical Research Institute, National Hospital Organization, 1-1 Fukakusa Mukaihatacho, Fushimi-ku, Kyoto 612-8555, Japan;
| | - Kiyoshi Mori
- Shizuoka General Hospital, 4-27-1 Kitaando, Aoi-ku, Shizuoka 420-8527, Japan;
| | - Hiromichi Wada
- Kyoto Medical Center, Clinical Research Institute, National Hospital Organization, 1-1 Fukakusa Mukaihatacho, Fushimi-ku, Kyoto 612-8555, Japan;
| | - Koji Hasegawa
- Division of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan; (M.F.); (Y.S.); (Y.K.); (K.S.); (Y.M.); (N.S.); (S.S.); (K.H.)
- Kyoto Medical Center, Clinical Research Institute, National Hospital Organization, 1-1 Fukakusa Mukaihatacho, Fushimi-ku, Kyoto 612-8555, Japan;
| | - Tatsuya Morimoto
- Division of Molecular Medicine, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan; (M.F.); (Y.S.); (Y.K.); (K.S.); (Y.M.); (N.S.); (S.S.); (K.H.)
- Kyoto Medical Center, Clinical Research Institute, National Hospital Organization, 1-1 Fukakusa Mukaihatacho, Fushimi-ku, Kyoto 612-8555, Japan;
- Shizuoka General Hospital, 4-27-1 Kitaando, Aoi-ku, Shizuoka 420-8527, Japan;
| |
Collapse
|
26
|
Wang X, Li W, Yue Q, Du W, Li Y, Liu F, Yang L, Xu L, Zhao R, Hu J. C-C chemokine receptor 5 signaling contributes to cardiac remodeling and dysfunction under pressure overload. Mol Med Rep 2020; 23:49. [PMID: 33200795 PMCID: PMC7716393 DOI: 10.3892/mmr.2020.11687] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 10/21/2020] [Indexed: 12/21/2022] Open
Abstract
Aortic stenosis (AS) leads to chronic pressure overload, cardiac remodeling and eventually heart failure. Chemokines and their receptors have been implicated in pressure overload‑induced cardiac remodeling and dysfunction. In the present study, the role of C‑C chemokine receptor 5 (CCR5) in pressure overload‑induced cardiac remodeling and dysfunction was investigated in mice subjected to transverse aortic constriction (TAC). Cardiac levels of CCR5 and C‑C motif chemokine ligands (CCLs)3, 4 and 5 were determined by western blotting and reverse transcription‑quantitative PCR, respectively. Cardiac functional parameters were evaluated by echocardiographic and hemodynamic measurements. Myocardial fibrosis was assessed by Masson's trichrome staining and α‑smooth muscle actin immunostaining. Myocardial hypertrophy and inflammatory cell infiltration were evaluated by hematoxylin and eosin staining. Angiotensin II (Ang II)‑induced hypertrophy of H9c2 cardiomyocytes was assessed by F‑actin immunostaining. ERK1/2 and P38 phosphorylation was examined by western blotting. TAC mice exhibited higher myocardial CCL3, CCL4, CCL5 and CCR5 levels compared with sham mice. Compared with sham mice, TAC mice also exhibited impaired cardiac function along with myocardial hypertrophy, fibrosis and inflammatory cell infiltration. TAC‑induced cardiac remodeling and dysfunction were effectively ameliorated by administration of anti‑CCR5 but not by IgG control antibody. Mechanistically, increased ERK1/2 and P38 phosphorylation was detected in TAC hearts and Ang II‑stimulated H9c2 cardiomyocytes. Treatment with anti‑CCR5 antibody decreased ERK1/2 and P38 phosphorylation and attenuated Ang II‑induced H9c2 cell hypertrophy. CCR5 inhibition protected against pressure overload‑induced cardiac abnormality. The findings of the present study indicate that ERK1/2 and P38 signaling pathways may be involved in the cardioprotective effects of CCR5 inhibition.
Collapse
Affiliation(s)
- Xiaomin Wang
- Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, P.R. China
| | - Wei Li
- Translational Medicine Center, Baotou Central Hospital, Donghe, Baotou 014040, P.R. China
| | - Qiang Yue
- Department of Cardiology, Baotou Central Hospital, Donghe, Baotou 014040, P.R. China
| | - Wei Du
- Department of Cardiology, Baotou Central Hospital, Donghe, Baotou 014040, P.R. China
| | - Yongming Li
- Department of Cardiology, Baotou Central Hospital, Donghe, Baotou 014040, P.R. China
| | - Fu Liu
- Department of Cardiology, Baotou Central Hospital, Donghe, Baotou 014040, P.R. China
| | - Liu Yang
- Department of Institution of Interventional and Vascular Surgery, Tongji University, Shanghai 200072, P.R. China
| | - Lijuan Xu
- Department of Institution of Interventional and Vascular Surgery, Tongji University, Shanghai 200072, P.R. China
| | - Ruiping Zhao
- Baotou Central Hospital (The Post-doctoral Research Station of Clinic Medicine, Tongji University), Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Jiang Hu
- Translational Medicine Center, Baotou Central Hospital, Donghe, Baotou 014040, P.R. China
| |
Collapse
|
27
|
Abstract
Heart failure exhibits remarkable pathophysiologic heterogeneity. A large body of evidence suggests that regardless of the underlying etiology, heart failure is associated with induction of cytokines and chemokines that may contribute to the pathogenesis of adverse remodeling, and systolic and diastolic dysfunction. The pro-inflammatory cytokines tumor necrosis factor (TNF)-α, interleukin (IL)-1, and IL-6 have been extensively implicated in the pathogenesis of heart failure. Inflammatory cytokines modulate phenotype and function of all myocardial cells, suppressing contractile function in cardiomyocytes, inducing inflammatory activation in macrophages, stimulating microvascular inflammation and dysfunction, and promoting a matrix-degrading phenotype in fibroblasts. Moreover, cytokine-induced growth factor synthesis may exert chronic fibrogenic actions contributing to the pathogenesis of heart failure with preserved ejection fraction (HFpEF). In addition to their role in adverse cardiac remodeling, some inflammatory cytokines may also exert protective actions on cardiomyocytes under conditions of stress. Chemokines, such as CCL2, are also upregulated in failing hearts and may stimulate recruitment of pro-inflammatory leukocytes, promoting myocardial injury, fibrotic remodeling, and dysfunction. Although experimental evidence suggests that cytokine and chemokine targeting may hold therapeutic promise in heart failure, clinical translation remains challenging. This review manuscript summarizes our knowledge on the role of TNF-α, IL-1, IL-6, and CCL2 in the pathogenesis of heart failure, and discusses the promises and challenges of targeted anti-cytokine therapy. Dissection of protective and maladaptive cellular actions of cytokines in the failing heart, and identification of patient subsets with overactive or dysregulated myocardial inflammatory responses are required for design of successful therapeutic approaches.
Collapse
|
28
|
Boehm M, Novoyatleva T, Kojonazarov B, Veit F, Weissmann N, Ghofrani HA, Seeger W, Schermuly RT. Nitric Oxide Synthase 2 Induction Promotes Right Ventricular Fibrosis. Am J Respir Cell Mol Biol 2019; 60:346-356. [PMID: 30277804 DOI: 10.1165/rcmb.2018-0069oc] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The ability of the right ventricle to compensate pressure overload determines survival in pulmonary arterial hypertension (PAH). Nitric oxide (NO) reduces the right ventricular afterload through pulmonary vasodilation, but excessive NO amounts cause oxidative stress. Oxidative stress drives remodeling of pulmonary arteries and the right ventricle. In the present study, we hypothesized that nitric oxide synthase 2 (NOS2) induction leads to excessive NO amounts that contribute to oxidative stress and impair right ventricular adaptation to PAH. We used a surgical pulmonary artery banding (PAB) mouse model in which right ventricular dysfunction and remodeling occur independently of changes in the pulmonary vasculature. Three weeks after PAB, NOS2 expression was increased twofold in the hypertrophied right ventricle on transcript and protein levels together with increased NO production. Histomorphology localized NOS2 in interstitial and perivascular cardiac fibroblasts after PAB, which was confirmed by cell isolation experiments. In the hypertrophied right ventricle, NOS2 induction was accompanied by an increased formation of reactive oxidants blocked by ex vivo NOS inhibition. We show that reactive oxidant formation in the hypertrophied right ventricle is in part NOS2 dependent (in NOS2-deficient mice [NOS2-/-]). Lack of NOS2 induction prevented superoxide scavenging and decreased reactive oxidant formation. Functional measures of cardiac function by noninvasive echocardiography together with intracardiac catheterization revealed no differences in heart function between both genotypes after PAB. However, reduced NO and reactive oxidant formation in the hypertrophied right ventricle of NOS2-/- mice was linked to reduced collagen accumulation through reduced collagen deposition from the cardiac fibroblast. Together, our data demonstrate a profibrotic role for NOS2 induction in the hypertrophied right ventricle.
