1
|
Kaplan A, El‐Samadi L, Zahreddine R, Amin G, Booz GW, Zouein FA. Canonical or non-canonical, all aspects of G protein-coupled receptor kinase 2 in heart failure. Acta Physiol (Oxf) 2025; 241:e70010. [PMID: 39960030 PMCID: PMC11831727 DOI: 10.1111/apha.70010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 01/12/2025] [Accepted: 01/20/2025] [Indexed: 02/20/2025]
Abstract
G protein-coupled receptor kinase 2 (GRK2) with its multidomain structure performs various crucial cellular functions under both normal and pathological conditions. Overexpression of GRK2 is linked to cardiovascular diseases, and its inhibition or deletion has been shown to be protective. The functions of GRK2 extend beyond G protein-coupled receptor (GPCR) signaling, influencing non-GPCR substrates as well. Increased GRK2 in heart failure (HF) initially may be protective but ultimately leads to maladaptive effects such as GPCR desensitization, insulin resistance, and apoptosis. The multifunctional nature of GRK2, including its action in hypertrophic gene expression, insulin signaling, and cardiac fibrosis, highlights its complex role in HF pathogenesis. Additionally, GRK2 is involved in mitochondrial biogenesis and lipid metabolism. GRK2 also regulates epinephrine secretion from the adrenal gland and its increase in circulating lymphocytes can be used to monitor HF status. Overall, GRK2 is a multifaceted protein with significant implications for HF and the regulation of GRK2 is crucial for understanding and treating cardiovascular diseases.
Collapse
Affiliation(s)
- Abdullah Kaplan
- Department of Pharmacology and ToxicologyAmerican University of Beirut Faculty of MedicineBeirutLebanon
- The Cardiovascular, Renal, and Metabolic Diseases Research Center of ExcellenceAmerican University of Beirut Medical CenterBeirutLebanon
- Cardiology ClinicKemer Public HospitalAntalyaTurkey
| | - Lana El‐Samadi
- Department of Pharmacology and ToxicologyAmerican University of Beirut Faculty of MedicineBeirutLebanon
- The Cardiovascular, Renal, and Metabolic Diseases Research Center of ExcellenceAmerican University of Beirut Medical CenterBeirutLebanon
| | - Rana Zahreddine
- Department of Pharmacology and ToxicologyAmerican University of Beirut Faculty of MedicineBeirutLebanon
- The Cardiovascular, Renal, and Metabolic Diseases Research Center of ExcellenceAmerican University of Beirut Medical CenterBeirutLebanon
| | - Ghadir Amin
- Department of Pharmacology and ToxicologyAmerican University of Beirut Faculty of MedicineBeirutLebanon
- The Cardiovascular, Renal, and Metabolic Diseases Research Center of ExcellenceAmerican University of Beirut Medical CenterBeirutLebanon
- Department of Pharmacology and Toxicology, School of MedicineUniversity of Mississippi Medical CenterJacksonMississippiUSA
| | - George W. Booz
- Department of Pharmacology and Toxicology, School of MedicineUniversity of Mississippi Medical CenterJacksonMississippiUSA
| | - Fouad A. Zouein
- Department of Pharmacology and ToxicologyAmerican University of Beirut Faculty of MedicineBeirutLebanon
- The Cardiovascular, Renal, and Metabolic Diseases Research Center of ExcellenceAmerican University of Beirut Medical CenterBeirutLebanon
- Department of Pharmacology and Toxicology, School of MedicineUniversity of Mississippi Medical CenterJacksonMississippiUSA
| |
Collapse
|
2
|
Xu Z, Li M, Lyu D, Xiao H, Li S, Li Z, Li M, Xiao J, Huang H. Cinnamaldehyde activates AMPK/PGC-1α pathway via targeting GRK2 to ameliorate heart failure. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 133:155894. [PMID: 39089090 DOI: 10.1016/j.phymed.2024.155894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 05/25/2024] [Accepted: 07/15/2024] [Indexed: 08/03/2024]
Abstract
BACKGROUND According to recent research, treating heart failure (HF) by inhibiting G protein-coupled receptor kinase 2 (GRK2) to improve myocardial energy metabolism has been identified as a potential approach. Cinnamaldehyde (CIN), a phenylpropyl aldehyde compound, has been demonstrated to exhibit beneficial effects in cardiovascular diseases. However, whether CIN inhibits GRK2 to ameliorate myocardial energy metabolism in HF is still unclear. PURPOSE This study examines the effects of CIN on GRK2 and myocardial energy metabolism to elucidate its underlying mechanism to treat HF. METHODS The isoproterenol (ISO) induced HF model in vivo and in vitro were constructed using Sprague-Dawley (SD) rats and primary neonatal rat cardiomyocytes (NRCMs). Based on this, the effects of CIN on myocardial energy metabolism and GRK2 were investigated. Additionally, validation experiments were conducted after interfering and over-expressing GRK2 in ISO-induced NRCMs to verify the regulatory effect of CIN on GRK2. Furthermore, binding capacity between GRK2 and CIN was explored by Cellular Thermal Shift Assay (CETSA) and Microscale Thermophoresis (MST). RESULTS In vivo and in vitro, CIN significantly improved HF as demonstrated by reversing abnormal changes in myocardial injury markers, inhibiting myocardial hypertrophy and decreasing myocardial fibrosis. Additionally, CIN promoted myocardial fatty acid metabolism to ameliorate myocardial energy metabolism disorder by activating AMPK/PGC-1α signaling pathway. Moreover, CIN reversed the inhibition of myocardial fatty acid metabolism and AMPK/PGC-1α signaling pathway by GRK2 over-expression in ISO-induced NRCMs. Meanwhile, CIN had no better impact on the stimulation of cardiac fatty acid metabolism and the AMPK/PGC-1α signaling pathway in ISO-induced NRCMs when GRK2 was disrupted. Noticeably, CETSA and MST confirmed that CIN binds to GRK2 directly. The binding of CIN and GRK2 promoted the ubiquitination degradation of GRK2 mediated by murine double mimute 2. CONCLUSION This study demonstrates that CIN exerts a protective intervention in HF by targeting GRK2 and promoting its ubiquitination degradation to activate AMPK/PGC-1α signaling pathway, ultimately improving myocardial fatty acid metabolism.
Collapse
Affiliation(s)
- Zhanchi Xu
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China; Guangzhou Hospital of Integrated Traditional and West Medicine, Guangzhou 510801, China
| | - Minghui Li
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Dongxin Lyu
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Haiming Xiao
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Shanshan Li
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Zhuoming Li
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Min Li
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Junhui Xiao
- Guangzhou Hospital of Integrated Traditional and West Medicine, Guangzhou 510801, China.
| | - Heqing Huang
- Guangzhou Hospital of Integrated Traditional and West Medicine, Guangzhou 510801, China.
| |
Collapse
|
3
|
Vora N, Patel P, Gajjar A, Ladani P, Konat A, Bhanderi D, Gadam S, Prajjwal P, Sharma K, Arunachalam SP. Gene therapy for heart failure: A novel treatment for the age old disease. Dis Mon 2024; 70:101636. [PMID: 37734966 DOI: 10.1016/j.disamonth.2023.101636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/23/2023]
Abstract
Across the globe, cardiovascular disease (CVD) is the leading cause of mortality. According to reports, around 6.2 million people in the United states have heart failure. Current standards of care for heart failure can delay but not prevent progression of disease. Gene therapy is one of the novel treatment modalities that promises to fill this limitation in the current standard of care for Heart Failure. In this paper we performed an extensive search of the literature on various advances made in gene therapy for heart failure till date. We review the delivery methods, targets, current applications, trials, limitations and feasibility of gene therapy for heart failure. Various methods have been employed till date for administering gene therapies including but not limited to arterial and venous infusion, direct myocardial injection and pericardial injection. Various strategies such as AC6 expression, S100A1 protein upregulation, VEGF-B and SDF-1 gene therapy have shown promise in recent preclinical trials. Furthermore, few studies even show that stimulation of cardiomyocyte proliferation such as through cyclin A2 overexpression is a realistic avenue. However, a considerable number of obstacles need to be overcome for gene therapy to be part of standard treatment of care such as definitive choice of gene, gene delivery systems and a suitable method for preclinical trials and clinical trials on patients. Considering the challenges and taking into account the recent advances in gene therapy research, there are encouraging signs to indicate gene therapy for heart failure to be a promising treatment modality for the future. However, the time and feasibility of this option remains in a situation of balance.
Collapse
Affiliation(s)
- Neel Vora
- B. J. Medical College, Ahmedabad, India
| | - Parth Patel
- Pramukhswami Medical College, Karamsad, India
| | | | | | - Ashwati Konat
- University School of Sciences, Gujarat University, Ahmedabad, India
| | | | | | | | - Kamal Sharma
- U. N. Mehta Institute of Cardiology and Research Centre, Ahmedabad, India.
| | | |
Collapse
|
4
|
Triposkiadis F, Briasoulis A, Sarafidis P, Magouliotis D, Athanasiou T, Paraskevaidis I, Skoularigis J, Xanthopoulos A. The Sympathetic Nervous System in Hypertensive Heart Failure with Preserved LVEF. J Clin Med 2023; 12:6486. [PMID: 37892623 PMCID: PMC10607346 DOI: 10.3390/jcm12206486] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 10/09/2023] [Accepted: 10/11/2023] [Indexed: 10/29/2023] Open
Abstract
The neurohormonal model of heart failure (HF) pathogenesis states that a reduction in cardiac output caused by cardiac injury results in sympathetic nervous system (SNS) activation, that is adaptive in the short-term and maladaptive in the long-term. This model has proved extremely valid and has been applied in HF with a reduced left ventricular (LV) ejection fraction (LVEF). In contrast, it has been undermined in HF with preserved LVEF (HFpEF), which is due to hypertension (HTN) in the vast majority of the cases. Erroneously, HTN, which is the leading cause of cardiovascular disease and premature death worldwide and is present in more than 90% of HF patients, is tightly linked with SNS overactivity. In this paper we provide a contemporary overview of the contribution of SNS overactivity to the development and progression of hypertensive HF (HHF) as well as the clinical implications resulting from therapeutic interventions modifying SNS activity. Throughout the manuscript the terms HHF with preserved LVEF and HfpEF will be used interchangeably, considering that the findings in most HFpEF studies are driven by HTN.
Collapse
Affiliation(s)
| | - Alexandros Briasoulis
- Department of Therapeutics, Heart Failure and Cardio-Oncology Clinic, National and Kapodistrian University of Athens, 115 27 Athens, Greece;
| | - Pantelis Sarafidis
- Department of Nephrology, Hippokration Hospital, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece;
| | - Dimitrios Magouliotis
- Unit of Quality Improvement, Department of Cardiothoracic Surgery, University of Thessaly, 411 10 Biopolis, Greece;
| | - Thanos Athanasiou
- Department of Surgery and Cancer, Imperial College London, St Mary’s Hospital, London W2 1NY, UK;
| | | | - John Skoularigis
- Department of Cardiology, University Hospital of Larissa, 411 10 Larissa, Greece;
| | - Andrew Xanthopoulos
- Department of Cardiology, University Hospital of Larissa, 411 10 Larissa, Greece;
| |
Collapse
|
5
|
Dhalla NS, Bhullar SK, Adameova A, Mota KO, de Vasconcelos CML. Status of β 1-Adrenoceptor Signal Transduction System in Cardiac Hypertrophy and Heart Failure. Rev Cardiovasc Med 2023; 24:264. [PMID: 39076390 PMCID: PMC11270071 DOI: 10.31083/j.rcm2409264] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 05/30/2023] [Accepted: 06/15/2023] [Indexed: 07/31/2024] Open
Abstract
Although β 1-adrenoceptor ( β 1-AR) signal transduction, which maintains cardiac function, is downregulated in failing hearts, the mechanisms for such a defect in heart failure are not fully understood. Since cardiac hypertrophy is invariably associated with heart failure, it is possible that the loss of β 1-AR mechanisms in failing heart occurs due to hypertrophic process. In this regard, we have reviewed the information from a rat model of adaptive cardiac hypertrophy and maladaptive hypertrophy at 4 and 24 weeks after inducing pressure overload as well as adaptive cardiac hypertrophy and heart failure at 4 and 24 weeks after inducing volume overload, respectively. Varying degrees of alterations in β 1-AR density as well as isoproterenol-induced increases in cardiac function, intracellular Ca 2 + -concentration in cardiomyocytes and adenylyl cyclase activity in crude membranes have been reported under these hypertrophic conditions. Adaptive hypertrophy at 4 weeks of pressure or volume overload showed unaltered or augmented increases in the activities of different components of β 1-AR signaling. On the other hand, maladaptive hypertrophy due to pressure overload and heart failure due to volume overload at 24 weeks revealed depressions in the activities of β 1-AR signal transduction pathway. These observations provide evidence that β 1-AR signal system is either unaltered or upregulated in adaptive cardiac hypertrophy and downregulated in maladaptive cardiac hypertrophy or heart failure. Furthermore, the information presented in this article supports the concept that downregulation of β 1-AR mechanisms in heart failure or maladaptive cardiac hypertrophy is not due to hypertrophic process per se. It is suggested that a complex mechanism involving the autonomic imbalance may be of a critical importance in determining differential alterations in non-failing and failing hearts.
Collapse
Affiliation(s)
- Naranjan S. Dhalla
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, and Department of Physiology and Pathophysiology, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R2H 2A6, Canada
| | - Sukhwinder K. Bhullar
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, and Department of Physiology and Pathophysiology, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R2H 2A6, Canada
| | - Adriana Adameova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University and Centre of Experimental Medicine, Institute for Heart Research, Slovak Academy of Sciences, 811 03 Bratislava, Slovakia
| | - Karina Oliveira Mota
- Heart Biophysics Laboratory, Department of Physiology, Center for Biological and Health Sciences, Federal University of Sergipe, 73330 Sergipe, Brazil
| | - Carla Maria Lins de Vasconcelos
- Heart Biophysics Laboratory, Department of Physiology, Center for Biological and Health Sciences, Federal University of Sergipe, 73330 Sergipe, Brazil
| |
Collapse
|
6
|
Abd Alla J, Quitterer U. The RAF Kinase Inhibitor Protein (RKIP): Good as Tumour Suppressor, Bad for the Heart. Cells 2022; 11:cells11040654. [PMID: 35203304 PMCID: PMC8869954 DOI: 10.3390/cells11040654] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 02/08/2022] [Accepted: 02/11/2022] [Indexed: 02/04/2023] Open
Abstract
The RAF kinase inhibitor protein, RKIP, is a dual inhibitor of the RAF1 kinase and the G protein-coupled receptor kinase 2, GRK2. By inhibition of the RAF1-MAPK (mitogen-activated protein kinase) pathway, RKIP acts as a beneficial tumour suppressor. By inhibition of GRK2, RKIP counteracts GRK2-mediated desensitisation of G protein-coupled receptor (GPCR) signalling. GRK2 inhibition is considered to be cardioprotective under conditions of exaggerated GRK2 activity such as heart failure. However, cardioprotective GRK2 inhibition and pro-survival RAF1-MAPK pathway inhibition counteract each other, because inhibition of the pro-survival RAF1-MAPK cascade is detrimental for the heart. Therefore, the question arises, what is the net effect of these apparently divergent functions of RKIP in vivo? The available data show that, on one hand, GRK2 inhibition promotes cardioprotective signalling in isolated cardiomyocytes. On the other hand, inhibition of the pro-survival RAF1-MAPK pathway by RKIP deteriorates cardiomyocyte viability. In agreement with cardiotoxic effects, endogenous RKIP promotes cardiac fibrosis under conditions of cardiac stress, and transgenic RKIP induces heart dysfunction. Supported by next-generation sequencing (NGS) data of the RKIP-induced cardiac transcriptome, this review provides an overview of different RKIP functions and explains how beneficial GRK2 inhibition can go awry by RAF1-MAPK pathway inhibition. Based on RKIP studies, requirements for the development of a cardioprotective GRK2 inhibitor are deduced.
Collapse
Affiliation(s)
- Joshua Abd Alla
- Molecular Pharmacology, Department of Chemistry and Applied Biosciences, ETH Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland;
| | - Ursula Quitterer
- Molecular Pharmacology, Department of Chemistry and Applied Biosciences, ETH Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland;
- Department of Medicine, Institute of Pharmacology and Toxicology, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
- Correspondence: ; Tel.: +41-44-632-9801
| |
Collapse
|
7
|
Ågren R, Sahlholm K. G protein-coupled receptor kinase-2 confers isoform-specific calcium sensitivity to dopamine D 2 receptor desensitization. FASEB J 2021; 35:e22013. [PMID: 34699610 DOI: 10.1096/fj.202100704rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 10/04/2021] [Accepted: 10/11/2021] [Indexed: 11/11/2022]
Abstract
The dopamine D2 receptor (D2 R) functions as an autoreceptor on dopaminergic cell bodies and terminals and as a postsynaptic receptor on a variety of neurons in the central nervous system. As a result of alternative splicing, the D2 R is expressed as two isoforms: long (D2L R) and short (D2S R) differing by a stretch of 29 residues in the third intracellular loop, with D2S R being the predominant presynaptic isoform. Recent reports described a Ca2+ sensitivity of the desensitization time course of potassium currents elicited via D2S R, but not via D2L R, when either isoform was selectively expressed in dopaminergic neurons. Here, we aimed to study the mechanism behind this subtype-specific Ca2+ sensitivity. Thus, we measured the desensitization of potassium channel responses evoked by D2L R and D2S R using two-electrode voltage clamp in Xenopus oocytes in the absence and presence of different amounts of β-arrestin2 and G protein-coupled receptor kinase-2 (GRK2), both of which are known to play important roles in D2 R desensitization in native cells. We found that co-expression of both GRK2 and β-arrestin2 was necessary for reconstitution of the Ca2+ sensitivity of D2S R desensitization, while D2L R did not display Ca2+ sensitivity under these conditions. The effect of Ca2+ chelation by BAPTA-AM to slow the rate of D2S R desensitization was mimicked by the GRK2 inhibitor, Cmpd101, and by the kinase-inactivating GRK2 mutation, K220R, but not by the PKC inhibitor, Gö6976, nor by the calmodulin antagonist, KN-93. Thus, Ca2+ -sensitive desensitization of D2S R appears to be mediated via a GRK2 phosphorylation-dependent mechanism.
