1
|
Xing Y, Tian T, Zhang X, Yang D, Zhang C, Wang M, Wang Y, Luo T, Wang Z, Wang H, Li H. ENDOGENOUS β 3 -ADRENERGIC RECEPTOR ACTIVATION ALLEVIATES SEPSIS-INDUCED CARDIOMYOCYTE APOPTOSIS VIA PI3K/AKT SIGNALING PATHWAY. Shock 2024; 61:915-923. [PMID: 38662592 DOI: 10.1097/shk.0000000000002354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
ABSTRACT β 3 -adrenergic receptor (β 3 -AR) has been proposed as a new therapy for several myocardial diseases. However, the effect of β 3 -AR activation on sepsis-induced myocardial apoptosis is unclear. Here, we investigated the effect of β 3 -AR activation on the cardiomyocyte apoptosis and cardiac dysfunction in cecal ligation and puncture (CLP)-operated rats and lipopolysaccharide (LPS)-treated cardiomyocytes. We found that β 3 -AR existed both in adult rat ventricular myocytes (ARVMs) and H9c2 cells. The expression of β 3 -AR was upregulated in LPS-treated ARVMs and the heart of CLP rats. Pretreatment with β 3 -AR agonist, BRL37344, inhibited LPS-induced cardiomyocyte apoptosis and caspase-3, -8, and -9 activation in ARVMs. BRL37344 also reduced apoptosis and increased the protein levels of PI3K, p-Akt Ser473 and p-eNOS Ser1177 in LPS-treated H9c2 cells. Inhibition of PI3K using LY294002 abolished the inhibitory effect of BRL37344 on LPS-induced caspase-3, -8, and -9 activation in H9c2 cells. Furthermore, administration of β 3 -AR antagonist, SR59230A (5 mg/kg), significantly decreased the maximum rate of left ventricular pressure rise (+dP/dt) in CLP-induced septic rats. SR59230A not only increased myocardial apoptosis, reduced p-Akt Ser473 and Bcl-2 contents, but also increased mitochondrial Bax, cytoplasm cytochrome c, cleaved caspase-9, and cleaved caspase-3 levels of the myocardium in septic rats. These results suggest that endogenous β 3 -AR activation alleviates sepsis-induced cardiomyocyte apoptosis via PI3K/Akt signaling pathway and maintains intrinsic myocardial systolic function in sepsis.
Collapse
Affiliation(s)
- Yun Xing
- Department of Pathophysiology, Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, Guangzhou, China
| | - Tian Tian
- Department of Pathophysiology, Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, Guangzhou, China
| | - Xue Zhang
- Department of Pathophysiology, Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, Guangzhou, China
| | - Duomeng Yang
- Department of Pathophysiology, Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, Guangzhou, China
| | - Chanjuan Zhang
- Department of Pathophysiology, Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, Guangzhou, China
| | - Miao Wang
- Department of Pathophysiology, Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, Guangzhou, China
| | - Yiyang Wang
- Department of Pathophysiology, Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, Guangzhou, China
| | - Tao Luo
- Department of Pathophysiology, Zhuhai Campus of Zunyi Medical University, Zhuhai, China
| | - Zhi Wang
- Key Laboratory of Occupational Environment and Health, Guangzhou Twelfth People's Hospital, Guangzhou, China
| | - Huadong Wang
- Department of Pathophysiology, Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, Guangzhou, China
| | - Hongmei Li
- Department of Pathophysiology, Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, Guangzhou, China
| |
Collapse
|
2
|
Odnoshivkina JG, Averin AS, Khakimov IR, Trusov NA, Trusova DA, Petrov AM. The mechanism of 25-hydroxycholesterol-mediated suppression of atrial β1-adrenergic responses. Pflugers Arch 2024; 476:407-421. [PMID: 38253680 DOI: 10.1007/s00424-024-02913-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 12/27/2023] [Accepted: 01/14/2024] [Indexed: 01/24/2024]
Abstract
25-Hydroxycholesterol (25HC) is a biologically active oxysterol, whose production greatly increases during inflammation by macrophages and dendritic cells. The inflammatory reactions are frequently accompanied by changes in heart regulation, such as blunting of the cardiac β-adrenergic receptor (AR) signaling. Here, the mechanism of 25HC-dependent modulation of responses to β-AR activation was studied in the atria of mice. 25HC at the submicromolar levels decreased the β-AR-mediated positive inotropic effect and enhancement of the Ca2+ transient amplitude, without changing NO production. Positive inotropic responses to β1-AR (but not β2-AR) activation were markedly attenuated by 25HC. The depressant action of 25HC on the β1-AR-mediated responses was prevented by selective β3-AR antagonists as well as inhibitors of Gi protein, Gβγ, G protein-coupled receptor kinase 2/3, or β-arrestin. Simultaneously, blockers of protein kinase D and C as well as a phosphodiesterase inhibitor did not preclude the negative action of 25HC on the inotropic response to β-AR activation. Thus, 25HC can suppress the β1-AR-dependent effects via engaging β3-AR, Gi protein, Gβγ, G protein-coupled receptor kinase, and β-arrestin. This 25HC-dependent mechanism can contribute to the inflammatory-related alterations in the atrial β-adrenergic signaling.
Collapse
Affiliation(s)
- Julia G Odnoshivkina
- Kazan State Medical University, 49 Butlerova St, Kazan, RT, Russia, 420012
- Laboratory of Biophysics of Synaptic Processes, Kazan Institute of Biochemistry and Biophysics, FRC Kazan Scientific Center of RAS, 2/31 Lobachevsky St, Kazan, RT, Russia, 420111
| | - Alexey S Averin
- Institute of Cell Biophysics, Federal Research Center "Pushchino Scientific Center of Biological Research", Pushchino Branch, Russian Academy of Sciences, Pushchino, 142290, Russia
| | - Ildar R Khakimov
- Kazan State Medical University, 49 Butlerova St, Kazan, RT, Russia, 420012
| | - Nazar A Trusov
- Kazan State Medical University, 49 Butlerova St, Kazan, RT, Russia, 420012
| | - Diliara A Trusova
- Kazan State Medical University, 49 Butlerova St, Kazan, RT, Russia, 420012
| | - Alexey M Petrov
- Kazan State Medical University, 49 Butlerova St, Kazan, RT, Russia, 420012.
- Laboratory of Biophysics of Synaptic Processes, Kazan Institute of Biochemistry and Biophysics, FRC Kazan Scientific Center of RAS, 2/31 Lobachevsky St, Kazan, RT, Russia, 420111.
- Kazan Federal University, 18 Kremlyovskaya Street, Kazan, Russia, 420008.
| |
Collapse
|
3
|
Liu Y, Shao Q, Cheng HJ, Li T, Zhang X, Callahan MF, Herrington D, Kitzman D, Zhao D, Cheng CP. Chronic Ca 2+/Calmodulin-Dependent Protein Kinase II Inhibition Rescues Advanced Heart Failure. J Pharmacol Exp Ther 2021; 377:316-325. [PMID: 33722881 PMCID: PMC8140392 DOI: 10.1124/jpet.120.000361] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 03/11/2021] [Indexed: 11/22/2022] Open
Abstract
Ca2+/calmodulin-dependent protein kinase II (CaMKII) is upregulated in congestive heart failure (CHF), contributing to electrical, structural, and functional remodeling. CaMKII inhibition is known to improve CHF, but its direct cardiac effects in CHF remain unclear. We hypothesized that CaMKII inhibition improves cardiomyocyte function, [Ca2+]i regulation, and β-adrenergic reserve, thus improving advanced CHF. In a 16-week study, we compared plasma neurohormonal levels and left ventricular (LV)- and myocyte-functional and calcium transient ([Ca2+]iT) responses in male Sprague-Dawley rats (10/group) with CHF induced by isoproterenol (170 mg/kg sq for 2 days). In rats with CHF, we studied the effects of the CaMKII inhibitor KN-93 or its inactive analog KN-92 (n = 4) (70 µg/kg per day, mini-pump) for 4 weeks. Compared with controls, isoproterenol-treated rats had severe CHF with 5-fold-increased plasma norepinephrine and about 50% decreases in ejection fraction (EF) and LV contractility [slope of LV end-systolic pressure-LV end-systolic volume relation (EES)] but increased time constant of LV relaxation (τ). They also showed significantly reduced myocyte contraction [maximum rate of myocyte shortening (dL/dtmax)], relaxation (dL/dtmax), and [Ca2+]iT Isoproterenol superfusion caused significantly fewer increases in dL/dtmax and [Ca2+]iT KN-93 treatment prevented plasma norepinephrine elevation, with increased basal and acute isoproterenol-stimulated increases in EF and EES and decreased τ in CHF. KN-93 treatment preserved normal myocyte contraction, relaxation, [Ca2+]iT, and β-adrenergic reserve, whereas KN-92 treatment failed to improve LV and myocyte function, and plasma norepinephrine remained high in CHF. Thus, chronic CaMKII inhibition prevented CHF-induced activation of the sympathetic nervous system, restoring normal LV and cardiomyocyte basal and β-adrenergic-stimulated contraction, relaxation, and [Ca2+]iT, thereby playing a rescue role in advanced CHF. SIGNIFICANCE STATEMENT: We investigated the therapeutic efficacy of late initiation of chronic Ca2+/calmodulin-dependent protein kinase II (CaMKII) inhibition on progression of advanced congestive heart failure (CHF). Chronic CaMKII inhibition prevented CHF-induced activation of the sympathetic nervous system and restored normal intrinsic cardiomyocyte basal and β-adrenergic receptor-stimulated relaxation, contraction, and [Ca2+]i regulation, leading to reversal of CHF progression. These data provide new evidence that CaMKII inhibition is able and sufficient to rescue a failing heart, and thus cardiac CaMKII inhibition is a promising target for improving CHF treatment.
Collapse
Affiliation(s)
- Yixi Liu
- Department of Cardiology, the First Affiliated Hospital of Kunming Medical University, Kunming, China (Y.L.); Department of Cardiology, Harbin Medical University Cancer Hospital, Harbin, China (Q.S.); Department of Internal Medicine, Cardiovascular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina (Y.L., Q.S., H.-J.C., T.L., X.Z., M.F.C., D.H., D.K., D.Z., C.-P.C.); Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin, China (T.L.); and Department of Cardiology, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China (X.Z.)
| | - Qun Shao
- Department of Cardiology, the First Affiliated Hospital of Kunming Medical University, Kunming, China (Y.L.); Department of Cardiology, Harbin Medical University Cancer Hospital, Harbin, China (Q.S.); Department of Internal Medicine, Cardiovascular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina (Y.L., Q.S., H.-J.C., T.L., X.Z., M.F.C., D.H., D.K., D.Z., C.-P.C.); Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin, China (T.L.); and Department of Cardiology, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China (X.Z.)
| | - Heng-Jie Cheng
- Department of Cardiology, the First Affiliated Hospital of Kunming Medical University, Kunming, China (Y.L.); Department of Cardiology, Harbin Medical University Cancer Hospital, Harbin, China (Q.S.); Department of Internal Medicine, Cardiovascular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina (Y.L., Q.S., H.-J.C., T.L., X.Z., M.F.C., D.H., D.K., D.Z., C.-P.C.); Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin, China (T.L.); and Department of Cardiology, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China (X.Z.)
| | - Tiankai Li
- Department of Cardiology, the First Affiliated Hospital of Kunming Medical University, Kunming, China (Y.L.); Department of Cardiology, Harbin Medical University Cancer Hospital, Harbin, China (Q.S.); Department of Internal Medicine, Cardiovascular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina (Y.L., Q.S., H.-J.C., T.L., X.Z., M.F.C., D.H., D.K., D.Z., C.-P.C.); Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin, China (T.L.); and Department of Cardiology, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China (X.Z.)
| | - Xiaowei Zhang
- Department of Cardiology, the First Affiliated Hospital of Kunming Medical University, Kunming, China (Y.L.); Department of Cardiology, Harbin Medical University Cancer Hospital, Harbin, China (Q.S.); Department of Internal Medicine, Cardiovascular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina (Y.L., Q.S., H.-J.C., T.L., X.Z., M.F.C., D.H., D.K., D.Z., C.-P.C.); Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin, China (T.L.); and Department of Cardiology, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China (X.Z.)
| | - Michael F Callahan
- Department of Cardiology, the First Affiliated Hospital of Kunming Medical University, Kunming, China (Y.L.); Department of Cardiology, Harbin Medical University Cancer Hospital, Harbin, China (Q.S.); Department of Internal Medicine, Cardiovascular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina (Y.L., Q.S., H.-J.C., T.L., X.Z., M.F.C., D.H., D.K., D.Z., C.-P.C.); Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin, China (T.L.); and Department of Cardiology, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China (X.Z.)