Collapse
Affiliation(s)
- Mario Boehm
- 1 Universities of Giessen and Marburg Lung Center, Giessen, Germany.,2 Excellence Cluster Cardio-Pulmonary System, Giessen, Germany.,3 German Center for Lung Research, Giessen, Germany; and
| | - Tatyana Novoyatleva
- 1 Universities of Giessen and Marburg Lung Center, Giessen, Germany.,2 Excellence Cluster Cardio-Pulmonary System, Giessen, Germany.,3 German Center for Lung Research, Giessen, Germany; and
| | - Baktybek Kojonazarov
- 1 Universities of Giessen and Marburg Lung Center, Giessen, Germany.,2 Excellence Cluster Cardio-Pulmonary System, Giessen, Germany.,3 German Center for Lung Research, Giessen, Germany; and
| | - Florian Veit
- 1 Universities of Giessen and Marburg Lung Center, Giessen, Germany.,2 Excellence Cluster Cardio-Pulmonary System, Giessen, Germany.,3 German Center for Lung Research, Giessen, Germany; and
| | - Norbert Weissmann
- 1 Universities of Giessen and Marburg Lung Center, Giessen, Germany.,2 Excellence Cluster Cardio-Pulmonary System, Giessen, Germany.,3 German Center for Lung Research, Giessen, Germany; and
| | - Hossein A Ghofrani
- 1 Universities of Giessen and Marburg Lung Center, Giessen, Germany.,2 Excellence Cluster Cardio-Pulmonary System, Giessen, Germany.,3 German Center for Lung Research, Giessen, Germany; and
| | - Werner Seeger
- 1 Universities of Giessen and Marburg Lung Center, Giessen, Germany.,2 Excellence Cluster Cardio-Pulmonary System, Giessen, Germany.,3 German Center for Lung Research, Giessen, Germany; and.,4 Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Ralph T Schermuly
- 1 Universities of Giessen and Marburg Lung Center, Giessen, Germany.,2 Excellence Cluster Cardio-Pulmonary System, Giessen, Germany.,3 German Center for Lung Research, Giessen, Germany; and
| |
Collapse
|
29
|
Martini E, Kunderfranco P, Peano C, Carullo P, Cremonesi M, Schorn T, Carriero R, Termanini A, Colombo FS, Jachetti E, Panico C, Faggian G, Fumero A, Torracca L, Molgora M, Cibella J, Pagiatakis C, Brummelman J, Alvisi G, Mazza EMC, Colombo MP, Lugli E, Condorelli G, Kallikourdis M. Single-Cell Sequencing of Mouse Heart Immune Infiltrate in Pressure Overload-Driven Heart Failure Reveals Extent of Immune Activation. Circulation 2019; 140:2089-2107. [PMID: 31661975 DOI: 10.1161/circulationaha.119.041694] [Citation(s) in RCA: 219] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Inflammation is a key component of cardiac disease, with macrophages and T lymphocytes mediating essential roles in the progression to heart failure. Nonetheless, little insight exists on other immune subsets involved in the cardiotoxic response. METHODS Here, we used single-cell RNA sequencing to map the cardiac immune composition in the standard murine nonischemic, pressure-overload heart failure model. By focusing our analysis on CD45+ cells, we obtained a higher resolution identification of the immune cell subsets in the heart, at early and late stages of disease and in controls. We then integrated our findings using multiparameter flow cytometry, immunohistochemistry, and tissue clarification immunofluorescence in mouse and human. RESULTS We found that most major immune cell subpopulations, including macrophages, B cells, T cells and regulatory T cells, dendritic cells, Natural Killer cells, neutrophils, and mast cells are present in both healthy and diseased hearts. Most cell subsets are found within the myocardium, whereas mast cells are found also in the epicardium. Upon induction of pressure overload, immune activation occurs across the entire range of immune cell types. Activation led to upregulation of key subset-specific molecules, such as oncostatin M in proinflammatory macrophages and PD-1 in regulatory T cells, that may help explain clinical findings such as the refractivity of patients with heart failure to anti-tumor necrosis factor therapy and cardiac toxicity during anti-PD-1 cancer immunotherapy, respectively. CONCLUSIONS Despite the absence of infectious agents or an autoimmune trigger, induction of disease leads to immune activation that involves far more cell types than previously thought, including neutrophils, B cells, Natural Killer cells, and mast cells. This opens up the field of cardioimmunology to further investigation by using toolkits that have already been developed to study the aforementioned immune subsets. The subset-specific molecules that mediate their activation may thus become useful targets for the diagnostics or therapy of heart failure.
Collapse
Affiliation(s)
- Elisa Martini
- Adaptive Immunity Laboratory (E.M., M.C., M.K.), Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Paolo Kunderfranco
- Bioinformatics Unit (P.K., R.C., A.T.), Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Clelia Peano
- Genomic Unit (C. Peano, J.C.), Humanitas Clinical and Research Center, Rozzano, Milan, Italy.,Institute of Genetic and Biomedical Research, UoS Milan, National Research Council, Rozzano, Italy (C. Peano, P.C., G.C.)
| | - Pierluigi Carullo
- Department of Cardiovascular Medicine (P.C., C. Panico, C. Pagiatakis, G.C.), Humanitas Clinical and Research Center, Rozzano, Milan, Italy.,Institute of Genetic and Biomedical Research, UoS Milan, National Research Council, Rozzano, Italy (C. Peano, P.C., G.C.)
| | - Marco Cremonesi
- Adaptive Immunity Laboratory (E.M., M.C., M.K.), Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Tilo Schorn
- Advanced Imaging Unit (T.S.), Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Roberta Carriero
- Bioinformatics Unit (P.K., R.C., A.T.), Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Alberto Termanini
- Bioinformatics Unit (P.K., R.C., A.T.), Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Federico Simone Colombo
- Flow Cytometry Core (F.S.C., E.L.), Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Elena Jachetti
- Molecular Immunology Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy (E.J., M.P.C.)
| | - Cristina Panico
- Department of Cardiovascular Medicine (P.C., C. Panico, C. Pagiatakis, G.C.), Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Giuseppe Faggian
- Department of Cardiac Surgery, University of Verona, Italy (G.F.)
| | - Andrea Fumero
- Cardiac Surgery Division, Department of Cardiovascular Medicine (A.F., L.T.), Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Lucia Torracca
- Cardiac Surgery Division, Department of Cardiovascular Medicine (A.F., L.T.), Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Martina Molgora
- Laboratory of Experimental Immunopathology (M.M.), Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Javier Cibella
- Genomic Unit (C. Peano, J.C.), Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Christina Pagiatakis
- Department of Cardiovascular Medicine (P.C., C. Panico, C. Pagiatakis, G.C.), Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Jolanda Brummelman
- Laboratory of Translational Immunology (J.B., G.A., E.M.C., E.L.), Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Giorgia Alvisi
- Laboratory of Translational Immunology (J.B., G.A., E.M.C., E.L.), Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Emilia Maria Cristina Mazza
- Laboratory of Translational Immunology (J.B., G.A., E.M.C., E.L.), Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Mario Paolo Colombo
- Molecular Immunology Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy (E.J., M.P.C.)