Collapse
Affiliation(s)
- Richard Ågren
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Kristoffer Sahlholm
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden.,Department of Integrative Medical Biology, Wallenberg Centre for Molecular Medicine, Umeå University, Umeå, Sweden
| |
Collapse
|
8
|
Rowe G, Tracy E, Beare JE, LeBlanc AJ. Cell therapy rescues aging-induced beta-1 adrenergic receptor and GRK2 dysfunction in the coronary microcirculation. GeroScience 2021; 44:329-348. [PMID: 34608562 DOI: 10.1007/s11357-021-00455-6] [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] [Received: 06/11/2021] [Accepted: 09/03/2021] [Indexed: 01/08/2023] Open
Abstract
Our past study showed that coronary arterioles isolated from adipose-derived stromal vascular fraction (SVF)-treated rats showed amelioration of the age-related decrease in vasodilation to beta-adrenergic receptor (β-AR) agonist and improved β-AR-dependent coronary flow and microvascular function in a model of advanced age. We hypothesized that intravenously (i.v.) injected SVF improves coronary microvascular function in aged rats by re-establishing the equilibrium of the negative regulators of the internal adrenergic signaling cascade, G-protein receptor kinase 2 (GRK2) and G-alpha inhibitory (Gαi) proteins, back to youthful levels. Female Fischer-344 rats aged young (3 months, n = 24), old (24 months, n = 26), and old animals that received 1 × 107 green fluorescent protein (GFP+) SVF cells (O + SVF, n = 11) 4 weeks prior to sacrifice were utilized. Overnight urine was collected prior to sacrifice for catecholamine measurements. Cardiac samples were used for western blotting while coronary arterioles were isolated for pressure myography studies, immunofluorescence staining, and RNA sequencing. Coronary microvascular levels of the β1 adrenergic receptor are decreased with advancing age, but this decreased expression was rescued by SVF treatment. Aging led to a decrease in phosphorylated GRK2 in cardiomyocytes vs. young control with restoration of phosphorylation status by SVF. In vessels, there was no change in genetic transcription (RNAseq) or protein expression (immunofluorescence); however, inhibition of GRK2 (paroxetine) led to improved vasodilation to norepinephrine in the old control (OC) and O + SVF, indicating greater GRK2 functional inhibition of β1-AR in aging. SVF works to improve adrenergic-mediated vasodilation by restoring the β1-AR population and mitigating signal cascade inhibitors to improve vasodilation.
Collapse
Affiliation(s)
- Gabrielle Rowe
- Cardiovascular Innovation Institute, University of Louisville, 302 E Muhammad Ali Blvd, Louisville, KY, 40202, USA
- Department of Physiology, University of Louisville, Louisville, KY, 40292, USA
| | - Evan Tracy
- Cardiovascular Innovation Institute, University of Louisville, 302 E Muhammad Ali Blvd, Louisville, KY, 40202, USA
- Department of Physiology, University of Louisville, Louisville, KY, 40292, USA
| | - Jason E Beare
- Cardiovascular Innovation Institute, University of Louisville, 302 E Muhammad Ali Blvd, Louisville, KY, 40202, USA
- Kentucky Spinal Cord Injury Research Center, University of Louisville, Louisville, KY, 40292, USA
| | - Amanda J LeBlanc
- Cardiovascular Innovation Institute, University of Louisville, 302 E Muhammad Ali Blvd, Louisville, KY, 40202, USA.
- Department of Physiology, University of Louisville, Louisville, KY, 40292, USA.
| |
Collapse
|
9
|
Marsico F, Paolillo S, Gargiulo P, Parisi V, Nappi C, Assante R, Dell'Aversana S, Esposito I, Renga F, Esposito L, Bardi L, Rengo G, Dellegrottaglie S, Marciano C, Leosco D, Cuocolo A, Filardi PP. Renal function and cardiac adrenergic impairment in patients affected by heart failure. J Nucl Cardiol 2021; 28:2112-2122. [PMID: 31808105 DOI: 10.1007/s12350-019-01975-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 11/20/2019] [Indexed: 01/17/2023]
Abstract
Although in heart failure (HF) there is a strict correlation between heart and kidney, no data are available on the potential relationship in HF between renal dysfunction (RD) and the impaired sympathetic innervation. Aim of the present study was to assess the relationship between RD and cardiac sympathetic innervation in HF patients with reduced ejection fraction. Two hundred and sixty-three patients with mild-to-severe HF underwent iodine-123 meta-iodobenzylguanidine myocardial scintigraphy to assess sympathetic innervation, evaluating early and late heart-to-mediastinum (H/M) ratios and washout rate. In all patients, glomerular filtration rate (eGFR) by Chronic Kidney Disease Epidemiology Collaboration (CKD-EPI) formula was assessed. A direct association was found between EPI-eGFR and late H/M (r = .215; P < .001). Dividing the population into moderate-to-severe eGFR reduction and normal-to-mildly reduced eGFR (cutoff ≤ 60 mL·min-1·1.73 m-2), a statistically significant reduction of late H/M value was found in patients with RD compared to patients with preserved eGFR (P = .030). By multivariable linear regression analysis, eGFR resulted in the prediction of impaired late H/M in patients with RD (P = .005). Patients with RD and HF show more impaired cardiac sympathetic activity than HF patients with preserved renal function, and reduced eGFR is a predictor of reduced late H/M.
Collapse
Affiliation(s)
- Fabio Marsico
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Via Pansini, 5, 80131, Naples, Italy
| | - Stefania Paolillo
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Via Pansini, 5, 80131, Naples, Italy
- Mediterranea Cardiocentro, Naples, Italy
| | - Paola Gargiulo
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Via Pansini, 5, 80131, Naples, Italy
| | - Valentina Parisi
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Carmela Nappi
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Via Pansini, 5, 80131, Naples, Italy
| | - Roberta Assante
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Via Pansini, 5, 80131, Naples, Italy
| | - Simona Dell'Aversana
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Via Pansini, 5, 80131, Naples, Italy
| | - Immacolata Esposito
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Via Pansini, 5, 80131, Naples, Italy
| | - Francesco Renga
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Via Pansini, 5, 80131, Naples, Italy
| | - Luca Esposito
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Via Pansini, 5, 80131, Naples, Italy
| | - Luca Bardi
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Via Pansini, 5, 80131, Naples, Italy
| | - Giuseppe Rengo
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | | | | | - Dario Leosco
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Alberto Cuocolo
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Via Pansini, 5, 80131, Naples, Italy
| | - Pasquale Perrone Filardi
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Via Pansini, 5, 80131, Naples, Italy.
| |
Collapse
|
10
|
Korpela H, Järveläinen N, Siimes S, Lampela J, Airaksinen J, Valli K, Turunen M, Pajula J, Nurro J, Ylä-Herttuala S. Gene therapy for ischaemic heart disease and heart failure. J Intern Med 2021; 290:567-582. [PMID: 34033164 DOI: 10.1111/joim.13308] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/29/2021] [Accepted: 03/30/2021] [Indexed: 12/27/2022]
Abstract
Gene therapy has been expected to become a novel treatment method since the structure of DNA was discovered in 1953. The morbidity from cardiovascular diseases remains remarkable despite the improvement of percutaneous interventions and pharmacological treatment, underlining the need for novel therapeutics. Gene therapy-mediated therapeutic angiogenesis could help those who have not gained sufficient symptom relief with traditional treatment methods. Especially patients with severe coronary artery disease and heart failure could benefit from gene therapy. Some clinical trials have reported improved myocardial perfusion and symptom relief in CAD patients, but few trials have come up with disappointing negative results. Translating preclinical success into clinical applications has encountered difficulties in successful transduction, study design, endpoint selection, and patient selection and recruitment. However, promising new methods for transducing the cells, such as retrograde delivery and cardiac-specific AAV vectors, hold great promise for myocardial gene therapy. This review introduces gene therapy for ischaemic heart disease and heart failure and discusses the current status and future developments in this field.
Collapse
Affiliation(s)
- H Korpela
- From the, A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - N Järveläinen
- From the, A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - S Siimes
- From the, A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - J Lampela
- From the, A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - J Airaksinen
- From the, A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - K Valli
- From the, A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - M Turunen
- From the, A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - J Pajula
- From the, A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - J Nurro
- From the, A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - S Ylä-Herttuala
- From the, A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
11
|
D'Assante R, Arcopinto M, Rengo G, Salzano A, Walser M, Gambino G, Monti MG, Bencivenga L, Marra AM, Åberg DN, De Vincentiis C, Ballotta A, Bossone E, Isgaard J, Cittadini A. Myocardial expression of somatotropic axis, adrenergic signalling, and calcium handling genes in heart failure with preserved ejection fraction and heart failure with reduced ejection fraction. ESC Heart Fail 2021; 8:1681-1686. [PMID: 33512777 PMCID: PMC8006736 DOI: 10.1002/ehf2.13067] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 09/17/2020] [Accepted: 09/25/2020] [Indexed: 12/28/2022] Open
Abstract
Aims Limited data are available regarding cardiac expression of molecules involved in heart failure (HF) pathophysiology. The majority of the studies have focused on end‐stage HF with reduced ejection fraction (HFrEF) without comparison with healthy subjects, while no data are available with regard to HF with preserved ejection fraction (HFpEF). HFpEF is a condition whose multiple pathophysiological mechanisms are still not fully defined, with many proposed hypotheses remaining speculative due to limited access to human heart tissue. This study aimed at evaluating cardiac expression levels of key genes of interest in human biopsy samples from patients affected with HFrEF and HFpEF in order to possibly point out distinct phenotypes. Methods and results Total RNA was extracted from left ventricular cardiac biopsies collected from stable patients with HFrEF (n = 6) and HFpEF (n = 7) and healthy subjects (n = 9) undergoing elective cardiac surgery for valvular replacement, mitral valvuloplasty, aortic surgery, or coronary artery bypass. Real‐time PCR was performed to evaluate the mRNA expression levels of genes involved in somatotropic axis regulation [IGF‐1, IGF‐1 receptor (IGF‐1R), and GH receptor (GHR)], in adrenergic signalling (GRK2, GRK5, ADRB1, and ADRB2), in myocardial calcium handling (SERCA2), and in TNF‐α. Patients with HFrEF and HFpEF showed reduced serum IGF‐1 circulating levels when compared with controls (102 ± 35.6, 138 ± 11.5, and 160 ± 13.2 ng/mL, P < 0.001, respectively). At myocardial level, HFrEF showed significant decreased GHR and increased IGF‐1R expressions when compared with HFpEF and controls (0.54 ± 0.27, 0.94 ± 0.25, and 0.84 ± 0.2, P < 0.05 and 1.52 ± 0.9, 1.06 ± 0.21, and 0.72 ± 0.12, P < 0.05, respectively), while no differences in the local expression of IGF‐1 mRNA were detected among the groups (0.80 ± 0.45, 0.97 ± 0.18, and 0.63 ± 0.23, P = 0.09, respectively). With regard to calcium handling and adrenergic signalling, HFrEF displayed significant decreased levels of SERCA2 (0.19 ± 0.39, 0.82 ± 0.15, and 0.87 ± 0.32, P < 0.01) and increased levels of GRK2 (3.45 ± 2.94, 0.93 ± 0.12, and 0.80 ± 0.14, P < 0.01) and GRK5 (1.32 ± 0.70, 0.71 ± 0.14, and 0.77 ± 0.15, P < 0.05), while no significant difference was found in ADRB1 (0.66 ± 0.4, 0.83 ± 0.3, and 0.86 ± 0.4) and ADRB2 mRNA expression (0.65 ± 0.3, 0.66 ± 0.2, and 0.68 ± 0.1) when compared with HFpEF and controls. Finally, no changes in the local expression of TNF‐α were detected among groups. Conclusions Heart failure with reduced ejection fraction and HFpEF patients with stable clinical condition display a distinct molecular milieu of genes involved in somatotropic axis regulation, calcium handling, and adrenergic derangement at a myocardial level. The unique opportunity to compare these results with a control group, as reference population, may contribute to better understand HF pathophysiology and to identify novel potential therapeutic targets that could be modulated to improve ventricular function in patients with HF.
Collapse
Affiliation(s)
- Roberta D'Assante
- Department of Translational Medical Sciences, Federico II University of Naples, Via Pansini 5, Naples, 80138, Italy
| | - Michele Arcopinto
- Department of Translational Medical Sciences, Federico II University of Naples, Via Pansini 5, Naples, 80138, Italy
| | - Giuseppe Rengo
- Department of Translational Medical Sciences, Federico II University of Naples, Via Pansini 5, Naples, 80138, Italy.,Istituti Clinici Scientifici Maugeri SpA Società Benefit (ICS Maugeri SpA SB) - IRCCS - Scientific Institute of Telese Terme, Telese Terme, Italy
| | - Andrea Salzano
- IRCCS SDN, Diagnostic and Nuclear Research Institute, Naples, Italy
| | - Marion Walser
- Department of Internal Medicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Giuseppina Gambino
- Department of Translational Medical Sciences, Federico II University of Naples, Via Pansini 5, Naples, 80138, Italy
| | - Maria Gaia Monti
- Department of Translational Medical Sciences, Federico II University of Naples, Via Pansini 5, Naples, 80138, Italy
| | - Leonardo Bencivenga
- Department of Translational Medical Sciences, Federico II University of Naples, Via Pansini 5, Naples, 80138, Italy
| | - Alberto M Marra
- Department of Translational Medical Sciences, Federico II University of Naples, Via Pansini 5, Naples, 80138, Italy
| | - David N Åberg
- Department of Internal Medicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Carlo De Vincentiis
- Department of Cardiothoracic and Vascular Anesthesia and Intensive Care, IRCCS Policlinico San Donato, Milan, Italy
| | - Andrea Ballotta
- Department of Cardiothoracic and Vascular Anesthesia and Intensive Care, IRCCS Policlinico San Donato, Milan, Italy
| | - Eduardo Bossone
- Division of Cardiology, A. Cardarelli Hospital, Naples, Italy
| | - Jörgen Isgaard
- Department of Internal Medicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Antonio Cittadini
- Department of Translational Medical Sciences, Federico II University of Naples, Via Pansini 5, Naples, 80138, Italy
| |
Collapse
|
12
|
GRKs and Epac1 Interaction in Cardiac Remodeling and Heart Failure. Cells 2021; 10:cells10010154. [PMID: 33466800 PMCID: PMC7830799 DOI: 10.3390/cells10010154] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 01/06/2021] [Accepted: 01/08/2021] [Indexed: 12/25/2022] Open
Abstract
β-adrenergic receptors (β-ARs) play a major role in the physiological regulation of cardiac function through signaling routes tightly controlled by G protein-coupled receptor kinases (GRKs). Although the acute stimulation of β-ARs and the subsequent production of cyclic AMP (cAMP) have beneficial effects on cardiac function, chronic stimulation of β-ARs as observed under sympathetic overdrive promotes the development of pathological cardiac remodeling and heart failure (HF), a leading cause of mortality worldwide. This is accompanied by an alteration in cAMP compartmentalization and the activation of the exchange protein directly activated by cAMP 1 (Epac1) signaling. Among downstream signals of β-ARs, compelling evidence indicates that GRK2, GRK5, and Epac1 represent attractive therapeutic targets for cardiac disease. Here, we summarize the pathophysiological roles of GRK2, GRK5, and Epac1 in the heart. We focus on their signalosome and describe how under pathological settings, these proteins can cross-talk and are part of scaffolded nodal signaling systems that contribute to a decreased cardiac function and HF development.
Collapse
|
13
|
Adenoviral βARKct Cardiac Gene Transfer Ameliorates Postresuscitation Myocardial Injury in a Porcine Model of Cardiac Arrest. Shock 2020; 52:631-638. [PMID: 31725109 DOI: 10.1097/shk.0000000000001320] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE The aim of the study was to determine whether the inhibition of the G-protein-coupled receptor kinase 2 by adenoviral βARKct cardiac gene transfer can ameliorate postresuscitation myocardial injury in pigs with cardiac arrest (CA) and explore the mechanism of myocardial protection. METHODS Male landrace domestic pigs were randomized into the sham group (anesthetized and instrumented, but ventricular fibrillation was not induced) (n = 4), control group (ventricular fibrillation 8 min, n = 8), and βARKct group (ventricular fibrillation 8 min, n = 8). Hemodynamic parameters were monitored continuously. Blood samples were collected at baseline, 30 min, 2 h, 4 h, and 6 h after the return of spontaneous circulation (ROSC). Left ventricular ejection fraction was assessed by echocardiography at baseline and 6 h after ROSC. These animals were euthanized, and the cardiac tissue was removed for analysis at 6 h after ROSC. RESULTS Compared with those in the sham group, left ventricular +dp/dtmax, -dp/dtmax, cardiac output (CO), and ejection fraction (EF) in the control group and the βARKct group were significantly decreased at 6 h after the restoration of spontaneous circulation. However, the βARKct treatment produced better left ventricular +dp/dtmax, -dp/dtmax, CO, and EF after ROSC. The βARKct treatment also produced lower serum cardiac troponin I, CK-MB, and lactate after ROSC. Furthermore, the adenoviral βARKct gene transfer significantly increased β1 adrenergic receptors, SERCA2a, RyR2 levels, and decreased GRK2 levels compared to control. CONCLUSIONS The inhibition of GRK2 by adenoviral βARKct cardiac gene transfer can ameliorate postresuscitation myocardial injury through beneficial effects on restoring the sarcoplasmic reticulum Ca-handling proteins expression and upregulating the β1-adrenergic receptor level after cardiac arrest.
Collapse
|
14
|
Design of substrates and inhibitors of G protein-coupled receptor kinase 2 (GRK2) based on its phosphorylation reaction. Amino Acids 2020; 52:863-870. [PMID: 32577910 DOI: 10.1007/s00726-020-02864-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 06/19/2020] [Indexed: 01/05/2023]
Abstract
The G protein-coupled receptor kinase (GRK) family consists of seven cytosolic serine/threonine (Ser/Thr) protein kinases, and among them, GRK2 is involved in the regulation of an enormous range of both G protein-coupled receptors (GPCRs) and non-GPCR substrates that participate in or regulate many critical cellular processes. GRK2 dysfunction is associated with multiple diseases, including cancers, brain diseases, cardiovascular and metabolic diseases, and therefore GRK2-specific substrates/inhibitors are needed not only for studies of GRK2-mediated cellular functions but also for GRK2-targeted drug development. Here, we first review the structure, regulation and functions of GRK2, and its synthetic substrates and inhibitors. We then highlight recent work on synthetic peptide substrates/inhibitors as promising tools for fundamental studies of the physiological functions of GRK2, and as candidates for applications in clinical diagnostics.
Collapse
|
15
|
Li X, Liao J, Jiang Z, Liu X, Chen S, He X, Zhu L, Duan X, Xu Z, Qi B, Guo X, Tong R, Shi J. A concise review of recent advances in anti-heart failure targets and its small molecules inhibitors in recent years. Eur J Med Chem 2020; 186:111852. [PMID: 31759729 DOI: 10.1016/j.ejmech.2019.111852] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 11/03/2019] [Accepted: 11/04/2019] [Indexed: 11/30/2022]
Abstract
Heart failure is a disease with high mortality and the risk of heart failure increases in magnitude with age. The patients of heart failure is increasing year by year. Hence, Pharmaceutical researchers have to develop new drugs with better pharmacological effects to coping with this phenomenon. In this article, we reviewed the small molecule compounds for heart failure that have been marketed in recent years or are promising to enter clinical research. We also reviewed the SAR (structure activity relationship) of these molecules, such as renin inhibitors, ROMK inhibitors, a kind of new diuretics, and some dual-targets inhibitors. These small molecules proven to be beneficial effect on heart failure patients. Which may provide ideas for the design of novel anti-heart failure therapeutic drugs.
