1
|
Heusch G, Andreadou I, Bell R, Bertero E, Botker HE, Davidson SM, Downey J, Eaton P, Ferdinandy P, Gersh BJ, Giacca M, Hausenloy DJ, Ibanez B, Krieg T, Maack C, Schulz R, Sellke F, Shah AM, Thiele H, Yellon DM, Di Lisa F. Health position paper and redox perspectives on reactive oxygen species as signals and targets of cardioprotection. Redox Biol 2023; 67:102894. [PMID: 37839355 PMCID: PMC10590874 DOI: 10.1016/j.redox.2023.102894] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 09/04/2023] [Accepted: 09/15/2023] [Indexed: 10/17/2023] Open
Abstract
The present review summarizes the beneficial and detrimental roles of reactive oxygen species in myocardial ischemia/reperfusion injury and cardioprotection. In the first part, the continued need for cardioprotection beyond that by rapid reperfusion of acute myocardial infarction is emphasized. Then, pathomechanisms of myocardial ischemia/reperfusion to the myocardium and the coronary circulation and the different modes of cell death in myocardial infarction are characterized. Different mechanical and pharmacological interventions to protect the ischemic/reperfused myocardium in elective percutaneous coronary interventions and coronary artery bypass grafting, in acute myocardial infarction and in cardiotoxicity from cancer therapy are detailed. The second part keeps the focus on ROS providing a comprehensive overview of molecular and cellular mechanisms involved in ischemia/reperfusion injury. Starting from mitochondria as the main sources and targets of ROS in ischemic/reperfused myocardium, a complex network of cellular and extracellular processes is discussed, including relationships with Ca2+ homeostasis, thiol group redox balance, hydrogen sulfide modulation, cross-talk with NAPDH oxidases, exosomes, cytokines and growth factors. While mechanistic insights are needed to improve our current therapeutic approaches, advancements in knowledge of ROS-mediated processes indicate that detrimental facets of oxidative stress are opposed by ROS requirement for physiological and protective reactions. This inevitable contrast is likely to underlie unsuccessful clinical trials and limits the development of novel cardioprotective interventions simply based upon ROS removal.
Collapse
Affiliation(s)
- Gerd Heusch
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Duisburg-Essen, Essen, Germany.
| | - Ioanna Andreadou
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Robert Bell
- The Hatter Cardiovascular Institute, University College London, London, United Kingdom
| | - Edoardo Bertero
- Chair of Cardiovascular Disease, Department of Internal Medicine and Specialties, University of Genova, Genova, Italy
| | - Hans-Erik Botker
- Department of Cardiology, Institute for Clinical Medicine, Aarhus University, Aarhus N, Denmark
| | - Sean M Davidson
- The Hatter Cardiovascular Institute, University College London, London, United Kingdom
| | - James Downey
- Department of Physiology, University of South Alabama, Mobile, AL, USA
| | - Philip Eaton
- William Harvey Research Institute, Queen Mary University of London, Heart Centre, Charterhouse Square, London, United Kingdom
| | - Peter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary; Pharmahungary Group, Szeged, Hungary
| | - Bernard J Gersh
- Department of Cardiovascular Medicine, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Mauro Giacca
- School of Cardiovascular and Metabolic Medicine & Sciences, King's College, London, United Kingdom
| | - Derek J Hausenloy
- The Hatter Cardiovascular Institute, University College London, London, United Kingdom; Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, National Heart Research Institute Singapore, National Heart Centre, Yong Loo Lin School of Medicine, National University Singapore, Singapore
| | - Borja Ibanez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), IIS-Fundación Jiménez Díaz University Hospital, and CIBERCV, Madrid, Spain
| | - Thomas Krieg
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Christoph Maack
- Department of Translational Research, Comprehensive Heart Failure Center, University Clinic Würzburg, Würzburg, Germany
| | - Rainer Schulz
- Institute for Physiology, Justus-Liebig -Universität, Giessen, Germany
| | - Frank Sellke
- Division of Cardiothoracic Surgery, Alpert Medical School of Brown University and Rhode Island Hospital, Providence, RI, USA
| | - Ajay M Shah
- King's College London British Heart Foundation Centre of Excellence, London, United Kingdom
| | - Holger Thiele
- Heart Center Leipzig at University of Leipzig and Leipzig Heart Science, Leipzig, Germany
| | - Derek M Yellon
- The Hatter Cardiovascular Institute, University College London, London, United Kingdom
| | - Fabio Di Lisa
- Dipartimento di Scienze Biomediche, Università degli studi di Padova, Padova, Italy.
| |
Collapse
|
2
|
Sun L, Ji D, Zhi F, Fang Y, Zhu Z, Ni T, Zhu Q, Bao J. MiR-494-3p Upregulation Exacerbates Cerebral Ischemia Injury by Targeting Bhlhe40. Yonsei Med J 2022; 63:389-398. [PMID: 35352891 PMCID: PMC8965425 DOI: 10.3349/ymj.2022.63.4.389] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 11/19/2021] [Accepted: 12/21/2021] [Indexed: 11/27/2022] Open
Abstract
PURPOSE Cerebral ischemia is related to insufficient blood supply and is characterized by abnormal reactive oxygen species (ROS) production and cell apoptosis. Previous studies have revealed a key role for basic helix-loop-helix family member e40 (Bhlhe40) in oxidative stress and cell apoptosis. This study aimed to investigate the roles of miR-494-3p in cerebral ischemia/reperfusion (I/R) injury. MATERIALS AND METHODS A mouse middle cerebral artery occlusion (MCAO/R) model was established to mimic cerebral ischemia in vivo. Brain infarct area was assessed using triphenyl tetrazolium chloride staining. Oxygen-glucose deprivation/reoxygenation (OGD/R) operation was adopted to mimic neuronal injury in vitro. Cell apoptosis was analyzed by flow cytometry. The relationship between miR-494-3p and Bhlhe40 was validated by luciferase reporter and RNA immunoprecipitation assays. RESULTS Bhlhe40 expression was downregulated both in MCAO/R animal models and OGD/R-induced SH-SY5Y cells. Bhlhe40 overexpression inhibited cell apoptosis and reduced ROS production in SH-SY5Y cells after OGD/R treatment. MiR-494-3p was verified to bind to Bhlhe40 and negatively regulate Bhlhe40 expression. Additionally, cell apoptosis and ROS production in OGD/R-treated SH-SY5Y cells were accelerated by miR-494-3p overexpression. Rescue experiments suggested that Bhlhe40 could reverse the effects of miR-494-3p overexpression on ROS production and cell apoptosis. CONCLUSION MiR-494-3p exacerbates brain injury and neuronal injury by regulating Bhlhe40 after I/R.