| | - Enrico Lugli
- Flow Cytometry Core (F.S.C., E.L.), Humanitas Clinical and Research Center, Rozzano, Milan, Italy.,Laboratory of Translational Immunology (J.B., G.A., E.M.C., E.L.), Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Gianluigi Condorelli
- Department of Cardiovascular Medicine (P.C., C. Panico, C. Pagiatakis, G.C.), Humanitas Clinical and Research Center, Rozzano, Milan, Italy.,Institute of Genetic and Biomedical Research, UoS Milan, National Research Council, Rozzano, Italy (C. Peano, P.C., G.C.).,Humanitas University, Pieve Emanuele, Italy (G.C., M.K.)
| | - Marinos Kallikourdis
- Adaptive Immunity Laboratory (E.M., M.C., M.K.), Humanitas Clinical and Research Center, Rozzano, Milan, Italy.,Humanitas University, Pieve Emanuele, Italy (G.C., M.K.)
| |
Collapse
|
30
|
Abstract
Chemokines are small secreted proteins with chemoattractant properties that play a key role in inflammation. One such chemokine, Stromal cell-derived factor-1 (SDF-1) also known as CXCL12, and its receptor, CXCR4, are expressed and functional in cardiac myocytes. SDF-1 both stimulates and enhances the cellular signal which attracts potentially beneficial stem cells for tissue repair within the ischemic heart. Paradoxically however, this chemokine is known to act in concert with the inflammatory cytokines of the innate immune response which contributes to cellular injury through the recruitment of inflammatory cells during ischemia. In the present study, we have demonstrated that SDF-1 has dose dependent effects on freshly isolated cardiomyocytes. Using Tunnel and caspase 3-activation assays, we have demonstrated that the treatment of isolated adult rat cardiac myocyte with SDF-1 at higher concentrations (pathological concentrations) induced apoptosis. Furthermore, ELISA data demonstrated that the treatment of isolated adult rat cardiac myocyte with SDF-1 at higher concentrations upregulated TNF-α protein expression which directly correlated with subsequent apoptosis. There was a significant reduction in SDF-1 mediated apoptosis when TNF-α expression was neutralized which suggests that SDF-1 mediated apoptosis is TNF-α-dependent. The fact that certain stimuli are capable of driving cardiomyocytes into apoptosis indicates that these cells are susceptible to clinically relevant apoptotic triggers. Our findings suggest that the elevated SDF-1 levels seen in a variety of clinical conditions, including ischemic myocardial infarction, may either directly or indirectly contribute to cardiac cell death via a TNF-α mediated pathway. This highlights the importance of this receptor/ligand in regulating the cardiomyocyte response to stress conditions.
Collapse
|
31
|
Abstract
Advanced heart failure (HF) is a progressive disease characterized by recurrent hospitalizations and high risk of mortality. Indeed, outcomes in late stages of HF approximate those seen in patients with various aggressive malignancies. Clinical trials assessing beneficial outcomes of new treatments in patients with cancer have used innovative approaches to measure impact on total disease burden or surrogates to assess treatment efficacy. Although most cardiovascular outcomes trials continue to use time-to-first event analyses to assess the primary efficacy end point, such analyses do not adequately reflect the impact of new treatments on the totality of the chronic disease burden. Consequently, patient enrichment and other strategies for ongoing clinical trial design, as well as new statistical methodologies, are important considerations, particularly when studying a population with advanced chronic HF. The DREAM-HF trial (Double-Blind Randomized Assessment of Clinical Events With Allogeneic Mesenchymal Precursor Cells in Advanced Heart Failure) is an ongoing, randomized, sham-controlled phase 3 study of the efficacy and safety of mesenchymal precursor cells as immunotherapy in patients with advanced chronic HF with reduced ejection fraction. Mesenchymal precursor cells have a unique multimodal mechanism of action that is believed to result in polarization of proinflammatory type 1 macrophages in the heart to an anti-inflammatory type 2 macrophage state, inhibition of maladaptive adverse left ventricular remodeling, reversal of cardiac and peripheral endothelial dysfunction, and recovery of deranged vasculature. The objective of DREAM-HF is to confirm earlier phase 2 results and evaluate whether mesenchymal precursor cells will reduce the rate of nonfatal recurrent HF-related major adverse cardiac events while delaying or preventing progression of HF to terminal cardiac events. DREAM-HF is an example of an ongoing contemporary events-driven cardiovascular cell-based immunotherapy study that has utilized the concepts of baseline disease enrichment, prognostic enrichment, and predictive enrichment to improve its efficiency by using accumulating data from within as well as external to the trial. Adaptive enrichment designs and strategies are important components of a rational approach to achieve clinical research objectives in shorter clinical trial timelines and with increased cost-effectiveness without compromising ethical standards or the overall statistical integrity of the study. The DREAM-HF trial also presents an alternative approach to traditional composite time-to-first event primary efficacy end points. Statistical methodologies such as the joint frailty model provide opportunities to expand the scope of events-driven HF with reduced ejection fraction clinical trials to utilize time to recurrent nonfatal HF-related major adverse cardiac events as the primary efficacy end point without compromising the integrity of the statistical analyses for terminal cardiac events. In advanced chronic HF with reduced ejection fraction studies, the joint frailty model is utilized to reflect characteristics of the high-risk patient population with important unmet therapeutic needs. In some cases, use of the joint frailty model may substantially reduce sample size requirements. In addition, using an end point that is acceptable to the Food and Drug Administration and the European Medicines Agency, such as recurrent nonfatal HF-related major adverse cardiac events, enables generation of clinically relevant pharmacoeconomic data while providing comprehensive views of the patient's overall cardiovascular disease burden. The major goal of this review is to provide lessons learned from the ongoing DREAM-HF trial that relate to biologic plausibility and flexible clinical trial design and are potentially applicable to other development programs of innovative therapies for patients with advanced cardiovascular disease. Clinical Trial Registration: URL: https://www.clinicaltrials.gov. Unique identifier: NCT02032004.
Collapse
Affiliation(s)
| | | | - Barry Greenberg
- University of California, San Diego School of Medicine, La Jolla (B.G.)
- Advanced Heart Failure Treatment Program, Sulpizio Cardiovascular Center, University of California, San Diego Healthcare System, La Jolla (B.G.)
| | - Emerson C. Perin
- Stem Cell Center and Adult Cardiology, Texas Heart Institute, Houston (E.C.P.)