Collapse
Affiliation(s)
- Xingxing Li
- School of Medicine, University of Electronic Science and Technology of China, Sichuan Provincial People's Hospital, Chengdu, 610054, China
| | - Jing Liao
- School of Medicine, University of Electronic Science and Technology of China, Sichuan Provincial People's Hospital, Chengdu, 610054, China; Pediatric Department Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, People's Republic of China, Chengdu, 610072, China
| | - Zhongliang Jiang
- Miller School of Medicine, University of Miami, Miami, FL, 33136, USA
| | - Xinyu Liu
- School of Medicine, University of Electronic Science and Technology of China, Sichuan Provincial People's Hospital, Chengdu, 610054, China
| | - Shan Chen
- School of Medicine, University of Electronic Science and Technology of China, Sichuan Provincial People's Hospital, Chengdu, 610054, China
| | - Xia He
- School of Medicine, University of Electronic Science and Technology of China, Sichuan Provincial People's Hospital, Chengdu, 610054, China; Department of Pharmacy, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Individual Key Laboratory, Chengdu, People's Republic of China, Chengdu, 610072, China
| | - Ling Zhu
- Eastern Hospital, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, 610072, China
| | - Xingmei Duan
- School of Medicine, University of Electronic Science and Technology of China, Sichuan Provincial People's Hospital, Chengdu, 610054, China; Department of Pharmacy, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Individual Key Laboratory, Chengdu, People's Republic of China, Chengdu, 610072, China
| | - Zhuyu Xu
- School of Medicine, University of Electronic Science and Technology of China, Sichuan Provincial People's Hospital, Chengdu, 610054, China
| | - Baowen Qi
- College of Pharmacy and Biological Engineering, Chengdu University, Chengdu, 610106, China
| | - Xiaoqiang Guo
- College of Pharmacy and Biological Engineering, Chengdu University, Chengdu, 610106, China
| | - Rongsheng Tong
- School of Medicine, University of Electronic Science and Technology of China, Sichuan Provincial People's Hospital, Chengdu, 610054, China; Department of Pharmacy, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Individual Key Laboratory, Chengdu, People's Republic of China, Chengdu, 610072, China.
| | - Jianyou Shi
- School of Medicine, University of Electronic Science and Technology of China, Sichuan Provincial People's Hospital, Chengdu, 610054, China; Department of Pharmacy, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Individual Key Laboratory, Chengdu, People's Republic of China, Chengdu, 610072, China.
| |
Collapse
|
16
|
Adenoviral.βARKct Cardiac Gene Therapy Ameliorates Cardiac Function Following Cardiopulmonary Bypass in A Swine Model. Shock 2019; 54:563-573. [PMID: 31895875 DOI: 10.1097/shk.0000000000001499] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
17
|
Keretsu S, Bhujbal SP, Joo Cho S. Computational study of paroxetine-like inhibitors reveals new molecular insight to inhibit GRK2 with selectivity over ROCK1. Sci Rep 2019; 9:13053. [PMID: 31506468 PMCID: PMC6736929 DOI: 10.1038/s41598-019-48949-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 07/19/2019] [Indexed: 01/08/2023] Open
Abstract
The G-protein coupled receptor kinase 2 (GRK2) regulates the desensitization of beta-adrenergic receptors (β-AR), and its overexpression has been implicated in heart failure. Hence, the inhibition of GRK2 is considered to be an important drug target for the treatment of heart failure. Due to the high sequence similarity of GRK2 with the A, G, and C family (AGC family) of kinases, the inhibition of GRK2 also leads to the inhibition of AGC kinases such as Rho-associated coiled-coil kinase 1 (ROCK1). Therefore, unraveling the mechanisms to selectively inhibit GRK2 poses an important challenge. We have performed molecular docking, three dimensional quantitative structure activity relationship (3D-QSAR), molecular dynamics (MD) simulation, and free energy calculations techniques on a series of 53 paroxetine-like compounds to understand the structural properties desirable for enhancing the inhibitory activity for GRK2 with selectivity over ROCK1. The formation of stable hydrogen bond interactions with the residues Phe202 and Lys220 of GRK2 seems to be important for selective inhibition of GRK2. Electropositive substituents at the piperidine ring and electronegative substituents near the amide linker between the benzene ring and pyrazole ring showed a higher inhibitory preference for GRK2 over ROCK1. This study may be used in designing more potent and selective GRK2 inhibitors for therapeutic intervention of heart failure.
Collapse
Affiliation(s)
- Seketoulie Keretsu
- Department of Biomedical Sciences, College of Medicine, Chosun University, Gwangju, 501-759, Republic of Korea
| | - Swapnil P Bhujbal
- Department of Biomedical Sciences, College of Medicine, Chosun University, Gwangju, 501-759, Republic of Korea
| | - Seung Joo Cho
- Department of Biomedical Sciences, College of Medicine, Chosun University, Gwangju, 501-759, Republic of Korea. .,Department of Cellular Molecular Medicine, College of Medicine, Chosun University, Gwangju, 501-759, Republic of Korea.
| |
Collapse
|
18
|
Bencivenga L, Liccardo D, Napolitano C, Visaggi L, Rengo G, Leosco D. β-Adrenergic Receptor Signaling and Heart Failure: From Bench to Bedside. Heart Fail Clin 2019; 15:409-419. [PMID: 31079699 DOI: 10.1016/j.hfc.2019.02.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Despite improvements in management and therapeutic approach in the last decades, heart failure is still associated with high mortality rates. The sustained enhancement in the sympathetic nervous system tone, observed in patients with heart failure, causes alteration in β-adrenergic receptor signaling and function. This latter phenomenon is the result of several heart failure-related molecular abnormalities involving adrenergic receptors, G-protein-coupled receptor kinases, and β-arrestins. This article summarizes novel encouraging preclinical strategies to reactivate β-adrenergic receptor signaling in heart failure, including pharmacologic and gene therapy approaches, and attempts to translate acquired notions into the clinical setting.
Collapse
Affiliation(s)
- Leonardo Bencivenga
- Department of Translational Medical Sciences, Division of Geriatrics, Federico II University, Via Sergio Pansini, 5, Naples 80131, Italy
| | - Daniela Liccardo
- Department of Translational Medical Sciences, Division of Geriatrics, Federico II University, Via Sergio Pansini, 5, Naples 80131, Italy
| | - Carmen Napolitano
- Department of Translational Medical Sciences, Division of Geriatrics, Federico II University, Via Sergio Pansini, 5, Naples 80131, Italy
| | - Lucia Visaggi
- Department of Translational Medical Sciences, Division of Geriatrics, Federico II University, Via Sergio Pansini, 5, Naples 80131, Italy
| | - Giuseppe Rengo
- Department of Translational Medical Sciences, Division of Geriatrics, Federico II University, Via Sergio Pansini, 5, Naples 80131, Italy; Istituti Clinici Scientifici Maugeri SpA Società Benefit (ICS Maugeri SpA SB), Telese Terme, Italy
| | - Dario Leosco
- Department of Translational Medical Sciences, Division of Geriatrics, Federico II University, Via Sergio Pansini, 5, Naples 80131, Italy.
| |
Collapse
|
19
|
Komici K, Femminella GD, de Lucia C, Cannavo A, Bencivenga L, Corbi G, Leosco D, Ferrara N, Rengo G. Predisposing factors to heart failure in diabetic nephropathy: a look at the sympathetic nervous system hyperactivity. Aging Clin Exp Res 2019; 31:321-330. [PMID: 29858985 DOI: 10.1007/s40520-018-0973-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Accepted: 05/17/2018] [Indexed: 12/30/2022]
Abstract
Diabetes mellitus (DM) and heart failure (HF) are frequent comorbidities among elderly patients. HF, a leading cause of mortality and morbidity worldwide, is characterized by sympathetic nervous system hyperactivity. The prevalence of diabetes mellitus (DM) is rapidly growing and the risk of developing HF is higher among DM patients. DM is responsible for several macro- and micro-angiopathies that contribute to the development of coronary artery disease (CAD), peripheral artery disease, retinopathy, neuropathy and diabetic nephropathy (DN) as well. Independently of CAD, chronic kidney disease (CKD) and DM increase the risk of HF. Individuals with diabetic nephropathy are likely to present a distinct pathological condition, defined as diabetic cardiomyopathy, even in the absence of hypertension or CAD, whose pathogenesis is only partially known. However, several hypotheses have been proposed to explain the mechanism of diabetic cardiomyopathy: increased oxidative stress, altered substrate metabolism, mitochondrial dysfunction, activation of renin-angiotensin-aldosterone system (RAAS), insulin resistance, and autonomic dysfunction. In this review, we will focus on the involvement of sympathetic system hyperactivity in the diabetic nephropathy.
Collapse
Affiliation(s)
- Klara Komici
- Department of Medicine and Health Sciences, University of Molise, Campobasso, Italy.
| | - Grazia Daniela Femminella
- Division of Geriatrics, Department of Translational Medical Sciences, Federico II University of Naples, Via Sergio Pansini, 5, 80131, Naples, Italy
| | - Claudio de Lucia
- Center for Translational Medicine and Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, USA
| | - Alessandro Cannavo
- Division of Geriatrics, Department of Translational Medical Sciences, Federico II University of Naples, Via Sergio Pansini, 5, 80131, Naples, Italy
| | - Leonardo Bencivenga
- Division of Geriatrics, Department of Translational Medical Sciences, Federico II University of Naples, Via Sergio Pansini, 5, 80131, Naples, Italy
| | - Graziamaria Corbi
- Department of Medicine and Health Sciences, University of Molise, Campobasso, Italy
| | - Dario Leosco
- Division of Geriatrics, Department of Translational Medical Sciences, Federico II University of Naples, Via Sergio Pansini, 5, 80131, Naples, Italy
| | - Nicola Ferrara
- Division of Geriatrics, Department of Translational Medical Sciences, Federico II University of Naples, Via Sergio Pansini, 5, 80131, Naples, Italy
- Istituti Clinici Scientifici Maugeri SPA - Società Benefit, IRCCS - Istituto Scientifico di Telese, Terme, BN, Italy
| | - Giuseppe Rengo
- Division of Geriatrics, Department of Translational Medical Sciences, Federico II University of Naples, Via Sergio Pansini, 5, 80131, Naples, Italy.
- Istituti Clinici Scientifici Maugeri SPA - Società Benefit, IRCCS - Istituto Scientifico di Telese, Terme, BN, Italy.
| |
Collapse
|
20
|
Rodriguez-Serrano M, Rueda J, Buendía F, Monto F, Aguero J, Osa A, Cano O, Martínez-Dolz L, D'Ocon P. β2-Adrenoceptors and GRK2 as Potential Biomarkers in Patients With Chronic Pulmonary Regurgitation. Front Pharmacol 2019; 10:93. [PMID: 30837872 PMCID: PMC6390728 DOI: 10.3389/fphar.2019.00093] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 01/24/2019] [Indexed: 01/11/2023] Open
Abstract
Pulmonary regurgitation (PR) is a frequent complication after repair of congenital heart disease. Three different GRK isoforms (GRK2, GRK5, and GRK3) and two β-adrenoceptors (β1-AR and β2-AR) are present in peripheral blood mononuclear cells (PBMC) and their expression changes as a consequence of the hemodynamic and neurohumoral alterations that occur in some cardiovascular diseases. Therefore, they could be useful as biomarkers in PR. A prospective study was conducted to describe the expression (TaqMan Gene Expression Assays) of β-ARs and GRKs in PBMC isolated (Ficoll® gradient) from patients with severe PR before and after pulmonary valve replacement and establish if this expression correlates to clinical status. 23 patients with severe PR were included and compared with 22 healthy volunteers (controls). PR patients before the PVR had a significantly lower expression of β2-AR (513.8 ± 261.2 mRNA copies) vs. controls (812.5 ± 497.2 mRNA copies), so as GRK2 expression (503.4 ± 364.9 copies vs. 858.1 ± 380.3 mRNA copies). The expression of β2-AR and GRK2 significantly decreases in symptomatic and asymptomatic patients, as well as in patients under treatment with beta-blockers and non-treated patients. The expression of β2-AR and GRK2 in PR patients recovers the normal values after pulmonary valve replacement (754,8 ± 77,1 and 897,8 ± 87,4 copies, respectively). Therefore, changes in the expression of β2-AR and GRK2 in PBMC of PR patients, could be considered as potential biomarkers to determine clinical decisions.
Collapse
Affiliation(s)
| | - Joaquín Rueda
- Servicio de Cardiología, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - Francisco Buendía
- Servicio de Cardiología, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - Fermi Monto
- Departamento de Farmacología, Facultad de Farmacia, Universitat de València, Valencia, Spain.,Estructura de Recerca Interdisciplinar en Biotecnologia i Biomedicina (ERI BIOTECMED), Universitat de València, Valencia, Spain
| | - Jaime Aguero
- Servicio de Cardiología, Hospital Universitario y Politécnico La Fe, Valencia, Spain.,Área de Fisiopatología del Miocardio, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Ana Osa
- Servicio de Cardiología, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - Oscar Cano
- Servicio de Cardiología, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - Luis Martínez-Dolz
- Servicio de Cardiología, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - Pilar D'Ocon
- Departamento de Farmacología, Facultad de Farmacia, Universitat de València, Valencia, Spain.,Estructura de Recerca Interdisciplinar en Biotecnologia i Biomedicina (ERI BIOTECMED), Universitat de València, Valencia, Spain
| |
Collapse
|
21
|
Parker BM, Wertz SL, Pollard CM, Desimine VL, Maning J, McCrink KA, Lymperopoulos A. Novel Insights into the Crosstalk between Mineralocorticoid Receptor and G Protein-Coupled Receptors in Heart Adverse Remodeling and Disease. Int J Mol Sci 2018; 19:3764. [PMID: 30486399 PMCID: PMC6320977 DOI: 10.3390/ijms19123764] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 11/21/2018] [Accepted: 11/23/2018] [Indexed: 12/16/2022] Open
Abstract
The mineralocorticoid hormone aldosterone regulates sodium and potassium homeostasis but also adversely modulates the maladaptive process of cardiac adverse remodeling post-myocardial infarction. Through activation of its mineralocorticoid receptor (MR), a classic steroid hormone receptor/transcription factor, aldosterone promotes inflammation and fibrosis of the heart, the vasculature, and the kidneys. This is why MR antagonists reduce morbidity and mortality of heart disease patients and are part of the mainstay pharmacotherapy of advanced human heart failure. A plethora of animal studies using cell type⁻specific targeting of the MR gene have established the importance of MR signaling and function in cardiac myocytes, vascular endothelial and smooth muscle cells, renal cells, and macrophages. In terms of its signaling properties, the MR is distinct from nuclear receptors in that it has, in reality, two physiological hormonal agonists: not only aldosterone but also cortisol. In fact, in several tissues, including in the myocardium, cortisol is the primary hormone activating the MR. There is a considerable amount of evidence indicating that the effects of the MR in each tissue expressing it depend on tissue- and ligand-specific engagement of molecular co-regulators that either activate or suppress its transcriptional activity. Identification of these co-regulators for every ligand that interacts with the MR in the heart (and in other tissues) is of utmost importance therapeutically, since it can not only help elucidate fully the pathophysiological ramifications of the cardiac MR's actions, but also help design and develop novel better MR antagonist drugs for heart disease therapy. Among the various proteins the MR interacts with are molecules involved in cardiac G protein-coupled receptor (GPCR) signaling. This results in a significant amount of crosstalk between GPCRs and the MR, which can affect the latter's activity dramatically in the heart and in other cardiovascular tissues. This review summarizes the current experimental evidence for this GPCR-MR crosstalk in the heart and discusses its pathophysiological implications for cardiac adverse remodeling as well as for heart disease therapy. Novel findings revealing non-conventional roles of GPCR signaling molecules, specifically of GPCR-kinase (GRK)-5, in cardiac MR regulation are also highlighted.
Collapse
Affiliation(s)
- Barbara M Parker
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences (Pharmacology), College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA.
| | - Shelby L Wertz
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences (Pharmacology), College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA.
| | - Celina M Pollard
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences (Pharmacology), College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA.
| | - Victoria L Desimine
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences (Pharmacology), College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA.
| | - Jennifer Maning
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences (Pharmacology), College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA.
- Present address: Jackson Memorial Hospital, Miami, FL 33136, USA.
| | - Katie A McCrink
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences (Pharmacology), College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA.
- Present address: Massachusetts General Hospital, Boston, MA 02114, USA.
| | - Anastasios Lymperopoulos
- Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences (Pharmacology), College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA.
| |
Collapse
|
22
|
Mangmool S, Parichatikanond W, Kurose H. Therapeutic Targets for Treatment of Heart Failure: Focus on GRKs and β-Arrestins Affecting βAR Signaling. Front Pharmacol 2018; 9:1336. [PMID: 30538631 PMCID: PMC6277550 DOI: 10.3389/fphar.2018.01336] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 10/30/2018] [Indexed: 12/19/2022] Open
Abstract
Heart failure (HF) is a heart disease that is classified into two main types: HF with reduced ejection fraction (HFrEF) and HF with preserved ejection fraction (HFpEF). Both types of HF lead to significant risk of mortality and morbidity. Pharmacological treatment with β-adrenergic receptor (βAR) antagonists (also called β-blockers) has been shown to reduce the overall hospitalization and mortality rates and improve the clinical outcomes in HF patients with HFrEF but not HFpEF. Although, the survival rate of patients suffering from HF continues to drop, the management of HF still faces several limitations and discrepancies highlighting the need to develop new treatment strategies. Overstimulation of the sympathetic nervous system is an adaptive neurohormonal response to acute myocardial injury and heart damage, whereas prolonged exposure to catecholamines causes defects in βAR regulation, including a reduction in the amount of βARs and an increase in βAR desensitization due to the upregulation of G protein-coupled receptor kinases (GRKs) in the heart, contributing in turn to the progression of HF. Several studies show that myocardial GRK2 activity and expression are raised in the failing heart. Furthermore, β-arrestins play a pivotal role in βAR desensitization and, interestingly, can mediate their own signal transduction without any G protein-dependent pathway involved. In this review, we provide new insight into the role of GRKs and β-arrestins on how they affect βAR signaling regarding the molecular and cellular pathophysiology of HF. Additionally, we discuss the therapeutic potential of targeting GRKs and β-arrestins for the treatment of HF.