| | - David Herrington
- Department of Cardiology, the First Affiliated Hospital of Kunming Medical University, Kunming, China (Y.L.); Department of Cardiology, Harbin Medical University Cancer Hospital, Harbin, China (Q.S.); Department of Internal Medicine, Cardiovascular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina (Y.L., Q.S., H.-J.C., T.L., X.Z., M.F.C., D.H., D.K., D.Z., C.-P.C.); Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin, China (T.L.); and Department of Cardiology, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China (X.Z.)
| | - Dalane Kitzman
- Department of Cardiology, the First Affiliated Hospital of Kunming Medical University, Kunming, China (Y.L.); Department of Cardiology, Harbin Medical University Cancer Hospital, Harbin, China (Q.S.); Department of Internal Medicine, Cardiovascular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina (Y.L., Q.S., H.-J.C., T.L., X.Z., M.F.C., D.H., D.K., D.Z., C.-P.C.); Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin, China (T.L.); and Department of Cardiology, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China (X.Z.)
| | - David Zhao
- Department of Cardiology, the First Affiliated Hospital of Kunming Medical University, Kunming, China (Y.L.); Department of Cardiology, Harbin Medical University Cancer Hospital, Harbin, China (Q.S.); Department of Internal Medicine, Cardiovascular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina (Y.L., Q.S., H.-J.C., T.L., X.Z., M.F.C., D.H., D.K., D.Z., C.-P.C.); Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin, China (T.L.); and Department of Cardiology, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China (X.Z.)
| | - Che-Ping Cheng
- Department of Cardiology, the First Affiliated Hospital of Kunming Medical University, Kunming, China (Y.L.); Department of Cardiology, Harbin Medical University Cancer Hospital, Harbin, China (Q.S.); Department of Internal Medicine, Cardiovascular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina (Y.L., Q.S., H.-J.C., T.L., X.Z., M.F.C., D.H., D.K., D.Z., C.-P.C.); Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin, China (T.L.); and Department of Cardiology, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China (X.Z.)
| |
Collapse
|
4
|
Joca HC, Santos-Miranda A, Joviano-Santos JV, Maia-Joca RPM, Brum PC, Williams GSB, Cruz JS. Chronic Sympathetic Hyperactivity Triggers Electrophysiological Remodeling and Disrupts Excitation-Contraction Coupling in Heart. Sci Rep 2020; 10:8001. [PMID: 32409748 PMCID: PMC7224293 DOI: 10.1038/s41598-020-64949-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 04/20/2020] [Indexed: 12/31/2022] Open
Abstract
The sympathetic nervous system is essential for maintenance of cardiac function via activation of post-junctional adrenergic receptors. Prolonged adrenergic receptor activation, however, has deleterious long-term effects leading to hypertrophy and the development of heart failure. Here we investigate the effect of chronic adrenergic receptors activation on excitation-contraction coupling (ECC) in ventricular cardiomyocytes from a previously characterized mouse model of chronic sympathetic hyperactivity, which are genetically deficient in the adrenoceptor α2A and α2C genes (ARDKO). When compared to wild-type (WT) cardiomyocytes, ARDKO displayed reduced fractional shortening (~33%) and slower relaxation (~20%). Furthermore, ARDKO cells exhibited several electrophysiological changes such as action potential (AP) prolongation (~50%), reduced L-type calcium channel (LCC) current (~33%), reduced outward potassium (K+) currents (~30%), and increased sodium/calcium exchanger (NCX) activity (~52%). Consistent with reduced contractility and calcium (Ca2+) currents, the cytosolic Ca2+ ([Ca2+]i) transient from ARDKO animals was smaller and decayed slower. Importantly, no changes were observed in membrane resting potential, AP amplitude, or the inward K+ current. Finally, we modified our existing cardiac ECC computational model to account for changes in the ARDKO heart. Simulations suggest that cellular changes in the ARDKO heart resulted in variable and dyssynchronous Ca2+-induced Ca2+ release therefore altering [Ca2+]i transient dynamics and reducing force generation. In conclusion, chronic sympathetic hyperactivity impairs ECC by changing the density of several ionic currents (and thus AP repolarization) causing altered Ca2+ dynamics and contractile activity. This demonstrates the important role of ECC remodeling in the cardiac dysfunction secondary to chronic sympathetic activity.
Collapse
Affiliation(s)
- Humberto C Joca
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Artur Santos-Miranda
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
- Department of Biophysics, Universidade Federal de Sao Paulo, Sao Paulo, SP, Brazil
| | | | - Rebeca P M Maia-Joca
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Patricia C Brum
- School of Physical Education and Sport, University of São Paulo, São Paulo, SP, Brazil
| | - George S B Williams
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Jader S Cruz
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil.
| |
Collapse
|
5
|
Pan R, Zhu X, Maretich P, Chen Y. Metabolic Improvement via Enhancing Thermogenic Fat-Mediated Non-shivering Thermogenesis: From Rodents to Humans. Front Endocrinol (Lausanne) 2020; 11:633. [PMID: 33013706 PMCID: PMC7511774 DOI: 10.3389/fendo.2020.00633] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 08/05/2020] [Indexed: 12/28/2022] Open
Abstract
Brown and beige adipose tissues play a large role in non-shivering thermogenesis (NST) in mammals, and subsequently have been studied for decades as potential therapeutic targets to treat obesity and its related metabolic diseases. However, the mechanistic regulation of brown/beige adipose tissue induction and maintenance in humans is very limited due to the ethical reasons. In fact, metabolic signaling has primarily been investigated using rodent models. A better understanding of non-shivering thermogenesis in humans is thus vital and urgent in order to treat obesity by targeting human brown adipose tissue (BAT). In this review, we summarize the anatomical and physiological differences between rodent and human BAT, current useful and mostly non-invasive methods in studying human BAT, as well as recent advancements targeting thermogenic adipocytes as a means to combat metabolic diseases in humans. Furthermore, we also discuss several novel relevant strategies of therapeutic interventions, which has been attempted in rodent experiments, and possible future investigations in humans in this field.
Collapse
Affiliation(s)
- Ruping Pan
- Department of Nuclear Medicine, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaohua Zhu
- Department of Nuclear Medicine, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Pema Maretich
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Yong Chen
- Department of Endocrinology, Internal Medicine, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Yong Chen
| |
Collapse
|
6
|
Pan R, Zhu X, Maretich P, Chen Y. Combating Obesity With Thermogenic Fat: Current Challenges and Advancements. Front Endocrinol (Lausanne) 2020; 11:185. [PMID: 32351446 PMCID: PMC7174745 DOI: 10.3389/fendo.2020.00185] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 03/16/2020] [Indexed: 12/12/2022] Open
Abstract
Brown fat and beige fat are known as thermogenic fat due to their contribution to non-shivering thermogenesis in mammals following cold stimulation. Beige fat is unique due to its origin and its development in white fat. Subsequently, both brown fat and beige fat have become viable targets to combat obesity. Over the last few decades, most therapeutic strategies have been focused on the canonical pathway of thermogenic fat activation via the β3-adrenergic receptor (AR). Notwithstanding, administering β3-AR agonists often leads to side effects including hypertension and particularly cardiovascular disease. It is thus imperative to search for alternative therapeutic approaches to combat obesity. In this review, we discuss the current challenges in the field with respect to stimulating brown/beige fat thermogenesis. Additionally, we include a summary of other newly discovered pathways, including non-AR signaling- and non-UCP1-dependent mechanisms, which could be potential targets for the treatment of obesity and its related metabolic diseases.
Collapse
MESH Headings
- Adipose Tissue, Beige/drug effects
- Adipose Tissue, Beige/metabolism
- Adipose Tissue, Beige/physiology
- Adipose Tissue, Brown/drug effects
- Adipose Tissue, Brown/metabolism
- Adipose Tissue, Brown/physiology
- Adrenergic beta-3 Receptor Agonists/pharmacology
- Adrenergic beta-3 Receptor Agonists/therapeutic use
- Animals
- Anti-Obesity Agents/pharmacology
- Anti-Obesity Agents/therapeutic use
- Humans
- Obesity/metabolism
- Obesity/therapy
- Receptors, Adrenergic, beta-3/metabolism
- Receptors, Adrenergic, beta-3/physiology
- Signal Transduction/drug effects
- Thermogenesis/drug effects
- Thermogenesis/physiology
Collapse
Affiliation(s)
- Ruping Pan
- Department of Nuclear Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaohua Zhu
- Department of Nuclear Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pema Maretich
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Yong Chen
- Department of Endocrinology, Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Yong Chen
| |
Collapse
|
7
|
Li T, Zhang Z, Zhang X, Chen Z, Cheng HJ, Ahmad S, Ferrario CM, Cheng CP. Reversal of angiotensin-(1-12)-caused positive modulation on left ventricular contractile performance in heart failure: Assessment by pressure-volume analysis. Int J Cardiol 2019; 301:135-141. [PMID: 31521437 DOI: 10.1016/j.ijcard.2019.09.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 08/19/2019] [Accepted: 09/04/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Angiotensin-(1-12) [Ang-(1-12)] is a renin-independent precursor for direct angiotensin-II production by chymase. Substantial evidence suggests that heart failure (HF) may alter cardiac Ang-(1-12) expression and activity; this novel Ang-(1-12)/chymase axis may be the main source for angiotensin-II deleterious actions in HF. We hypothesized that HF alters cardiac response to Ang-(1-12). Its stimulation may produce cardiac negative modulation and exacerbate left ventricle (LV) systolic and diastolic dysfunction. METHODS AND RESULTS We assessed the effects of Ang-(1-12) (2 nmol/kg/min, iv, 10 min) on LV contractility, LV diastolic filling, and LV-arterial coupling (AVC) in 16 SD male rats with HF-induced by isoproterenol (3 mo after 170 mg/kg sq. for 2 consecutive days) and 10 age-matched male controls. In normal controls, versus baseline, Ang-(1-12) increased LV end-systolic pressure, without altering heart rate, arterial elastance (EA), LV end-diastolic pressure (PED), the time constant of LV relaxation (τ) and ejection fraction (EF). Ang-(1-12) significantly increased the slopes (EES) of LV end-systolic pressure (P)-volume (V) relations and the slopes (MSW) of LV stroke wok-end-diastolic V relations, indicating increased LV contractility. AVC (quantified as EES/EA) improved. In contrast, in HF, versus HF baseline, Ang-(1-12) produced a similar increase in PES, but significantly increased τ, EA, and PED. The early diastolic portion of LV PV loop was shifted upward with reduced in EF. Moreover, Ang-(1-12) significantly decreased EES and MSW, demonstrating decreased LV contractility. AVC was decreased by 43%. CONCLUSIONS In both normal and HF rats, Ang-(1-12) causes similar vasoconstriction. In normal, Ang-(1-12) increases LV contractile function. In HF, Ang-(1-12) has adverse effects and depresses LV systolic and diastolic functional performance.