Collapse
Affiliation(s)
- Lingjiang Sun
- Department of Critical Care Medicine, Wuxi Second People's Hospital, Wuxi, Jiangsu, China
| | - Dandan Ji
- Department of Critical Care Medicine, Wuxi Second People's Hospital, Wuxi, Jiangsu, China
| | - Feng Zhi
- Department of Critical Care Medicine, Wuxi Second People's Hospital, Wuxi, Jiangsu, China
| | - Yu Fang
- Department of Critical Care Medicine, Wuxi Second People's Hospital, Wuxi, Jiangsu, China
| | - Zigang Zhu
- Department of Critical Care Medicine, Wuxi Second People's Hospital, Wuxi, Jiangsu, China
| | - Tong Ni
- Department of Critical Care Medicine, Wuxi Second People's Hospital, Wuxi, Jiangsu, China
| | - Qin Zhu
- Department of Stomatology, Taixing Third People's Hospital, Taizhou, Jiangsu, China.
| | - Jie Bao
- Department of Critical Care Medicine, Wuxi Second People's Hospital, Wuxi, Jiangsu, China.
| |
Collapse
|
3
|
Lee SY, Hsin LW, Su MJ, ChangChien CC, Ku HC. A novel isoquinoline derivative exhibits anti-inflammatory properties and improves the outcomes of endotoxemia. Pharmacol Rep 2019; 71:1281-1288. [PMID: 31683199 DOI: 10.1016/j.pharep.2019.06.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 05/27/2019] [Accepted: 06/28/2019] [Indexed: 12/25/2022]
Abstract
BACKGROUND Sepsis initiates an inflammatory response that causes widespread injury, and candidates for related myocardial depressant factors include cytokines and nitric oxide (NO). Nuclear factor kappa-B (NF-κB) stimulated by toll-like receptor 4 activation in sepsis mediates the transcription of multiple proinflammatory genes. These inflammatory mediators can cause myocardial dysfunction, which may deteriorate sepsis outcomes. To address this risk, we investigated the potential beneficial effects of a novel isoquinolines derivative, CYY054c, in LPS-induced inflammatory response leading to endotoxemia. METHODS The effects of CYY054c on cytokine and inflammatory-related protein production were evaluated in lipopolysaccharide (LPS)-stimulated macrophages. To determine whether CYY054c alleviates inflammatory storm-induced myocardial dysfunction in vivo, LPS was injected in rats, and cardiac function was measured by a pressure-volume loop. RESULTS CYY054c inhibited LPS-induced NF-κB expression in macrophages and reduced the release of tumor necrosis factor-alpha (TNF-α), interleukin-1β (IL-1β), and interleukin-6 (IL-6), as well as the expression of inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2). In the animal studies, CYY054c alleviated LPS-upregulated plasma TNF-α, IL-1β, IL-6, and NO concentrations, as well as cardiac monocyte chemotactic protein-1, iNOS, and COX-2 expression in rats, contributing to the improvement of cardiac function during endotoxemia. CONCLUSIONS The reduction of NF-κB-mediated inflammatory mediators and the maintenance of hemodynamic performance by CYY054c improved the outcomes during endotoxemia. CYY054c may be a potential therapeutic agent for sepsis.
Collapse
Affiliation(s)
- Shih-Yi Lee
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Taitung MacKay Memorial Hospital, Taiwan; Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, MacKay Memorial Hospital, Taiwan; Mackay Junior College of Medicine, Nursing, and Management, Taipei, Taiwan
| | - Ling-Wei Hsin
- School of Pharmacy, Molecular Probes Development Core, Molecular Imaging Center, Taipei, Taiwan; Center for Innovative Therapeutics Discovery, National Taiwan University, Taipei, Taiwan
| | - Ming-Jai Su
- Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ching-Chia ChangChien
- Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Hui-Chun Ku
- Department of Life Science, Fu Jen Catholic University, New Taipei City, Taiwan.
| |
Collapse
|
4
|
Urmaliya V, Franchelli G. A multidimensional sight on cardiac failure: uncovered from structural to molecular level. Heart Fail Rev 2018; 22:357-370. [PMID: 28474325 DOI: 10.1007/s10741-017-9610-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Heart failure is one of the leading causes of death, with high mortality rate within 5 years after diagnosis. Treatment and prognosis options for heart failure primarily targeted on hemodynamic and neurohumoral components that drive progressive deterioration of the heart. However, given the multifactorial background that eventually leads to the "phenotype" named heart failure, better insight into the various components may lead to personalized treatment opportunities. Indeed, currently used criteria to diagnose and/or classify heart failure are possibly too focused on phenotypic improvement rather than the molecular driver of the disease and could therefore be further refined by integrating the leap of molecular and cellular knowledge. The ambiguity of the ejection fraction-based classification criteria became evident with development of advanced molecular techniques and the dawn of omics disciplines which introduced the idea that disease is caused by a myriad of cellular and molecular processes rather than a single event or pathway. The fact that different signaling pathways may underlie similar clinical manifestations calls for a more holistic study of heart failure. In this context, the systems biology approach can offer a better understanding of how different components of a system are altered during disease and how they interact with each other, potentially leading to improved diagnosis and classification of this condition. This review is aimed at addressing heart failure through a multilayer approach that covers individually some of the anatomical, morphological, functional, and tissue aspects, with focus on cellular and subcellular features as an alternative insight into new therapeutic opportunities.
Collapse
Affiliation(s)
- Vijay Urmaliya
- Discovery Sciences, Janssen Research & Development, Beerse, Belgium.
| | | |
Collapse
|
5
|
Song HK, Kim J, Lee JS, Nho KJ, Jeong HC, Kim J, Ahn Y, Park WJ, Kim DH. Pik3ip1 modulates cardiac hypertrophy by inhibiting PI3K pathway. PLoS One 2015; 10:e0122251. [PMID: 25826393 PMCID: PMC4380398 DOI: 10.1371/journal.pone.0122251] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 02/10/2015] [Indexed: 01/19/2023] Open
Abstract
Cardiac hypertrophy is an adaptive response to various physiological and pathological stimuli. Phosphoinositide-3 kinase (PI3K) is a highly conserved lipid kinase involved in physiological cardiac hypertrophy (PHH). PI3K interacting protein1 (Pik3ip1) shares homology with the p85 regulatory subunit of PI3K and is known to interact with the p110 catalytic subunit of PI3K, leading to attenuation of PI3K activity in liver and immune cells. However, the role of Pik3ip1 in the heart remains unknown. In the present study, the effects of Pik3ip1 on cardiac hypertrophy were examined. We found that the expression level of Pik3ip1 was markedly higher in cardiomyocytes than in fibroblasts. The interaction of Pik3ip1 with the p110a subunit of PI3K in the heart was identified by immunoprecipitation using neonatal rat cardiomyocytes (NRCM). Approximately 35% knockdown of Pik3ip1 was sufficient to induce myocardial hypertrophy. Pik3ip1 deficiency was shown to lead to activation of PI3K/protein kinase B (AKT)/ mammalian target of rapamycin (mTOR) signaling pathway, increasing protein synthesis and cell size. However, adenovirus-mediated overexpression of Pik3ip1 attenuated PI3K-mediated cardiac hypertrophy. Pik3ip1 was upregulated by PHH due to swimming training, but not by pathological cardiac hypertrophy (PAH) due to pressure-overload, suggesting that Pik3ip1 plays a compensatory negative role for PHH. Collectively, our results elucidate the mechanisms for the roles of Pik3ip1 in PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Hong Ki Song
- School of Life Sciences and Systems Biology Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea
| | - Jiyeon Kim
- School of Life Sciences and Systems Biology Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea
| | - Jong Sub Lee
- School of Life Sciences and Systems Biology Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea
| | - Kyoung Jin Nho
- School of Life Sciences and Systems Biology Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea
| | - Hae Chang Jeong
- Department of Cardiology, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Jihwa Kim
- School of Life Sciences and Systems Biology Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea
| | - Youngkeun Ahn
- Department of Cardiology, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Woo Jin Park
- School of Life Sciences and Systems Biology Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea
| | - Do Han Kim
- School of Life Sciences and Systems Biology Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea
- * E-mail:
| |
Collapse
|
6
|
ROS and endothelial nitric oxide synthase (eNOS)-dependent trafficking of angiotensin II type 2 receptor begets neuronal NOS in cardiac myocytes. Basic Res Cardiol 2015; 110:21. [PMID: 25804308 DOI: 10.1007/s00395-015-0477-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Revised: 03/09/2015] [Accepted: 03/11/2015] [Indexed: 12/14/2022]
Abstract
Angiotensin II (Ang II), a potent precursor of hypertrophy and heart failure, upregulates neuronal nitric oxide synthase (nNOS or NOS1) in the myocardium. Here, we investigate the involvement of type 1 and 2 angiotensin receptors (AT1R and AT2R) and molecular mechanisms mediating Ang II-upregulation of nNOS. Our results showed that pre-treatment of left ventricular (LV) myocytes with antagonists of AT1R or AT2R (losartan, PD123319) and ROS scavengers (apocynin, tiron or PEG-catalase) blocked Ang II-upregulation of nNOS. Surface biotinylation or immunocytochemistry experiments demonstrated that AT1R expression in plasma membrane was progressively decreased (internalization), whereas AT2R was increased (membrane trafficking) by Ang II. Inhibition of AT1R or ROS scavengers prevented Ang II-induced translocation of AT2R to plasma membrane, suggesting an alignment of AT1R-ROS-AT2R. Furthermore, Ang II increased eNOS-Ser(1177) but decreased eNOS-Thr(495), indicating concomitant activation of eNOS. Intriguingly, ROS scavengers but not AT2R antagonist prevented Ang II-activation of eNOS. NOS inhibitor (L-NG-Nitroarginine Methyl Ester, L-NAME) or eNOS gene deletion (eNOS(-/-)) abolished Ang II-induced membrane trafficking of AT2R, nNOS protein expression and activity. Mechanistically, S-nitrosation of AT2R was increased by sodium nitroprusside (SNP), a NO donor. Site-specific mutagenesis analysis reveals that C-terminal cysteine 349 in AT2R is essential in AT2R translocation to plasma membrane. Taken together, we demonstrate, for the first time, that Ang II upregulates nNOS protein expression and activity via AT1R/ROS/eNOS-dependent S-nitrosation and membrane translocation of AT2R. Our results suggest a novel crosstalk between AT1R and AT2R in regulating nNOS via eNOS in the myocardium under pathogenic stimuli.