Collapse
Affiliation(s)
- Supachoke Mangmool
- Department of Pharmacology, Faculty of Pharmacy, Mahidol University, Bangkok, Thailand
| | | | - Hitoshi Kurose
- Department of Pharmacology and Toxicology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
23
|
Razzaque MA, Xu X, Han M, Badami A, Akhter SA. Inhibition of Postinfarction Ventricular Remodeling by High Molecular Weight Polyethylene Glycol. J Surg Res 2018; 232:171-178. [PMID: 30463715 DOI: 10.1016/j.jss.2018.05.035] [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: 02/26/2018] [Revised: 05/09/2018] [Accepted: 05/23/2018] [Indexed: 10/28/2022]
Abstract
BACKGROUND Myocardial infarction (MI) is a major etiology for the development of heart failure. We have previously shown that high molecular weight polyethylene glycol (PEG) can protect cardiac myocytes from hypoxia-reoxygenation injury in vitro. In this study, we investigated the potential protective effects of 15-20 kD PEG postinfarction without reperfusion. METHODS One milliliter of PEG 15-20 was delivered intravenously following permanent left anterior descending ligation in adult male rats with phosphate buffer saline (PBS) as control (n = 9 in each group). Echocardiography was performed at baseline and at 8 wk post-MI. Left ventricles (LVs) were harvested to quantify fibrosis, apoptosis, cell survival signaling, regulation of β-adrenergic signaling, and caveolin (Cav) expression. RESULTS The PEG group had significant recovery of LV function at 8 wk compared with the PBS group. There was less LV fibrosis in both the infarct and remote territory. Cell survival signaling was upregulated in the PEG group with increased Akt and ERK phosphorylation. PEG inhibited apoptosis as measured by terminal deoxynucleotidyl transferase [TdT]-mediated dUTP nick-end labeling positive nuclei and caspase-3 activity. There was maintenance of Cav-1, Cav-2, and Cav-3 expression following PEG treatment versus a decline in the PBS group. Negative regulators of β-adrenergic signaling, G protein-coupled receptor kinase-2, and β-arrestin 1 and 2 were all upregulated in PBS-treated samples compared to normal control; however, PEG treatment led to decreased expression. CONCLUSIONS These data suggest that PEG 15-20 may have significant protective effects post-MI even in the setting of no acute reperfusion. Upregulation of Cav expression appears to be a key mechanism for the beneficial effects of PEG on ventricular remodeling and function.
Collapse
Affiliation(s)
- Md Abdur Razzaque
- Department of Surgery and Cardiovascular Center of Excellence, Louisiana State University, New Orleans, Louisiana; Division of Cardiothoracic Surgery, Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin.
| | - Xianyao Xu
- Division of Cardiothoracic Surgery, Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Mei Han
- Division of Cardiothoracic Surgery, Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Abbas Badami
- Division of Cardiothoracic Surgery, Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Shahab A Akhter
- Division of Cardiothoracic Surgery, Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin; Division of Cardiac Surgery, Department of Cardiovascular Sciences, East Carolina Heart Institute at East Carolina University, Greenville, North Carolina
| |
Collapse
|
24
|
Abstract
G protein-coupled receptors (GPCRs) are the largest class of drug targets, largely owing to their druggability, diversity and physiological efficacy. Many drugs selectively target specific subtypes of GPCRs, but high specificity for individual GPCRs may not be desirable in complex multifactorial disease states in which multiple receptors may be involved. One approach is to target G protein subunits rather than the GPCRs directly. This approach has the potential to achieve broad efficacy by blocking pathways shared by multiple GPCRs. Additionally, because many GPCRs couple to multiple G protein signalling pathways, blocking specific G protein subunits can 'bias' GPCR signals by inhibiting only a subset of these signals. Molecules that target G protein α or βγ-subunits have been developed and show strong efficacy in multiple preclinical disease models and biased inhibition of G protein signalling. In this Review, we discuss the development and characterization of G protein α and βγ-subunit ligands and the preclinical evidence that this exciting new approach has potential for therapeutic efficacy in a number of indications, such as pain, thrombosis, asthma and heart failure.
Collapse
|
25
|
GRK2 knockdown in mice exacerbates kidney injury and alters renal mechanisms of blood pressure regulation. Sci Rep 2018; 8:11415. [PMID: 30061705 PMCID: PMC6065385 DOI: 10.1038/s41598-018-29876-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 07/18/2018] [Indexed: 02/07/2023] Open
Abstract
The renin-angiotensin system regulates blood pressure and fluid balance in the body primarily via angiotensin receptor 1 (AT1R). Renal AT1R was found to be primarily responsible for Ang II-mediated hypertension. G protein-coupled receptor kinase 2 (GRK2) modulates AT1R desensitization and increased GRK2 protein expression is reported in hypertensive patients. However, the consequences of GRK2 inhibition on kidney functions remain unknown. We employed shGRK2 knockdown mice (shGRK2 mice) to test the role of GRK2 in kidney development and function that can be ultimately linked to the hypertensive phenotype detected in shGRK2 mice. GRK2 knockdown reduced kidney size, nephrogenesis and glomerular count, and impaired glomerular filtration. Glomerular damage in adult shGRK2 mice was associated with increased renin- and AT1R-mediated production of reactive oxygen species. The AT1R blocker, Losartan, normalized elevated blood pressure and markedly improved glomerular filtration in the shGRK2 knockdown mice. Our findings provide evidence for the crucial role of GRK2 in renal regulation of blood pressure. It also suggests that the detrimental outcomes of GRK2 inhibitors on the kidney should be carefully examined when used as antihypertensive.
Collapse
|
26
|
Sun F, Lu Z, Zhang Y, Geng S, Xu M, Xu L, Huang Y, Zhuang P, Zhang Y. Stage‑dependent changes of β2‑adrenergic receptor signaling in right ventricular remodeling in monocrotaline‑induced pulmonary arterial hypertension. Int J Mol Med 2018; 41:2493-2504. [PMID: 29393391 PMCID: PMC5846663 DOI: 10.3892/ijmm.2018.3449] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 01/10/2018] [Indexed: 11/29/2022] Open
Abstract
Right ventricular (RV) remodeling coupled with extensive apoptosis in response to unrestrained biomechanical stress may lead to RV failure (RVF), which is the immediate cause of death in the majority of patients with pulmonary arterial hypertension (PAH). Overexpression of β2-adrenergic receptor (β2-AR) signaling has been reported to induce myocardiotoxicity in patients with left heart failure. However, the role of β2-AR signaling in the pathophysiology of PAH development has remained elusive. To address this issue, the present study investigated the changes in cardiopulmonary function and structure, as well as the expression of regulators of fibrosis and apoptosis in RVF following monocrotaline (MCT; 60 mg/kg, i.p.)-induced PAH in rats. Cardiopulmonary function and structure, remodeling and apoptosis, as well as G protein-coupled receptor (GPCR) and β2-AR signaling, were documented over a period of 6 weeks. In the early stages, elevated pulmonary arterial pressure, pulmonary lesions, RV hypertrophy, evidence of left ventricular (LV) hyperfunction and accelerated heart rate were observed in animals with MCT-induced PAH. The levels of angiotensin II receptor type 1b (Agtr1b), Agtr2 and Agt were markedly upregulated and the expression of β2-AR phospho-Ser(355,356) steadily decreased in the right heart. As the disease progressed, LV dysfunction was observed, as evidenced by decreased LV systolic pressure and increased LV end-diastolic pressure, which was accompanied by a sustained increase in circulating brain natriuretic peptide levels. Of note, increased levels of cardiomyocyte apoptosis and concomitant RV remodeling, including hypertrophy, dilatation, inflammation and fibrosis, were observed, despite the enhanced RV contractility. Furthermore, alterations in GPCR signaling and activation in β2-AR-Gs-protein kinase A/Ca2+/calmodulin-dependent kinase II signaling were observed in the late stages of PAH. These results suggested that treatment with MCT results in adaptive and maladaptive RV remodeling and apoptosis during the progression of PAH, which is accompanied by distinct changes in the β2-AR signaling. Therefore, these results enable researchers to better understand of pathophysiology of MCT-induced PAH, as well as to determine the effects of novel therapies.
Collapse
Affiliation(s)
- Fengjiao Sun
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, P.R. China
| | - Zhiqiang Lu
- Department of Pharmacology, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, P.R. China
| | - Yidan Zhang
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, P.R. China
| | - Shihan Geng
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, P.R. China
| | - Mengxi Xu
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, P.R. China
| | - Liman Xu
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, P.R. China
| | - Yingying Huang
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, P.R. China
| | - Pengwei Zhuang
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, P.R. China
| | - Yanjun Zhang
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, P.R. China
| |
Collapse
|
27
|
Cook RF, Bussey CT, Mellor KM, Cragg PA, Lamberts RR. β1-Adrenoceptor, but not β2-adrenoceptor, subtype regulates heart rate in type 2 diabetic ratsin vivo. Exp Physiol 2017; 102:911-923. [DOI: 10.1113/ep086293] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 04/16/2017] [Indexed: 11/08/2022]
Affiliation(s)
- Rosalind F. Cook
- Department of Physiology, Otago School of Medical Sciences, HeartOtago; University of Otago; Dunedin New Zealand
| | - Carol T. Bussey
- Department of Physiology, Otago School of Medical Sciences, HeartOtago; University of Otago; Dunedin New Zealand
| | - Kimberley M. Mellor
- Department of Physiology, Faculty of Medical and Health Sciences; University of Auckland; Auckland New Zealand
| | - Patricia A. Cragg
- Department of Physiology, Otago School of Medical Sciences, HeartOtago; University of Otago; Dunedin New Zealand
| | - Regis R. Lamberts
- Department of Physiology, Otago School of Medical Sciences, HeartOtago; University of Otago; Dunedin New Zealand
| |
Collapse
|
28
|
Lymperopoulos A, Aukszi B. Angiotensin receptor blocker drugs and inhibition of adrenal beta-arrestin-1-dependent aldosterone production: Implications for heart failure therapy. World J Cardiol 2017; 9:200-206. [PMID: 28400916 PMCID: PMC5368669 DOI: 10.4330/wjc.v9.i3.200] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 11/29/2016] [Accepted: 12/16/2016] [Indexed: 02/06/2023] Open
Abstract
Aldosterone mediates many of the physiological and pathophysiological/cardio-toxic effects of angiotensin II (AngII). Its synthesis and secretion from the zona glomerulosa cells of the adrenal cortex, elevated in chronic heart failure (HF), is induced by AngII type 1 receptors (AT1Rs). The AT1R is a G protein-coupled receptor, mainly coupling to Gq/11 proteins. However, it can also signal through β-arrestin-1 (βarr1) or -2 (βarr2), both of which mediate G protein-independent signaling. Over the past decade, a second, Gq/11 protein-independent but βarr1-dependent signaling pathway emanating from the adrenocortical AT1R and leading to aldosterone production has become appreciated. Thus, it became apparent that AT1R antagonists that block both pathways equally well are warranted for fully effective aldosterone suppression in HF. This spurred the comparison of all of the currently marketed angiotensin receptor blockers (ARBs, AT1R antagonists or sartans) at blocking activation of the two signaling modes (G protein-, and βarr1-dependent) at the AngII-activated AT1R and hence, at suppression of aldosterone in vitro and in vivo. Although all agents are very potent inhibitors of G protein activation at the AT1R, candesartan and valsartan were uncovered to be the most potent ARBs at blocking βarr activation by AngII and at suppressing aldosterone in vitro and in vivo in post-myocardial infarction HF animals. In contrast, irbesartan and losartan are virtually G protein-"biased" blockers at the human AT1R, with very low efficacy for βarr inhibition and aldosterone suppression. Therefore, candesartan and valsartan (and other, structurally similar compounds) may be the most preferred ARB agents for HF pharmacotherapy, as well as for treatment of other conditions characterized by elevated aldosterone.
Collapse
Affiliation(s)
- Anastasios Lymperopoulos
- Anastasios Lymperopoulos, Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, United States
| | - Beatrix Aukszi
- Anastasios Lymperopoulos, Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, United States
| |
Collapse
|
29
|
Mody R, Hernandez Y, Lymperopoulos A. Assays of adrenal GPCR signaling and regulation: Measuring adrenal β-arrestin activity in vivo through plasma membrane recruitment. Methods Cell Biol 2017:79-87. [DOI: 10.1016/bs.mcb.2017.07.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
30
|
Powell JM, Ebin E, Borzak S, Lymperopoulos A, Hennekens CH. Hypothesis: Paroxetine, a G Protein-Coupled Receptor Kinase 2 (GRK2) Inhibitor Reduces Morbidity and Mortality in Patients With Heart Failure. J Cardiovasc Pharmacol Ther 2017; 22:51-53. [PMID: 27222484 DOI: 10.1177/1074248416644350] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The hypothesis that paroxetine decreases morbidity and mortality in patients with heart failure (HF) is plausible but unproven. Basic research demonstrates that inhibition of G protein-coupled receptor kinase 2 (GRK2) both in vitro and in vivo in the myocardium may be beneficial. G protein-coupled receptor kinase 2 antagonism is purported to exert cardioprotective effects immediately following myocardial injury by blunting toxic overstimulation on a recently injured heart. In addition, chronic overexpression of GRK2 inhibits catecholamine induction of vital positive chronotropic and ionotropic effects required to preserve cardiac output leading to worsening of congestive HF. In cardiac-specific GRK2 conditional knockout mice, there is significant improvement in left ventricular wall thickness, left ventricular end-diastolic diameter (LVEDD), and ejection fraction (EF) compared to controls. Paroxetine is a selective serotonin reuptake inhibitor which was recently shown to have the ability to directly inhibit GRK2 both in vitro and in vivo. At physiologic temperatures, paroxetine inhibits GRK2-dependent phosphorylation of an activated G-protein-coupled receptor with a half maximal inhibitory concentration of 35 micromoles, a substantially greater affinity than for other G protein-coupled receptor kinases. In a randomized trial in mice with systolic HF and depressed EF postmyocardial infarction, those treated with paroxetine had a 30% increase in EF, improved contractility, and LVEDD and wall thickness compared to those treated with medical therapy alone. While further basic research may continue to elucidate plausible mechanisms of benefit and observational studies will contribute important relevant information, large scale randomized trials designed a priori to do so are necessary to test the hypothesis.
Collapse
Affiliation(s)
- Jonathan M Powell
- 1 Charles E. Schmidt College of Medicine and Graduate Medical Education Consortium (Bethesda Hospital, Boca Raton Regional Hospital, Delray Medical Center, St. Mary's Medical Center, West Boca Raton Hospital), Florida Atlantic University, Boca Raton, FL, USA
| | - Emanuel Ebin
- 1 Charles E. Schmidt College of Medicine and Graduate Medical Education Consortium (Bethesda Hospital, Boca Raton Regional Hospital, Delray Medical Center, St. Mary's Medical Center, West Boca Raton Hospital), Florida Atlantic University, Boca Raton, FL, USA
| | - Steven Borzak
- 1 Charles E. Schmidt College of Medicine and Graduate Medical Education Consortium (Bethesda Hospital, Boca Raton Regional Hospital, Delray Medical Center, St. Mary's Medical Center, West Boca Raton Hospital), Florida Atlantic University, Boca Raton, FL, USA
| | - Anastasios Lymperopoulos
- 2 Department of Pharmaceutical Sciences, University College of Pharmacy, Nova Southeastern University, Lakeland, FL, USA
| | - Charles H Hennekens
- 1 Charles E. Schmidt College of Medicine and Graduate Medical Education Consortium (Bethesda Hospital, Boca Raton Regional Hospital, Delray Medical Center, St. Mary's Medical Center, West Boca Raton Hospital), Florida Atlantic University, Boca Raton, FL, USA
| |
Collapse
|
31
|
cAMP-PKA-CaMKII signaling pathway is involved in aggravated cardiotoxicity during Fuzi and Beimu Combination Treatment of Experimental Pulmonary Hypertension. Sci Rep 2016; 6:34903. [PMID: 27739450 PMCID: PMC5064387 DOI: 10.1038/srep34903] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 09/20/2016] [Indexed: 01/03/2023] Open
Abstract
Aconiti Lateralis Radix Praeparata (Fuzi) and Fritillariae Thunbergii bulbus (Beimu) have been widely used clinically to treat cardiopulmonary related diseases in China. However, according to the classic rules of traditional Chinese medicine, Fuzi and Beimu should be prohibited to use as a combination for their incompatibility. Therefore, it is critical to elucidate the paradox on the use of Fuzi and Beimu combination therapy. Monocrotaline-induced pulmonary hypertension rats were treated with either Fuzi, Beimu, or their combination at different stages of PH. We demonstrated that at the early stage of PH, Fuzi and Beimu combination significantly improved lung function and reduced pulmonary histopathology. However, as the disease progressed, when Fuzi and Beimu combination were used at the late stage of PH, right ventricular chamber dilation was histologically apparent and myocardial apoptosis was significantly increased compared with each drug alone. Western-blotting results indicated that the main chemical ingredient of Beimu could down-regulate the protein phosphorylation levels of Akt and PDE4D, whereas the combination of Fuzi and Beimu could up-regulate PKA and CaMKII signaling pathways. Therefore, we concluded that Fuzi and Beimu combination potentially aggravated the heart injury due to the inhibition of PDK1/Akt/PDE4D axis and subsequent synergistic activation of βAR-Gs-PKA/CaMKII signaling pathway.
Collapse
|
32
|
Végh AMD, Duim SN, Smits AM, Poelmann RE, Ten Harkel ADJ, DeRuiter MC, Goumans MJ, Jongbloed MRM. Part and Parcel of the Cardiac Autonomic Nerve System: Unravelling Its Cellular Building Blocks during Development. J Cardiovasc Dev Dis 2016; 3:jcdd3030028. [PMID: 29367572 PMCID: PMC5715672 DOI: 10.3390/jcdd3030028] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2016] [Revised: 09/05/2016] [Accepted: 09/07/2016] [Indexed: 02/06/2023] Open
Abstract
The autonomic nervous system (cANS) is essential for proper heart function, and complications such as heart failure, arrhythmias and even sudden cardiac death are associated with an altered cANS function. A changed innervation state may underlie (part of) the atrial and ventricular arrhythmias observed after myocardial infarction. In other cardiac diseases, such as congenital heart disease, autonomic dysfunction may be related to disease outcome. This is also the case after heart transplantation, when the heart is denervated. Interest in the origin of the autonomic nerve system has renewed since the role of autonomic function in disease progression was recognized, and some plasticity in autonomic regeneration is evident. As with many pathological processes, autonomic dysfunction based on pathological innervation may be a partial recapitulation of the early development of innervation. As such, insight into the development of cardiac innervation and an understanding of the cellular background contributing to cardiac innervation during different phases of development is required. This review describes the development of the cANS and focuses on the cellular contributions, either directly by delivering cells or indirectly by secretion of necessary factors or cell-derivatives.
Collapse
Affiliation(s)
- Anna M D Végh
- Department of Molecular Cell Biology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, The Netherlands.
| | - Sjoerd N Duim
- Department of Molecular Cell Biology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, The Netherlands.
| | - Anke M Smits
- Department of Molecular Cell Biology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, The Netherlands.
| | - Robert E Poelmann
- Department of Cardiology, Leiden University Medical Center, Albinusdreef 2, 2333 ZC Leiden, The Netherlands.