Collapse
Affiliation(s)
- Tiankai Li
- Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin, China; Department of Internal Medicine, Section on Cardiovascular Medicine, Wake Forest School of Medicine, Winston Salem, NC, United States of America
| | - Zhi Zhang
- Department of Internal Medicine, Section on Cardiovascular Medicine, Wake Forest School of Medicine, Winston Salem, NC, United States of America; Department of cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine (originally named "Shanghai First People's Hospital"), Shanghai, China
| | - Xiaowei Zhang
- Department of Internal Medicine, Section on Cardiovascular Medicine, Wake Forest School of Medicine, Winston Salem, NC, United States of America; Department of Cardiology, The Second Hospital of Shandong University, Jinan, Shandong, China
| | - Zhe Chen
- Department of Internal Medicine, Section on Cardiovascular Medicine, Wake Forest School of Medicine, Winston Salem, NC, United States of America; Department of Endocrinology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Heng-Jie Cheng
- Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin, China; Department of Internal Medicine, Section on Cardiovascular Medicine, Wake Forest School of Medicine, Winston Salem, NC, United States of America
| | - Sarfaraz Ahmad
- Department of Surgery, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America; Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC, United States of America
| | - Carlos M Ferrario
- Department of Surgery, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America; Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC, United States of America
| | - Che Ping Cheng
- Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin, China; Department of Internal Medicine, Section on Cardiovascular Medicine, Wake Forest School of Medicine, Winston Salem, NC, United States of America.
| |
Collapse
|
8
|
Arioglu-Inan E, Kayki-Mutlu G, Michel MC. Cardiac β 3 -adrenoceptors-A role in human pathophysiology? Br J Pharmacol 2019; 176:2482-2495. [PMID: 30801686 DOI: 10.1111/bph.14635] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 01/11/2019] [Accepted: 01/29/2019] [Indexed: 01/06/2023] Open
Abstract
As β3 -adrenoceptors were first demonstrated to be expressed in adipose tissue they have received much attention for their metabolic effects in obesity and diabetes. After the existence of this subtype had been suggested to be present in the heart, studies focused on its role in cardiac function. While the presence and functional role of β3 -adrenoceptors in the heart has not uniformly been detected, there is a broad consensus that they become up-regulated in pathological conditions associated with increased sympathetic activity such as heart failure and diabetes. When detected, the β3 -adrenceptor has been demonstrated to mediate negative inotropic effects in an inhibitory G protein-dependent manner through the NO-cGMP-PKG signalling pathway. Whether these negative inotropic effects provide protection from the adverse effects induced by overstimulation of β1 /β2 -adrenoceptors or in themselves are potentially harmful is controversial, but ongoing clinical studies in patients with congestive heart failure are testing the hypothesis that β3 -adrenceptor agonism has a beneficial effect. LINKED ARTICLES: This article is part of a themed section on Adrenoceptors-New Roles for Old Players. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.14/issuetoc.
Collapse
Affiliation(s)
- Ebru Arioglu-Inan
- Department of Pharmacology, Faculty of Pharmacy, Ankara University, Ankara, Turkey
| | - Gizem Kayki-Mutlu
- Department of Pharmacology, Faculty of Pharmacy, Ankara University, Ankara, Turkey
| | - Martin C Michel
- Department of Pharmacology, Johannes Gutenberg University, Mainz, Germany
| |
Collapse
|
9
|
Okeke K, Angers S, Bouvier M, Michel MC. Agonist-induced desensitisation of β 3 -adrenoceptors: Where, when, and how? Br J Pharmacol 2019; 176:2539-2558. [PMID: 30809805 DOI: 10.1111/bph.14633] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 01/27/2019] [Accepted: 02/11/2019] [Indexed: 12/13/2022] Open
Abstract
β3 -Adrenoceptor agonists have proven useful in the treatment of overactive bladder syndrome, but it is not known whether their efficacy during chronic administration may be limited by receptor-induced desensitisation. Whereas the β2 -adrenoceptor has phosphorylation sites that are important for desensitisation, the β3 -adrenoceptor lacks these; therefore, it had been assumed that β3 -adrenoceptors are largely resistant to agonist-induced desensitisation. While all direct comparative studies demonstrate that β3 -adrenoceptors are less susceptible to desensitisation than β2 -adrenoceptors, desensitisation of β3 -adrenoceptors has been observed in many models and treatment settings. Chimeric β2 - and β3 -adrenoceptors have demonstrated that the C-terminal tail of the receptor plays an important role in the relative resistance to desensitisation but is not the only relevant factor. While the evidence from some models, such as transfected CHO cells, is inconsistent, it appears that desensitisation is observed more often after long-term (hours to days) than short-term (minutes to hours) agonist exposure. When it occurs, desensitisation of β3 -adrenoceptors can involve multiple levels including down-regulation of its mRNA and the receptor protein and alterations in post-receptor signalling events. The relative contributions of these mechanistic factors apparently depend on the cell type under investigation. Which if any of these factors is applicable to the human urinary bladder remains to be determined. LINKED ARTICLES: This article is part of a themed section on Adrenoceptors-New Roles for Old Players. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.14/issuetoc.
Collapse
Affiliation(s)
- Katerina Okeke
- Department of Pharmacology, Johannes Gutenberg University, Mainz, Germany
| | - Stephane Angers
- Leslie Dan Faculty of Pharmacy and Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - Michel Bouvier
- Institute for Research in Immunology and Cancer, Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, QC, Canada
| | - Martin C Michel
- Department of Pharmacology, Johannes Gutenberg University, Mainz, Germany
| |
Collapse
|
10
|
Yang LK, Tao YX. Physiology and pathophysiology of the β 3-adrenergic receptor. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2018; 161:91-112. [PMID: 30711031 DOI: 10.1016/bs.pmbts.2018.09.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The β3-adrenergic receptor (β3-AR) is an important regulator of various physiological functions, such as thermogenesis in brown adipose tissue, lipolysis in white adipose tissue, negative inotropic effect in cardiomyocyte, and relaxation in blood vessel. The activation of β3-AR by its agonists is shown to have metabolic (antiobesity and antidiabetic) and cardiovascular effects in animal models, highlighting β3-AR as a potential therapeutic target in the treatment of several human diseases. Moreover, a substantial number of studies performed on different populations have identified some β3-AR polymorphic variants associated with obesity, diabetes, cardiovascular diseases, and other disorders. The clinical phenotypes and functional characteristics of these variants provide insights into potential pathophysiological roles of β3-AR in the development of these diseases.
Collapse
Affiliation(s)
- Li-Kun Yang
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
| | - Ya-Xiong Tao
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States.
| |
Collapse
|
11
|
Li T, Zhang X, Cheng HJ, Zhang Z, Ahmad S, Varagic J, Li W, Cheng CP, Ferrario CM. Critical role of the chymase/angiotensin-(1-12) axis in modulating cardiomyocyte contractility. Int J Cardiol 2018; 264:137-144. [PMID: 29685688 DOI: 10.1016/j.ijcard.2018.03.066] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 02/23/2018] [Accepted: 03/13/2018] [Indexed: 12/22/2022]
Abstract
BACKGROUND Angiotensin-(1-12) [Ang-(1-12)] is a chymase-dependent source for angiotensin II (Ang II) cardiac activity. The direct contractile effects of Ang-(1-12) in normal and heart failure (HF) remain to be demonstrated. We assessed the hypothesis that Ang-(1-12) may modulate [Ca2+]i regulation and alter cardiomyocyte contractility in normal and HF rats. METHODS AND RESULTS We compared left ventricle (LV) myocyte contractile and calcium transient ([Ca2+]iT) responses to angiotensin peptides in 16 SD rats with isoproterenol-induced HF and 16 age-matched controls. In normal myocytes, versus baseline, Ang II (10-6 M) superfusion significantly increased myocyte contractility (dL/dtmax: 40%) and [Ca2+]iT (29%). Ang-(1-12) (4 × 10-6 M) caused similar increases in dL/dtmax (34%) and [Ca2+]iT (25%). Compared with normal myocytes, superfusion of Ang II and Ang-(1-12) in myocytes obtained from rats with isoproterenol-induced HF caused similar but significantly attenuated positive inotropic actions with about 42% to 50% less increases in dL/dtmax and [Ca2+]iT. Chymostatin abolished Ang-(1-12)-mediated effects in normal and HF myocytes. The presence of an inhibitory cAMP analog, Rp-cAMPS prevented Ang-(1-12)-induced inotropic effects in both normal and HF myocytes. Incubation of HF myocytes with pertussis toxin (PTX) further augmented Ang II-mediated contractility. CONCLUSIONS Ang-(1-12) stimulates cardiomyocyte contractile function and [Ca2+]iT in both normal and HF rats through a chymase mediated action. Altered inotropic responses to Ang-(1-12) and Ang II in HF myocytes are mediated through a cAMP-dependent mechanism that is coupled to both stimulatory G and inhibitory PTX-sensitive G proteins.
Collapse
Affiliation(s)
- Tiankai Li
- Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin, China; Department of Internal Medicine, Section on Cardiovascular Medicine, Wake Forest School of Medicine, Winston Salem, NC, United States
| | - Xiaowei Zhang
- Department of Internal Medicine, Section on Cardiovascular Medicine, Wake Forest School of Medicine, Winston Salem, NC, United States; Department of Cardiology, the Second Affiliated Hospital of Nantong University, Nantong, China
| | - Heng-Jie Cheng
- Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin, China; Department of Internal Medicine, Section on Cardiovascular Medicine, Wake Forest School of Medicine, Winston Salem, NC, United States
| | - Zhi Zhang
- Department of Internal Medicine, Section on Cardiovascular Medicine, Wake Forest School of Medicine, Winston Salem, NC, United States; Cardiovascular Department, Shanghai First People's Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - Sarfaraz Ahmad
- Departments of Surgery, Internal Medicine-Nephrology, and Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Jasmina Varagic
- Departments of Surgery, Internal Medicine-Nephrology, and Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Weimin Li
- Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Che Ping Cheng
- Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin, China; Department of Internal Medicine, Section on Cardiovascular Medicine, Wake Forest School of Medicine, Winston Salem, NC, United States.
| | - Carlos M Ferrario
- Departments of Surgery, Internal Medicine-Nephrology, and Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| |
Collapse
|
12
|
Yang N, Shi XL, Zhang BL, Rong J, Zhang TN, Xu W, Liu CF. The Trend of β3-Adrenergic Receptor in the Development of Septic Myocardial Depression: A Lipopolysaccharide-Induced Rat Septic Shock Model. Cardiology 2018; 139:234-244. [PMID: 29566368 DOI: 10.1159/000487126] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 01/18/2018] [Indexed: 01/08/2023]
Abstract
Septic shock with low cardiac output is very common in children. However, the mechanism underlying myocardial depression is unclear. The role of β3-AR in the development of myocardial depression in sepsis is unknown. In the present study, we generated an adolescent rat model of hypodynamic septic shock induced by lipopolysaccharide (LPS). Neonatal cardiomyocytes were also treated with LPS to mimic myocardial depression in sepsis, which was confirmed via an in vivo left ventricular hemodynamic study, and measurements of contractility and the Ca2+ transient in isolated adolescent and neonatal cardiomyocytes. After 16 h of LPS treatment, cultured neonatal cardiomyocytes showed a diminished Ca2+ transient amplitude associated with an increase in the β3-AR level. With the addition of a β3-AR agonist, the Ca2+ transient in LPS-treated neonatal rat cardiomyocytes gradually decreased over time; such a change was absent in cells treated with nitric oxide synthase (NOS) inhibitors prior to treatment with a β3-AR agonist. In adolescent rats with septic myocardial depression, cardiac function declined as indicated by decreased MAP, dP/dtmax, and dP/dtmix for 6 h after LPS injection; however, the β3-AR level first increased 2 h after LPS treatment and then decreased 6 h after LPS treatment in the absence of exogenous catecholamines. The results indicate that, in vitro, at the cellular level β3-AR may be involved in the development of myocardial depression (Ca2+ transient depression) in sepsis through NOS signaling pathways; however, in vivo, a complicated mechanism for modulating β3-AR may exist.
Collapse
Affiliation(s)
- Ni Yang
- Department of Pediatrics, PICU, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xiao-Lu Shi
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China
| | - Bing-Lun Zhang
- Department of Pediatrics, PICU, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jian Rong
- Department of Pediatrics, PICU, Shengjing Hospital of China Medical University, Shenyang, China
| | - Tie-Ning Zhang
- Department of Pediatrics, PICU, Shengjing Hospital of China Medical University, Shenyang, China
| | - Wei Xu
- Department of Pediatrics, PICU, Shengjing Hospital of China Medical University, Shenyang, China
| | - Chun-Feng Liu
- Department of Pediatrics, PICU, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
13
|
Urmaliya V, Franchelli G. A multidimensional sight on cardiac failure: uncovered from structural to molecular level. Heart Fail Rev 2018; 22:357-370. [PMID: 28474325 DOI: 10.1007/s10741-017-9610-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Heart failure is one of the leading causes of death, with high mortality rate within 5 years after diagnosis. Treatment and prognosis options for heart failure primarily targeted on hemodynamic and neurohumoral components that drive progressive deterioration of the heart. However, given the multifactorial background that eventually leads to the "phenotype" named heart failure, better insight into the various components may lead to personalized treatment opportunities. Indeed, currently used criteria to diagnose and/or classify heart failure are possibly too focused on phenotypic improvement rather than the molecular driver of the disease and could therefore be further refined by integrating the leap of molecular and cellular knowledge. The ambiguity of the ejection fraction-based classification criteria became evident with development of advanced molecular techniques and the dawn of omics disciplines which introduced the idea that disease is caused by a myriad of cellular and molecular processes rather than a single event or pathway. The fact that different signaling pathways may underlie similar clinical manifestations calls for a more holistic study of heart failure. In this context, the systems biology approach can offer a better understanding of how different components of a system are altered during disease and how they interact with each other, potentially leading to improved diagnosis and classification of this condition. This review is aimed at addressing heart failure through a multilayer approach that covers individually some of the anatomical, morphological, functional, and tissue aspects, with focus on cellular and subcellular features as an alternative insight into new therapeutic opportunities.