| |
Collapse
|
32
|
Matsushita N, Ishida N, Ibi M, Saito M, Takahashi M, Taniguchi S, Iwakura Y, Morino Y, Taira E, Sawa Y, Hirose M. IL-1β Plays an Important Role in Pressure Overload-Induced Atrial Fibrillation in Mice. Biol Pharm Bull 2019; 42:543-546. [DOI: 10.1248/bpb.b18-00363] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Naoko Matsushita
- Division of Molecular and Cellular Pharmacology, Department of Pathophysiology and Pharmacology, School of Pharmaceutical Sciences, Iwate Medical University
- Division of Cardiology, Department of Internal Medicine, School of Medicine, Iwate Medical University
| | - Nanae Ishida
- Division of Molecular and Cellular Pharmacology, Department of Pathophysiology and Pharmacology, School of Pharmaceutical Sciences, Iwate Medical University
| | - Miho Ibi
- Division of Molecular and Cellular Pharmacology, Department of Pathophysiology and Pharmacology, School of Pharmaceutical Sciences, Iwate Medical University
| | - Maki Saito
- Division of Molecular and Cellular Pharmacology, Department of Pathophysiology and Pharmacology, School of Pharmaceutical Sciences, Iwate Medical University
| | - Masafumi Takahashi
- Division of Inflammation Research, Center for Molecular Medicine, Jichi Medical University
| | - Shunichiro Taniguchi
- Department of Molecular Oncology, Graduate School of Medicine, Shinshu University
| | - Yoichiro Iwakura
- Division of Experimental Animal Immunology, Research Institute for Biological Sciences, Tokyo University of Science
| | - Yoshihiro Morino
- Division of Cardiology, Department of Internal Medicine, School of Medicine, Iwate Medical University
| | - Eiichi Taira
- Department of Molecular Oncology, Graduate School of Medicine, Shinshu University
| | - Yohei Sawa
- Division of Cardiology, Department of Internal Medicine, School of Medicine, Iwate Medical University
| | - Masamichi Hirose
- Division of Molecular and Cellular Pharmacology, Department of Pathophysiology and Pharmacology, School of Pharmaceutical Sciences, Iwate Medical University
| |
Collapse
|
33
|
Lock MC, Darby JRT, Soo JY, Brooks DA, Perumal SR, Selvanayagam JB, Seed M, Macgowan CK, Porrello ER, Tellam RL, Morrison JL. Differential Response to Injury in Fetal and Adolescent Sheep Hearts in the Immediate Post-myocardial Infarction Period. Front Physiol 2019; 10:208. [PMID: 30890961 PMCID: PMC6412108 DOI: 10.3389/fphys.2019.00208] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Accepted: 02/18/2019] [Indexed: 12/11/2022] Open
Abstract
Aim: Characterizing the response to myocardial infarction (MI) in the regenerative sheep fetus heart compared to the post-natal non-regenerative adolescent heart may reveal key morphological and molecular differences that equate to the response to MI in humans. We hypothesized that the immediate response to injury in (a) infarct compared with sham, and (b) infarct, border, and remote tissue, in the fetal sheep heart would be fundamentally different to the adolescent, allowing for repair after damage. Methods: We used a sheep model of MI induced by ligating the left anterior descending coronary artery. Surgery was performed on fetuses (105 days) and adolescent sheep (6 months). Sheep were randomly separated into MI (n = 5) or Sham (n = 5) surgery groups at both ages. We used magnetic resonance imaging (MRI), histological/immunohistochemical staining, and qRT-PCR to assess the morphological and molecular differences between the different age groups in response to infarction. Results: Magnetic resonance imaging showed no difference in fetuses for key functional parameters; however there was a significant decrease in left ventricular ejection fraction and cardiac output in the adolescent sheep heart at 3 days post-infarction. There was no significant difference in functional parameters between MRI sessions at Day 0 and Day 3 after surgery. Expression of genes involved in glucose transport and fatty acid metabolism, inflammatory cytokines as well as growth factors and cell cycle regulators remained largely unchanged in the infarcted compared to sham ventricular tissue in the fetus, but were significantly dysregulated in the adolescent sheep. Different cardiac tissue region-specific gene expression profiles were observed between the fetal and adolescent sheep. Conclusion: Fetuses demonstrated a resistance to cardiac damage not observed in the adolescent animals. The manipulation of specific gene expression profiles to a fetal-like state may provide a therapeutic strategy to treat patients following an infarction.
Collapse
Affiliation(s)
- Mitchell C Lock
- Early Origins of Adult Health Research Group, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia
| | - Jack R T Darby
- Early Origins of Adult Health Research Group, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia
| | - Jia Yin Soo
- Early Origins of Adult Health Research Group, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia
| | - Doug A Brooks
- Mechanisms in Cell Biology and Disease Research Group, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia
| | - Sunthara Rajan Perumal
- Preclinical, Imaging and Research Laboratories, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Joseph B Selvanayagam
- Cardiac Imaging Research Group, Department of Heart Health, South Australian Health and Medical Research Institute, Flinders University, Adelaide, SA, Australia
| | - Mike Seed
- The Hospital for Sick Children, Division of Cardiology, Toronto, ON, Canada
| | | | - Enzo R Porrello
- Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, VIC, Australia.,Department of Physiology, School of Biomedical Sciences, University of Melbourne, Parkville, VIC, Australia
| | - Ross L Tellam
- Early Origins of Adult Health Research Group, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia
| | - Janna L Morrison
- Early Origins of Adult Health Research Group, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia
| |
Collapse
|
34
|
Wu X, Chen Z, Yang Y, Dong Y, Liu H, Kuang S, Luo K. Impact of proteasome inhibitor MG-132 on expression of NF-κB, IL-1β and histological remodeling after myocardial infarction. Exp Ther Med 2018; 16:1365-1372. [PMID: 30112065 DOI: 10.3892/etm.2018.6308] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Accepted: 09/01/2017] [Indexed: 11/06/2022] Open
Abstract
The aim of the present study was to investigate the impact of carbobenzoxy-Leu-Leu-leucinal (MG-132) on myocardial remodeling in rats with myocardial infarction (MI) and investigate the possible underlying mechanisms. The rat model of MI was established, followed by administration of MG-132 (MG group), pyrrolidine dithiocarbamic acid (PDTC group) or normal saline (MI group) for 28 days. The expression of nuclear factor-κB (NF-κB) p65, interleukin 1β (IL-1β) and matrix metalloproteinase 2 (MMP-2), as well as the total volume of collagen and the ratio of type I/III collagen were then detected. Total collagen, including type I and III collagen, and the ratio of type I/III collagen were significantly increased in MI rats compared with those in the sham group (P<0.01), while it was significantly decreased in the PDTC and MG groups compared with that in the MI group (P<0.01). A similar trend was identified for the expression of NF-κB, IL-1β and MMP-2, which was significantly increased in the MI group compared with that in the sham group (P<0.01), while it was significantly decreased in the MG and PDTC groups compared with that in the MI group (P<0.01). In conclusion, MG-132 was demonstrated to improve post-MI tissue remodeling, and the mechanism may be associated with the inhibition of NF-κB activation and the downregulation of inflammatory cytokines, such as IL-1β.
Collapse
Affiliation(s)
- Xinhua Wu
- Department of Cardiology, The First Affiliated Hospital of Dali University, Dali, Yunnan 671000, P.R. China
| | - Zhangrong Chen
- Department of Cardiology, The First Affiliated Hospital of Dali University, Dali, Yunnan 671000, P.R. China
| | - Ying Yang
- Department of Cardiology, The First Affiliated Hospital of Dali University, Dali, Yunnan 671000, P.R. China
| | - Yu Dong
- Department of Cardiology, The First Affiliated Hospital of Dali University, Dali, Yunnan 671000, P.R. China
| | - Hong Liu
- Department of Cardiology, The First Affiliated Hospital of Dali University, Dali, Yunnan 671000, P.R. China
| | - Shiquan Kuang
- Department of Cardiology, The First Affiliated Hospital of Dali University, Dali, Yunnan 671000, P.R. China
| | - Kailiang Luo
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 404100, P.R. China
| |
Collapse
|
35
|
Carubelli V, Bonadei I, Castrini AI, Gorga E, Ravera A, Lombardi C, Metra M. Prognostic value of the absolute lymphocyte count in patients admitted for acute heart failure. J Cardiovasc Med (Hagerstown) 2018; 18:859-865. [PMID: 27541359 DOI: 10.2459/jcm.0000000000000428] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Low relative lymphocyte count is an important prognostic marker in acute heart failure (AHF); however, it could be influenced by other abnormalities in white cells count. Our purpose is to evaluate if low absolute lymphocyte count (ALC) is an independent predictor of events in patients with AHF. METHODS In a retrospective analysis, we included 309 patients with AHF, divided into two groups according to the median value of ALC at admission (1410 cells/μl). The primary end point was all-cause mortality or urgent heart transplantation within 1 year. RESULTS Patients with low ALC were older and had more comorbidity, namely atrial fibrillation, chronic kidney disease, chronic obstructive pulmonary disease and anemia. Low ALC was associated with higher all-cause mortality or urgent heart transplantation at 1 year (24.3 vs 13.0%; P = 0.012). In a multivariable model, the independent predictors of mortality at 1 year were ALC 1410 cells/μl or less at admission [hazard ratio 2.04; CI (confidence interval) 95% (1.06-3.95); P = 0.033], age [hazard ratio 1.08; CI 95% (1.04-1.12); P < 0.001], baseline serum creatinine [hazard ratio 1.25; CI 95% (1.05-1.50); P = 0.012] and baseline serum Na [hazard ratio 0.91; CI 95% (0.85-0.98); P = 0.013]. CONCLUSION Low ALC in patients with AHF is associated with higher in-hospital mortality during the hospitalization and is an independent predictor of long-term mortality.