Collapse
|
7
|
Effects of pseudo-phosphorylated rat cardiac troponin T are differently modulated by α- and β-myosin heavy chain isoforms. Basic Res Cardiol 2014; 109:442. [PMID: 25301196 DOI: 10.1007/s00395-014-0442-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Revised: 08/31/2014] [Accepted: 09/18/2014] [Indexed: 10/24/2022]
Abstract
Interplay between the protein kinase C (PKC)-mediated phosphorylation of troponin T (TnT)- and myosin heavy chain (MHC)-mediated effects on thin filaments takes on a new significance because: (1) there is significant interaction between the TnT- and MHC-mediated effects on cardiac thin filaments; (2) although the phosphorylation of TnT by PKC isoforms is common to both human and rodent hearts, human hearts predominantly express β-MHC while rodent hearts predominantly express α-MHC. Therefore, we tested how α- and β-MHC isoforms differently affected the functional effects of phosphorylated TnT. Contractile measurements were made on cardiac muscle fibers from normal rats (α-MHC) and propylthiouracil-treated rats (β-MHC), reconstituted with the recombinant phosphomimetic-TnT (T204E; threonine 204 replaced by glutamate). Ca2+ -activated maximal tension decreased differently in α-MHC + T204E (~68%) and β-MHC + T204E (~35%). However, myofilament Ca2+ sensitivity decreased similarly in α-MHC + T204E and β-MHC + T204E, demonstrating that a decrease in Ca2+ sensitivity alone cannot explain the greater attenuation of tension in α-MHC + T204E. Interestingly, dynamic contractile parameters (rates of tension redevelopment, crossbridge (XB) recruitment dynamics, XB distortion dynamics, and XB detachment kinetics) decreased only in α-MHC + T204E. Thus, the transition of thin filaments from the blocked- to closed-state was attenuated in α-MHC + T204E and β-MHC + T204E, but the closed- to open-state transition was attenuated only in α-MHC + T204E. Our study demonstrates that the effects of phosphorylated TnT and MHC isoforms interact to bring about different functional states of cardiac thin filaments.
Collapse
|
8
|
Redox balance and cardioprotection. Basic Res Cardiol 2013; 108:392. [DOI: 10.1007/s00395-013-0392-7] [Citation(s) in RCA: 101] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2013] [Revised: 09/24/2013] [Accepted: 10/14/2013] [Indexed: 12/11/2022]
|
9
|
Balogh A, Santer D, Pásztor ET, Tóth A, Czuriga D, Podesser BK, Trescher K, Jaquet K, Erdodi F, Edes I, Papp Z. Myofilament protein carbonylation contributes to the contractile dysfunction in the infarcted LV region of mouse hearts. Cardiovasc Res 2013; 101:108-19. [PMID: 24127233 DOI: 10.1093/cvr/cvt236] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
AIMS The region-specific mechanical function of left ventricular (LV) murine cardiomyocytes and the role of phosphorylation and oxidative modifications of myofilament proteins were investigated in the process of post-myocardial infarction (MI) remodelling 10 weeks after ligation of the left anterior descending (LAD) coronary artery. METHODS AND RESULTS Permeabilized murine cardiomyocytes from the remaining anterior and a remote non-infarcted inferior LV area were compared with those of non-infarcted age-matched controls. Myofilament phosphorylation, sulfhydryl (SH) oxidation, and carbonylation were also assayed. Ca(2+) sensitivity of force production was significantly lower in the anterior wall (pCa50: 5.81 ± 0.03, means ± SEM, at 2.3 µm sarcomere length) than that in the controls (pCa50: 5.91 ± 0.02) or in the MI inferior area (pCa50: 5.88 ± 0.02). The level of troponin I phosphorylation was lower and that of myofilament protein SH oxidation was higher in the anterior location relative to controls, but these changes did not explain the differences in Ca(2+) sensitivities. On the other hand, significantly higher carbonylation levels, [e.g. in myosin heavy chain (MHC) and actin] were observed in the MI anterior wall [carbonylation index (CI), CIMHC: 2.06 ± 0.46, CIactin: 1.46 ± 0.18] than in the controls (CI: 1). In vitro Fenton-based myofilament carbonylation in the control cardiomyocytes also decreased the Ca(2+) sensitivity of force production irrespective of the phosphorylation status of the myofilaments. Furthermore, the Ca(2+) sensitivity correlated strongly with myofilament carbonylation levels in all investigated samples. CONCLUSION Post-MI myocardial remodelling involves increased myofibrillar protein carbonylation and decreased Ca(2+) sensitivity of force production, leading potentially to contractile dysfunction in the remaining cardiomyocytes of the infarcted area.