Collapse
Affiliation(s)
- Vijay Urmaliya
- Discovery Sciences, Janssen Research & Development, Beerse, Belgium.
| | | |
Collapse
|
14
|
Role of the β 3-adrenergic receptor subtype in catecholamine-induced myocardial remodeling. Mol Cell Biochem 2018; 446:149-160. [PMID: 29363058 DOI: 10.1007/s11010-018-3282-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 01/16/2018] [Indexed: 10/18/2022]
Abstract
β3-Adrenoceptors (AR) stimulate cardiac Na+/K+ pump in healthy hearts. β3-ARs are upregulated by persistent sympathetic hyperactivity; however, their effect on Na+/K+ ATPase activity and ventricular function in this condition is still unknown. Here, we investigate preventive effects of additional β3-AR activation (BRL) on Na+/K+ ATPase activity and in vivo hemodynamics in a model of noradrenaline-induced hypertrophy. Rats received NA or NA plus simultaneously administered BRL in vivo infusion for 14 days; their cardiac function was investigated by left ventricular pressure-volume analysis. Moreover, fibrosis and apoptosis were also assessed histologically. NA induced an hypertrophic pattern, as detected by morphological, histological, and biochemical markers. Additional BRL exposure reversed the hypertrophic pattern and restored Na+/K+ ATPase activity. NA treatment increased systolic function and depressed diastolic function (slowed relaxation). Additional BRL treatment reversed most NA-induced hemodynamic changes. NA decreased Na+/K+ pump α2 subunit expression selectively, a change also reversed by additional BRL treatment. Increasing β3-AR stimulation may prevent the consequences of chronic NA exposure on Na+/K+ pump and in vivo hemodynamics. β3-AR agonism may thus represent a new therapeutic strategy for pharmacological modulation of hypertrophy under conditions of chronically enhanced sympathetic activity.
Collapse
|
15
|
Bond RC, Bryant SM, Watson JJ, Hancox JC, Orchard CH, James AF. Reduced density and altered regulation of rat atrial L-type Ca 2+ current in heart failure. Am J Physiol Heart Circ Physiol 2017; 312:H384-H391. [PMID: 27923791 PMCID: PMC5402008 DOI: 10.1152/ajpheart.00528.2016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 11/21/2016] [Accepted: 11/23/2016] [Indexed: 11/22/2022]
Abstract
Constitutive regulation by PKA has recently been shown to contribute to L-type Ca2+ current (ICaL) at the ventricular t-tubule in heart failure. Conversely, reduction in constitutive regulation by PKA has been proposed to underlie the downregulation of atrial ICaL in heart failure. The hypothesis that downregulation of atrial ICaL in heart failure involves reduced channel phosphorylation was examined. Anesthetized adult male Wistar rats underwent surgical coronary artery ligation (CAL, N=10) or equivalent sham-operation (Sham, N=12). Left atrial myocytes were isolated ~18 wk postsurgery and whole cell currents recorded (holding potential=-80 mV). ICaL activated by depolarizing pulses to voltages from -40 to +50 mV were normalized to cell capacitance and current density-voltage relations plotted. CAL cell capacitances were ~1.67-fold greater than Sham (P ≤ 0.0001). Maximal ICaL conductance (Gmax ) was downregulated more than 2-fold in CAL vs. Sham myocytes (P < 0.0001). Norepinephrine (1 μmol/l) increased Gmax >50% more effectively in CAL than in Sham so that differences in ICaL density were abolished. Differences between CAL and Sham Gmax were not abolished by calyculin A (100 nmol/l), suggesting that increased protein dephosphorylation did not account for ICaL downregulation. Treatment with either H-89 (10 μmol/l) or AIP (5 μmol/l) had no effect on basal currents in Sham or CAL myocytes, indicating that, in contrast to ventricular myocytes, neither PKA nor CaMKII regulated basal ICaL Expression of the L-type α1C-subunit, protein phosphatases 1 and 2A, and inhibitor-1 proteins was unchanged. In conclusion, reduction in PKA-dependent regulation did not contribute to downregulation of atrial ICaL in heart failure.NEW & NOTEWORTHY Whole cell recording of L-type Ca2+ currents in atrial myocytes from rat hearts subjected to coronary artery ligation compared with those from sham-operated controls reveals marked reduction in current density in heart failure without change in channel subunit expression and associated with altered phosphorylation independent of protein kinase A.
Collapse
Affiliation(s)
- Richard C Bond
- Cardiovascular Research Laboratories, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
| | - Simon M Bryant
- Cardiovascular Research Laboratories, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
| | - Judy J Watson
- Cardiovascular Research Laboratories, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
| | - Jules C Hancox
- Cardiovascular Research Laboratories, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
| | - Clive H Orchard
- Cardiovascular Research Laboratories, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
| | - Andrew F James
- Cardiovascular Research Laboratories, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
16
|
Zhang X, Cheng HJ, Zhou P, Kitzman DW, Ferrario CM, Li WM, Cheng CP. Cellular basis of angiotensin-(1-7)-induced augmentation of left ventricular functional performance in heart failure. Int J Cardiol 2017; 236:405-412. [PMID: 28096047 DOI: 10.1016/j.ijcard.2017.01.071] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 01/06/2017] [Indexed: 10/20/2022]
Abstract
BACKGROUND Angiotensin-(1-7) [Ang-(1-7)] exhibits cardiovascular effects opposite those of angiotensin II (Ang II), thus providing protection against heart disease. However, how Ang-(1-7) imparts cardioprotection is unclear, and its direct cardiac effects are controversial. Whether heart failure (HF) alters cardiac contractile responses to Ang-(1-7) remains undetermined. We tested the hypothesis that in HF, Ang-(1-7) may produce positive modulation on [Ca2+]i regulation, enhancing left ventricular (LV) and myocyte contraction and relaxation via Ang-(1-7) Mas receptor coupled with nitric oxide (NO)/bradykinin (BK)-mediated mechanism. METHODS AND RESULTS We measured LV contractility changes after Ang-(1-7) (650ng/kg, iv) and compared myocyte functional and [Ca2+]i transient ([Ca2+]iT) responses to Ang-(1-7) superfusion in 24 normal rats and 34 rats with isoproterenol-induced HF (3months after 170mg/kg, s.q. for 2days). To assess the mechanisms of altered HF responses to Ang-(1-7), subsets of HF myocytes were pretreated to inhibit NO synthase (L-NAME), BK (HOE-140), and Mas receptor (A-779) followed with Ang-(1-7). In normal rats, Ang-(1-7) produced no significant changes in LV and myocyte function. In HF rats, Ang-(1-7) significantly augmented LV contractility and relaxation with increased EES (51%), but decreased τ compared to baseline. Ang-(1-7) also significantly increased myocyte contraction (dL/dtmax, 30%), relaxation (dR/dtmax, 41%), and [Ca2+]iT. L-NAME increased, HOE-140 decreased, and A-779 prevented HF myocyte contractile responses to Ang-(1-7). CONCLUSIONS In a rat model of HF, Ang-(1-7) increases [Ca2+]iT, and produces positive inotropic and lusitropic effects in the LV and myocytes. These effects are mediated by the Mas receptor and involve activation of NO/BK pathways.
Collapse
Affiliation(s)
- Xiaowei Zhang
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Heng-Jie Cheng
- Section on Cardiovascular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Peng Zhou
- Section on Cardiovascular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Dalane W Kitzman
- Section on Cardiovascular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Carlos M Ferrario
- Department of Surgery, Wake Forest School of Medicine, Winston-Salem, NC, United States; Department of Internal Medicine-Nephrology, Wake Forest School of Medicine, Winston-Salem, NC, United States; Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Wei-Min Li
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
| | - Che Ping Cheng
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China; Section on Cardiovascular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States.
| |
Collapse
|
17
|
Bundgaard H, Axelsson A, Hartvig Thomsen J, Sørgaard M, Kofoed KF, Hasselbalch R, Fry NAS, Valeur N, Boesgaard S, Gustafsson F, Køber L, Iversen K, Rasmussen HH. The first-in-man randomized trial of a beta3 adrenoceptor agonist in chronic heart failure: the BEAT-HF trial. Eur J Heart Fail 2016; 19:566-575. [PMID: 27990717 DOI: 10.1002/ejhf.714] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2016] [Revised: 10/21/2016] [Accepted: 11/14/2016] [Indexed: 12/22/2022] Open
Abstract
AIMS The third isotype of beta adrenergic receptors (β3 ARs) has distinctly different effects on cardiomyocytes compared with β1 and β2 ARs. Stimulation of β3 ARs may reduce cardiomyocyte Na+ overload and reduce oxidative stress in heart failure (HF). We examined if treatment with the β3 AR agonist mirabegron increases LVEF in patients with HF. METHODS AND RESULTS In a double-blind trial we randomly assigned 70 patients with NYHA class II-III HF and LVEF <40% at screening-echocardiography to receive mirabegron or placebo for 6 months as add-on to optimized standard therapy. The primary endpoint was an increase in LVEF after 6 months as measured by computed tomography (CT). Changes in LVEF after 6 months between treatment groups were not significantly different (0.4%, -3.5 to 3.8%, P = 0.82). In an exploratory analysis, based on an expectation that the pathophysiological substrate targeted with treatment is dependent on the baseline LVEF, patients with LVEF <40% by CT given mirabegron had a significant increase in LVEF while no increase was seen in patients given placebo. The changes were significantly different between groups (5.5%, 0.6-10.4%, P < 0.03). Additionally, there was interaction between baseline LVEF and change in LVEF in the entire group of patients treated with mirabegron (R2 = 0.40, β = -0.63, P < 0.001), but not in the placebo group (R2 = 0.00, β = -0.01, P = 0.95). Treatment was generally well tolerated. Three patients in each group had fatal or life-threatening events. CONCLUSIONS The primary endpoint was not reached. Exploratory analysis indicated that β3 AR stimulation by mirabegron increased LVEF in patients with severe HF. Treatment appeared safe. Additional studies in severe HF are needed. TRIAL REGISTRATION NCT01876433.
Collapse
Affiliation(s)
- Henning Bundgaard
- Department of Cardiology, The Heart Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Anna Axelsson
- Department of Cardiology, The Heart Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Jakob Hartvig Thomsen
- Department of Cardiology, The Heart Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Mathias Sørgaard
- Department of Cardiology, The Heart Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Klaus F Kofoed
- Department of Cardiology, The Heart Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark.,Department of Radiology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Rasmus Hasselbalch
- Department of Cardiology, Herlev Hospital, University of Copenhagen, Copenhagen, Denmark
| | | | - Nana Valeur
- Department of Cardiology, Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Søren Boesgaard
- Department of Cardiology, The Heart Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Finn Gustafsson
- Department of Cardiology, The Heart Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Lars Køber
- Department of Cardiology, The Heart Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Kasper Iversen
- Department of Cardiology, Herlev Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Helge H Rasmussen
- Department of Cardiology, Royal North Shore Hospital and University of Sydney, Sydney, Australia
| |
Collapse
|
18
|
Michel MC, Korstanje C. β3-Adrenoceptor agonists for overactive bladder syndrome: Role of translational pharmacology in a repositioning clinical drug development project. Pharmacol Ther 2016; 159:66-82. [PMID: 26808167 DOI: 10.1016/j.pharmthera.2016.01.007] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
β3-Adrenoceptor agonists were originally considered as a promising drug class for the treatment of obesity and/or type 2 diabetes. When these development efforts failed, they were repositioned for the treatment of the overactive bladder syndrome. Based on the example of the β3-adrenoceptor agonist mirabegron, but also taking into consideration evidence obtained with ritobegron and solabegron, we discuss challenges facing a translational pharmacology program accompanying clinical drug development for a first-in-class molecule. Challenges included generic ones such as ligand selectivity, species differences and drug target gene polymorphisms. Challenges that are more specific included changing concepts of the underlying pathophysiology of the target condition while clinical development was under way; moreover, a paucity of public domain tools for the study of the drug target and aspects of receptor agonists as drugs had to be addressed. Nonetheless, a successful first-in-class launch was accomplished. Looking back at this translational pharmacology program, we conclude that a specifically tailored and highly flexible approach is required. However, several of the lessons learned may also be applicable to translational pharmacology programs in other indications.