- Institute of Biology Leiden, Leiden University, Sylviusweg 20, 2311 EZ Leiden, The Netherlands.
| | - Arend D J Ten Harkel
- Department of Pediatric Cardiology, Leiden University Medical Center, Albinusdreef 2, 2333 ZC Leiden, The Netherlands.
| | - Marco C DeRuiter
- Department of Anatomy & Embryology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, The Netherlands.
| | - Marie José Goumans
- Department of Molecular Cell Biology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, The Netherlands.
| | - Monique R M Jongbloed
- Department of Cardiology, Leiden University Medical Center, Albinusdreef 2, 2333 ZC Leiden, The Netherlands.
- Department of Pediatric Cardiology, Leiden University Medical Center, Albinusdreef 2, 2333 ZC Leiden, The Netherlands.
| |
Collapse
|
33
|
Lymperopoulos A, Brill A, McCrink KA. GPCRs of adrenal chromaffin cells & catecholamines: The plot thickens. Int J Biochem Cell Biol 2016; 77:213-219. [PMID: 26851510 DOI: 10.1016/j.biocel.2016.02.003] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2015] [Revised: 02/01/2016] [Accepted: 02/02/2016] [Indexed: 12/14/2022]
Abstract
The circulating catecholamines (CAs) epinephrine (Epi) and norepinephrine (NE) derive from two major sources in the whole organism: the sympathetic nerve endings, which release NE on effector organs, and the chromaffin cells of the adrenal medulla, which are cells that synthesize, store and release Epi (mainly) and NE. All of the Epi in the body and a significant amount of circulating NE derive from the adrenal medulla. The secretion of CAs from adrenal chromaffin cells is regulated in a complex way by a variety of membrane receptors, the vast majority of which are G protein-coupled receptors (GPCRs), including adrenergic receptors (ARs), which act as "presynaptic autoreceptors" in this regard. There is a plethora of CA-secretagogue signals acting on these receptors but some of them, most notably the α2ARs, inhibit CA secretion. Over the past few years, however, a few new proteins present in chromaffin cells have been uncovered to participate in CA secretion regulation. Most prominent among these are GRK2 and β-arrestin1, which are known to interact with GPCRs regulating receptor signaling and function. The present review will discuss the molecular and signaling mechanisms by which adrenal chromaffin cell-residing GPCRs and their regulatory proteins modulate CA synthesis and secretion. Particular emphasis will be given to the newly discovered roles of GRK2 and β-arrestins in these processes and particular points of focus for future research will be highlighted, as well.
Collapse
Affiliation(s)
- Anastasios Lymperopoulos
- From the Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences, Nova Southeastern University, College of Pharmacy, 3200 S. University Dr., Fort Lauderdale, FL 33328-2018, USA.
| | - Ava Brill
- From the Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences, Nova Southeastern University, College of Pharmacy, 3200 S. University Dr., Fort Lauderdale, FL 33328-2018, USA
| | - Katie A McCrink
- From the Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences, Nova Southeastern University, College of Pharmacy, 3200 S. University Dr., Fort Lauderdale, FL 33328-2018, USA
| |
Collapse
|
34
|
Lother A, Hein L. Pharmacology of heart failure: From basic science to novel therapies. Pharmacol Ther 2016; 166:136-49. [PMID: 27456554 DOI: 10.1016/j.pharmthera.2016.07.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 07/08/2016] [Indexed: 01/10/2023]
Abstract
Chronic heart failure is one of the leading causes for hospitalization in the United States and Europe, and is accompanied by high mortality. Current pharmacological therapy of chronic heart failure with reduced ejection fraction is largely based on compounds that inhibit the detrimental action of the adrenergic and the renin-angiotensin-aldosterone systems on the heart. More than one decade after spironolactone, two novel therapeutic principles have been added to the very recently released guidelines on heart failure therapy: the HCN-channel inhibitor ivabradine and the combined angiotensin and neprilysin inhibitor valsartan/sacubitril. New compounds that are in phase II or III clinical evaluation include novel non-steroidal mineralocorticoid receptor antagonists, guanylate cyclase activators or myosine activators. A variety of novel candidate targets have been identified and the availability of gene transfer has just begun to accelerate translation from basic science to clinical application. This review provides an overview of current pharmacology and pharmacotherapy in chronic heart failure at three stages: the updated clinical guidelines of the American Heart Association and the European Society of Cardiology, new drugs which are in clinical development, and finally innovative drug targets and their mechanisms in heart failure which are emerging from preclinical studies will be discussed.
Collapse
Affiliation(s)
- Achim Lother
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Heart Center, Department of Cardiology and Angiology I, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| | - Lutz Hein
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg, Germany; BIOSS Centre for Biological Signaling Studies, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
35
|
Gomez-Monterrey I, Carotenuto A, Cipolletta E, Sala M, Vernieri E, Limatola A, Bertamino A, Musella S, Grieco P, Trimarco B, Novellino E, Iaccarino G, Campiglia P. SAR study and conformational analysis of a series of novel peptide G protein-coupled receptor kinase 2 inhibitors. Biopolymers 2016; 101:121-8. [PMID: 23733420 DOI: 10.1002/bip.22295] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Revised: 05/22/2013] [Accepted: 05/22/2013] [Indexed: 01/09/2023]
Abstract
G protein-coupled receptor kinase 2 (GRK2) plays a central role in the cellular transduction network. In particular, during chronic heart failure GRK2 is upregulated and believed to contribute to disease progression. Thereby, its inhibition offers a potential therapeutic solution to several pathological conditions. In the present study, we performed a SAR study and a NMR conformational analysis of peptides derived from HJ loop of GRK2 and able to selectively inhibit GRK2. From Ala-scan and D-Ala point replacement, we found that Arg residues don't affect the inhibitory properties, while a D-amino acid at position 5 is key to the activity. Conformational analysis identified two β-turns that involve N-terminal residues, followed by a short extended region. These information can help the design of peptides and peptido-mimetics with enhanced GRK2 inhibition properties.
Collapse
|
36
|
Rossi F, Mascolo A, Mollace V. The pathophysiological role of natriuretic peptide-RAAS cross talk in heart failure. Int J Cardiol 2016; 226:121-125. [PMID: 27062428 DOI: 10.1016/j.ijcard.2016.03.080] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 03/19/2016] [Indexed: 12/22/2022]
Abstract
Chronic Heart Failure (HF) is still a disease state characterized by elevated morbidity and mortality and represents an unresolved problem for its socio-economic impact. Besides many of the pathophysiological events leading to advanced HF have been widely disclosed in the past decades, the role of neuro-hormonal dysregulation accompanying HF has to be clearly assessed with the objective of better therapeutic approaches in treating such a disease. In the present review article, alongside with a brief re-evaluation of general aspects of HF physiopathology, we summarize recent advances in the cross talk between renin-angiotensin-aldosterone system (RAAS) with natriuretic peptides (NPs) which have been shown to play a relevant role in the development of severe HF. The role of RAAS-NPs interplay has been shown to be crucial in both hemodynamic and tissue remodeling associated to cardiomyocyte dysfunction, leading to advanced impairment of left ventricular performance. On the basis of these results, the development of drugs resetting both RAAS and NPs system seems to be promising for a successful long term treatment of chronic HF.
Collapse
Affiliation(s)
- Francesco Rossi
- Second University of Naples, Campania Regional Centre for Pharmacovigilance and Pharmacoepidemiology, Department of Experimental Medicine, Section of Pharmacology L. Donatelli, Napoli, Italy
| | - Annamaria Mascolo
- Second University of Naples, Campania Regional Centre for Pharmacovigilance and Pharmacoepidemiology, Department of Experimental Medicine, Section of Pharmacology L. Donatelli, Napoli, Italy.
| | - Vincenzo Mollace
- Institute of Research for Food Safety & Health (IRC-FSH), University "Magna Graecia" of Catanzaro, Italy
| |
Collapse
|
37
|
Parisi V, Rengo G, Perrone-Filardi P, Pagano G, Femminella GD, Paolillo S, Petraglia L, Gambino G, Caruso A, Grimaldi MG, Baldascino F, Nolano M, Elia A, Cannavo A, De Bellis A, Coscioni E, Pellegrino T, Cuocolo A, Ferrara N, Leosco D. Increased Epicardial Adipose Tissue Volume Correlates With Cardiac Sympathetic Denervation in Patients With Heart Failure. Circ Res 2016; 118:1244-53. [PMID: 26926470 DOI: 10.1161/circresaha.115.307765] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 02/26/2016] [Indexed: 12/31/2022]
Abstract
RATIONALE It has been reported that epicardial adipose tissue (EAT) may affect myocardial autonomic function. OBJECTIVE The aim of this study was to explore the relationship between EAT and cardiac sympathetic nerve activity in patients with heart failure. METHODS AND RESULTS In 110 patients with systolic heart failure, we evaluated the correlation between echocardiographic EAT thickness and cardiac adrenergic nerve activity assessed by (123)I-metaiodobenzylguanidine ((123)I-MIBG). The predictive value of EAT thickness on cardiac sympathetic denervation ((123)I-MIBG early and late heart:mediastinum ratio and single-photon emission computed tomography total defect score) was tested in a multivariate analysis. Furthermore, catecholamine levels, catecholamine biosynthetic enzymes, and sympathetic nerve fibers were measured in EAT and subcutaneous adipose tissue biopsies obtained from patients with heart failure who underwent cardiac surgery. EAT thickness correlated with (123)I-MIBG early and late heart:mediastinum ratio and single-photon emission computed tomography total defect score, but not with left ventricular ejection fraction. Moreover, EAT resulted as an independent predictor of (123)I-MIBG early and late heart:mediastinum ratio and single-photon emission computed tomography total defect score and showed a significant additive predictive value on (123)I-MIBG planar and single-photon emission computed tomography results over demographic and clinical data. Although no differences were found in sympathetic innervation between EAT and subcutaneous adipose tissue, EAT showed an enhanced adrenergic activity demonstrated by the increased catecholamine levels and expression of catecholamine biosynthetic enzymes. CONCLUSIONS This study provides the first evidence of a direct correlation between increased EAT thickness and cardiac sympathetic denervation in heart failure.
Collapse
Affiliation(s)
- Valentina Parisi
- From the Department of Translational Medical Sciences (V.P., G.R., G.P., G.D.F., L.P., G.G., A.C., N.F., D.L.) and Department of Advanced Biomedical Science (P.P.-F., T.P., A.C.), University Federico II, Naples, Italy; Department of Cardiology (G.R., G.G.) and Department of Neurology (M.N., A.E.), Salvatore Maugeri Foundation, IRCCS, Istituto di Telese, Benevento, Italy (G.R., G.G., M.N., A.E.); SDN Foundation, Institute of Diagnostic and Nuclear Development, Naples, Italy (S.P.); Department of Cardiology and Cardiac Surgery, Casa di Cura San Michele, Maddaloni (CE), Italy (A.C., M.G.G., F.B., A.D.B.); Department of Cardiac Surgery, Ruggi D'Aragona Hospital, Salerno, Italy (E.C.); and Institute of Biostructure and Bioimaging Italian National Research Council (CNR), Naples, Italy (T.P.)
| | - Giuseppe Rengo
- From the Department of Translational Medical Sciences (V.P., G.R., G.P., G.D.F., L.P., G.G., A.C., N.F., D.L.) and Department of Advanced Biomedical Science (P.P.-F., T.P., A.C.), University Federico II, Naples, Italy; Department of Cardiology (G.R., G.G.) and Department of Neurology (M.N., A.E.), Salvatore Maugeri Foundation, IRCCS, Istituto di Telese, Benevento, Italy (G.R., G.G., M.N., A.E.); SDN Foundation, Institute of Diagnostic and Nuclear Development, Naples, Italy (S.P.); Department of Cardiology and Cardiac Surgery, Casa di Cura San Michele, Maddaloni (CE), Italy (A.C., M.G.G., F.B., A.D.B.); Department of Cardiac Surgery, Ruggi D'Aragona Hospital, Salerno, Italy (E.C.); and Institute of Biostructure and Bioimaging Italian National Research Council (CNR), Naples, Italy (T.P.).
| | - Pasquale Perrone-Filardi
- From the Department of Translational Medical Sciences (V.P., G.R., G.P., G.D.F., L.P., G.G., A.C., N.F., D.L.) and Department of Advanced Biomedical Science (P.P.-F., T.P., A.C.), University Federico II, Naples, Italy; Department of Cardiology (G.R., G.G.) and Department of Neurology (M.N., A.E.), Salvatore Maugeri Foundation, IRCCS, Istituto di Telese, Benevento, Italy (G.R., G.G., M.N., A.E.); SDN Foundation, Institute of Diagnostic and Nuclear Development, Naples, Italy (S.P.); Department of Cardiology and Cardiac Surgery, Casa di Cura San Michele, Maddaloni (CE), Italy (A.C., M.G.G., F.B., A.D.B.); Department of Cardiac Surgery, Ruggi D'Aragona Hospital, Salerno, Italy (E.C.); and Institute of Biostructure and Bioimaging Italian National Research Council (CNR), Naples, Italy (T.P.)
| | - Gennaro Pagano
- From the Department of Translational Medical Sciences (V.P., G.R., G.P., G.D.F., L.P., G.G., A.C., N.F., D.L.) and Department of Advanced Biomedical Science (P.P.-F., T.P., A.C.), University Federico II, Naples, Italy; Department of Cardiology (G.R., G.G.) and Department of Neurology (M.N., A.E.), Salvatore Maugeri Foundation, IRCCS, Istituto di Telese, Benevento, Italy (G.R., G.G., M.N., A.E.); SDN Foundation, Institute of Diagnostic and Nuclear Development, Naples, Italy (S.P.); Department of Cardiology and Cardiac Surgery, Casa di Cura San Michele, Maddaloni (CE), Italy (A.C., M.G.G., F.B., A.D.B.); Department of Cardiac Surgery, Ruggi D'Aragona Hospital, Salerno, Italy (E.C.); and Institute of Biostructure and Bioimaging Italian National Research Council (CNR), Naples, Italy (T.P.)
| | - Grazia Daniela Femminella
- From the Department of Translational Medical Sciences (V.P., G.R., G.P., G.D.F., L.P., G.G., A.C., N.F., D.L.) and Department of Advanced Biomedical Science (P.P.-F., T.P., A.C.), University Federico II, Naples, Italy; Department of Cardiology (G.R., G.G.) and Department of Neurology (M.N., A.E.), Salvatore Maugeri Foundation, IRCCS, Istituto di Telese, Benevento, Italy (G.R., G.G., M.N., A.E.); SDN Foundation, Institute of Diagnostic and Nuclear Development, Naples, Italy (S.P.); Department of Cardiology and Cardiac Surgery, Casa di Cura San Michele, Maddaloni (CE), Italy (A.C., M.G.G., F.B., A.D.B.); Department of Cardiac Surgery, Ruggi D'Aragona Hospital, Salerno, Italy (E.C.); and Institute of Biostructure and Bioimaging Italian National Research Council (CNR), Naples, Italy (T.P.)
| | - Stefania Paolillo
- From the Department of Translational Medical Sciences (V.P., G.R., G.P., G.D.F., L.P., G.G., A.C., N.F., D.L.) and Department of Advanced Biomedical Science (P.P.-F., T.P., A.C.), University Federico II, Naples, Italy; Department of Cardiology (G.R., G.G.) and Department of Neurology (M.N., A.E.), Salvatore Maugeri Foundation, IRCCS, Istituto di Telese, Benevento, Italy (G.R., G.G., M.N., A.E.); SDN Foundation, Institute of Diagnostic and Nuclear Development, Naples, Italy (S.P.); Department of Cardiology and Cardiac Surgery, Casa di Cura San Michele, Maddaloni (CE), Italy (A.C., M.G.G., F.B., A.D.B.); Department of Cardiac Surgery, Ruggi D'Aragona Hospital, Salerno, Italy (E.C.); and Institute of Biostructure and Bioimaging Italian National Research Council (CNR), Naples, Italy (T.P.)
| | - Laura Petraglia
- From the Department of Translational Medical Sciences (V.P., G.R., G.P., G.D.F., L.P., G.G., A.C., N.F., D.L.) and Department of Advanced Biomedical Science (P.P.-F., T.P., A.C.), University Federico II, Naples, Italy; Department of Cardiology (G.R., G.G.) and Department of Neurology (M.N., A.E.), Salvatore Maugeri Foundation, IRCCS, Istituto di Telese, Benevento, Italy (G.R., G.G., M.N., A.E.); SDN Foundation, Institute of Diagnostic and Nuclear Development, Naples, Italy (S.P.); Department of Cardiology and Cardiac Surgery, Casa di Cura San Michele, Maddaloni (CE), Italy (A.C., M.G.G., F.B., A.D.B.); Department of Cardiac Surgery, Ruggi D'Aragona Hospital, Salerno, Italy (E.C.); and Institute of Biostructure and Bioimaging Italian National Research Council (CNR), Naples, Italy (T.P.)
| | - Giuseppina Gambino
- From the Department of Translational Medical Sciences (V.P., G.R., G.P., G.D.F., L.P., G.G., A.C., N.F., D.L.) and Department of Advanced Biomedical Science (P.P.-F., T.P., A.C.), University Federico II, Naples, Italy; Department of Cardiology (G.R., G.G.) and Department of Neurology (M.N., A.E.), Salvatore Maugeri Foundation, IRCCS, Istituto di Telese, Benevento, Italy (G.R., G.G., M.N., A.E.); SDN Foundation, Institute of Diagnostic and Nuclear Development, Naples, Italy (S.P.); Department of Cardiology and Cardiac Surgery, Casa di Cura San Michele, Maddaloni (CE), Italy (A.C., M.G.G., F.B., A.D.B.); Department of Cardiac Surgery, Ruggi D'Aragona Hospital, Salerno, Italy (E.C.); and Institute of Biostructure and Bioimaging Italian National Research Council (CNR), Naples, Italy (T.P.)
| | - Aurelio Caruso
- From the Department of Translational Medical Sciences (V.P., G.R., G.P., G.D.F., L.P., G.G., A.C., N.F., D.L.) and Department of Advanced Biomedical Science (P.P.-F., T.P., A.C.), University Federico II, Naples, Italy; Department of Cardiology (G.R., G.G.) and Department of Neurology (M.N., A.E.), Salvatore Maugeri Foundation, IRCCS, Istituto di Telese, Benevento, Italy (G.R., G.G., M.N., A.E.); SDN Foundation, Institute of Diagnostic and Nuclear Development, Naples, Italy (S.P.); Department of Cardiology and Cardiac Surgery, Casa di Cura San Michele, Maddaloni (CE), Italy (A.C., M.G.G., F.B., A.D.B.); Department of Cardiac Surgery, Ruggi D'Aragona Hospital, Salerno, Italy (E.C.); and Institute of Biostructure and Bioimaging Italian National Research Council (CNR), Naples, Italy (T.P.)