Collapse
Affiliation(s)
- Agnes Balogh
- Division of Clinical Physiology, Research Center for Molecular Medicine, Institute of Cardiology, Faculty of Medicine, Medical and Health Science Center, University of Debrecen, Móricz Zs. krt. 22, Debrecen H-4032, Hungary
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Ionizing radiation regulates cardiac Ca handling via increased ROS and activated CaMKII. Basic Res Cardiol 2013; 108:385. [PMID: 24068185 PMCID: PMC3898380 DOI: 10.1007/s00395-013-0385-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Revised: 08/29/2013] [Accepted: 08/30/2013] [Indexed: 11/08/2022]
Abstract
Ionizing radiation (IR) is an integral part of modern multimodal anti-cancer therapies. IR involves the formation of reactive oxygen species (ROS) in targeted tissues. This is associated with subsequent cardiac dysfunction when applied during chest radiotherapy. We hypothesized that IR (i.e., ROS)-dependently impaired cardiac myocytes’ Ca handling might contribute to IR-dependent cardiocellular dysfunction. Isolated ventricular mouse myocytes and the mediastinal area of anaesthetized mice (that included the heart) were exposed to graded doses of irradiation (sham 4 and 20 Gy) and investigated acutely (after ~1 h) as well as chronically (after ~1 week). IR induced a dose-dependent effect on myocytes’ systolic function with acutely increased, but chronically decreased Ca transient amplitudes, which was associated with an acutely unaltered but chronically decreased sarcoplasmic reticulum (SR) Ca load. Likewise, in vivo echocardiography of anaesthetized mice revealed acutely enhanced left ventricular contractility (strain analysis) that declined after 1 week. Irradiated myocytes showed persistently increased diastolic SR Ca leakage, which was acutely compensated by an increase in SR Ca reuptake. This was reversed in the chronic setting in the face of slowed relaxation kinetics. As underlying cause, acutely increased ROS levels were identified to activate Ca/calmodulin-dependent protein kinase II (CaMKII). Accordingly, CaMKII-, but not PKA-dependent phosphorylation sites of the SR Ca release channels (RyR2, at Ser-2814) and phospholamban (at Thr-17) were found to be hyperphosphorylated following IR. Conversely, ROS-scavenging as well as CaMKII-inhibition significantly attenuated CaMKII-activation, disturbed Ca handling, and subsequent cellular dysfunction upon irradiation. Targeted cardiac irradiation induces a biphasic effect on cardiac myocytes Ca handling that is associated with chronic cardiocellular dysfunction. This appears to be mediated by increased oxidative stress and persistently activated CaMKII. Our findings suggest impaired cardiac myocytes Ca handling as a so far unknown mediator of IR-dependent cardiac damage that might be of relevance for radiation-induced cardiac dysfunction.
Collapse
|
11
|
Myocardial energetics in heart failure. Basic Res Cardiol 2013; 108:358. [PMID: 23740216 DOI: 10.1007/s00395-013-0358-9] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2013] [Revised: 04/24/2013] [Accepted: 05/09/2013] [Indexed: 12/12/2022]
Abstract
It has become common sense that the failing heart is an "engine out of fuel". However, undisputable evidence that, indeed, the failing heart is limited by insufficient ATP supply is currently lacking. Over the last couple of years, an increasingly complex picture of mechanisms evolved that suggests that potentially metabolic intermediates and redox state could play the more dominant roles for signaling that eventually results in left ventricular remodeling and contractile dysfunction. In the pathophysiology of heart failure, mitochondria emerge in the crossfire of defective excitation-contraction coupling and increased energetic demand, which may provoke oxidative stress as an important upstream mediator of cardiac remodeling and cell death. Thus, future therapies may be guided towards restoring defective ion homeostasis and mitochondrial redox shifts rather than aiming solely at improving the generation of ATP.
Collapse
|
12
|
Neef S, Maier LS. Novel aspects of excitation-contraction coupling in heart failure. Basic Res Cardiol 2013; 108:360. [PMID: 23740218 DOI: 10.1007/s00395-013-0360-2] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Revised: 03/18/2013] [Accepted: 03/28/2013] [Indexed: 12/19/2022]
Abstract
Excitation-contraction coupling is the process by which electrical activation is translated into contraction of a cardiac myocyte and thus the heart. In heart failure, expression, phosphorylation, and function of several intracellular proteins that are involved in excitation-contraction coupling are altered. The present review article summarizes central principles and highlights novel aspects of alterations in heart failure, focusing especially on recent findings regarding altered sarcoplasmic reticulum Ca2+ -leak and late Na+ -current without being able to cover all changes in full detail. These two pathomechanisms seem to play interesting roles with respect to systolic and diastolic dysfunction and may also be important for cardiac arrhythmias. Furthermore, the article outlines the translation of these novel findings into potential therapeutic approaches.
Collapse
Affiliation(s)
- Stefan Neef
- Abt. Kardiologie und Pneumologie/Herzzentrum, Georg-August-Universität Göttingen, Robert-Koch-Str. 40, 37075, Göttingen, Germany
| | | |
Collapse
|
13
|
Hofmann U, Frantz S. How can we cure a heart "in flame"? A translational view on inflammation in heart failure. Basic Res Cardiol 2013; 108:356. [PMID: 23740214 PMCID: PMC3709073 DOI: 10.1007/s00395-013-0356-y] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Revised: 04/03/2013] [Accepted: 04/08/2013] [Indexed: 01/12/2023]
Abstract
The prevalence of chronic heart failure is still increasing making it a major health issue in the 21st century. Tremendous evidence has emerged over the past decades that heart failure is associated with a wide array of mechanisms subsumed under the term “inflammation”. Based on the great success of immuno-suppressive treatments in auto-immunity and transplantation, clinical trials were launched targeting inflammatory mediators in patients with chronic heart failure. However, they widely lacked positive outcomes. The failure of the initial study program directed against tumor necrosis factor-α led to the search for alternative therapeutic targets involving a broader spectrum of mechanisms besides cytokines. We here provide an overview of the current knowledge on immune activation in chronic heart failure of different etiologies, summarize clinical studies in the field, address unresolved key questions, and highlight some promising novel therapeutic targets for clinical trials from a translational basic science and clinical perspective.
Collapse
Affiliation(s)
- Ulrich Hofmann
- Department of Internal Medicine I, University Hospital Würzburg, Oberdürrbacher Straße 6, 97080, Würzburg, Germany.
| | | |
Collapse
|
14
|
Novel aspects of ROS signalling in heart failure. Basic Res Cardiol 2013; 108:359. [PMID: 23740217 DOI: 10.1007/s00395-013-0359-8] [Citation(s) in RCA: 119] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Revised: 05/03/2013] [Accepted: 05/05/2013] [Indexed: 12/20/2022]
Abstract
Heart failure and many of the conditions that predispose to heart failure are associated with oxidative stress. This is considered to be important in the pathophysiology of the condition but clinical trials of antioxidant approaches to prevent cardiovascular morbidity and mortality have been unsuccessful. Part of the reason for this may be the failure to appreciate the complexity of the effects of reactive oxygen species. At one extreme, excessive oxidative stress damages membranes, proteins and DNA but lower levels of reactive oxygen species may exert much more subtle and specific regulatory effects (termed redox signalling), even on physiological signalling pathways. In this article, we review our current understanding of the roles of such redox signalling pathways in the pathophysiology of heart failure, including effects on cardiomyocyte hypertrophy signalling, excitation-contraction coupling, arrhythmia, cell viability and energetics. Reactive oxygen species generated by NADPH oxidase proteins appear to be especially important in redox signalling. The delineation of specific redox-sensitive pathways and mechanisms that contribute to different components of the failing heart phenotype may facilitate the development of newer targeted therapies as opposed to the failed general antioxidant approaches of the past.