Collapse
Affiliation(s)
- Martin C Michel
- Department of Pharmacology, Johannes Gutenberg University, Mainz, Germany.
| | - Cees Korstanje
- Department of Drug Discovery Science & Management-Europe, Astellas Pharma Europe R&D, Leiden, The Netherlands
| |
Collapse
|
19
|
Shao Q, Cheng HJ, Callahan MF, Kitzman DW, Li WM, Cheng CP. Overexpression myocardial inducible nitric oxide synthase exacerbates cardiac dysfunction and beta-adrenergic desensitization in experimental hypothyroidism. Int J Cardiol 2015; 204:229-41. [PMID: 26681542 DOI: 10.1016/j.ijcard.2015.11.040] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Revised: 10/15/2015] [Accepted: 11/04/2015] [Indexed: 01/15/2023]
Abstract
BACKGROUND Altered nitric oxide synthase (NOS) has been implicated in the pathophysiology of heart failure (HF). Recent evidence links hypothyroidism to the pathology of HF. However, the precise mechanisms are incompletely understood. The alterations and functional effects of cardiac NOS in hypothyroidism are unknown. We tested the hypothesis that hypothyroidism increases cardiomyocyte inducible NOS (iNOS) expression, which plays an important role in hypothyroidism-induced depression of cardiomyocyte contractile properties, [Ca(2+)]i transient ([Ca(2+)]iT), and β-adrenergic hyporesponsiveness. METHODS AND RESULTS We simultaneously evaluated LV functional performance and compared myocyte three NOS, β-adrenergic receptors (AR) and SERCA2a expressions and assessed cardiomyocyte contractile and [Ca(2+)]iT responses to β-AR stimulation with and without pretreatment of iNOS inhibitor (1400 W, 10(-5)mol/L) in 26 controls and 26 rats with hypothyroidism induced by methimazole (~30 mg/kg/day for 8 weeks in the drinking water). Compared with controls, in hypothyroidism, total serum T3 and T4 were significantly reduced followed by significantly decreased LV contractility (EES) with increased LV time constant of relaxation. These LV abnormalities were accompanied by concomitant significant decreases in myocyte contraction (dL/dtmax), relaxation (dR/dtmax), and [Ca(2+)]iT. In hypothyroidism, isoproterenol (10(-8)M) produced significantly smaller increases in dL/dtmax, dR/dtmax and [Ca(2+)]iT. These changes were associated with decreased β1-AR and SERCA2a, but significantly increased iNOS. Moreover, only in hypothyroidism, pretreatment with iNOS inhibitor significantly improved basal and isoproterenol-stimulated myocyte contraction, relaxation and [Ca(2+)]iT. CONCLUSIONS Hypothyroidism produces intrinsic defects of LV myocyte force-generating capacity and relaxation with β-AR desensitization. Up-regulation of cardiomyocyte iNOS may promote progressive cardiac dysfunction in hypothyroidism.
Collapse
Affiliation(s)
- Qun Shao
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Heng-Jie Cheng
- Section on Cardiovascular Medicine, Wake Forest School of Medicine Winston-Salem, North Carolina, United States; Wake Forest, Institute for Regenerative Medicine, Winston-Salem, North Carolina, United States
| | - Michael F Callahan
- Department of Orthopaedic Surgery, Wake Forest School of Medicine Winston-Salem, North Carolina, United States
| | - Dalane W Kitzman
- Section on Cardiovascular Medicine, Wake Forest School of Medicine Winston-Salem, North Carolina, United States
| | - Wei-Min Li
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
| | - Che Ping Cheng
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China; Section on Cardiovascular Medicine, Wake Forest School of Medicine Winston-Salem, North Carolina, United States.
| |
Collapse
|
20
|
NOS1 induces NADPH oxidases and impairs contraction kinetics in aged murine ventricular myocytes. Basic Res Cardiol 2015; 110:506. [PMID: 26173391 DOI: 10.1007/s00395-015-0506-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 06/20/2015] [Accepted: 07/07/2015] [Indexed: 01/08/2023]
Abstract
Nitric oxide (NO) modulates calcium transients and contraction of cardiomyocytes. However, it is largely unknown whether NO contributes also to alterations in the contractile function of cardiomyocytes during aging. Therefore, we analyzed the putative role of nitric oxide synthases and NO for the age-related alterations of cardiomyocyte contraction. We used C57BL/6 mice, nitric oxide synthase 1 (NOS1)-deficient mice (NOS1(-/-)) and mice with cardiomyocyte-specific NOS1-overexpression to analyze contractions, calcium transients (Indo-1 fluorescence), acto-myosin ATPase activity (malachite green assay), NADPH oxidase activity (lucigenin chemiluminescence) of isolated ventricular myocytes and cardiac gene expression (Western blots, qPCR). In C57BL/6 mice, cardiac expression of NOS1 was upregulated by aging. Since we found a negative regulation of NOS1 expression by cAMP in isolated cardiomyocytes, we suggest that reduced efficacy of β-adrenergic signaling that is evident in aged hearts promotes upregulation of NOS1. Shortening and relengthening of cardiomyocytes from aged C57BL/6 mice were decelerated, but were normalized by pharmacological inhibition of NOS1/NO. Cardiomyocytes from NOS1(-/-) mice displayed no age-related changes in contraction, calcium transients or acto-myosin ATPase activity. Aging increased cardiac expression of NADPH oxidase subunits NOX2 and NOX4 in C57BL/6 mice, but not in NOS1(-/-) mice. Similarly, cardiac expression of NOX2 and NOX4 was upregulated in a murine model with cardiomyocyte-specific overexpression of NOS1. We conclude that age-dependently upregulated NOS1, putatively via reduced efficacy of β-adrenergic signaling, induces NADPH oxidases. By increasing nitrosative and oxidative stress, both enzyme systems act synergistically to decelerate contraction of aged cardiomyocytes.
Collapse
|
21
|
Petersen L, Burleson M, Huggett D. Temperature and species-specific effects on ß3-adrenergic receptor cardiac regulation in two freshwater teleosts: Channel catfish (Ictalurus punctatus) and common carp (Cyprinus carpio). Comp Biochem Physiol A Mol Integr Physiol 2015; 185:132-41. [DOI: 10.1016/j.cbpa.2015.04.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2014] [Revised: 04/03/2015] [Accepted: 04/05/2015] [Indexed: 11/17/2022]
|
22
|
Zhou P, Cheng CP, Li T, Ferrario CM, Cheng HJ. Modulation of cardiac L-type Ca2+ current by angiotensin-(1-7): normal versus heart failure. Ther Adv Cardiovasc Dis 2015; 9:342-53. [PMID: 26082338 DOI: 10.1177/1753944715587424] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
OBJECTIVE Recent evidence has shown that, in heart failure (HF), clinically relevant concentrations of angiotensin-(1-7) [Ang-(1-7)] counteracts angiotensin II induced cardiac depression and produces positive inotropic effects in both left ventricle (LV) and myocytes. However, the underlying electrophysiological mechanism is unclear. We investigated the role and mechanism of Ang-(1-7) on LV myocyte L-type calcium current (ICa,L) responses in normal state and in HF. METHOD We compared the effect of Ang-(1-7) (10(-5) M) on ICa,L responses in isolated LV myocytes obtained from 11 rats with isoproterenol (ISO) induced HF (3 months after 170 mg/kg subcutaneous for 2 days) and from 8 age-matched normal control rats by patch clamp technique. RESULTS In normal myocytes, compared with baseline, superfusion of Ang-(1-7) caused no significant changes in ICa,L (8.2 ± 0.2 versus 8.0 ± 0.3 pA/pF, p= not significant). In HF myocytes, the baseline ICa,L was significantly reduced (5.3 ± 0.1 versus 8.0 ± 0.3 pA/pF, p < 0.01). Ang-(1-7) produced a 21% increase in ICa,L (6.4±0.1 versus 5.3±0.1 pA/pF, p < 0.01). Pretreatment of HF myocytes with a nitric oxide (NO) synthase inhibitor (L-NAME, 10(-5) M) resulted in a significantly greater increase in ICa,L (28%, 8.4 ± 0.1 versus 6.5 ± 0.1 pA/pF, p < 0.01) during Ang-(1-7) superfusion. In contrast, during incubation with the bradykinin (BK) inhibitor HOE 140 (10(-6) M), Ang-(1-7) induced increase in ICa,L was significantly decreased. The Ang-(1-7) induced increase in ICa,L was abolished by [D-Ala(7)]-Ang-(1-7) (A-779, 10(-5) M). CONCLUSIONS HF alters the response of ICa,L to Ang-(1-7). In normal myocytes, Ang-(1-7) has no significant effect on ICa,L. However, in HF myocytes, Ang-(1-7) increases ICa,L. These effects are mediated by the Ang-(1-7) Mas receptors and involve activation of NO/BK pathways.
Collapse
Affiliation(s)
- Peng Zhou
- Section on Cardiovascular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Che Ping Cheng
- Section on Cardiovascular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Tiankai Li
- Section on Cardiovascular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USADepartment of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Carlos M Ferrario
- Department of Surgery, Internal Medicine-Nephrology, and Physiology-Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Heng-Jie Cheng
- Section on Cardiovascular Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157-1045, USAInstitute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
23
|
Lu C, Sun Z, Wang L. Inhibition of L-type Ca(2+) current by ginsenoside Rd in rat ventricular myocytes. J Ginseng Res 2015; 39:169-77. [PMID: 26045691 PMCID: PMC4452530 DOI: 10.1016/j.jgr.2014.11.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Revised: 10/01/2014] [Accepted: 11/02/2014] [Indexed: 11/28/2022] Open
Abstract
Background Ginsenoside Rd (GSRd), one of the most abundant ingredients of Panax ginseng, protects the heart via multiple mechanisms including the inhibition of Ca2+ influx. We intended to explore the effects of GSRd on L-type Ca2+ current (ICa,L) and define the mechanism of the suppression of ICa,L by GSRd. Methods Perforated-patch recording and whole-cell voltage clamp techniques were applied in isolated rat ventricular myocytes. Results (1) GSRd reduced ICa,L peak amplitude in a concentration-dependent manner [half-maximal inhibitory concentration (IC50) = 32.4 ± 7.1 μmol/L] and up-shifted the current–voltage (I–V) curve. (2) GSRd (30 μmol/L) significantly changed the steady-state activation curve of ICa,L (V0.5: −19.12 ± 0.68 vs. −16.26 ± 0.38 mV; n = 5, p < 0.05) and slowed down the recovery of ICa,L from inactivation [the time content (ζ) from 91 ms to 136 ms, n = 5, p < 0.01]. (3) A more significant inhibitive effect of GSRd (100 μmol/L) was identified in perforated-patch recording when compared with whole-cell recording [65.7 ± 3.2% (n = 10) vs. 31.4 ± 5.2% (n = 5), p < 0.01]. (4) Pertussis toxin (Giprotein inhibitor) completely abolished the ICa,L inhibition induced by GSRd. There was a significant difference in inhibition potency between the two cyclic adenosine monophosphate elevating agents (isoprenaline and forskolin) prestimulation [55 ± 7.8% (n = 5) vs. 17.2 ± 3.5% (n = 5), p < 0.01]. (5) 1H-[1,2,4]Oxadiazolo[4,3-a]-quinoxalin-1-one (a guanylate cyclase inhibitor) and N-acetyl-l-cysteine (a nitric oxide scavenger) partly recovered the ICa,L inhibition induced by GSRd. (6) Phorbol-12-myristate-13-acetate (a protein kinase C activator) and GF109203X (a protein kinase C inhibitor) did not contribute to the inhibition of GSRd. Conclusion These findings suggest that GSRd could inhibit ICa,L through pertussis toxin-sensitive G protein (Gi) and a nitric oxide–cyclic guanosine monophosphate-dependent mechanism.