Collapse
Affiliation(s)
- Valentina Carubelli
- Division of Cardiology, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University and Civil Hospital of Brescia, Brescia, Italy
| | | | | | | | | | | | | |
Collapse
|
36
|
Sun XQ, Abbate A, Bogaard HJ. Role of cardiac inflammation in right ventricular failure. Cardiovasc Res 2018; 113:1441-1452. [PMID: 28957536 DOI: 10.1093/cvr/cvx159] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 08/09/2017] [Indexed: 12/18/2022] Open
Abstract
Right ventricular failure (RVF) is the main determinant of mortality in patients with pulmonary arterial hypertension (PAH). Although the exact pathophysiology underlying RVF remains unclear, inflammation may play an important role, as it does in left heart failure. Perivascular pulmonary artery and systemic inflammation is relatively well studied and known to contribute to the initiation and maintenance of the pulmonary vascular insult in PAH. However, less attention has been paid to the role of cardiac inflammation in RVF and PAH. Consistent with many other types of heart failure, cardiac inflammation, triggered by systemic and local stressors, has been shown in RVF patients as well as in RVF animal models. RV inflammation likely contributes to impaired RV contractility, maladaptive remodelling and a vicious circle between RV and pulmonary vascular injury. Although the potential to improve RV function through anti-inflammatory therapy has not been tested, this approach has been applied clinically in left ventricular failure patients, with variable success. Because inflammation plays a dual role in the development of both pulmonary vascular pathology and RVF, anti-inflammatory therapies may have a potential double benefit in patients with PAH and associated RVF.
Collapse
Affiliation(s)
- Xiao-Qing Sun
- Department of Pulmonology, VU University Medical Center/Institute for Cardiovascular Research, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Antonio Abbate
- Department of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Harm-Jan Bogaard
- Department of Pulmonology, VU University Medical Center/Institute for Cardiovascular Research, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| |
Collapse
|
37
|
Frangogiannis NG. Fibroblasts and the extracellular matrix in right ventricular disease. Cardiovasc Res 2018; 113:1453-1464. [PMID: 28957531 DOI: 10.1093/cvr/cvx146] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 08/01/2017] [Indexed: 12/17/2022] Open
Abstract
Right ventricular failure predicts adverse outcome in patients with pulmonary hypertension (PH), and in subjects with left ventricular heart failure and is associated with interstitial fibrosis. This review manuscript discusses the cellular effectors and molecular mechanisms implicated in right ventricular fibrosis. The right ventricular interstitium contains vascular cells, fibroblasts, and immune cells, enmeshed in a collagen-based matrix. Right ventricular pressure overload in PH is associated with the expansion of the fibroblast population, myofibroblast activation, and secretion of extracellular matrix proteins. Mechanosensitive transduction of adrenergic signalling and stimulation of the renin-angiotensin-aldosterone cascade trigger the activation of right ventricular fibroblasts. Inflammatory cytokines and chemokines may contribute to expansion and activation of macrophages that may serve as a source of fibrogenic growth factors, such as transforming growth factor (TGF)-β. Endothelin-1, TGF-βs, and matricellular proteins co-operate to activate cardiac myofibroblasts, and promote synthesis of matrix proteins. In comparison with the left ventricle, the RV tolerates well volume overload and ischemia; whether the right ventricular interstitial cells and matrix are implicated in these favourable responses remains unknown. Expansion of fibroblasts and extracellular matrix protein deposition are prominent features of arrhythmogenic right ventricular cardiomyopathies and may be implicated in the pathogenesis of arrhythmic events. Prevailing conceptual paradigms on right ventricular remodelling are based on extrapolation of findings in models of left ventricular injury. Considering the unique embryologic, morphological, and physiologic properties of the RV and the clinical significance of right ventricular failure, there is a need further to dissect RV-specific mechanisms of fibrosis and interstitial remodelling.
Collapse
Affiliation(s)
- Nikolaos G Frangogiannis
- Department of Medicine (Cardiology), The Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Forchheimer G46B Bronx, 10461 NY, USA
| |
Collapse
|
38
|
Affiliation(s)
- Farhan Shahid
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Gregory Y H Lip
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, United Kingdom.,Aalborg Thrombosis Research Unit, Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Eduard Shantsila
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
39
|
Martini E, Stirparo GG, Kallikourdis M. Immunotherapy for cardiovascular disease. J Leukoc Biol 2017; 103:493-500. [PMID: 29345361 DOI: 10.1002/jlb.5mr0717-306r] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 11/16/2017] [Accepted: 11/24/2017] [Indexed: 12/11/2022] Open
Abstract
Heart failure (HF), the final stage of pathological cardiac hypertrophy, is a major cause of hospitalization and mortality. The role of inflammation in the pathogenesis of HF has been extensively studied, with great emphasis on proinflammatory cytokines. Yet, clinical trials targeting these cytokines failed to become a credible therapeutic strategy for HF. More recent studies are increasingly highlighting an active role for T cells in the progression of HF pathology. As a result, a number of novel immunotherapy strategies are emerging for the treatment of HF and other cardiovascular diseases, via the targeting of adaptive immunity. Here we provide an overview of the background, details, and expected outcomes of these attempts.
Collapse
Affiliation(s)
- Elisa Martini
- Adaptive Immunity Laboratory, Humanitas Clinical and Research Center, Via Manzoni 56, Rozzano, Milan, Italy
| | - Giuliano Giuseppe Stirparo
- Department of Cardiovascular Medicine, Humanitas Clinical and Research Center, Via Manzoni 56, Rozzano, Milan, Italy
| | - Marinos Kallikourdis
- Adaptive Immunity Laboratory, Humanitas Clinical and Research Center, Via Manzoni 56, Rozzano, Milan, Italy.,Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, Milan, Italy
| |
Collapse
|
40
|
Gröschel C, Sasse A, Röhrborn C, Monecke S, Didié M, Elsner L, Kruse V, Bunt G, Lichtman AH, Toischer K, Zimmermann WH, Hasenfuß G, Dressel R. T helper cells with specificity for an antigen in cardiomyocytes promote pressure overload-induced progression from hypertrophy to heart failure. Sci Rep 2017; 7:15998. [PMID: 29167489 PMCID: PMC5700082 DOI: 10.1038/s41598-017-16147-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 11/08/2017] [Indexed: 12/13/2022] Open
Abstract
We investigated whether CD4+-T cells with specificity for an antigen in cardiomyocytes promote the progression from hypertrophy to heart failure in mice with increased pressure load due to transverse aortic constriction (TAC). OT-II mice expressing a transgenic T cell receptor (TCR) with specificity for ovalbumin (OVA) on CD4+-T cells and cMy-mOVA mice expressing OVA on cardiomyocytes were crossed. The resulting cMy-mOVA-OT-II mice did not display signs of spontaneous autoimmunity despite the fact that their OVA-specific CD4+-T cells were not anergic. After TAC, progression to heart failure was significantly accelerated in cMy-mOVA-OT-II compared to cMy-mOVA mice. No OVA-specific antibodies were induced in response to TAC in cMy-mOVA-OT-II mice, yet more CD3+ T cells infiltrated their myocardium when compared with TAC-operated cMy-mOVA mice. Systemically, the proportion of activated CD4+-T cells with a Th1 and Th17 cytokine profile was increased in cMy-mOVA-OT-II mice after TAC. Thus, T helper cells with specificity for an antigen in cardiomyocytes can directly promote the progression of heart failure in response to pressure overload independently of autoantibodies.