Collapse
|
15
|
Perrino C, Schiattarella GG, Sannino A, Pironti G, Petretta MP, Cannavo A, Gargiulo G, Ilardi F, Magliulo F, Franzone A, Carotenuto G, Serino F, Altobelli GG, Cimini V, Cuocolo A, Lombardi A, Goglia F, Indolfi C, Trimarco B, Esposito G. Genetic deletion of uncoupling protein 3 exaggerates apoptotic cell death in the ischemic heart leading to heart failure. J Am Heart Assoc 2013; 2:e000086. [PMID: 23688674 PMCID: PMC3698767 DOI: 10.1161/jaha.113.000086] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
BACKGROUND Uncoupling protein 3 (ucp3) is a member of the mitochondrial anion carrier superfamily of proteins uncoupling mitochondrial respiration. In this study, we investigated the effects of ucp3 genetic deletion on mitochondrial function and cell survival under low oxygen conditions in vitro and in vivo. METHODS AND RESULTS To test the effects of ucp3 deletion in vitro, murine embryonic fibroblasts and adult cardiomyocytes were isolated from wild-type (WT, n=67) and ucp3 knockout mice (ucp3(-/-), n=70). To test the effects of ucp3 genetic deletion in vivo, myocardial infarction (MI) was induced by permanent coronary artery ligation in WT and ucp3(-/-) mice. Compared with WT, ucp3(-/-) murine embryonic fibroblasts and cardiomyocytes exhibited mitochondrial dysfunction and increased mitochondrial reactive oxygen species generation and apoptotic cell death under hypoxic conditions in vitro (terminal deoxynucleotidyl transferase-dUTP nick end labeling-positive nuclei: WT hypoxia, 70.3 ± 1.2%; ucp3(-/-) hypoxia, 85.3 ± 0.9%; P<0.05). After MI, despite similar areas at risk in the 2 groups, ucp3(-/-) hearts demonstrated a significantly larger infarct size compared with WT (infarct area/area at risk: WT, 48.2 ± 3.7%; ucp3(-/-), 65.0 ± 2.9%; P<0.05). Eight weeks after MI, cardiac function was significantly decreased in ucp3(-/-) mice compared with WT (fractional shortening: WT MI, 42.7 ± 3.1%; ucp3(-/-) MI, 24.4 ± 2.9; P<0.05), and this was associated with heightened apoptotic cell death (terminal deoxynucleotidyl transferase-dUTP nick end labeling-positive nuclei: WT MI, 0.7 ± 0.04%; ucp3(-/-) MI, 1.1 ± 0.09%, P<0.05). CONCLUSIONS Our data indicate that ucp3 levels regulate reactive oxygen species levels and cell survival during hypoxia, modulating infarct size in the ischemic heart.
Collapse
Affiliation(s)
- Cinzia Perrino
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy (C.P., G.G.S., A.S., G.P., M.P.P., A.C., G.G., F.I., F.M., A.F., G.C., F.S., G.G.A., V.C., A.C., B.T., G.E.)
| | - Gabriele G. Schiattarella
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy (C.P., G.G.S., A.S., G.P., M.P.P., A.C., G.G., F.I., F.M., A.F., G.C., F.S., G.G.A., V.C., A.C., B.T., G.E.)
| | - Anna Sannino
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy (C.P., G.G.S., A.S., G.P., M.P.P., A.C., G.G., F.I., F.M., A.F., G.C., F.S., G.G.A., V.C., A.C., B.T., G.E.)
| | - Gianluigi Pironti
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy (C.P., G.G.S., A.S., G.P., M.P.P., A.C., G.G., F.I., F.M., A.F., G.C., F.S., G.G.A., V.C., A.C., B.T., G.E.)
| | - Maria Piera Petretta
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy (C.P., G.G.S., A.S., G.P., M.P.P., A.C., G.G., F.I., F.M., A.F., G.C., F.S., G.G.A., V.C., A.C., B.T., G.E.)
| | - Alessandro Cannavo
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy (C.P., G.G.S., A.S., G.P., M.P.P., A.C., G.G., F.I., F.M., A.F., G.C., F.S., G.G.A., V.C., A.C., B.T., G.E.)
| | - Giuseppe Gargiulo
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy (C.P., G.G.S., A.S., G.P., M.P.P., A.C., G.G., F.I., F.M., A.F., G.C., F.S., G.G.A., V.C., A.C., B.T., G.E.)
| | - Federica Ilardi
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy (C.P., G.G.S., A.S., G.P., M.P.P., A.C., G.G., F.I., F.M., A.F., G.C., F.S., G.G.A., V.C., A.C., B.T., G.E.)
| | - Fabio Magliulo
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy (C.P., G.G.S., A.S., G.P., M.P.P., A.C., G.G., F.I., F.M., A.F., G.C., F.S., G.G.A., V.C., A.C., B.T., G.E.)
| | - Anna Franzone
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy (C.P., G.G.S., A.S., G.P., M.P.P., A.C., G.G., F.I., F.M., A.F., G.C., F.S., G.G.A., V.C., A.C., B.T., G.E.)
| | - Giuseppe Carotenuto
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy (C.P., G.G.S., A.S., G.P., M.P.P., A.C., G.G., F.I., F.M., A.F., G.C., F.S., G.G.A., V.C., A.C., B.T., G.E.)
| | - Federica Serino
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy (C.P., G.G.S., A.S., G.P., M.P.P., A.C., G.G., F.I., F.M., A.F., G.C., F.S., G.G.A., V.C., A.C., B.T., G.E.)
| | - Giovanna G. Altobelli
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy (C.P., G.G.S., A.S., G.P., M.P.P., A.C., G.G., F.I., F.M., A.F., G.C., F.S., G.G.A., V.C., A.C., B.T., G.E.)
| | - Vincenzo Cimini
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy (C.P., G.G.S., A.S., G.P., M.P.P., A.C., G.G., F.I., F.M., A.F., G.C., F.S., G.G.A., V.C., A.C., B.T., G.E.)
| | - Alberto Cuocolo
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy (C.P., G.G.S., A.S., G.P., M.P.P., A.C., G.G., F.I., F.M., A.F., G.C., F.S., G.G.A., V.C., A.C., B.T., G.E.)
| | - Assunta Lombardi
- Department of Biology, Federico II University, Naples, Italy (A.L.)
| | - Fernando Goglia
- Department of Biology Sciences, Geology and Environment, Sannio University, Benevento, Italy (F.G.)
| | - Ciro Indolfi
- Department of Cardiology, Magna Graecia University, Catanzaro, Italy (C.I.)
| | - Bruno Trimarco
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy (C.P., G.G.S., A.S., G.P., M.P.P., A.C., G.G., F.I., F.M., A.F., G.C., F.S., G.G.A., V.C., A.C., B.T., G.E.)
| | - Giovanni Esposito
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy (C.P., G.G.S., A.S., G.P., M.P.P., A.C., G.G., F.I., F.M., A.F., G.C., F.S., G.G.A., V.C., A.C., B.T., G.E.)