Collapse
Affiliation(s)
- Cheng Lu
- Department of Otorhinolaryngology, Beijing Friendship Hospital Affiliated to Capital Medical University, Beijing, China
| | - Zhijun Sun
- Department of Cardiology, Beijing Friendship Hospital Affiliated to Capital Medical University, Beijing, China
| | - Line Wang
- Department of Otorhinolaryngology, Beijing Friendship Hospital Affiliated to Capital Medical University, Beijing, China
| |
Collapse
|
24
|
Imbrogno S, Gattuso A, Mazza R, Angelone T, Cerra MC. β3 -AR and the vertebrate heart: a comparative view. Acta Physiol (Oxf) 2015; 214:158-75. [PMID: 25809182 DOI: 10.1111/apha.12493] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Revised: 11/04/2014] [Accepted: 03/16/2015] [Indexed: 01/13/2023]
Abstract
Recent cardiovascular research showed that, together with β1- and β2-adrenergic receptors (ARs), β3-ARs contribute to the catecholamine (CA)-dependent control of the heart. β3-ARs structure, function and ligands were investigated in mammals because of their applicative potential in human cardiovascular diseases. Only recently, the concept of a β3-AR-dependent cardiac modulation was extended to non-mammalian vertebrates, although information is still scarce and fragmentary. β3-ARs were structurally described in fish, showing a closer relationship to mammalian β1-AR than β2-AR. Functional β3-ARs are present in the cardiac tissue of teleosts and amphibians. As in mammals, activation of these receptors elicits a negative modulation of the inotropic performance through the involvement of the endothelium endocardium (EE), Gi/0 proteins and the nitric oxide (NO) signalling. This review aims to comparatively analyse data from literature on β3-ARs in mammals, with those on teleosts and amphibians. The purpose is to highlight aspects of uniformity and diversity of β3-ARs structure, ligands activity, function and signalling cascades throughout vertebrates. This may provide new perspectives aimed to clarify the biological relevance of β3-ARs in the context of the nervous and humoral control of the heart and its functional plasticity.
Collapse
Affiliation(s)
- S. Imbrogno
- Department of Biology, Ecology and Earth Sciences; University of Calabria; Arcavacata di Rende Italy
| | - A. Gattuso
- Department of Biology, Ecology and Earth Sciences; University of Calabria; Arcavacata di Rende Italy
| | - R. Mazza
- Department of Biology, Ecology and Earth Sciences; University of Calabria; Arcavacata di Rende Italy
| | - T. Angelone
- Department of Biology, Ecology and Earth Sciences; University of Calabria; Arcavacata di Rende Italy
- National Institute of Cardiovascular Research; Bologna Italy
| | - M. C. Cerra
- Department of Biology, Ecology and Earth Sciences; University of Calabria; Arcavacata di Rende Italy
- National Institute of Cardiovascular Research; Bologna Italy
| |
Collapse
|
25
|
Haley JM, Thackeray JT, Kolajova M, Thorn SL, DaSilva JN. Insulin therapy normalizes reduced myocardial β-adrenoceptors at both the onset and after sustained hyperglycemia in diabetic rats. Life Sci 2015; 132:101-7. [PMID: 25934520 DOI: 10.1016/j.lfs.2015.03.024] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Revised: 03/25/2015] [Accepted: 03/27/2015] [Indexed: 01/09/2023]
Abstract
AIMS Reduced cardiac β-adrenoceptors (β-AR) and cardiovascular (CV) dysfunction occur in diabetes mellitus (DM) and can be normalized by insulin. It is unclear how the duration of untreated hyperglycemia prior to intervention impacts insulin's effects. This study assesses insulin's effect on reduced myocardial β-AR and CV function, comparing insulin therapy at the onset of hyperglycemia and after a sustained period of hyperglycemia in streptozotocin (STZ) rats. MAIN METHODS Ex vivo biodistribution experiments with [(3)H]CGP12177 were performed in high-fat fed STZ rats after 8 weeks of hyperglycemia evaluating cardiac β-AR expression. Western blotting of β-AR subtypes was completed in parallel. Serial echocardiography at 0, 6, and 8 weeks post-STZ investigated CV function. Sub-groups of hyperglycemic rats were treated with insulin early, at 1 week post-STZ (InsE) for 7 weeks, or late at 6 weeks post-STZ (InsL) for 2 weeks to observe how the duration of hyperglycemia prior to insulin impacts its effects. KEY FINDINGS Reduced myocardial [(3)H]CGP12177 binding occurred 8 weeks post-STZ in hyperglycemics, but was normal in both insulin treatments. Western blotting supported reduced β1-AR in hyperglycemics, but not in either treatment. InsE and InsL treatments improved prolonged mitral valve deceleration (MVD) observed in hyperglycemic animals, but hyperglycemic and InsL still displayed reduced heart rates (HR). SIGNIFICANCE This work supports that glycemic control with insulin normalizes cardiac β-AR effectively regardless of prior hyperglycemia but HR may not recover as readily, indirectly supporting the utility of [(11)C]CGP12177 positron emission tomography (PET) in assessing cardiac β-AR and their modulation with glycemic therapy.
Collapse
Affiliation(s)
- James M Haley
- Molecular Function & Imaging Program, National Cardiac PET Centre, University of Ottawa Heart Institute, 40 Ruskin Street, Ottawa, Ontario K1Y4W7, Canada; Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Roger Guindon Hall, 451 Smyth Road, Ottawa, Ontario K1H8M5, Canada
| | - James T Thackeray
- Molecular Function & Imaging Program, National Cardiac PET Centre, University of Ottawa Heart Institute, 40 Ruskin Street, Ottawa, Ontario K1Y4W7, Canada; Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Roger Guindon Hall, 451 Smyth Road, Ottawa, Ontario K1H8M5, Canada; Department of Nuclear Medicine, Hannover Medical School, Carl Neuberg Street 1, 30625 Hannover, Germany
| | - Maria Kolajova
- Molecular Function & Imaging Program, National Cardiac PET Centre, University of Ottawa Heart Institute, 40 Ruskin Street, Ottawa, Ontario K1Y4W7, Canada
| | - Stephanie L Thorn
- Molecular Function & Imaging Program, National Cardiac PET Centre, University of Ottawa Heart Institute, 40 Ruskin Street, Ottawa, Ontario K1Y4W7, Canada; Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Roger Guindon Hall, 451 Smyth Road, Ottawa, Ontario K1H8M5, Canada; Yale Translational Research Imaging Center, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Jean N DaSilva
- Molecular Function & Imaging Program, National Cardiac PET Centre, University of Ottawa Heart Institute, 40 Ruskin Street, Ottawa, Ontario K1Y4W7, Canada; Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Roger Guindon Hall, 451 Smyth Road, Ottawa, Ontario K1H8M5, Canada; Department of Radiology, Radio-Oncology and Nuclear Medicine, University of Montreal, Montréal, Québec H2X 0A9, Canada; University of Montreal Hospital Research Centre (CRCHUM), 900 Rue Saint-Denis, Montréal, Québec H2X 0A9, Canada.
| |
Collapse
|
26
|
Al Kury LT, Voitychuk OI, Yang KHS, Thayyullathil FT, Doroshenko P, Ramez AM, Shuba YM, Galadari S, Howarth FC, Oz M. Effects of the endogenous cannabinoid anandamide on voltage-dependent sodium and calcium channels in rat ventricular myocytes. Br J Pharmacol 2015; 171:3485-98. [PMID: 24758718 DOI: 10.1111/bph.12734] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Revised: 02/17/2014] [Accepted: 03/14/2014] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND AND PURPOSE The endocannabinoid anandamide (N-arachidonoyl ethanolamide; AEA) exerts negative inotropic and antiarrhythmic effects in ventricular myocytes. EXPERIMENTAL APPROACH Whole-cell patch-clamp technique and radioligand-binding methods were used to analyse the effects of anandamide in rat ventricular myocytes. KEY RESULTS In the presence of 1-10 μM AEA, suppression of both Na(+) and L-type Ca(2+) channels was observed. Inhibition of Na(+) channels was voltage and Pertussis toxin (PTX) - independent. Radioligand-binding studies indicated that specific binding of [(3) H] batrachotoxin (BTX) to ventricular muscle membranes was also inhibited significantly by 10 μM metAEA, a non-metabolized AEA analogue, with a marked decrease in Bmax values but no change in Kd . Further studies on L-type Ca(2+) channels indicated that AEA potently inhibited these channels (IC50 0.1 μM) in a voltage- and PTX-independent manner. AEA inhibited maximal amplitudes without affecting the kinetics of Ba(2+) currents. MetAEA also inhibited Na(+) and L-type Ca(2+) currents. Radioligand studies indicated that specific binding of [(3) H]isradipine, was inhibited significantly by metAEA. (10 μM), changing Bmax but not Kd . CONCLUSION AND IMPLICATIONS Results indicate that AEA inhibited the function of voltage-dependent Na(+) and L-type Ca(2+) channels in rat ventricular myocytes, independent of CB1 and CB2 receptor activation.
Collapse
Affiliation(s)
- Lina T Al Kury
- Laboratory of Functional Lipidomics, Department of Pharmacology, UAE University, Al Ain, UAE
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Laukova M, Tillinger A, Novakova M, Krizanova O, Kvetnansky R, Myslivecek J. Repeated immobilization stress increases expression of β3 -adrenoceptor in the left ventricle and atrium of the rat heart. Stress Health 2014; 30:301-9. [PMID: 23878066 DOI: 10.1002/smi.2515] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2012] [Revised: 05/24/2013] [Accepted: 06/21/2013] [Indexed: 11/12/2022]
Abstract
Stress is a contributor of many cardiovascular diseases. Positive inotropic and chronotropic effects of catecholamines are regulated via β-adrenergic receptors (ARs). Many reports exist concerning changes of cardiac β1 - and β2 -ARs in stress, but only a few deal with modulation of cardiac β3 -AR. Our aim was to analyze the expression and binding sites of β1 -, β2 - and β3 -ARs and adenylyl cyclase activity in the left ventricle, and β3 -AR expression and binding in the left atrium of rats exposed to acute and chronic immobilization stress (IMO). The concentration of noradrenaline in the ventricle decreased, while adrenaline increased, especially after repeated IMO. The mRNA and protein levels, and binding sites of β3 -subtype significantly rose following chronic IMO, while all parameters for β2 -AR dropped after single and repeated exposure. Similarly, the mRNA levels and binding sites for β3 -subtype increased in the left atrium as a consequence of chronic IMO. The rise in β3 -subtypes and a drop in β2 -subtypes resulted in inhibition of adenylyl cyclase activity within the left ventricle. Taken together, among other factors, up-regulation of β3 -AR could represent an adaptation mechanism, which might be related to altered physiological function of the left ventricle and atrium during prolonged emotional stress and might serve cardioprotective function during catecholamine overload.
Collapse
Affiliation(s)
- Marcela Laukova
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, NY, USA; Institute of Experimental Endocrinology, Slovak Academy of Sciences, Bratislava, Slovakia
| | | | | | | | | | | |
Collapse
|
28
|
Abstract
Cardiomyocyte necrosis with attendant microscopic scarring is a pathological feature of human hypertensive heart disease (HHD). Understanding the pathophysiological origins of necrosis is integral to its prevention. In a rat model of HHD associated with aldosterone/salt treatment (ALDOST), myocyte necrosis is attributable to oxidative stress induced by cytosolic-free [Ca]i and mitochondrial [Ca]m overloading in which the rate of reactive oxygen species generation overwhelms their rate of detoxification by endogenous Zn-based antioxidant defenses. We hypothesized that nebivolol (Neb), unlike another β1 adrenergic receptor antagonist atenolol (Aten), would have a multifaceted antioxidant potential based on its dual property as a β3 receptor agonist, which activates endothelial nitric oxide synthase to stimulate nitric oxide (NO) generation. NO promotes the release of cytosolic Zn sequestered inactive by its binding protein, metallothionein. Given the reciprocal regulation between these cations, increased [Zn]i reduces Ca entry and attendant rise in [Ca]i and [Ca]m. Herein, we examined the antioxidant and cardioprotectant properties of Neb and Aten in rats receiving 4 weeks ALDOST. Compared with untreated age-/sex-matched controls, ALDOST alone or ALDOST with Aten, Neb cotreatment induced endothelial nitric oxide synthase activation, NO generation and a marked increase in [Zn]i with associated decline in [Ca]i and [Ca]m. Attendant antioxidant profile at subcellular and cellular levels included attenuation of mitochondrial H2O2 production and lipid peroxidation expressed as reduced 8-isoprostane concentrations in both mitochondria and cardiac tissue. Myocyte salvage was expressed as reduced microscopic scarring and tissue collagen volume fraction. Neb is a multifaceted antioxidant with unique properties as cardioprotectant in HHD.