| | - Maria Gabriella Grimaldi
- From the Department of Translational Medical Sciences (V.P., G.R., G.P., G.D.F., L.P., G.G., A.C., N.F., D.L.) and Department of Advanced Biomedical Science (P.P.-F., T.P., A.C.), University Federico II, Naples, Italy; Department of Cardiology (G.R., G.G.) and Department of Neurology (M.N., A.E.), Salvatore Maugeri Foundation, IRCCS, Istituto di Telese, Benevento, Italy (G.R., G.G., M.N., A.E.); SDN Foundation, Institute of Diagnostic and Nuclear Development, Naples, Italy (S.P.); Department of Cardiology and Cardiac Surgery, Casa di Cura San Michele, Maddaloni (CE), Italy (A.C., M.G.G., F.B., A.D.B.); Department of Cardiac Surgery, Ruggi D'Aragona Hospital, Salerno, Italy (E.C.); and Institute of Biostructure and Bioimaging Italian National Research Council (CNR), Naples, Italy (T.P.)
| | - Francesco Baldascino
- From the Department of Translational Medical Sciences (V.P., G.R., G.P., G.D.F., L.P., G.G., A.C., N.F., D.L.) and Department of Advanced Biomedical Science (P.P.-F., T.P., A.C.), University Federico II, Naples, Italy; Department of Cardiology (G.R., G.G.) and Department of Neurology (M.N., A.E.), Salvatore Maugeri Foundation, IRCCS, Istituto di Telese, Benevento, Italy (G.R., G.G., M.N., A.E.); SDN Foundation, Institute of Diagnostic and Nuclear Development, Naples, Italy (S.P.); Department of Cardiology and Cardiac Surgery, Casa di Cura San Michele, Maddaloni (CE), Italy (A.C., M.G.G., F.B., A.D.B.); Department of Cardiac Surgery, Ruggi D'Aragona Hospital, Salerno, Italy (E.C.); and Institute of Biostructure and Bioimaging Italian National Research Council (CNR), Naples, Italy (T.P.)
| | - Maria Nolano
- From the Department of Translational Medical Sciences (V.P., G.R., G.P., G.D.F., L.P., G.G., A.C., N.F., D.L.) and Department of Advanced Biomedical Science (P.P.-F., T.P., A.C.), University Federico II, Naples, Italy; Department of Cardiology (G.R., G.G.) and Department of Neurology (M.N., A.E.), Salvatore Maugeri Foundation, IRCCS, Istituto di Telese, Benevento, Italy (G.R., G.G., M.N., A.E.); SDN Foundation, Institute of Diagnostic and Nuclear Development, Naples, Italy (S.P.); Department of Cardiology and Cardiac Surgery, Casa di Cura San Michele, Maddaloni (CE), Italy (A.C., M.G.G., F.B., A.D.B.); Department of Cardiac Surgery, Ruggi D'Aragona Hospital, Salerno, Italy (E.C.); and Institute of Biostructure and Bioimaging Italian National Research Council (CNR), Naples, Italy (T.P.)
| | - Andrea Elia
- From the Department of Translational Medical Sciences (V.P., G.R., G.P., G.D.F., L.P., G.G., A.C., N.F., D.L.) and Department of Advanced Biomedical Science (P.P.-F., T.P., A.C.), University Federico II, Naples, Italy; Department of Cardiology (G.R., G.G.) and Department of Neurology (M.N., A.E.), Salvatore Maugeri Foundation, IRCCS, Istituto di Telese, Benevento, Italy (G.R., G.G., M.N., A.E.); SDN Foundation, Institute of Diagnostic and Nuclear Development, Naples, Italy (S.P.); Department of Cardiology and Cardiac Surgery, Casa di Cura San Michele, Maddaloni (CE), Italy (A.C., M.G.G., F.B., A.D.B.); Department of Cardiac Surgery, Ruggi D'Aragona Hospital, Salerno, Italy (E.C.); and Institute of Biostructure and Bioimaging Italian National Research Council (CNR), Naples, Italy (T.P.)
| | - Alessandro Cannavo
- From the Department of Translational Medical Sciences (V.P., G.R., G.P., G.D.F., L.P., G.G., A.C., N.F., D.L.) and Department of Advanced Biomedical Science (P.P.-F., T.P., A.C.), University Federico II, Naples, Italy; Department of Cardiology (G.R., G.G.) and Department of Neurology (M.N., A.E.), Salvatore Maugeri Foundation, IRCCS, Istituto di Telese, Benevento, Italy (G.R., G.G., M.N., A.E.); SDN Foundation, Institute of Diagnostic and Nuclear Development, Naples, Italy (S.P.); Department of Cardiology and Cardiac Surgery, Casa di Cura San Michele, Maddaloni (CE), Italy (A.C., M.G.G., F.B., A.D.B.); Department of Cardiac Surgery, Ruggi D'Aragona Hospital, Salerno, Italy (E.C.); and Institute of Biostructure and Bioimaging Italian National Research Council (CNR), Naples, Italy (T.P.)
| | - Antonio De Bellis
- From the Department of Translational Medical Sciences (V.P., G.R., G.P., G.D.F., L.P., G.G., A.C., N.F., D.L.) and Department of Advanced Biomedical Science (P.P.-F., T.P., A.C.), University Federico II, Naples, Italy; Department of Cardiology (G.R., G.G.) and Department of Neurology (M.N., A.E.), Salvatore Maugeri Foundation, IRCCS, Istituto di Telese, Benevento, Italy (G.R., G.G., M.N., A.E.); SDN Foundation, Institute of Diagnostic and Nuclear Development, Naples, Italy (S.P.); Department of Cardiology and Cardiac Surgery, Casa di Cura San Michele, Maddaloni (CE), Italy (A.C., M.G.G., F.B., A.D.B.); Department of Cardiac Surgery, Ruggi D'Aragona Hospital, Salerno, Italy (E.C.); and Institute of Biostructure and Bioimaging Italian National Research Council (CNR), Naples, Italy (T.P.)
| | - Enrico Coscioni
- From the Department of Translational Medical Sciences (V.P., G.R., G.P., G.D.F., L.P., G.G., A.C., N.F., D.L.) and Department of Advanced Biomedical Science (P.P.-F., T.P., A.C.), University Federico II, Naples, Italy; Department of Cardiology (G.R., G.G.) and Department of Neurology (M.N., A.E.), Salvatore Maugeri Foundation, IRCCS, Istituto di Telese, Benevento, Italy (G.R., G.G., M.N., A.E.); SDN Foundation, Institute of Diagnostic and Nuclear Development, Naples, Italy (S.P.); Department of Cardiology and Cardiac Surgery, Casa di Cura San Michele, Maddaloni (CE), Italy (A.C., M.G.G., F.B., A.D.B.); Department of Cardiac Surgery, Ruggi D'Aragona Hospital, Salerno, Italy (E.C.); and Institute of Biostructure and Bioimaging Italian National Research Council (CNR), Naples, Italy (T.P.)
| | - Teresa Pellegrino
- From the Department of Translational Medical Sciences (V.P., G.R., G.P., G.D.F., L.P., G.G., A.C., N.F., D.L.) and Department of Advanced Biomedical Science (P.P.-F., T.P., A.C.), University Federico II, Naples, Italy; Department of Cardiology (G.R., G.G.) and Department of Neurology (M.N., A.E.), Salvatore Maugeri Foundation, IRCCS, Istituto di Telese, Benevento, Italy (G.R., G.G., M.N., A.E.); SDN Foundation, Institute of Diagnostic and Nuclear Development, Naples, Italy (S.P.); Department of Cardiology and Cardiac Surgery, Casa di Cura San Michele, Maddaloni (CE), Italy (A.C., M.G.G., F.B., A.D.B.); Department of Cardiac Surgery, Ruggi D'Aragona Hospital, Salerno, Italy (E.C.); and Institute of Biostructure and Bioimaging Italian National Research Council (CNR), Naples, Italy (T.P.)
| | - Alberto Cuocolo
- From the Department of Translational Medical Sciences (V.P., G.R., G.P., G.D.F., L.P., G.G., A.C., N.F., D.L.) and Department of Advanced Biomedical Science (P.P.-F., T.P., A.C.), University Federico II, Naples, Italy; Department of Cardiology (G.R., G.G.) and Department of Neurology (M.N., A.E.), Salvatore Maugeri Foundation, IRCCS, Istituto di Telese, Benevento, Italy (G.R., G.G., M.N., A.E.); SDN Foundation, Institute of Diagnostic and Nuclear Development, Naples, Italy (S.P.); Department of Cardiology and Cardiac Surgery, Casa di Cura San Michele, Maddaloni (CE), Italy (A.C., M.G.G., F.B., A.D.B.); Department of Cardiac Surgery, Ruggi D'Aragona Hospital, Salerno, Italy (E.C.); and Institute of Biostructure and Bioimaging Italian National Research Council (CNR), Naples, Italy (T.P.)
| | - Nicola Ferrara
- From the Department of Translational Medical Sciences (V.P., G.R., G.P., G.D.F., L.P., G.G., A.C., N.F., D.L.) and Department of Advanced Biomedical Science (P.P.-F., T.P., A.C.), University Federico II, Naples, Italy; Department of Cardiology (G.R., G.G.) and Department of Neurology (M.N., A.E.), Salvatore Maugeri Foundation, IRCCS, Istituto di Telese, Benevento, Italy (G.R., G.G., M.N., A.E.); SDN Foundation, Institute of Diagnostic and Nuclear Development, Naples, Italy (S.P.); Department of Cardiology and Cardiac Surgery, Casa di Cura San Michele, Maddaloni (CE), Italy (A.C., M.G.G., F.B., A.D.B.); Department of Cardiac Surgery, Ruggi D'Aragona Hospital, Salerno, Italy (E.C.); and Institute of Biostructure and Bioimaging Italian National Research Council (CNR), Naples, Italy (T.P.)
| | - Dario Leosco
- From the Department of Translational Medical Sciences (V.P., G.R., G.P., G.D.F., L.P., G.G., A.C., N.F., D.L.) and Department of Advanced Biomedical Science (P.P.-F., T.P., A.C.), University Federico II, Naples, Italy; Department of Cardiology (G.R., G.G.) and Department of Neurology (M.N., A.E.), Salvatore Maugeri Foundation, IRCCS, Istituto di Telese, Benevento, Italy (G.R., G.G., M.N., A.E.); SDN Foundation, Institute of Diagnostic and Nuclear Development, Naples, Italy (S.P.); Department of Cardiology and Cardiac Surgery, Casa di Cura San Michele, Maddaloni (CE), Italy (A.C., M.G.G., F.B., A.D.B.); Department of Cardiac Surgery, Ruggi D'Aragona Hospital, Salerno, Italy (E.C.); and Institute of Biostructure and Bioimaging Italian National Research Council (CNR), Naples, Italy (T.P.)
| |
Collapse
|
38
|
Rengo G, Pagano G, Filardi PP, Femminella GD, Parisi V, Cannavo A, Liccardo D, Komici K, Gambino G, D'Amico ML, de Lucia C, Paolillo S, Trimarco B, Vitale DF, Ferrara N, Koch WJ, Leosco D. Prognostic Value of Lymphocyte G Protein-Coupled Receptor Kinase-2 Protein Levels in Patients With Heart Failure. Circ Res 2016; 118:1116-24. [PMID: 26884616 DOI: 10.1161/circresaha.115.308207] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 02/16/2016] [Indexed: 01/08/2023]
Abstract
RATIONALE Sympathetic nervous system hyperactivity is associated with poor prognosis in patients with heart failure (HF), yet routine assessment of sympathetic nervous system activation is not recommended for clinical practice. Myocardial G protein-coupled receptor kinase-2 (GRK2) is upregulated in HF patients, causing dysfunctional β-adrenergic receptor signaling. Importantly, myocardial GRK2 levels correlate with levels found in peripheral lymphocytes of HF patients. OBJECTIVE The independent prognostic value of blood GRK2 measurements in HF patients has never been investigated; thus, the purpose of this study was to evaluate whether lymphocyte GRK2 levels predict clinical outcome in HF patients. METHODS AND RESULTS We prospectively studied 257 HF patients with mean left ventricular ejection fraction of 31.4±8.5%. At the time of enrollment, plasma norepinephrine, serum NT-proBNP, and lymphocyte GRK2 levels, as well as clinical and instrumental variables were measured. The prognostic value of GRK2 to predict cardiovascular (CV) death and all-cause mortality was assessed using the Cox proportional hazard model including demographic, clinical, instrumental, and laboratory data. Over a mean follow-up period of 37.5±20.2 months (range, 3-60 months), there were 102 CV deaths. Age, left ventricular ejection fraction, New York Heart Association class, chronic obstructive pulmonary disease, chronic kidney disease, N-terminal-pro brain natriuretic peptide, and lymphocyte GRK2 protein levels were independent predictors of CV mortality in HF patients. GRK2 levels showed an additional prognostic and clinical value over demographic and clinical variables. The independent prognostic value of lymphocyte GRK2 levels was also confirmed for all-cause mortality. CONCLUSIONS Lymphocyte GRK2 protein levels can independently predict prognosis in patients with HF.
Collapse
Affiliation(s)
- Giuseppe Rengo
- From the Division of Cardiology, Salvatore Maugeri Foundation, IRCCS, Scientific Institute of Telese Terme (BN), Italy (G.R., G.G., D.F.V., N.F.); Division of Geriatrics, Department of Translational Medical Sciences (G.R., G.P., G.D.F., V.P., A.C., D. Liccardo, K.K., G.G., M.L.D.'A., C.d.L., N.F., D. Leosco), Division of Cardiology, Department of Advanced Biomedical Sciences (P.P.F., B.T.), Federico II University of Naples, Naples, Italy; SDN Foundation IRCCS, Institute of Diagnostic and Nuclear Development, Naples, Italy (S.P.); and Department of Pharmacology, Center of Translational Medicine, Temple University, Philadelphia, PA (A.C., D. Liccardo, W.J.K.)
| | - Gennaro Pagano
- From the Division of Cardiology, Salvatore Maugeri Foundation, IRCCS, Scientific Institute of Telese Terme (BN), Italy (G.R., G.G., D.F.V., N.F.); Division of Geriatrics, Department of Translational Medical Sciences (G.R., G.P., G.D.F., V.P., A.C., D. Liccardo, K.K., G.G., M.L.D.'A., C.d.L., N.F., D. Leosco), Division of Cardiology, Department of Advanced Biomedical Sciences (P.P.F., B.T.), Federico II University of Naples, Naples, Italy; SDN Foundation IRCCS, Institute of Diagnostic and Nuclear Development, Naples, Italy (S.P.); and Department of Pharmacology, Center of Translational Medicine, Temple University, Philadelphia, PA (A.C., D. Liccardo, W.J.K.)
| | - Pasquale Perrone Filardi
- From the Division of Cardiology, Salvatore Maugeri Foundation, IRCCS, Scientific Institute of Telese Terme (BN), Italy (G.R., G.G., D.F.V., N.F.); Division of Geriatrics, Department of Translational Medical Sciences (G.R., G.P., G.D.F., V.P., A.C., D. Liccardo, K.K., G.G., M.L.D.'A., C.d.L., N.F., D. Leosco), Division of Cardiology, Department of Advanced Biomedical Sciences (P.P.F., B.T.), Federico II University of Naples, Naples, Italy; SDN Foundation IRCCS, Institute of Diagnostic and Nuclear Development, Naples, Italy (S.P.); and Department of Pharmacology, Center of Translational Medicine, Temple University, Philadelphia, PA (A.C., D. Liccardo, W.J.K.)
| | - Grazia Daniela Femminella
- From the Division of Cardiology, Salvatore Maugeri Foundation, IRCCS, Scientific Institute of Telese Terme (BN), Italy (G.R., G.G., D.F.V., N.F.); Division of Geriatrics, Department of Translational Medical Sciences (G.R., G.P., G.D.F., V.P., A.C., D. Liccardo, K.K., G.G., M.L.D.'A., C.d.L., N.F., D. Leosco), Division of Cardiology, Department of Advanced Biomedical Sciences (P.P.F., B.T.), Federico II University of Naples, Naples, Italy; SDN Foundation IRCCS, Institute of Diagnostic and Nuclear Development, Naples, Italy (S.P.); and Department of Pharmacology, Center of Translational Medicine, Temple University, Philadelphia, PA (A.C., D. Liccardo, W.J.K.)
| | - Valentina Parisi
- From the Division of Cardiology, Salvatore Maugeri Foundation, IRCCS, Scientific Institute of Telese Terme (BN), Italy (G.R., G.G., D.F.V., N.F.); Division of Geriatrics, Department of Translational Medical Sciences (G.R., G.P., G.D.F., V.P., A.C., D. Liccardo, K.K., G.G., M.L.D.'A., C.d.L., N.F., D. Leosco), Division of Cardiology, Department of Advanced Biomedical Sciences (P.P.F., B.T.), Federico II University of Naples, Naples, Italy; SDN Foundation IRCCS, Institute of Diagnostic and Nuclear Development, Naples, Italy (S.P.); and Department of Pharmacology, Center of Translational Medicine, Temple University, Philadelphia, PA (A.C., D. Liccardo, W.J.K.)
| | - Alessandro Cannavo
- From the Division of Cardiology, Salvatore Maugeri Foundation, IRCCS, Scientific Institute of Telese Terme (BN), Italy (G.R., G.G., D.F.V., N.F.); Division of Geriatrics, Department of Translational Medical Sciences (G.R., G.P., G.D.F., V.P., A.C., D. Liccardo, K.K., G.G., M.L.D.'A., C.d.L., N.F., D. Leosco), Division of Cardiology, Department of Advanced Biomedical Sciences (P.P.F., B.T.), Federico II University of Naples, Naples, Italy; SDN Foundation IRCCS, Institute of Diagnostic and Nuclear Development, Naples, Italy (S.P.); and Department of Pharmacology, Center of Translational Medicine, Temple University, Philadelphia, PA (A.C., D. Liccardo, W.J.K.)
| | - Daniela Liccardo
- From the Division of Cardiology, Salvatore Maugeri Foundation, IRCCS, Scientific Institute of Telese Terme (BN), Italy (G.R., G.G., D.F.V., N.F.); Division of Geriatrics, Department of Translational Medical Sciences (G.R., G.P., G.D.F., V.P., A.C., D. Liccardo, K.K., G.G., M.L.D.'A., C.d.L., N.F., D. Leosco), Division of Cardiology, Department of Advanced Biomedical Sciences (P.P.F., B.T.), Federico II University of Naples, Naples, Italy; SDN Foundation IRCCS, Institute of Diagnostic and Nuclear Development, Naples, Italy (S.P.); and Department of Pharmacology, Center of Translational Medicine, Temple University, Philadelphia, PA (A.C., D. Liccardo, W.J.K.)
| | - Klara Komici
- From the Division of Cardiology, Salvatore Maugeri Foundation, IRCCS, Scientific Institute of Telese Terme (BN), Italy (G.R., G.G., D.F.V., N.F.); Division of Geriatrics, Department of Translational Medical Sciences (G.R., G.P., G.D.F., V.P., A.C., D. Liccardo, K.K., G.G., M.L.D.'A., C.d.L., N.F., D. Leosco), Division of Cardiology, Department of Advanced Biomedical Sciences (P.P.F., B.T.), Federico II University of Naples, Naples, Italy; SDN Foundation IRCCS, Institute of Diagnostic and Nuclear Development, Naples, Italy (S.P.); and Department of Pharmacology, Center of Translational Medicine, Temple University, Philadelphia, PA (A.C., D. Liccardo, W.J.K.)