Collapse
Affiliation(s)
- Carina Gröschel
- Institute of Cellular and Molecular Immunology, University Medical Center Göttingen, Göttingen, Germany.,DZHK (German Center for Cardiovascular Research), partner site Göttingen, Göttingen, Germany
| | - André Sasse
- Institute of Cellular and Molecular Immunology, University Medical Center Göttingen, Göttingen, Germany
| | - Charlotte Röhrborn
- Institute of Cellular and Molecular Immunology, University Medical Center Göttingen, Göttingen, Germany
| | - Sebastian Monecke
- Institute of Cellular and Molecular Immunology, University Medical Center Göttingen, Göttingen, Germany.,DZHK (German Center for Cardiovascular Research), partner site Göttingen, Göttingen, Germany
| | - Michael Didié
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Göttingen, Germany.,Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Göttingen, Germany.,Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany
| | - Leslie Elsner
- Institute of Cellular and Molecular Immunology, University Medical Center Göttingen, Göttingen, Germany
| | - Vanessa Kruse
- Institute of Cellular and Molecular Immunology, University Medical Center Göttingen, Göttingen, Germany
| | - Gertrude Bunt
- Clinical Optical Microscopy, Department of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
| | - Andrew H Lichtman
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Karl Toischer
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Göttingen, Germany.,Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany
| | - Wolfram-Hubertus Zimmermann
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Göttingen, Germany.,Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Göttingen, Germany
| | - Gerd Hasenfuß
- DZHK (German Center for Cardiovascular Research), partner site Göttingen, Göttingen, Germany.,Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany
| | - Ralf Dressel
- Institute of Cellular and Molecular Immunology, University Medical Center Göttingen, Göttingen, Germany. .,DZHK (German Center for Cardiovascular Research), partner site Göttingen, Göttingen, Germany.
| |
Collapse
|
41
|
Proinflammatory Cytokines Are Soluble Mediators Linked with Ventricular Arrhythmias and Contractile Dysfunction in a Rat Model of Metabolic Syndrome. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:7682569. [PMID: 29201273 PMCID: PMC5671748 DOI: 10.1155/2017/7682569] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2017] [Accepted: 08/27/2017] [Indexed: 12/15/2022]
Abstract
Metabolic syndrome (MS) increases cardiovascular risk and is associated with cardiac dysfunction and arrhythmias, although the precise mechanisms are still under study. Chronic inflammation in MS has emerged as a possible cause of adverse cardiac events. Male Wistar rats fed with 30% sucrose in drinking water and standard chow for 25–27 weeks were compared to a control group. The MS group showed increased weight, visceral fat, blood pressure, and serum triglycerides. The most important increases in serum cytokines included IL-1β (7-fold), TNF-α (84%), IL-6 (41%), and leptin (2-fold), the latter also showing increased gene expression in heart tissue (35-fold). Heart function ex vivo in MS group showed a decreased mechanical performance response to isoproterenol challenge (ISO). Importantly, MS hearts under ISO showed nearly twofold the incidence of ventricular fibrillation. Healthy rat cardiomyocytes exposed to MS group serum displayed impaired contractile function and Ca2+ handling during ISO treatment, showing slightly decreased cell shortening and Ca2+ transient amplitude (23%), slower cytosolic calcium removal (17%), and more frequent spontaneous Ca2+ release events (7.5-fold). As spontaneous Ca2+ releases provide a substrate for ventricular arrhythmias, our study highlights the possible role of serum proinflammatory mediators in the development of arrhythmic events during MS.
Collapse
|
42
|
Myocardial Expression of Macrophage Migration Inhibitory Factor in Patients with Heart Failure. J Clin Med 2017; 6:jcm6100095. [PMID: 29027966 PMCID: PMC5664010 DOI: 10.3390/jcm6100095] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 09/26/2017] [Accepted: 10/10/2017] [Indexed: 12/28/2022] Open
Abstract
Macrophage migration inhibitory factor (MIF) is a pleiotropic inflammatory protein and contributes to several different inflammatory and ischemic/hypoxic diseases. MIF was shown to be cardioprotective in experimental myocardial ischemia/reperfusion injury and its expression is regulated by the transcription factor hypoxia-inducible factor (HIF)-1α. We here report on MIF expression in the failing human heart and assess myocardial MIF in different types of cardiomyopathy. Myocardial tissue samples from n = 30 patients were analyzed by quantitative Real-Time PCR. MIF and HIF-1α mRNA expression was analyzed in myocardial samples from patients with ischemic (ICM) and non-ischemic cardiomyopathy (NICM) and from patients after heart transplantation (HTX). MIF expression was elevated in myocardial samples from patients with ICM compared to NICM. Transplanted hearts showed lower MIF levels compared to hearts from patients with ICM. Expression of HIF-1α was analyzed and was shown to be significantly increased in ICM patients compared to patients with NICM. MIF and HIF-1α mRNA is expressed in the human heart. MIF and HIF-1α expression depends on the underlying type of cardiomyopathy. Patients with ICM show increased myocardial MIF and HIF-1α expression.
Collapse
|
43
|
Zhang Y, Bauersachs J, Langer HF. Immune mechanisms in heart failure. Eur J Heart Fail 2017; 19:1379-1389. [DOI: 10.1002/ejhf.942] [Citation(s) in RCA: 115] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2017] [Revised: 05/26/2017] [Accepted: 06/19/2017] [Indexed: 12/15/2022] Open
Affiliation(s)
- Yingying Zhang
- University Hospital, Department of Cardiology and Cardiovascular Medicine; Eberhard Karls University Tuebingen; Tuebingen Germany
- Section for Cardioimmunology; Eberhard Karls University Tuebingen; Tübingen Germany
- Affiliated Hospital of Qingdao University, Department of Cardiology and Cardiovascular Medicine; Qingdao University; Qingdao China
| | - Johann Bauersachs
- Department of Cardiology and Angiology; Hannover Medical School; Hannover Germany
| | - Harald F. Langer
- University Hospital, Department of Cardiology and Cardiovascular Medicine; Eberhard Karls University Tuebingen; Tuebingen Germany
- Section for Cardioimmunology; Eberhard Karls University Tuebingen; Tübingen Germany
| |
Collapse
|
44
|
Ablation of IL-33 gene exacerbate myocardial remodeling in mice with heart failure induced by mechanical stress. Biochem Pharmacol 2017; 138:73-80. [DOI: 10.1016/j.bcp.2017.04.022] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 04/20/2017] [Indexed: 01/19/2023]
|
45
|
Kallikourdis M, Martini E, Carullo P, Sardi C, Roselli G, Greco CM, Vignali D, Riva F, Ormbostad Berre AM, Stølen TO, Fumero A, Faggian G, Di Pasquale E, Elia L, Rumio C, Catalucci D, Papait R, Condorelli G. T cell costimulation blockade blunts pressure overload-induced heart failure. Nat Commun 2017; 8:14680. [PMID: 28262700 PMCID: PMC5343521 DOI: 10.1038/ncomms14680] [Citation(s) in RCA: 136] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 01/23/2017] [Indexed: 02/07/2023] Open
Abstract
Heart failure (HF) is a leading cause of mortality. Inflammation is implicated in HF, yet clinical trials targeting pro-inflammatory cytokines in HF were unsuccessful, possibly due to redundant functions of individual cytokines. Searching for better cardiac inflammation targets, here we link T cells with HF development in a mouse model of pathological cardiac hypertrophy and in human HF patients. T cell costimulation blockade, through FDA-approved rheumatoid arthritis drug abatacept, leads to highly significant delay in progression and decreased severity of cardiac dysfunction in the mouse HF model. The therapeutic effect occurs via inhibition of activation and cardiac infiltration of T cells and macrophages, leading to reduced cardiomyocyte death. Abatacept treatment also induces production of anti-inflammatory cytokine interleukin-10 (IL-10). IL-10-deficient mice are refractive to treatment, while protection could be rescued by transfer of IL-10-sufficient B cells. These results suggest that T cell costimulation blockade might be therapeutically exploited to treat HF. Abatacept is an FDA-approved drug used for treatment of rheumatoid arthritis. Here the authors show that abatacept reduces cardiomyocyte death in a mouse model of heart failure by inhibiting activation and heart infiltration of T cells and macrophages, an effect mediated by IL-10, suggesting a potential therapy for heart failure.