Collapse
|
29
|
Al Kury LT, Yang KHS, Thayyullathil FT, Rajesh M, Ali RM, Shuba YM, Howarth FC, Galadari S, Oz M. Effects of endogenous cannabinoid anandamide on cardiac Na⁺/Ca²⁺ exchanger. Cell Calcium 2014; 55:231-7. [PMID: 24674601 DOI: 10.1016/j.ceca.2014.02.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Revised: 02/17/2014] [Accepted: 02/23/2014] [Indexed: 10/25/2022]
Abstract
Endocannabinoid anandamide (N-arachidonoyl ethanolamide; AEA) has been shown to cause negative inotropic and antiarrhythmic effects in ventricular myocytes. In this study, using whole-cell patch clamp technique, we have investigated the effects of AEA on cardiac Na(+)/Ca(2+) exchanger (NCX1)-mediated currents. AEA suppressed NCX1 with an IC50 value of 4.7 μM. Both inward and outward components of exchanger currents were suppressed by AEA equally. AEA inhibition was mimicked by the metabolically stable analogue, methanandamide (metAEA, 10 μM) while it was not influenced by inhibition of fatty acid amide hydrolase with 1 μM URB597 incubation. The effect of AEA, was not altered in the presence of cannabinoid receptor 1 and 2 antagonists AM251 (1 μM) and AM630 (1 μM), respectively. In addition, inhibition by AEA remained unchanged after pertussis toxin (PTX, 2 μg/ml) treatment or following the inclusion of GDP-β-S (1 mM) in pipette solution. Currents mediated by NCX1 expressed in HEK-293 cells were also inhibited by 10 μM AEA a partially reversible manner. Confocal microscopy images indicated that the intensity of YFP-NCX1 expression on cell surface was not altered by AEA. Collectively, the results indicate that AEA directly inhibits the function of NCX1 in rat ventricular myocytes and in HEK-293 cells expressing NCX1.
Collapse
Affiliation(s)
- Lina T Al Kury
- Laboratory of Functional Lipidomics, Department of Pharmacology, Faculty of Medicine and Health Sciences, UAE University, Al Ain, United Arab Emirates
| | - Keun-Hang Susan Yang
- Department of Biological Sciences, Schmid College of Science and Engineering, Chapman University, One University Drive, Orange, CA 92866, USA
| | - Faisal T Thayyullathil
- Department of Biochemistry, Faculty of Medicine and Health Sciences, UAE University, Al Ain, United Arab Emirates
| | - Mohanraj Rajesh
- Laboratory of Functional Lipidomics, Department of Pharmacology, Faculty of Medicine and Health Sciences, UAE University, Al Ain, United Arab Emirates
| | - Ramez M Ali
- Laboratory of Functional Lipidomics, Department of Pharmacology, Faculty of Medicine and Health Sciences, UAE University, Al Ain, United Arab Emirates
| | - Yaroslav M Shuba
- Bogomoletz Institute of Physiology and International Center of Molecular Physiology, National Academy of Sciences of Ukraine, Kyiv 24, Ukraine
| | - Frank Christopher Howarth
- Department of Physiology, Faculty of Medicine and Health Sciences, UAE University, Al Ain, United Arab Emirates
| | - Sehamuddin Galadari
- Department of Biochemistry, Faculty of Medicine and Health Sciences, UAE University, Al Ain, United Arab Emirates
| | - Murat Oz
- Laboratory of Functional Lipidomics, Department of Pharmacology, Faculty of Medicine and Health Sciences, UAE University, Al Ain, United Arab Emirates.
| |
Collapse
|
30
|
van Wieringen JP, Michel-Reher MB, Hatanaka T, Ueshima K, Michel MC. The new radioligand [(3)H]-L 748,337 differentially labels human and rat β3-adrenoceptors. Eur J Pharmacol 2013; 720:124-30. [PMID: 24183974 DOI: 10.1016/j.ejphar.2013.10.039] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2013] [Revised: 10/16/2013] [Accepted: 10/17/2013] [Indexed: 10/26/2022]
Abstract
As no suitable radioligand exists for the detection of β3-adrenoceptors, we have explored the radioligand binding properties of a tritiated version of the selective β3-adrenoceptor antagonist L 748,337. Kinetic and equilibrium saturation and competition binding experiments were performed with [(3)H]-L 748,337 on membrane fractions of HEK and CHO cells stably transfected with human and rat β-adrenoceptor subtypes. Based on both association/dissociation kinetic and equilibrium saturation binding studies in transfected HEK cells, [(3)H]-L 748,337 exhibited an affinity of approximately 2 nM for human β3-adrenoceptors. Competition studies with agonists and subtype-selective antagonists validated its binding to β3-adrenoceptors. In CHO cells transfected with human β3-adrenoceptors similar saturable high-affinity of [(3)H]-L 748,337 was observed. While some isoprenaline-sensitive [(3)H]-L 748,337 binding was also observed in CHO cells transfected with human β1- or β2-adrenoceptors, this was not saturable in a similar concentration range and/or not sensitive to the antagonists propranolol and SR 59,230, indicating that it did not primarily involve β-adrenoceptors. In CHO cells transfected with rat β3-adrenoceptors [(3)H]-L 748,337 exhibited a considerably lower affinity than with the human subtype (12-95 nM). Low affinity for the rat β3-adrenoceptor was also found with unlabelled L 748,337 in rat bladder strip relaxation experiments. We conclude that L 748,337 apparently has lower affinity for the rat than the human β3-adrenoceptors and that [(3)H]-L 748,337 can bind to a low-affinity site distinct from the orthosteric pocket of β-adrenoceptors. Nevertheless, [(3)H]-L 748,337 appears to be the most promising radioligand for the selective labelling of human β3-adrenoceptors reported to date.
Collapse
|
31
|
Wang J, Li M, Ma X, Bai K, Wang L, Yan Z, Lv T, Zhao Z, Zhao R, Liu H. Autoantibodies against the β3-adrenoceptor protect from cardiac dysfunction in a rat model of pressure overload. PLoS One 2013; 8:e78207. [PMID: 24147120 PMCID: PMC3795652 DOI: 10.1371/journal.pone.0078207] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2012] [Accepted: 09/17/2013] [Indexed: 11/29/2022] Open
Abstract
β3-adrenoceptors (β3-ARs) mediate a negative inotropic effect in human ventricular cardiomyocytes, which is opposite to that of β1- and β2-ARs. It has been previously demonstrated that autoantibodies against the β1/β2-AR exist in the sera of some patients with heart failure (HF) and these autoantibodies display agonist-like effects. Our aim in this study was to observe whether autoantibodies against the β3-AR (β3-AR Abs) exist in the sera of patients with HF and to assess the effects of β3-AR Abs on rat model of pressure overload cardiomyopthy. In the present study, the level of β3-AR Abs in the sera of HF patients was screened by ELISA. β3-AR Abs from HF patients were administrated to male adult rats with abdominal aortic banding (AAB), and the cardiac function was measured by echocardiographic examination and hemodynamic studies. The biological effects of this autoantibody on cardiomyocytes were evaluated using a motion-edge detection system, intracellular calcium transient assay, and patch clamp techniques. Compared to healthy subjects, the frequency of occurrence and titer of β3-AR Abs in the sera of HF patients were greatly increased, and β3-AR Abs could prevent LV dilation and improve the cardiac function of rats with AAB. β3-AR Abs exhibited negative chronotropic and inotropic effects and were accompanied by a decreased intracellular Ca2+ transient and membrane L-type Ca2+ current in cardiomyocytes. Our results demonstrated the existence of β3-AR Abs in the sera of patients with HF and found that this autoantibody could alleviate the cardiac dysfunction induced by pressure-overload in AAB rats.
Collapse
Affiliation(s)
- Jin Wang
- Department of Physiology, Shanxi Medical University, Taiyuan, Shanxi, P. R. China
| | - Meixia Li
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Science, Beijing, P. R. China
| | - Xiurui Ma
- Shanxi Cardiovascular Diseases Hospital, Taiyuan, Shanxi, P. R. China
| | - Kehua Bai
- Department of Physiology, Shanxi Medical University, Taiyuan, Shanxi, P. R. China
| | - Li Wang
- Department of Physiology, Shanxi Medical University, Taiyuan, Shanxi, P. R. China
| | - Zi Yan
- Department of Physiology, Shanxi Medical University, Taiyuan, Shanxi, P. R. China
| | - Tingting Lv
- School of Basic Medical Sciences, Cardiovascular Research Institute, Capital Medical University, Beijing, P. R. China
| | - Zhiqing Zhao
- Department of Physiology, Shanxi Medical University, Taiyuan, Shanxi, P. R. China
- Department of Basic Biomedical Sciences, Mercer University School of Medicine, Savannah, Georgia, United States of America
| | - Rongrui Zhao
- Department of Physiology, Shanxi Medical University, Taiyuan, Shanxi, P. R. China
| | - Huirong Liu
- Department of Physiology, Shanxi Medical University, Taiyuan, Shanxi, P. R. China
- School of Basic Medical Sciences, Cardiovascular Research Institute, Capital Medical University, Beijing, P. R. China
- * E-mail:
| |
Collapse
|
32
|
Effect of β3-adrenergic receptor on atrial L-type Ca(2+) current in rats with chronic heart failure. Heart Lung Circ 2013; 23:369-77. [PMID: 24055266 DOI: 10.1016/j.hlc.2013.08.008] [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: 03/02/2013] [Revised: 08/18/2013] [Accepted: 08/20/2013] [Indexed: 11/22/2022]
Abstract
OBJECTIVES To investigate the effect of selective β3-adrenoreceptor agonist BRL-37344 on L-type Ca(2+) current (Ica,L) and mRNA expression of L-type Ca(2+) channel α2δ-2 (Cacna2d2) in rats with chronic heart failure (CHF). METHODS Twenty-four male Wistar rats were divided into normal control (n=6) and CHF group (n=18), which were further divided into CHF control and BRL group (0.4nmol/kg, IV, twice weekly for four weeks). Echocardiography was performed to assess the structure and function of the left atrium (LA). RESULTS The LA in the BRL group (4.4 ± 0.2mm) was larger than in the normal control (3.5 ± 0.3mm, P<0.01) or CHF control (4.0 ± 0.2mm, P<0.05) group. The LA ejection fraction in the BRL group (36.2 ± 4.2%) was lower than in the normal control (58.0 ± 3.1%, P<0.01) or CHF control group (42.3 ± 4.8%, P<0.05). There was no difference in Ica,L density between the BRL group and CHF control group (8.3 ± 1.7 vs. 8.2 ± 2.6 pA/pF, P>0.05), which was higher than in the normal control group (6.0 ± 1.8 pA/pF, P<0.01). There was no difference in the mRNA expression of α2δ-2 (Cacna2d2) between the BRL group and CHF control group (0.264 ± 0.005 vs. 0.243 ± 0.017, P>0.05), which was also higher than in the normal control group (0.137 ± 0.013, P<0.01). CONCLUSION β3-Adrenoreceptor stimulation with BRL-37344 was associated with an increase in LA diameter and a decrease in LA function in chronic heart failure. These structural and function changes were not related to Ica,L or L-type Ca(2+) channel α2δ-2 (Cacna2d2) subunit in the LA myocytes.
Collapse
|
33
|
Miao G, Chen Z, Fang X, Liu M, Hao G, An H, Zhang Z, Lu L, Zhang J, Zhang L. Relationship between the autoantibody and expression of β3-adrenoceptor in lung and heart. PLoS One 2013; 8:e68747. [PMID: 23861938 PMCID: PMC3702604 DOI: 10.1371/journal.pone.0068747] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2012] [Accepted: 06/03/2013] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Evidences suggest that β3 -adrenoceptor (β3-AR) plays an important role in heart failure (HF), although no data is reported indicating how these effects may change with the increasing age. Pulmonary congestion and edema are the major life-threatening complications associated with HF. The purpose of this study is to explore the relationship between the anti-β3-AR autoantibody and the expression of β3-AR in the lungs and heart for both aged patients and rats with HF. METHODS Synthetic β3-AR peptides served as the target antigens in ELISA were used to screen the anti-β3-AR autoantibody in aged patients and rats. Two aged rat models were constructed based on aortic banding and sham-operation. The expression of β3-AR mRNA and protein in the lung and heart was measured in intervention and non-intervention groups by Western blot analysis at the baseline, 5(th), 7(th), 9(th) and 11(th) week, respectively. RESULTS The frequency and titer of anti-β3-AR autoantibody in aged patients and rats with HF were higher than those in the control group (p<0.05). The expression of β3-AR mRNA and protein in pulmonary tissues decreased continually from the 7(th) week (p<0.05), followed by HF observed during the 9(th) week. The expression of β3-AR in myocardial tissues continued to increase after the 9(th) week (p<0.05), and the expression of both β3-AR mRNA and protein in the BRL group [HF group with BRL37344 (4-[-[2-hydroxy-(3-chlorophenyl)ethyl-amino] phenoxyacetic acid) (a β3-AR agonist) injection] was positively correlated with BRL37344 when compared with non-BRL group (HF group without BRL37344 injection) (p<0.05). CONCLUSION Anti-β3-AR autoantibody was detected in aged patients and rats with HF. The expression of β3-AR mRNA and protein in pulmonary tissues decreased continually, and began earlier than in the heart, but its expression in myocardial tissues increased continually and could be further promoted by β3-AR agonist.