- Correspondence to: Giovanni Esposito, MD, PhD, or Cinzia Perrino, MD, PhD, Division of Cardiology, Federico II University, Via Pansini 5, 80131 Naples, Italy. E‐mail: ,
| |
Collapse
|
16
|
Cialdella P, Pedicino D, Gustapane M, Santangeli P, Narducci ML, Pelargonio G, Basile E, Giglio AF, Pazzano V, Vitulano N, Bellocci F. Inflammatory markers in heart failure. J Cardiovasc Med (Hagerstown) 2013; 14:342-50. [DOI: 10.2459/jcm.0b013e328349afb6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
17
|
Manning JR, Perkins SO, Sinclair EA, Gao X, Zhang Y, Newman G, Pyle WG, Schultz JEJ. Low molecular weight fibroblast growth factor-2 signals via protein kinase C and myofibrillar proteins to protect against postischemic cardiac dysfunction. Am J Physiol Heart Circ Physiol 2013; 304:H1382-96. [PMID: 23479264 DOI: 10.1152/ajpheart.00613.2012] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Among its many biological roles, fibroblast growth factor-2 (FGF2) acutely protects the heart from dysfunction associated with ischemia/reperfusion (I/R) injury. Our laboratory has demonstrated that this is due to the activity of the low molecular weight (LMW) isoform of FGF2 and that FGF2-mediated cardioprotection relies on the activity of protein kinase C (PKC); however, which PKC isoforms are responsible for LMW FGF2-mediated cardioprotection, and their downstream targets, remain to be elucidated. To identify the PKC pathway(s) that contributes to postischemic cardiac recovery by LMW FGF2, mouse hearts expressing only LMW FGF2 (HMWKO) were bred to mouse hearts not expressing PKCα (PKCαKO) or subjected to a selective PKCε inhibitor (εV(1-2)) before and during I/R. Hearts only expressing LMW FGF2 showed significantly improved postischemic recovery of cardiac function following I/R (P < 0.05), which was significantly abrogated in the absence of PKCα (P < 0.05) or presence of PKCε inhibition (P < 0.05). Hearts only expressing LMW FGF2 demonstrated differences in actomyosin ATPase activity as well as increases in the phosphorylation of troponin I and T during I/R compared with wild-type hearts; several of these effects were dependent on PKCα activity. This evidence indicates that both PKCα and PKCε play a role in LMW FGF2-mediated protection from cardiac dysfunction and that PKCα signaling to the contractile apparatus is a key step in the mechanism of LMW FGF2-mediated protection against myocardial dysfunction.
Collapse
Affiliation(s)
- Janet R Manning
- Department of Pharmacology and Cell Biophysics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Vinexin-β protects against cardiac hypertrophy by blocking the Akt-dependent signalling pathway. Basic Res Cardiol 2013; 108:338. [PMID: 23429936 DOI: 10.1007/s00395-013-0338-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2012] [Revised: 01/15/2013] [Accepted: 02/05/2013] [Indexed: 12/20/2022]
Abstract
Cardiac hypertrophy is the heart's response to hypertrophic stimuli and is associated with increased mortality. Vinexin-β is a vinculin-binding protein that belongs to a family of adaptor proteins and mediates signal transduction and actin cytoskeleton organisation. A previous study has shown that Vinexin-β is ubiquitously expressed and that it is highly expressed in the heart. However, a critical role for Vinexin-β in cardiac hypertrophy has not been investigated. Therefore, to examine the role of Vinexin-β in pathological cardiac hypertrophy, we used Vinexin-β knockout mice and transgenic mice that overexpress human Vinexin-β in the heart. Cardiac hypertrophy was induced by aortic banding (AB). The extent of cardiac hypertrophy was quantitated by echocardiography and pathological and molecular analyses of heart samples. Our results demonstrated that Vinexin-β overexpression in the heart markedly attenuated cardiac hypertrophy, fibrosis, and cardiac dysfunction, whereas loss of Vinexin-β exaggerated the pathological cardiac remodelling and fibrosis response to pressure overload. Further analysis of the in vitro and in vivo signalling events indicated that beneficial Vinexin-β effects were associated with AKT signalling abrogation. Our findings demonstrate for the first time that Vinexin-β is a novel mediator that protects against cardiac hypertrophy by blocking the AKT signalling pathway.
Collapse
|
19
|
Abstract
Heart rate variability (HRV) non-invasively assesses the activity of the autonomic nervous system. During the past 30 years, an increasing number of studies have related the imbalance of the autonomic nervous system (as assessed by HRV) to several pathophysiogical conditions, particularly in the setting of cardiovascular disease. Sudden death, coronary artery disease, heart failure, or merely cardiovascular risk factors (smoking, diabetes, hyperlipidemia, and hypertension) are the best-known clinical circumstances that can affect and/or be affected by the autonomic nervous system. Analyses of HRV variables have been proposed as a component of the clinical evaluation for patient risk stratification due to its independent prognostic information. Yet the potential for HRV to be used widely in clinical practice remains to be established.
Collapse
Affiliation(s)
- Borejda Xhyheri
- Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale. University of Bologna, Bologna, Italy
| | | | | | | | | |
Collapse
|
20
|
Lu J, Bian ZY, Zhang R, Zhang Y, Liu C, Yan L, Zhang SM, Jiang DS, Wei X, Zhu XH, Chen M, Wang AB, Chen Y, Yang Q, Liu PP, Li H. Interferon regulatory factor 3 is a negative regulator of pathological cardiac hypertrophy. Basic Res Cardiol 2013; 108:326. [PMID: 23307144 DOI: 10.1007/s00395-012-0326-9] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2012] [Revised: 12/13/2012] [Accepted: 12/20/2012] [Indexed: 11/25/2022]
Abstract
Interferon regulatory factor (IRF) 3, a member of the highly conserved IRF family transcription factors, plays a pivotal role in innate immune response, apoptosis, and oncogenesis. Recent studies have implicated IRF3 in a wide range of host defense. However, whether IRF3 induces defensive responses to hypertrophic stresses such as biomechanical stress and neurohumoral factors remains unclear. Herein, we employed an IRF3-deficient mouse model, cardiac-specific IRF3-overexpression mouse model and isolated cardiomyocytes to investigate the role of IRF3 in cardiac hypertrophy induced by aortic banding (AB) or isoproterenol (ISO). The extent of cardiac hypertrophy was quantitated by echocardiography as well as by pathological and molecular analysis. Our results demonstrate that IRF3 deficiency profoundly exacerbated cardiac hypertrophy, whereas overexpression of IRF3 in the heart significantly blunted pathological cardiac remodeling induced by pressure overload. Similar results were also observed in cultured cardiomyocytes upon the treatment with ISO. Mechanistically, we discovered that IRF3 interacted with ERK2 and thereby inhibited the ERK1/2 signaling. Furthermore, inactivation of ERK1/2 by U0126 offset the IRF3-deficient-mediated hypertrophic response induced by aortic banding. Altogether, these data demonstrate that IRF3 plays a protective role in AB-induced hypertrophic response by inactivating ERK1/2 in the heart. Therefore, IRF3 could be a new target for the prevention and therapy of cardiac hypertrophy and failure.
Collapse
Affiliation(s)
- Jing Lu
- Department of Cardiology, Renmin Hospital, Cardiovascular Research Institute, Wuhan University, Wuhan, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Perjés Á, Kubin A, Kónyi A, Szabados S, Cziráki A, Skoumal R, Ruskoaho H, Szokodi I. Physiological regulation of cardiac contractility by endogenous reactive oxygen species. Acta Physiol (Oxf) 2012; 205:26-40. [PMID: 22463609 DOI: 10.1111/j.1748-1716.2012.02391.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Increased production of reactive oxygen species (ROS) has been linked to the pathogenesis of congestive heart failure. However, emerging evidence suggests the involvement of ROS in the regulation of various physiological cellular processes in the myocardium. In this review, we summarize the latest findings regarding the role of ROS in the acute regulation of cardiac contractility. We discuss ROS-dependent modulation of the inotropic responses to G protein-coupled receptor agonists (e.g. β-adrenergic receptor agonists and endothelin-1), the potential cellular sources of ROS (e.g. NAD(P)H oxidases and mitochondria) and the proposed end-targets and signalling pathways by which ROS affect contractility. Accumulating new data supports the fundamental role of endogenously generated ROS to regulate cardiac function under physiological conditions.