| | - Giuseppina Gambino
- From the Division of Cardiology, Salvatore Maugeri Foundation, IRCCS, Scientific Institute of Telese Terme (BN), Italy (G.R., G.G., D.F.V., N.F.); Division of Geriatrics, Department of Translational Medical Sciences (G.R., G.P., G.D.F., V.P., A.C., D. Liccardo, K.K., G.G., M.L.D.'A., C.d.L., N.F., D. Leosco), Division of Cardiology, Department of Advanced Biomedical Sciences (P.P.F., B.T.), Federico II University of Naples, Naples, Italy; SDN Foundation IRCCS, Institute of Diagnostic and Nuclear Development, Naples, Italy (S.P.); and Department of Pharmacology, Center of Translational Medicine, Temple University, Philadelphia, PA (A.C., D. Liccardo, W.J.K.)
| | - Maria Loreta D'Amico
- From the Division of Cardiology, Salvatore Maugeri Foundation, IRCCS, Scientific Institute of Telese Terme (BN), Italy (G.R., G.G., D.F.V., N.F.); Division of Geriatrics, Department of Translational Medical Sciences (G.R., G.P., G.D.F., V.P., A.C., D. Liccardo, K.K., G.G., M.L.D.'A., C.d.L., N.F., D. Leosco), Division of Cardiology, Department of Advanced Biomedical Sciences (P.P.F., B.T.), Federico II University of Naples, Naples, Italy; SDN Foundation IRCCS, Institute of Diagnostic and Nuclear Development, Naples, Italy (S.P.); and Department of Pharmacology, Center of Translational Medicine, Temple University, Philadelphia, PA (A.C., D. Liccardo, W.J.K.)
| | - Claudio de Lucia
- From the Division of Cardiology, Salvatore Maugeri Foundation, IRCCS, Scientific Institute of Telese Terme (BN), Italy (G.R., G.G., D.F.V., N.F.); Division of Geriatrics, Department of Translational Medical Sciences (G.R., G.P., G.D.F., V.P., A.C., D. Liccardo, K.K., G.G., M.L.D.'A., C.d.L., N.F., D. Leosco), Division of Cardiology, Department of Advanced Biomedical Sciences (P.P.F., B.T.), Federico II University of Naples, Naples, Italy; SDN Foundation IRCCS, Institute of Diagnostic and Nuclear Development, Naples, Italy (S.P.); and Department of Pharmacology, Center of Translational Medicine, Temple University, Philadelphia, PA (A.C., D. Liccardo, W.J.K.)
| | - Stefania Paolillo
- From the Division of Cardiology, Salvatore Maugeri Foundation, IRCCS, Scientific Institute of Telese Terme (BN), Italy (G.R., G.G., D.F.V., N.F.); Division of Geriatrics, Department of Translational Medical Sciences (G.R., G.P., G.D.F., V.P., A.C., D. Liccardo, K.K., G.G., M.L.D.'A., C.d.L., N.F., D. Leosco), Division of Cardiology, Department of Advanced Biomedical Sciences (P.P.F., B.T.), Federico II University of Naples, Naples, Italy; SDN Foundation IRCCS, Institute of Diagnostic and Nuclear Development, Naples, Italy (S.P.); and Department of Pharmacology, Center of Translational Medicine, Temple University, Philadelphia, PA (A.C., D. Liccardo, W.J.K.)
| | - Bruno Trimarco
- From the Division of Cardiology, Salvatore Maugeri Foundation, IRCCS, Scientific Institute of Telese Terme (BN), Italy (G.R., G.G., D.F.V., N.F.); Division of Geriatrics, Department of Translational Medical Sciences (G.R., G.P., G.D.F., V.P., A.C., D. Liccardo, K.K., G.G., M.L.D.'A., C.d.L., N.F., D. Leosco), Division of Cardiology, Department of Advanced Biomedical Sciences (P.P.F., B.T.), Federico II University of Naples, Naples, Italy; SDN Foundation IRCCS, Institute of Diagnostic and Nuclear Development, Naples, Italy (S.P.); and Department of Pharmacology, Center of Translational Medicine, Temple University, Philadelphia, PA (A.C., D. Liccardo, W.J.K.)
| | - Dino Franco Vitale
- From the Division of Cardiology, Salvatore Maugeri Foundation, IRCCS, Scientific Institute of Telese Terme (BN), Italy (G.R., G.G., D.F.V., N.F.); Division of Geriatrics, Department of Translational Medical Sciences (G.R., G.P., G.D.F., V.P., A.C., D. Liccardo, K.K., G.G., M.L.D.'A., C.d.L., N.F., D. Leosco), Division of Cardiology, Department of Advanced Biomedical Sciences (P.P.F., B.T.), Federico II University of Naples, Naples, Italy; SDN Foundation IRCCS, Institute of Diagnostic and Nuclear Development, Naples, Italy (S.P.); and Department of Pharmacology, Center of Translational Medicine, Temple University, Philadelphia, PA (A.C., D. Liccardo, W.J.K.)
| | - Nicola Ferrara
- From the Division of Cardiology, Salvatore Maugeri Foundation, IRCCS, Scientific Institute of Telese Terme (BN), Italy (G.R., G.G., D.F.V., N.F.); Division of Geriatrics, Department of Translational Medical Sciences (G.R., G.P., G.D.F., V.P., A.C., D. Liccardo, K.K., G.G., M.L.D.'A., C.d.L., N.F., D. Leosco), Division of Cardiology, Department of Advanced Biomedical Sciences (P.P.F., B.T.), Federico II University of Naples, Naples, Italy; SDN Foundation IRCCS, Institute of Diagnostic and Nuclear Development, Naples, Italy (S.P.); and Department of Pharmacology, Center of Translational Medicine, Temple University, Philadelphia, PA (A.C., D. Liccardo, W.J.K.)
| | - Walter J Koch
- From the Division of Cardiology, Salvatore Maugeri Foundation, IRCCS, Scientific Institute of Telese Terme (BN), Italy (G.R., G.G., D.F.V., N.F.); Division of Geriatrics, Department of Translational Medical Sciences (G.R., G.P., G.D.F., V.P., A.C., D. Liccardo, K.K., G.G., M.L.D.'A., C.d.L., N.F., D. Leosco), Division of Cardiology, Department of Advanced Biomedical Sciences (P.P.F., B.T.), Federico II University of Naples, Naples, Italy; SDN Foundation IRCCS, Institute of Diagnostic and Nuclear Development, Naples, Italy (S.P.); and Department of Pharmacology, Center of Translational Medicine, Temple University, Philadelphia, PA (A.C., D. Liccardo, W.J.K.).
| | - Dario Leosco
- From the Division of Cardiology, Salvatore Maugeri Foundation, IRCCS, Scientific Institute of Telese Terme (BN), Italy (G.R., G.G., D.F.V., N.F.); Division of Geriatrics, Department of Translational Medical Sciences (G.R., G.P., G.D.F., V.P., A.C., D. Liccardo, K.K., G.G., M.L.D.'A., C.d.L., N.F., D. Leosco), Division of Cardiology, Department of Advanced Biomedical Sciences (P.P.F., B.T.), Federico II University of Naples, Naples, Italy; SDN Foundation IRCCS, Institute of Diagnostic and Nuclear Development, Naples, Italy (S.P.); and Department of Pharmacology, Center of Translational Medicine, Temple University, Philadelphia, PA (A.C., D. Liccardo, W.J.K.).
| |
Collapse
|
39
|
Cannavo A, Liccardo D, Lymperopoulos A, Gambino G, D'Amico ML, Rengo F, Koch WJ, Leosco D, Ferrara N, Rengo G. β Adrenergic Receptor Kinase C-Terminal Peptide Gene-Therapy Improves β2-Adrenergic Receptor-Dependent Neoangiogenesis after Hindlimb Ischemia. J Pharmacol Exp Ther 2016; 356:503-513. [PMID: 26604244 PMCID: PMC6047230 DOI: 10.1124/jpet.115.228411] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2015] [Accepted: 11/18/2015] [Indexed: 01/15/2023] Open
Abstract
After hindlimb ischemia (HI), increased catecholamine levels within the ischemic muscle can cause dysregulation of β2-adrenergic receptor (β2AR) signaling, leading to reduced revascularization. Indeed, in vivo β2AR overexpression via gene therapy enhances angiogenesis in a rat model of HI. G protein-coupled receptor kinase 2 (GRK2) is a key regulator of βAR signaling, and β adrenergic receptor kinase C-terminal peptide (βARKct), a peptide inhibitor of GRK2, has been shown to prevent βAR down-regulation and to protect cardiac myocytes and stem cells from ischemic injury through restoration of β2AR protective signaling (i.e., protein kinase B/endothelial nitric oxide synthase). Herein, we tested the potential therapeutic effects of adenoviral-mediated βARKct gene transfer in an experimental model of HI and its effects on βAR signaling and on endothelial cell (EC) function in vitro. Accordingly, in this study, we surgically induced HI in rats by femoral artery resection (FAR). Fifteen days of ischemia resulted in significant βAR down-regulation that was paralleled by an approximately 2-fold increase in GRK2 levels in the ischemic muscle. Importantly, in vivo gene transfer of the βARKct in the hindlimb of rats at the time of FAR resulted in a marked improvement of hindlimb perfusion, with increased capillary and βAR density in the ischemic muscle, compared with control groups. The effect of βARKct expression was also assessed in vitro in cultured ECs. Interestingly, ECs expressing the βARKct fenoterol, a β2AR-agonist, induced enhanced β2AR proangiogenic signaling and increased EC function. Our results suggest that βARKct gene therapy and subsequent GRK2 inhibition promotes angiogenesis in a model of HI by preventing ischemia-induced β2AR down-regulation.
Collapse
Affiliation(s)
- Alessandro Cannavo
- Division of Geriatrics, Department of Translational Medical Sciences, Federico II University of Naples, Italy (A.C., D.Li., G.G., M.L.D.A., D.Le., N.F., G.R.); Center for Translational Medicine, Temple University, Philadelphia, Pennsylvania (A.C., D.Li., W.J.K.); Department of Pharmaceutical Sciences, Nova Southeastern University College of Pharmacy, Fort Lauderdale, Florida (A.L.); Salvatore Maugeri Foundation, IRCCS, Scientific Institute of Telese Terme BN, Italy (F.R., G.R.)
| | - Daniela Liccardo
- Division of Geriatrics, Department of Translational Medical Sciences, Federico II University of Naples, Italy (A.C., D.Li., G.G., M.L.D.A., D.Le., N.F., G.R.); Center for Translational Medicine, Temple University, Philadelphia, Pennsylvania (A.C., D.Li., W.J.K.); Department of Pharmaceutical Sciences, Nova Southeastern University College of Pharmacy, Fort Lauderdale, Florida (A.L.); Salvatore Maugeri Foundation, IRCCS, Scientific Institute of Telese Terme BN, Italy (F.R., G.R.)
| | - Anastasios Lymperopoulos
- Division of Geriatrics, Department of Translational Medical Sciences, Federico II University of Naples, Italy (A.C., D.Li., G.G., M.L.D.A., D.Le., N.F., G.R.); Center for Translational Medicine, Temple University, Philadelphia, Pennsylvania (A.C., D.Li., W.J.K.); Department of Pharmaceutical Sciences, Nova Southeastern University College of Pharmacy, Fort Lauderdale, Florida (A.L.); Salvatore Maugeri Foundation, IRCCS, Scientific Institute of Telese Terme BN, Italy (F.R., G.R.)
| | - Giuseppina Gambino
- Division of Geriatrics, Department of Translational Medical Sciences, Federico II University of Naples, Italy (A.C., D.Li., G.G., M.L.D.A., D.Le., N.F., G.R.); Center for Translational Medicine, Temple University, Philadelphia, Pennsylvania (A.C., D.Li., W.J.K.); Department of Pharmaceutical Sciences, Nova Southeastern University College of Pharmacy, Fort Lauderdale, Florida (A.L.); Salvatore Maugeri Foundation, IRCCS, Scientific Institute of Telese Terme BN, Italy (F.R., G.R.)
| | - Maria Loreta D'Amico
- Division of Geriatrics, Department of Translational Medical Sciences, Federico II University of Naples, Italy (A.C., D.Li., G.G., M.L.D.A., D.Le., N.F., G.R.); Center for Translational Medicine, Temple University, Philadelphia, Pennsylvania (A.C., D.Li., W.J.K.); Department of Pharmaceutical Sciences, Nova Southeastern University College of Pharmacy, Fort Lauderdale, Florida (A.L.); Salvatore Maugeri Foundation, IRCCS, Scientific Institute of Telese Terme BN, Italy (F.R., G.R.)
| | - Franco Rengo
- Division of Geriatrics, Department of Translational Medical Sciences, Federico II University of Naples, Italy (A.C., D.Li., G.G., M.L.D.A., D.Le., N.F., G.R.); Center for Translational Medicine, Temple University, Philadelphia, Pennsylvania (A.C., D.Li., W.J.K.); Department of Pharmaceutical Sciences, Nova Southeastern University College of Pharmacy, Fort Lauderdale, Florida (A.L.); Salvatore Maugeri Foundation, IRCCS, Scientific Institute of Telese Terme BN, Italy (F.R., G.R.)
| | - Walter J Koch
- Division of Geriatrics, Department of Translational Medical Sciences, Federico II University of Naples, Italy (A.C., D.Li., G.G., M.L.D.A., D.Le., N.F., G.R.); Center for Translational Medicine, Temple University, Philadelphia, Pennsylvania (A.C., D.Li., W.J.K.); Department of Pharmaceutical Sciences, Nova Southeastern University College of Pharmacy, Fort Lauderdale, Florida (A.L.); Salvatore Maugeri Foundation, IRCCS, Scientific Institute of Telese Terme BN, Italy (F.R., G.R.)
| | - Dario Leosco
- Division of Geriatrics, Department of Translational Medical Sciences, Federico II University of Naples, Italy (A.C., D.Li., G.G., M.L.D.A., D.Le., N.F., G.R.); Center for Translational Medicine, Temple University, Philadelphia, Pennsylvania (A.C., D.Li., W.J.K.); Department of Pharmaceutical Sciences, Nova Southeastern University College of Pharmacy, Fort Lauderdale, Florida (A.L.); Salvatore Maugeri Foundation, IRCCS, Scientific Institute of Telese Terme BN, Italy (F.R., G.R.)
| | - Nicola Ferrara
- Division of Geriatrics, Department of Translational Medical Sciences, Federico II University of Naples, Italy (A.C., D.Li., G.G., M.L.D.A., D.Le., N.F., G.R.); Center for Translational Medicine, Temple University, Philadelphia, Pennsylvania (A.C., D.Li., W.J.K.); Department of Pharmaceutical Sciences, Nova Southeastern University College of Pharmacy, Fort Lauderdale, Florida (A.L.); Salvatore Maugeri Foundation, IRCCS, Scientific Institute of Telese Terme BN, Italy (F.R., G.R.)
| | - Giuseppe Rengo
- Division of Geriatrics, Department of Translational Medical Sciences, Federico II University of Naples, Italy (A.C., D.Li., G.G., M.L.D.A., D.Le., N.F., G.R.); Center for Translational Medicine, Temple University, Philadelphia, Pennsylvania (A.C., D.Li., W.J.K.); Department of Pharmaceutical Sciences, Nova Southeastern University College of Pharmacy, Fort Lauderdale, Florida (A.L.); Salvatore Maugeri Foundation, IRCCS, Scientific Institute of Telese Terme BN, Italy (F.R., G.R.)
| |
Collapse
|
40
|
β2- and β1-Adrenoceptor Expression Exhibits a Common Regulatory Pattern With GRK2 and GRK5 in Human and Animal Models of Cardiovascular Diseases. J Cardiovasc Pharmacol 2015; 66:478-86. [DOI: 10.1097/fjc.0000000000000299] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
41
|
McCrink KA, Brill A, Lymperopoulos A. Adrenal G protein-coupled receptor kinase-2 in regulation of sympathetic nervous system activity in heart failure. World J Cardiol 2015; 7:539-543. [PMID: 26413230 PMCID: PMC4577680 DOI: 10.4330/wjc.v7.i9.539] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Revised: 06/24/2015] [Accepted: 07/11/2015] [Indexed: 02/06/2023] Open
Abstract
Heart failure (HF), the number one cause of death in the western world, is caused by the insufficient performance of the heart leading to tissue underperfusion in response to an injury or insult. It comprises complex interactions between important neurohormonal mechanisms that try but ultimately fail to sustain cardiac output. The most prominent such mechanism is the sympathetic (adrenergic) nervous system (SNS), whose activity and outflow are greatly elevated in HF. SNS hyperactivity confers significant toxicity to the failing heart and markedly increases HF morbidity and mortality via excessive activation of adrenergic receptors, which are G protein-coupled receptors. Thus, ligand binding induces their coupling to heterotrimeric G proteins that transduce intracellular signals. G protein signaling is turned-off by the agonist-bound receptor phosphorylation courtesy of G protein-coupled receptor kinases (GRKs), followed by βarrestin binding, which prevents the GRK-phosphorylated receptor from further interaction with the G proteins and simultaneously leads it inside the cell (receptor sequestration). Recent evidence indicates that adrenal GRK2 and βarrestins can regulate adrenal catecholamine secretion, thereby modulating SNS activity in HF. The present review gives an account of all these studies on adrenal GRKs and βarrestins in HF and discusses the exciting new therapeutic possibilities for chronic HF offered by targeting these proteins pharmacologically.
Collapse
Affiliation(s)
- Katie A McCrink
- Katie A McCrink, Ava Brill, Anastasios Lymperopoulos, Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences, Nova Southeastern University College of Pharmacy, Ft. Lauderdale, FL 33328-2018, United States
| | - Ava Brill
- Katie A McCrink, Ava Brill, Anastasios Lymperopoulos, Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences, Nova Southeastern University College of Pharmacy, Ft. Lauderdale, FL 33328-2018, United States
| | - Anastasios Lymperopoulos
- Katie A McCrink, Ava Brill, Anastasios Lymperopoulos, Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences, Nova Southeastern University College of Pharmacy, Ft. Lauderdale, FL 33328-2018, United States
| |
Collapse
|
42
|
Capote LA, Mendez Perez R, Lymperopoulos A. GPCR signaling and cardiac function. Eur J Pharmacol 2015; 763:143-148. [PMID: 25981298 DOI: 10.1016/j.ejphar.2015.05.019] [Citation(s) in RCA: 116] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 03/30/2015] [Accepted: 05/11/2015] [Indexed: 12/27/2022]
Abstract
G protein-coupled receptors (GPCRs), such as β-adrenergic and angiotensin II receptors, located in the membranes of all three major cardiac cell types, i.e. myocytes, fibroblasts and endothelial cells, play crucial roles in regulating cardiac function and morphology. Their importance in cardiac physiology and disease is reflected by the fact that, collectively, they represent the direct targets of over a third of the currently approved cardiovascular drugs used in clinical practice. Over the past few decades, advances in elucidation of their structure, function and the signaling pathways they elicit, specifically in the heart, have led to identification of an increasing number of new molecular targets for heart disease therapy. Here, we review these signaling modalities employed by GPCRs known to be expressed in the cardiac myocyte membranes and to directly modulate cardiac contractility. We also highlight drugs and drug classes that directly target these GPCRs to modulate cardiac function, as well as molecules involved in cardiac GPCR signaling that have the potential of becoming novel drug targets for modulation of cardiac function in the future.