Collapse
Affiliation(s)
- Marinos Kallikourdis
- Adaptive Immunity Laboratory, Humanitas Clinical and Research Center, Via Manzoni 56, Rozzano, 20089 Milan, Italy.,Department of Biomedical Sciences, Humanitas University, Via Manzoni 113, Rozzano, 20089 Milan, Italy
| | - Elisa Martini
- Adaptive Immunity Laboratory, Humanitas Clinical and Research Center, Via Manzoni 56, Rozzano, 20089 Milan, Italy
| | - Pierluigi Carullo
- Department of Cardiovascular Medicine, Humanitas Clinical and Research Center, Via Manzoni 56, Rozzano, 20089 Milan, Italy.,Institute of Genetic and Biomedical Research (IRGB)-UOS of Milan, National Research Council of Italy, Via Manzoni 56, Rozzano, 20089 Milan, Italy
| | - Claudia Sardi
- Adaptive Immunity Laboratory, Humanitas Clinical and Research Center, Via Manzoni 56, Rozzano, 20089 Milan, Italy
| | - Giuliana Roselli
- Adaptive Immunity Laboratory, Humanitas Clinical and Research Center, Via Manzoni 56, Rozzano, 20089 Milan, Italy
| | - Carolina M Greco
- Department of Cardiovascular Medicine, Humanitas Clinical and Research Center, Via Manzoni 56, Rozzano, 20089 Milan, Italy
| | - Debora Vignali
- Adaptive Immunity Laboratory, Humanitas Clinical and Research Center, Via Manzoni 56, Rozzano, 20089 Milan, Italy
| | - Federica Riva
- Department of Veterinary Medicine (DIMEVET), Università degli Studi di Milano, Via Celoria 10, 20133 Milan, Italy
| | - Anne Marie Ormbostad Berre
- KG Jebsen Centre of Medicine, Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Postboks 8905, 7491 Trondheim, Norway
| | - Tomas O Stølen
- KG Jebsen Centre of Medicine, Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Postboks 8905, 7491 Trondheim, Norway.,Norwegian Health Association, Oscars gate 36A, 0258 Oslo, Norway
| | - Andrea Fumero
- Cardiac Surgery, Humanitas Clinical and Research Center, Via Manzoni 56, Rozzano, 20089 Milan, Italy
| | - Giuseppe Faggian
- Department of Cardiac Surgery, University of Verona, 37129 Verona, Italy
| | - Elisa Di Pasquale
- Department of Cardiovascular Medicine, Humanitas Clinical and Research Center, Via Manzoni 56, Rozzano, 20089 Milan, Italy.,Institute of Genetic and Biomedical Research (IRGB)-UOS of Milan, National Research Council of Italy, Via Manzoni 56, Rozzano, 20089 Milan, Italy
| | - Leonardo Elia
- Department of Cardiovascular Medicine, Humanitas Clinical and Research Center, Via Manzoni 56, Rozzano, 20089 Milan, Italy.,Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Cristiano Rumio
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Via Trentacoste 2, 20133 Milan, Italy
| | - Daniele Catalucci
- Institute of Genetic and Biomedical Research (IRGB)-UOS of Milan, National Research Council of Italy, Via Manzoni 56, Rozzano, 20089 Milan, Italy.,Laboratory of Signal Transduction in Cardiac Pathologies, Humanitas Clinical and Research Center, Via Manzoni 56, Rozzano, 20089 Milan, Italy
| | - Roberto Papait
- Department of Cardiovascular Medicine, Humanitas Clinical and Research Center, Via Manzoni 56, Rozzano, 20089 Milan, Italy.,Institute of Genetic and Biomedical Research (IRGB)-UOS of Milan, National Research Council of Italy, Via Manzoni 56, Rozzano, 20089 Milan, Italy
| | - Gianluigi Condorelli
- Department of Biomedical Sciences, Humanitas University, Via Manzoni 113, Rozzano, 20089 Milan, Italy.,Department of Cardiovascular Medicine, Humanitas Clinical and Research Center, Via Manzoni 56, Rozzano, 20089 Milan, Italy
| |
Collapse
|
46
|
Lu J, Liu F, Chen F, Jin Y, Chen H, Liu D, Cui W. Amlodipine and atorvastatin improve ventricular hypertrophy and diastolic function via inhibiting TNF-α, IL-1β and NF-κB inflammatory cytokine networks in elderly spontaneously hypertensive rats. Biomed Pharmacother 2016; 83:330-339. [DOI: 10.1016/j.biopha.2016.06.034] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 06/20/2016] [Accepted: 06/20/2016] [Indexed: 10/21/2022] Open
|
47
|
Kazanski V, Mitrokhin VM, Mladenov MI, Kamkin AG. Cytokine Effects on Mechano-Induced Electrical Activity in Atrial Myocardium. Immunol Invest 2016; 46:22-37. [PMID: 27617892 DOI: 10.1080/08820139.2016.1208220] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The role of cytokines as regulators of stretch-related mechanisms is of special importance since mechano-sensitivity plays an important role in a wide variety of biological processes. Here, we elucidate the influence of cytokine application on mechano-sensitivity and mechano-transduction. The atrial myocardial stretch induces production of interleukin (IL)-2, IL-6, IL-13, IL-17A, and IL-18 with exception of tumor necrosis factor α (TNF-α), IL-1β, and vascular endothelial growth factor B (VEGF-B). Positive ionotropic effect was specific for VEGF-B, negative ionotropic effects were specific for TNF-α, IL-1β, IL-2, IL-6, IL-13, IL-17A and IL-18, while IL-1α doesn't show direct ionotropic effect. The IL-2, IL-6, IL-17A, IL-18, and VEGF-B cause elongation of the APD, in comparison with the reduced APD caused by the IL-13. The TNF-α, IL-1β, and IL-18 influences L-type Ca2+ channels, IL-2 has an inhibitory effect on the fast Na+ channels while IL-17A and VEGF-B were specific for Kir channels. With exception of the IL-1α, IL-2, and VEGF-B, all analyzed cytokines include nitric oxide dependent signaling with resultant combined effects on mechano-gated and Ca2+ channels. The relationships between these pathways and the time-dependence of their activation are of important considerations in the evaluation of cytokine-induced electrical abnormality, specific for cardiac dysfunctions. In general, the discussion presented in this review covers research devoted to counterbalance between different cytokines in the regulation of stretch-induced effects in rat atrial myocardium. ABBREVIATIONS APs: action potentials; APD25: action potential durations to 25% of re-polarization; APD50: action potential durations to 50% of repolarization; APD90: action potential durations to 90% of repolarization; MGCs: mechanically gated channels.