Collapse
Affiliation(s)
| | - A.M. Kubin
- Department of Pharmacology and Toxicology; Institute of Biomedicine; Biocenter Oulu; University of Oulu; Oulu; Finland
| | - A. Kónyi
- Heart Institute; Medical School; University of Pécs; Pécs; Hungary
| | - S. Szabados
- Heart Institute; Medical School; University of Pécs; Pécs; Hungary
| | - A. Cziráki
- Heart Institute; Medical School; University of Pécs; Pécs; Hungary
| | - R. Skoumal
- Department of Pharmacology and Toxicology; Institute of Biomedicine; Biocenter Oulu; University of Oulu; Oulu; Finland
| | - H. Ruskoaho
- Department of Pharmacology and Toxicology; Institute of Biomedicine; Biocenter Oulu; University of Oulu; Oulu; Finland
| | - I. Szokodi
- Heart Institute; Medical School; University of Pécs; Pécs; Hungary
| |
Collapse
|
22
|
Schwartz BG, Kloner RA. Coronary no reflow. J Mol Cell Cardiol 2012; 52:873-82. [DOI: 10.1016/j.yjmcc.2011.06.009] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2011] [Revised: 06/10/2011] [Accepted: 06/13/2011] [Indexed: 10/18/2022]
|
23
|
Perjés Á, Kubin A, Kónyi A, Szabados S, Cziráki A, Skoumal R, Ruskoaho H, Szokodi I. Physiological regulation of cardiac contractility by endogenous reactive oxygen species. Acta Physiol (Oxf) 2012. [DOI: 10.1111/j.1748-1716.2011.02391.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
| | - A.M. Kubin
- Department of Pharmacology and Toxicology; Institute of Biomedicine; Biocenter Oulu; University of Oulu; Oulu; Finland
| | - A. Kónyi
- Heart Institute; Medical School; University of Pécs; Pécs; Hungary
| | - S. Szabados
- Heart Institute; Medical School; University of Pécs; Pécs; Hungary
| | - A. Cziráki
- Heart Institute; Medical School; University of Pécs; Pécs; Hungary
| | - R. Skoumal
- Department of Pharmacology and Toxicology; Institute of Biomedicine; Biocenter Oulu; University of Oulu; Oulu; Finland
| | - H. Ruskoaho
- Department of Pharmacology and Toxicology; Institute of Biomedicine; Biocenter Oulu; University of Oulu; Oulu; Finland
| | - I. Szokodi
- Heart Institute; Medical School; University of Pécs; Pécs; Hungary
| |
Collapse
|
24
|
de Souza MDSS, Barbalho SM, Damasceno DC, Rudge MVC, de Campos KE, Madi ACG, Coelho BR, Oliveira RC, de Melo RC, Donda VC. Effects ofPassiflora edulis(Yellow Passion) on Serum Lipids and Oxidative Stress Status of Wistar Rats. J Med Food 2012; 15:78-82. [DOI: 10.1089/jmf.2011.0056] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Affiliation(s)
- Maricelma da Silva Soares de Souza
- Department of Pharmacology, University of Marília, Marília, São Paulo, Brazil
- Laboratory of Experimental Research of Gynecology and Obstetrics, Department of Gynecology and Obstetrics, Botucatu Medicine School, São Paulo State University, Botucatu, São Paulo, Brazil
| | - Sandra Maria Barbalho
- Department of Biochemistry, School of Medicine and Health Sciences, University of Marília, Marília, São Paulo, Brazil
- Department of Food Research, Faculty of Food and Technology of Marília, Marília, São Paulo, Brazil
| | - Débora Cristina Damasceno
- Laboratory of Experimental Research of Gynecology and Obstetrics, Department of Gynecology and Obstetrics, Botucatu Medicine School, São Paulo State University, Botucatu, São Paulo, Brazil
| | - Marilza Vieira Cunha Rudge
- Laboratory of Experimental Research of Gynecology and Obstetrics, Department of Gynecology and Obstetrics, Botucatu Medicine School, São Paulo State University, Botucatu, São Paulo, Brazil
| | | | | | | | | | | | | |
Collapse
|
25
|
Agarwal D, Welsch MA, Keller JN, Francis J. Chronic exercise modulates RAS components and improves balance between pro- and anti-inflammatory cytokines in the brain of SHR. Basic Res Cardiol 2011; 106:1069-85. [PMID: 22124756 DOI: 10.1007/s00395-011-0231-7] [Citation(s) in RCA: 122] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2011] [Revised: 11/14/2011] [Accepted: 11/15/2011] [Indexed: 02/07/2023]
Abstract
Recently, exercise has been recommended as a part of lifestyle modification for all hypertensive patients; however, the precise mechanisms of its effects on hypertension are largely unknown. Therefore, this study aimed to investigate the mechanisms within the brain that can influence exercise-induced effects in an animal model of human essential hypertension. Young normotensive WKY rats and SHR were given moderate-intensity exercise for 16 weeks. Blood pressure was measured bi-weekly by tail-cuff method. Animals were then euthanized; paraventricular nucleus (PVN) and rostral ventrolateral medulla (RVLM), important cardiovascular regulatory centers in the brain, were collected and analyzed by real-time RT-PCR, Western blot, EIA, and fluorescent microscopy. Exercise of 16-week duration attenuated systolic, diastolic, and mean arterial pressure in SHR. Sedentary SHR exhibited increased pro-inflammatory cytokines (PICs) and decreased anti-inflammatory IL-10 levels in the PVN and RVLM. Furthermore, SHR(sed) rats exhibited elevated levels of ACE, AT1R, and decreased levels of ACE2 and receptor Mas in the PVN and RVLM. Chronic exercise not only prevented the increase in PICs (TNF-α, IL-1β), ACE, and AT1R protein expression in the brain of SHR, but also dramatically upregulated IL-10, ACE2, and Mas receptor expression in SHR. In addition, these changes were associated with reduced plasma AngII levels, reduced neuronal activity, reduced NADPH-oxidase subunit gp91(phox) and inducible NO synthase in trained SHRs indicating reduced oxidative stress. These results suggest that chronic exercise not only attenuates PICs and the vasoconstrictor axis of the RAS but also improves the anti-inflammatory defense mechanisms and vasoprotective axis of the RAS in the brain, which, at least in part, explains the blood pressure-lowering effects of exercise in hypertension.
Collapse
Affiliation(s)
- Deepmala Agarwal
- Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, 1909 Skip Bertman Drive, Baton Rouge, LA 70803, USA
| | | | | | | |
Collapse
|
26
|
Left ventricular remodeling in swine after myocardial infarction: a transcriptional genomics approach. Basic Res Cardiol 2011; 106:1269-81. [PMID: 22057716 PMCID: PMC3228945 DOI: 10.1007/s00395-011-0229-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2011] [Revised: 10/04/2011] [Accepted: 10/20/2011] [Indexed: 01/06/2023]
Abstract
Despite the apparent appropriateness of left ventricular (LV) remodeling following myocardial infarction (MI), it poses an independent risk factor for development of heart failure. There is a paucity of studies into the molecular mechanisms of LV remodeling in large animal species. We took an unbiased molecular approach to identify candidate transcription factors (TFs) mediating the genetic reprogramming involved in post-MI LV remodeling in swine. Left ventricular tissue was collected from remote, non-infarcted myocardium, 3 weeks after MI-induction or sham-surgery. Microarray analysis identified 285 upregulated and 278 downregulated genes (FDR < 0.05). Of these differentially expressed genes, the promoter regions of the human homologs were searched for common TF binding sites (TFBS). Eighteen TFBS were overrepresented >two-fold (p < 0.01) in upregulated and 13 in downregulated genes. Left ventricular nuclear protein extracts were assayed for DNA-binding activity by protein/DNA array. Out of 345 DNA probes, 30 showed signal intensity changes >two-fold. Five TFs were identified in both TFBS and protein/DNA array analyses, which showed matching changes for COUP-TFII and glucocorticoid receptor (GR) only. Treatment of swine with the GR antagonist mifepristone after MI reduced the post-MI increase in LV mass, but LV dilation remained unaffected. Thus, using an unbiased approach to study post-MI LV remodeling in a physiologically relevant large animal model, we identified COUP-TFII and GR as potential key mediators of post-MI remodeling.