Collapse
Affiliation(s)
- Leany A Capote
- From the Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences, Nova Southeastern University College of Pharmacy, Fort Lauderdale, FL 33328, USA
| | - Roberto Mendez Perez
- From the Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences, Nova Southeastern University College of Pharmacy, Fort Lauderdale, FL 33328, USA
| | - Anastasios Lymperopoulos
- From the Laboratory for the Study of Neurohormonal Control of the Circulation, Department of Pharmaceutical Sciences, Nova Southeastern University College of Pharmacy, Fort Lauderdale, FL 33328, USA.
| |
Collapse
|
43
|
Thaung HPA, Baldi JC, Wang HY, Hughes G, Cook RF, Bussey CT, Sheard PW, Bahn A, Jones PP, Schwenke DO, Lamberts RR. Increased Efferent Cardiac Sympathetic Nerve Activity and Defective Intrinsic Heart Rate Regulation in Type 2 Diabetes. Diabetes 2015; 64:2944-56. [PMID: 25784543 DOI: 10.2337/db14-0955] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Accepted: 03/09/2015] [Indexed: 11/13/2022]
Abstract
Elevated sympathetic nerve activity (SNA) coupled with dysregulated β-adrenoceptor (β-AR) signaling is postulated as a major driving force for cardiac dysfunction in patients with type 2 diabetes; however, cardiac SNA has never been assessed directly in diabetes. Our aim was to measure the sympathetic input to and the β-AR responsiveness of the heart in the type 2 diabetic heart. In vivo recording of SNA of the left efferent cardiac sympathetic branch of the stellate ganglion in Zucker diabetic fatty rats revealed an elevated resting cardiac SNA and doubled firing rate compared with nondiabetic rats. Ex vivo, in isolated denervated hearts, the intrinsic heart rate was markedly reduced. Contractile and relaxation responses to β-AR stimulation with dobutamine were compromised in externally paced diabetic hearts, but not in diabetic hearts allowed to regulate their own heart rate. Protein levels of left ventricular β1-AR and Gs (guanine nucleotide binding protein stimulatory) were reduced, whereas left ventricular and right atrial β2-AR and Gi (guanine nucleotide binding protein inhibitory regulatory) levels were increased. The elevated resting cardiac SNA in type 2 diabetes, combined with the reduced cardiac β-AR responsiveness, suggests that the maintenance of normal cardiovascular function requires elevated cardiac sympathetic input to compensate for changes in the intrinsic properties of the diabetic heart.
Collapse
Affiliation(s)
- H P Aye Thaung
- HeartOtago, Department of Physiology, Otago School of Medical Sciences, University of Otago, Dunedin, New Zealand
| | - J Chris Baldi
- Department of Medicine, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Heng-Yu Wang
- HeartOtago, Department of Physiology, Otago School of Medical Sciences, University of Otago, Dunedin, New Zealand
| | - Gillian Hughes
- HeartOtago, Department of Physiology, Otago School of Medical Sciences, University of Otago, Dunedin, New Zealand
| | - Rosalind F Cook
- HeartOtago, Department of Physiology, Otago School of Medical Sciences, University of Otago, Dunedin, New Zealand
| | - Carol T Bussey
- HeartOtago, Department of Physiology, Otago School of Medical Sciences, University of Otago, Dunedin, New Zealand
| | - Phil W Sheard
- HeartOtago, Department of Physiology, Otago School of Medical Sciences, University of Otago, Dunedin, New Zealand
| | - Andrew Bahn
- HeartOtago, Department of Physiology, Otago School of Medical Sciences, University of Otago, Dunedin, New Zealand
| | - Peter P Jones
- HeartOtago, Department of Physiology, Otago School of Medical Sciences, University of Otago, Dunedin, New Zealand
| | - Daryl O Schwenke
- HeartOtago, Department of Physiology, Otago School of Medical Sciences, University of Otago, Dunedin, New Zealand
| | - Regis R Lamberts
- HeartOtago, Department of Physiology, Otago School of Medical Sciences, University of Otago, Dunedin, New Zealand
| |
Collapse
|
44
|
Alterations of left ventricular deformation and cardiac sympathetic derangement in patients with systolic heart failure: a 3D speckle tracking echocardiography and cardiac ¹²³I-MIBG study. Eur J Nucl Med Mol Imaging 2015; 42:1601-11. [PMID: 25947572 DOI: 10.1007/s00259-015-3054-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Accepted: 03/19/2015] [Indexed: 12/29/2022]
Abstract
PURPOSE Myocardial contractile function is under the control of cardiac sympathetic activity. Three-dimensional speckle tracking echocardiography (3D-STE) and cardiac imaging with (123)I-metaiodobenzylguanidine ((123)I-MIBG) are two sophisticated techniques for the assessment of left ventricular (LV) deformation and sympathetic innervation, respectively, which offer important prognostic information in patients with heart failure (HF). The purpose of this investigation was to explore, in patients with systolic HF, the relationship between LV deformation assessed by 3D-STE and cardiac sympathetic derangement evaluated by (123)I-MIBG imaging. METHODS We prospectively studied 75 patients with systolic HF. All patients underwent a 3D-STE study (longitudinal, circumferential, area and radial) and (123)I-MIBG planar and SPECT cardiac imaging. RESULTS 3D-STE longitudinal, circumferential and area strain values were correlated with (123)I-MIBG late heart to mediastinum (H/M) ratio and late SPECT total defect score. After stratification of the patients according to ischaemic or nonischaemic HF aetiology, we observed a good correlation of all 3D-STE measurements with late H/M ratio and SPECT data in the ischaemic group, but in patients with HF of nonischaemic aetiology, no correlation was found between LV deformation and cardiac sympathetic activity. At the regional level, the strongest correlation between LV deformation and adrenergic innervation was found for the left anterior descending coronary artery distribution territory for all four 3D-STE values. In multivariate linear regression analyses, including age, gender, LV ejection fraction, NYHA class, body mass index, heart rate and HF aetiology, only 3D-STE area and radial strain values significantly predicted cardiac sympathetic derangement on (123)I-MIBG late SPECT. CONCLUSION This study indicated that 3D-STE measurements are correlated with (123)I-MIBG planar and SPECT data. Furthermore, 3D-STE area and radial strain values, but not LVEF, predict cardiac sympathetic derangement in human postischaemic HF.
Collapse
|
45
|
Malik S, deRubio RG, Trembley M, Irannejad R, Wedegaertner PB, Smrcka AV. G protein βγ subunits regulate cardiomyocyte hypertrophy through a perinuclear Golgi phosphatidylinositol 4-phosphate hydrolysis pathway. Mol Biol Cell 2015; 26:1188-98. [PMID: 25609085 PMCID: PMC4357516 DOI: 10.1091/mbc.e14-10-1476] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Gβγ regulation of the perinuclear Golgi PI4P pathway and a separate pathway at the PM is required for ET-1–stimulated hypertrophy, and the efficacy of Gβγ inhibition in preventing heart failure may be due, in part, to its blocking both of these pathways. We recently identified a novel GPCR-dependent pathway for regulation of cardiac hypertrophy that depends on Golgi phosphatidylinositol 4-phosphate (PI4P) hydrolysis by a specific isoform of phospholipase C (PLC), PLCε, at the nuclear envelope. How stimuli are transmitted from cell surface GPCRs to activation of perinuclear PLCε is not clear. Here we tested the role of G protein βγ subunits. Gβγ inhibition blocked ET-1–stimulated Golgi PI4P depletion in neonatal and adult ventricular myocytes. Blocking Gβγ at the Golgi inhibited ET-1–dependent PI4P depletion and nuclear PKD activation. Translocation of Gβγ to the Golgi stimulated perinuclear Golgi PI4P depletion and nuclear PKD activation. Finally, blocking Gβγ at the Golgi or PM blocked ET-1–dependent cardiomyocyte hypertrophy. These data indicate that Gβγ regulation of the perinuclear Golgi PI4P pathway and a separate pathway at the PM is required for ET-1–stimulated hypertrophy, and the efficacy of Gβγ inhibition in preventing heart failure maybe due in part to its blocking both these pathways.
Collapse
Affiliation(s)
- S Malik
- Department of Pharmacology and Physiology, University of Rochester, Rochester, NY 14642
| | - R G deRubio
- Department of Pharmacology and Physiology, University of Rochester, Rochester, NY 14642
| | - M Trembley
- Department of Pharmacology and Physiology, University of Rochester, Rochester, NY 14642
| | - R Irannejad
- Department of Psychiatry, University of California, San Francisco, San Francisco, CA 94158
| | - P B Wedegaertner
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, PA 19107
| | - A V Smrcka
- Department of Pharmacology and Physiology, University of Rochester, Rochester, NY 14642
| |
Collapse
|
46
|
Woo AYH, Song Y, Xiao RP, Zhu W. Biased β2-adrenoceptor signalling in heart failure: pathophysiology and drug discovery. Br J Pharmacol 2014; 172:5444-56. [PMID: 25298054 DOI: 10.1111/bph.12965] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Revised: 08/27/2014] [Accepted: 09/28/2014] [Indexed: 12/27/2022] Open
Abstract
The body is constantly faced with a dynamic requirement for blood flow. The heart is able to respond to these changing needs by adjusting cardiac output based on cues emitted by circulating catecholamine levels. Cardiac β-adrenoceptors transduce the signal produced by catecholamine stimulation via Gs proteins to their downstream effectors to increase heart contractility. During heart failure, cardiac output is insufficient to meet the needs of the body; catecholamine levels are high and β-adrenoceptors become hyperstimulated. The hyperstimulated β1-adrenoceptors induce a cardiotoxic effect, which could be counteracted by the cardioprotective effect of β2-adrenoceptor-mediated Gi signalling. However, β2-adrenoceptor-Gi signalling negates the stimulatory effect of the Gs signalling on cardiomyocyte contraction and further exacerbates cardiodepression. Here, further to the localization of β1- and β2-adrenoceptors and β2-adrenoceptor-mediated β-arrestin signalling in cardiomyocytes, we discuss features of the dysregulation of β-adrenoceptor subtype signalling in the failing heart, and conclude that Gi-biased β2-adrenoceptor signalling is a pathogenic pathway in heart failure that plays a crucial role in cardiac remodelling. In contrast, β2-adrenoceptor-Gs signalling increases cardiomyocyte contractility without causing cardiotoxicity. Finally, we discuss a novel therapeutic approach for heart failure using a Gs-biased β2-adrenoceptor agonist and a β1-adrenoceptor antagonist in combination. This combination treatment normalizes the β-adrenoceptor subtype signalling in the failing heart and produces therapeutic effects that outperform traditional heart failure therapies in animal models. The present review illustrates how the concept of biased signalling can be applied to increase our understanding of the pathophysiology of diseases and in the development of novel therapies.
Collapse
Affiliation(s)
- Anthony Yiu-Ho Woo
- Institute of Molecular Medicine, Centre for Life Sciences, Peking University, Beijing, China.,Key Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Ying Song
- Institute of Molecular Medicine, Centre for Life Sciences, Peking University, Beijing, China
| | - Rui-Ping Xiao
- Institute of Molecular Medicine, Centre for Life Sciences, Peking University, Beijing, China.,Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, China
| | - Weizhong Zhu
- Department of Pharmacology, Nantong University School of Pharmacy, Nantong, China
| |
Collapse
|
47
|
Kaldara E, Sanoudou D, Adamopoulos S, Nanas JN. Outpatient management of chronic heart failure. Expert Opin Pharmacother 2014; 16:17-41. [PMID: 25480690 DOI: 10.1517/14656566.2015.978286] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Heart failure (HF) treatment attracts a share of intensive research because of its poor HF prognosis. In the past decades, the prognosis of HF has improved considerably, mainly as a consequence of the progress that has been made in the pharmacological management of HF. AREAS COVERED This article reviews the outpatient pharmacological management of chronic HF due to left ventricular systolic dysfunction and offers recommendations on the use of various drugs. In addition, the present article attempts to provide practical therapeutic algorithms based on current clinical strategies. EXPERT OPINION Continued research directed toward identifying factors associated with high pharmacotherapy guideline adherence and understanding of variants that influence response to drugs will hopefully halt or reverse the major pathophysiological mechanisms involved in this syndrome.
Collapse
Affiliation(s)
- Elisabeth Kaldara
- University of Athens, Medical School, 3rd Cardiology Department , Mikras Asias 67, 11527 Attiki, Athens , Greece +30 2108236877 ; +30 2107789901 ;
| | | | | | | |
Collapse
|
48
|
Rivas V, Nogués L, Reglero C, Mayor F, Penela P. Role of G protein-coupled receptor kinase 2 in tumoral angiogenesis. Mol Cell Oncol 2014; 1:e969166. [PMID: 27308373 PMCID: PMC4905215 DOI: 10.4161/23723548.2014.969166] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Revised: 08/22/2014] [Accepted: 08/23/2014] [Indexed: 12/26/2022]
Abstract
Downregulation of G protein-coupled receptor kinase 2 (GRK2) in endothelial cells has recently been identified as a relevant event in the tumoral angiogenic switch. Based on the effects of altering GRK2 dosage in cell and animal models, this kinase appears to act as a hub in key signaling pathways involved in vascular stabilization and remodeling. Accordingly, decreased GRK2 expression in endothelial cells accelerates tumor growth in mice by impairing the pericytes ensheathing the vessels, thereby promoting hypoxia and macrophage infiltration. These results raise new questions regarding the mechanisms by which transformed cells trigger the decrease in GRK2 observed in human breast cancer vessels and how GRK2 modulates the interactions between different cell types that occur in the tumor microenvironment.
Collapse
Affiliation(s)
- Verónica Rivas
- Departamento de Biología Molecular and Centro de Biología Molecular "Severo Ochoa" (Consejo Superior de Investigaciones Científicas - Universidad Autónoma de Madrid); Universidad Autónoma de Madrid; Madrid, Spain; Instituto de Investigación Sanitaria La Princesa; Madrid, Spain
| | - Laura Nogués
- Departamento de Biología Molecular and Centro de Biología Molecular "Severo Ochoa" (Consejo Superior de Investigaciones Científicas - Universidad Autónoma de Madrid); Universidad Autónoma de Madrid; Madrid, Spain; Instituto de Investigación Sanitaria La Princesa; Madrid, Spain
| | - Clara Reglero
- Departamento de Biología Molecular and Centro de Biología Molecular "Severo Ochoa" (Consejo Superior de Investigaciones Científicas - Universidad Autónoma de Madrid); Universidad Autónoma de Madrid; Madrid, Spain; Instituto de Investigación Sanitaria La Princesa; Madrid, Spain
| | - Federico Mayor
- Departamento de Biología Molecular and Centro de Biología Molecular "Severo Ochoa" (Consejo Superior de Investigaciones Científicas - Universidad Autónoma de Madrid); Universidad Autónoma de Madrid; Madrid, Spain; Instituto de Investigación Sanitaria La Princesa; Madrid, Spain
| | - Petronila Penela
- Departamento de Biología Molecular and Centro de Biología Molecular "Severo Ochoa" (Consejo Superior de Investigaciones Científicas - Universidad Autónoma de Madrid); Universidad Autónoma de Madrid; Madrid, Spain; Instituto de Investigación Sanitaria La Princesa; Madrid, Spain
| |
Collapse
|
49
|
Femminella GD, Barrese V, Ferrara N, Rengo G. Tailoring therapy for heart failure: the pharmacogenomics of adrenergic receptor signaling. PHARMACOGENOMICS & PERSONALIZED MEDICINE 2014; 7:267-73. [PMID: 25276090 PMCID: PMC4175026 DOI: 10.2147/pgpm.s49799] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Heart failure is one of the leading causes of mortality in Western countries, and β-blockers are a cornerstone of its treatment. However, the response to these drugs is variable among individuals, which might be explained, at least in part, by genetic differences. Pharmacogenomics is the study of genetic contributions to drug response variability in order to provide evidence for a tailored therapy in an individual patient. Several studies have investigated the pharmacogenomics of the adrenergic receptor system and its role in the context of the use of β-blockers in treating heart failure. In this review, we will focus on the most significant polymorphisms described in the literature involving adrenergic receptors and adrenergic receptor-related proteins, as well as genetic variations influencing β-blocker metabolism.
Collapse
Affiliation(s)
| | - Vincenzo Barrese
- Department of Neuroscience, Reproductive Science and Odontostomatology, Federico II University, Naples, Italy ; Division of Biomedical Sciences, St George's University of London, London, UK
| | - Nicola Ferrara
- Department of Translational Medical Sciences, Federico II University, Naples, Italy ; "Salvatore Maugeri" Foundation - IRCCS - Scientific Institute of Telese Terme, Telese Terme, Benevento, Italy
| | - Giuseppe Rengo
- "Salvatore Maugeri" Foundation - IRCCS - Scientific Institute of Telese Terme, Telese Terme, Benevento, Italy
| |
Collapse
|
50
|
Abstract
The pathophysiology of heart failure (HF) is characterized by hemodynamic abnormalities that result in neurohormonal activation and autonomic imbalance with increase in sympathetic activity and withdrawal of vagal activity. Alterations in receptor activation from this autonomic imbalance may have profound effects on cardiac function and structure. Inhibition of the sympathetic drive to the heart through β-receptor blockade has become a standard component of therapy for HF with a dilated left ventricle because of its effectiveness in inhibiting the ventricular structural remodeling process and in prolonging life. Several devices for selective modulation of sympathetic and vagal activity have recently been developed in an attempt to alter the natural history of HF. The optimal counteraction of the excessive sympathetic activity is still unclear. A profound decrease in adrenergic support with excessive blockade of the sympathetic nervous system may result in adverse outcomes in clinical HF. In this review, we analyze the data supporting a contributory role of the autonomic functional alterations on the course of HF, the techniques used to assess autonomic nervous system activity, the evidence for clinical effectiveness of pharmacological and device interventions, and the potential future role of autonomic nervous system modifiers in the management of this syndrome.
Collapse
Affiliation(s)
- Viorel G Florea
- From the Minneapolis VA Health Care System, Section of Cardiology (V.G.F.) and Rasmussen Center for Cardiovascular Disease Prevention, Department of Medicine (J.N.C.), University of Minnesota Medical School
| | - Jay N Cohn
- From the Minneapolis VA Health Care System, Section of Cardiology (V.G.F.) and Rasmussen Center for Cardiovascular Disease Prevention, Department of Medicine (J.N.C.), University of Minnesota Medical School.
| |
Collapse
|