Collapse
Affiliation(s)
- V Kazanski
- a Department of Fundamental and Applied Physiology , Russian National Research Medical University , Moscow , Russia
| | - V M Mitrokhin
- a Department of Fundamental and Applied Physiology , Russian National Research Medical University , Moscow , Russia
| | - M I Mladenov
- a Department of Fundamental and Applied Physiology , Russian National Research Medical University , Moscow , Russia.,b Faculty of Natural Sciences and Mathematics, Institute of Biology , "Ss. Cyril and Methodius" University , Skopje , Macedonia
| | - A G Kamkin
- a Department of Fundamental and Applied Physiology , Russian National Research Medical University , Moscow , Russia
| |
Collapse
|
48
|
Westphal E, Pilowski C, Koch S, Ebelt H, Müller-Werdan U, Werdan K, Loppnow H. Endotoxin-activated cultured neonatal rat cardiomyocytes express functional surface-associated interleukin-1α. ACTA ACUST UNITED AC 2016; 13:25-34. [PMID: 17621543 DOI: 10.1177/0968051907078609] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Interleukin-1 (IL-1) is a potent regulator of cardiovascular proliferation, apoptosis, contraction or production of inflammatory mediators. Thus, we investigated expression and function of IL-1 in cultured neonatal rat heart cells upon endotoxin stimulation. We show that cultured neonatal rat cardiomyocytes expressed IL—1α and IL—1β mRNA. The cells expressed functional cell-associated IL—1 activity and a specific anti-IL—1α-antibody inhibited the activity. Biologically active IL—1α was present at the cell surface of the cardiomyocytes, as indicated in co-culture experiments. Immunohistochemistry showed IL—1α-staining of the neonatal cardiomyocytes. Although the cells also expressed IL—1β mRNA, we did not detect IL—1β in the supernatants of cultured cardiomyocytes by ELISA or in immunohistochemical staining. Furthermore, neonatal and adult rat heart tissues expressed IL—1α mRNA, whereas fetal, but not adult, human cardiac tissues expressed detectable IL—1α mRNA. In contrast, IL-1β mRNA was present in rat and human fetal and adult samples. Furthermore, in patients with dilated or ischemic cardiomyopathy, we measured IL—1β, but not IL—1α, mRNA. These results provide evidence for the presence of functionally active IL—1α on the cell surface of neonatal rat cardiomyocytes and may suggest a differential role of IL—1α in regulation of cellular functions during development, aging and disease in rat and human heart cells.
Collapse
Affiliation(s)
- Elena Westphal
- Martin-Luther-Universität Halle-Wittenberg, Universitätsklinik und Poliklinik für Innere Medizin III, Halle, Germany
| | | | | | | | | | | | | |
Collapse
|
49
|
Jahng JWS, Song E, Sweeney G. Crosstalk between the heart and peripheral organs in heart failure. Exp Mol Med 2016; 48:e217. [PMID: 26964833 PMCID: PMC4892881 DOI: 10.1038/emm.2016.20] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Revised: 12/10/2015] [Accepted: 12/11/2015] [Indexed: 12/31/2022] Open
Abstract
Mediators from peripheral tissues can influence the development and progression of heart failure (HF). For example, in obesity, an altered profile of adipokines secreted from adipose tissue increases the incidence of myocardial infarction (MI). Less appreciated is that heart remodeling releases cardiokines, which can strongly impact various peripheral tissues. Inflammation, and, in particular, activation of the nucleotide-binding oligomerization domain-like receptors with pyrin domain (NLRP3) inflammasome are likely to have a central role in cardiac remodeling and mediating crosstalk with other organs. Activation of the NLRP3 inflammasome in response to cardiac injury induces the production and secretion of the inflammatory cytokines interleukin (IL)-1β and IL-18. In addition to having local effects in the myocardium, these pro-inflammatory cytokines are released into circulation and cause remodeling in the spleen, kidney, skeletal muscle and adipose tissue. The collective effects of various cardiokines on peripheral organs depend on the degree and duration of myocardial injury, with systematic inflammation and peripheral tissue damage observed as HF progresses. In this article, we review mechanisms regulating myocardial inflammation in HF and the role of factors secreted by the heart in communication with peripheral tissues.
Collapse
Affiliation(s)
| | - Erfei Song
- Department of Biology, York University, Toronto, ON, Canada
| | - Gary Sweeney
- Department of Biology, York University, Toronto, ON, Canada
| |
Collapse
|
50
|
Xu X, Si L, Xu J, Yi C, Wang F, Gu W, Zhang Y, Wang X. Asiatic acid inhibits cardiac hypertrophy by blocking interleukin-1β-activated nuclear factor-κB signaling in vitro and in vivo. J Thorac Dis 2015; 7:1787-97. [PMID: 26623102 DOI: 10.3978/j.issn.2072-1439.2015.10.41] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Activated interleukin (IL)-1β signaling pathway is closely associated with pathological cardiac hypertrophy. This study investigated whether asiatic acid (AA) could inhibit IL-1β-related hypertrophic signaling, and thus suppressing the development of cardiac hypertrophy. METHODS Transverse aortic constriction (TAC) induced cardiac hypertrophy in C57BL/6 mice and cultured neonatal cardiac myocytes stimulated with IL-1β were used to evaluate the role of AA in cardiac hypertrophy. The expression of atrial natriuretic peptide (ANP) was evaluated by quantitative polymerase chain reaction (qPCR) and the nuclear factor (NF)-κB binding activity was measured by electrophoretic mobility shift assays (EMSA). RESULTS AA pretreatment significantly attenuated the IL-1β-induced hypertrophic response of cardiomyocytes as reflected by reduction in the cardiomyocyte surface area and the inhibition of ANP mRNA expression. The protective effect of AA on IL-1β-stimulated cardiomyocytes was associated with the reduction of NF-κB binding activity. In addition, AA prevented TAC-induced cardiac hypertrophy in vivo. It was found that AA markedly reduced the excessive expression of IL-1β and ANP, and inhibited the activation of NF-κB in the hypertrophic myocardium. CONCLUSIONS Our data suggest that AA may be a novel therapeutic agent for cardiac hypertrophy. The inhibition of IL-1β-activated NF-κB signaling may be the mechanism through which AA prevents cardiac hypertrophy.
Collapse
Affiliation(s)
- Xiaohan Xu
- 1 Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China ; 2 Institute of Integrated Medicine, Nanjing Medical University, Nanjing 210029, China ; 3 Department of Intensive Care Medicine, The First People's Hospital of Yancheng, Yancheng 224005, China ; 4 Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China ; 5 Department of Cardiology, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing 211100, China
| | - Linjie Si
- 1 Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China ; 2 Institute of Integrated Medicine, Nanjing Medical University, Nanjing 210029, China ; 3 Department of Intensive Care Medicine, The First People's Hospital of Yancheng, Yancheng 224005, China ; 4 Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China ; 5 Department of Cardiology, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing 211100, China
| | - Jing Xu
- 1 Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China ; 2 Institute of Integrated Medicine, Nanjing Medical University, Nanjing 210029, China ; 3 Department of Intensive Care Medicine, The First People's Hospital of Yancheng, Yancheng 224005, China ; 4 Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China ; 5 Department of Cardiology, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing 211100, China
| | - Chenlong Yi
- 1 Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China ; 2 Institute of Integrated Medicine, Nanjing Medical University, Nanjing 210029, China ; 3 Department of Intensive Care Medicine, The First People's Hospital of Yancheng, Yancheng 224005, China ; 4 Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China ; 5 Department of Cardiology, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing 211100, China
| | - Fang Wang
- 1 Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China ; 2 Institute of Integrated Medicine, Nanjing Medical University, Nanjing 210029, China ; 3 Department of Intensive Care Medicine, The First People's Hospital of Yancheng, Yancheng 224005, China ; 4 Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China ; 5 Department of Cardiology, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing 211100, China
| | - Weijuan Gu
- 1 Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China ; 2 Institute of Integrated Medicine, Nanjing Medical University, Nanjing 210029, China ; 3 Department of Intensive Care Medicine, The First People's Hospital of Yancheng, Yancheng 224005, China ; 4 Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China ; 5 Department of Cardiology, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing 211100, China
| | - Yuqing Zhang
- 1 Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China ; 2 Institute of Integrated Medicine, Nanjing Medical University, Nanjing 210029, China ; 3 Department of Intensive Care Medicine, The First People's Hospital of Yancheng, Yancheng 224005, China ; 4 Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China ; 5 Department of Cardiology, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing 211100, China
| | - Xiaowei Wang
- 1 Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China ; 2 Institute of Integrated Medicine, Nanjing Medical University, Nanjing 210029, China ; 3 Department of Intensive Care Medicine, The First People's Hospital of Yancheng, Yancheng 224005, China ; 4 Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China ; 5 Department of Cardiology, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing 211100, China
| |
Collapse
|