Collapse
|
27
|
Heusch G. Obesity and inflammatory vasculopathy: a surgical solution as ultima ratio? Arterioscler Thromb Vasc Biol 2011; 31:1953-4. [PMID: 21849703 DOI: 10.1161/atvbaha.111.232264] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
28
|
Murdoch CE, Alom-Ruiz SP, Wang M, Zhang M, Walker S, Yu B, Brewer A, Shah AM. Role of endothelial Nox2 NADPH oxidase in angiotensin II-induced hypertension and vasomotor dysfunction. Basic Res Cardiol 2011; 106:527-38. [PMID: 21528437 PMCID: PMC3105229 DOI: 10.1007/s00395-011-0179-7] [Citation(s) in RCA: 124] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2010] [Revised: 03/17/2011] [Accepted: 04/11/2011] [Indexed: 02/07/2023]
Abstract
NADPH oxidase (Nox)-derived reactive oxygen species (ROS) are known to be involved in angiotensin II-induced hypertension and endothelial dysfunction. Several Nox isoforms are expressed in the vessel wall, among which Nox2 is especially abundant in the endothelium. Endothelial Nox2 levels rise during hypertension but little is known about the cell-specific role of endothelial Nox2 in vivo. To address this question, we generated transgenic mice with endothelial-specific overexpression of Nox2 (Tg) and studied the effects on endothelial function and blood pressure. Tg had an about twofold increase in endothelial Nox2 levels which was accompanied by an increase in p22phox levels but no change in levels of other Nox isoforms or endothelial nitric oxide synthase (eNOS). Basal NADPH oxidase activity, endothelial function and blood pressure were unaltered in Tg compared to wild-type littermates. Angiotensin II caused a greater increase in ROS production in Tg compared to wild-type aorta and attenuated acetylcholine-induced vasorelaxation. Both low and high dose chronic angiotensin II infusion increased telemetric ambulatory blood pressure more in Tg compared to wild-type, but with different patterns of BP change and aortic remodeling depending upon the dose of angiotensin II dose. These results indicate that an increase in endothelial Nox2 levels contributes to angiotensin II-induced endothelial dysfunction, vascular remodeling and hypertension.
Collapse
Affiliation(s)
- Colin E. Murdoch
- Cardiovascular Division, King’s College London British Heart Foundation Centre, London, SE5 9PJ UK
| | - Sara P. Alom-Ruiz
- Cardiovascular Division, King’s College London British Heart Foundation Centre, London, SE5 9PJ UK
| | - Minshu Wang
- Cardiovascular Division, King’s College London British Heart Foundation Centre, London, SE5 9PJ UK
| | - Min Zhang
- Cardiovascular Division, King’s College London British Heart Foundation Centre, London, SE5 9PJ UK
| | - Simon Walker
- Cardiovascular Division, King’s College London British Heart Foundation Centre, London, SE5 9PJ UK
| | - Bin Yu
- Cardiovascular Division, King’s College London British Heart Foundation Centre, London, SE5 9PJ UK
| | - Alison Brewer
- Cardiovascular Division, King’s College London British Heart Foundation Centre, London, SE5 9PJ UK
| | - Ajay M. Shah
- Cardiovascular Division, King’s College London British Heart Foundation Centre, London, SE5 9PJ UK
- Cardiovascular Division, The James Black Centre, King’s College London, 125 Coldharbour Lane, London, SE5 9NU UK
| |
Collapse
|
29
|
Sirker A, Zhang M, Shah AM. NADPH oxidases in cardiovascular disease: insights from in vivo models and clinical studies. Basic Res Cardiol 2011; 106:735-47. [PMID: 21598086 PMCID: PMC3149671 DOI: 10.1007/s00395-011-0190-z] [Citation(s) in RCA: 150] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2011] [Revised: 04/11/2011] [Accepted: 04/28/2011] [Indexed: 02/07/2023]
Abstract
NADPH oxidase family enzymes (or NOXs) are the major sources of reactive oxygen species (ROS) that are implicated in the pathophysiology of many cardiovascular diseases. These enzymes appear to be especially important in the modulation of redox-sensitive signalling pathways that underlie key cellular functions such as growth, differentiation, migration and proliferation. Seven distinct members of the family have been identified of which four (namely NOX1, 2, 4 and 5) may have cardiovascular functions. In this article, we review our current understanding of the roles of NOX enzymes in several common cardiovascular disease states, with a focus on data from genetic studies and clinical data where available.
Collapse
Affiliation(s)
- Alexander Sirker
- Cardiovascular Division, King's College London British Heart Foundation Centre, London SE5 9PJ, UK
| | | | | |
Collapse
|
30
|
Morawietz H. Endothelial NADPH oxidases: friends or foes? Basic Res Cardiol 2011; 106:521-5. [DOI: 10.1007/s00395-011-0188-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2011] [Accepted: 04/26/2011] [Indexed: 11/29/2022]
|
31
|
Zhu Y, Li T, Song J, Liu C, Hu Y, Que L, Ha T, Kelley J, Chen Q, Li C, Li Y. The TIR/BB-loop mimetic AS-1 prevents cardiac hypertrophy by inhibiting IL-1R-mediated MyD88-dependent signaling. Basic Res Cardiol 2011; 106:787-99. [PMID: 21533832 DOI: 10.1007/s00395-011-0182-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2010] [Revised: 03/29/2011] [Accepted: 04/15/2011] [Indexed: 11/25/2022]
Abstract
Activation of NF-κB contributes to cardiac hypertrophy and the interleukin-1 receptor (IL-1R)-mediated MyD88-dependent signaling pathway predominately activates NF-κB. Recent studies have shown that the TIR/BB-Loop mimetic (AS-1) disrupted the interaction of MyD88 with the IL-1R, resulting in blunting of NF-κB activation. We have examined the effects of AS-1 on the IL-1β-induced hypertrophic response using cultured neonatal cardiac myocytes in vitro and transverse aortic constriction (TAC) pressure overload-induced cardiac hypertrophy in vivo. Neonatal cardiac myocytes were treated with AS-1 15 min prior to IL-1β stimulation for 24 h. AS-1 treatment significantly attenuated IL-1β-induced hypertrophic responses of cardiac myocytes. In vivo experiments showed that AS-1 administration prevented cardiac hypertrophy and dysfunction induced by pressure overload. AS-1 administration disrupted the interaction of IL-1R with MyD88 in the pressure overloaded hearts and prevented activation of NF-κB. In addition, AS-1 prevented increases in activation of the MAPK pathway (p38 and p-ERK) in TAC-induced hypertrophic hearts. Our data suggest that the IL-1R-mediated MyD88-dependent signaling pathway plays a role in the development of cardiac hypertrophy and AS-1 attenuation of cardiac hypertrophy is mediated by blocking the interaction between IL-1R and MyD88, resulting in decreased NF-κB binding activity and decreased MAPK activation.
Collapse
Affiliation(s)
- Yun Zhu
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Pathophysiology, Nanjing Medical University, 140 Hanzhong Road, Nanjing 210029, Jiangsu, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|