Collapse
Affiliation(s)
- Guobin Miao
- Department of Internal Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Zhe Chen
- Department of Internal Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Xiangyang Fang
- Department of Internal Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Miaobing Liu
- Department of Internal Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Gang Hao
- Department of Pathophysiology, Capital Medical University, Beijing, China
| | - Huiling An
- Department of Internal Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Zhiyong Zhang
- Heart Failure Center, Department of Cardiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Lingqiao Lu
- Department of Pathophysiology, Capital Medical University, Beijing, China
| | - Jian Zhang
- Medical and Health Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- * E-mail: (JZ); (LZ)
| | - Lin Zhang
- Heart Failure Center, Department of Cardiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
- * E-mail: (JZ); (LZ)
| |
Collapse
|
34
|
Krandycheva V, Kharin S, Strelkova M, Shumikhin K, Sobolev A, Shmakov D. Ventricular repolarization in a rat model of global heart failure. Clin Exp Pharmacol Physiol 2013; 40:431-7. [DOI: 10.1111/1440-1681.12104] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Revised: 04/02/2013] [Accepted: 04/29/2013] [Indexed: 11/28/2022]
Affiliation(s)
- Valeria Krandycheva
- Laboratory of Cardiac Physiology; Institute of Physiology of the Komi Science Centre of the Ural Branch of the Russian Academy of Sciences; Syktyvkar; Russia
| | - Sergey Kharin
- Laboratory of Cardiac Physiology; Institute of Physiology of the Komi Science Centre of the Ural Branch of the Russian Academy of Sciences; Syktyvkar; Russia
| | - Marina Strelkova
- Laboratory of Cardiac Physiology; Institute of Physiology of the Komi Science Centre of the Ural Branch of the Russian Academy of Sciences; Syktyvkar; Russia
| | - Konstantin Shumikhin
- Department of Biomedical Disciplines; Komi Branch of Kirov State Medical Academy; Syktyvkar; Russia
| | - Aleksey Sobolev
- Department of Physiology; Komi Branch of Kirov State Medical Academy; Syktyvkar; Russia
| | - Dmitry Shmakov
- Laboratory of Cardiac Physiology; Institute of Physiology of the Komi Science Centre of the Ural Branch of the Russian Academy of Sciences; Syktyvkar; Russia
| |
Collapse
|
35
|
Flynn R, Altier C. A macromolecular trafficking complex composed of β₂-adrenergic receptors, A-Kinase Anchoring Proteins and L-type calcium channels. J Recept Signal Transduct Res 2013; 33:172-6. [PMID: 23557075 DOI: 10.3109/10799893.2013.782219] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Abstract Sympathetic modulation of cardiac L-type calcium channels is an important mechanism for regulating heart rate and cardiac contractility. At the molecular level, activation of β-adrenergic receptors (βAR) increases calcium influx into cardiac myocytes by activating protein kinase A (PKA), leading to subsequent phosphorylation of L-type calcium channels. In the case of the β2AR, this process is facilitated by the presence of A-Kinase Anchoring Proteins (AKAPs) that serve as scaffolding proteins for the L-type calcium channel and the β2AR complex. Our work has shown that, in addition to facilitating PKA phosphorylation of the channel, AKAPs also promote an increase in the Cav1.2 channel surface expression. Here we review the molecular mechanisms of β2AR/AKAP/L-type channel interactions and trafficking.
Collapse
Affiliation(s)
- Robyn Flynn
- Department of Physiology and Pharmacology, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | | |
Collapse
|
36
|
Kim SJ, Zhang H, Khaliulin I, Choisy SCM, Bond R, Lin H, El Haou S, Milnes JT, Hancox JC, Suleiman MS, James AF. Activation of glibenclamide-sensitive ATP-sensitive K+ channels during β-adrenergically induced metabolic stress produces a substrate for atrial tachyarrhythmia. Circ Arrhythm Electrophysiol 2012; 5:1184-92. [PMID: 23159416 DOI: 10.1161/circep.112.975425] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Cardiac ATP-sensitive K(+) channels have been suggested to contribute to the adaptive physiological response to metabolic challenge after β-adrenoceptor stimulation. However, an increased atrial K(+)-conductance might be expected to be proarrhythmic. We investigated the effect of ATP-sensitive K(+) channel blockade on the electrophysiological responses to β-adrenoceptor-induced metabolic challenge in intact atria. METHODS AND RESULTS Atrial electrograms were recorded from the left atrial epicardial surface of Langendorff-perfused rat hearts using a 5×5 electrode array. Atrial effective refractory period and conduction velocity were measured using an S(1)-S(2) protocol. The proportion of hearts in which atrial tachyarrhythmia was produced by burst-pacing was used as an index of atrial tachyarrhythmia-inducibility. Atrial nucleotide concentrations were measured by high performance liquid chromatography. Perfusion with ≥10(-9) mol/L of the β-adrenoceptor agonist, isoproterenol (ISO), resulted in a concentration-dependent reduction of atrial effective refractory period and conduction velocity. The ISO-induced changes produced a proarrhythmic substrate such that atrial tachyarrhythmia could be induced by burst-pacing. Atrial [ATP] was significantly reduced by ISO (10(-6) mol/L). Perfusion with either of the ATP-sensitive K(+) channel blockers, glibenclamide (10(-5) mol/L) or tolbutamide (10(-3) mol/L), in the absence of ISO had no effect on basal atrial electrophysiology. On the other hand, the proarrhythmic substrate induced by 10(-6) mol/L ISO was abolished by either of the sulfonylureas, which prevented induction of atrial tachyarrhythmia. CONCLUSIONS Atrial ATP-sensitive K(+) channels activate in response to β-adrenergic metabolic stress in Langendorff-perfused rat hearts, resulting in a proarrhythmic substrate.
Collapse
Affiliation(s)
- Shang-Jin Kim
- Department of Pharmacology and Toxicology, College of Veterinary Medicine, Chonbuk National University, Jeonju-City, South Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Cyclic AMP-dependent protein kinase A regulates the alternative splicing of CaMKIIδ. PLoS One 2011; 6:e25745. [PMID: 22132070 PMCID: PMC3222655 DOI: 10.1371/journal.pone.0025745] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2011] [Accepted: 09/09/2011] [Indexed: 01/08/2023] Open
Abstract
Ca(2+)/calmodulin-dependent protein kinase (CaMK) IIδ is predominantly expressed in the heart. There are three isoforms of CaMKIIδ resulting from the alternative splicing of exons 14, 15, and 16 of its pre-mRNA, which is regulated by the splicing factor SF2/ASF. Inclusion of exons 15 and 16 or of exon 14 generates δA or δB isoform. The exclusion of all three exons gives rise to δC isoform, which is selectively increased in pressure-overload-induced hypertrophy. Overexpression of either δB or δC induces hypertrophy and heart failure, suggesting their specific role in the pathogenesis of hypertrophy and heart failure. It is well known that the β-adrenergic-cyclic AMP-dependent protein kinase A (PKA) pathway is implicated in heart failure. To determine the role of PKA in the alternative splicing of CaMKIIδ, we constructed mini-CaMKIIδ genes and used these genes to investigate the regulation of the alternative splicing of CaMKIIδ by PKA in cultured cells. We found that PKA promoted the exclusion of exons 14, 15, and 16 of CaMKIIδ, resulting in an increase in δC isoform. PKA interacted with and phosphorylated SF2/ASF, and enhanced SF2/ASF's activity to promote the exclusion of exons 14, 15, and 16 of CaMKIIδ, leading to a further increase in the expression of δC isoform. These findings suggest that abnormality in β-adrenergic-PKA signaling may contribute to cardiomyopathy and heart failure through dysregulation in the alternative splicing of CaMKIIδ exons 14, 15, and 16 and up-regulation of CaMKIIδC.
Collapse
|
38
|
von Homeyer P, Schwinn DA. Pharmacogenomics of β-adrenergic receptor physiology and response to β-blockade. Anesth Analg 2011; 113:1305-18. [PMID: 21965354 DOI: 10.1213/ane.0b013e31822b887e] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Myocardial β-adrenergic receptors (βARs) are important in altering heart rate, inotropic state, and myocardial relaxation (lusitropy). The β1AR and β2AR stimulation increases cyclic adenosine monophosphate concentration with the net result of myocyte contraction, whereas β3AR stimulation results in decreased inotropy. Downregulation of β1ARs in heart failure, as well as an increased β3AR activity and density, lead to decreased cyclic adenosine monophosphate production and reduced inotropy. The βAR antagonists are commonly used in patients with coronary artery disease and heart failure; however, perioperative use of βAR antagonists is controversial. Individual patient's response to beta-blocker therapy is an area of intensive research, and apart from pharmacokinetics, pharmacodynamics, and ethnic differences, genetic alterations have become more important in the last 20 years. The most common genetic variants in humans are single nucleotide polymorphisms (SNPs). There are 2 clinically relevant SNPs for the β1AR (Ser49Gly, Arg389Gly), 3 for the β2AR (Arg16Gly, Gln27Glu, Thr164Ile), and 1 for the β3AR (Trp64Arg). Although results are somewhat controversial, generally large datasets have the potential to show a relationship between βAR SNPs and outcomes such as development and progression of heart failure, coronary artery disease, vascular reactivity, hypertension, asthma, obesity, and diabetes. Although βAR SNPs may not directly cause disease, they appear to be risk factors for, and modifiers of, disease and the response to stress and drugs. In the perioperative setting, this has specifically been demonstrated for the Arg389Gly β1AR polymorphism with which patients with the Gly variant had a higher incidence of adverse perioperative events. Knowing that genetic variants play an important role, perioperative medicine will likely change from simple therapeutic intervention to a more personalized way of adrenergic receptor modulation.
Collapse
Affiliation(s)
- Peter von Homeyer
- Department of Anesthesiology & Pain Medicine, University of Washington, Seattle, WA 98195-6540, USA.
| | | |
Collapse
|
39
|
Yu J, Li W, Li Y, Zhao J, Wang L, Dong D, Pan Z, Yang B. Activation of β(3)-adrenoceptor promotes rapid pacing-induced atrial electrical remodeling in rabbits. Cell Physiol Biochem 2011; 28:87-96. [PMID: 21865851 DOI: 10.1159/000331717] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/26/2011] [Indexed: 01/24/2023] Open
Abstract
Cardiac electrophysiological function is under the regulatory control of the sympathetic nervous system. In addition to classical β-adrenoceptors (β-AR, including β(1)- and β(2)- subtypes), β(3)-AR is also expressed in human heart and shows its distinctive functions. This study is aimed to elucidate the role of β(3)-AR in the regulation of atrial fibrillation (AF), especially its role in rapid pacing-induced atrial electrical remodeling in rabbits. The rapid atrial pacing model was established by embedding electrodes in the right atrium pacing at a speed of 600 beats per minute. The protein level of β(3)-AR in the atria was found significantly upregulated by western blot. The atrial effective refractory period (AERP) and its rate adaptation were decreased after pacing which were further shortened by BRL37344, a selective β(3)-AR agonist, leading to the increase of AF inducibility and duration. Similarly, β(3)-AR activation induced time-dependent shortening of action potential duration (APD), together with decrease of L-type calcium current (I(Ca,L)) and increase of inward rectifier potassium current (I(K1)) and transient outward potassium current (I(to)) in rapid pacing atrial myocytes. Meanwhile, all the effects were abolished by specific β(3)-AR antagonist, SR59230A. In summary, our study represents that activation of β(3)-AR promotes the atrial electrical remodeling process by altering the balance of ion channels in atrial myocytes, which provides new insights into the pharmacological role of β(3)-AR in heart diseases.
Collapse
Affiliation(s)
- Jiahui Yu
- Department of Cardiology, the First Affiliated Hospital, Harbin Medical University, Harbin, RP China
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Carll AP, Willis MS, Lust RM, Costa DL, Farraj AK. Merits of non-invasive rat models of left ventricular heart failure. Cardiovasc Toxicol 2011; 11:91-112. [PMID: 21279739 DOI: 10.1007/s12012-011-9103-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Heart failure (HF) is characterized as a limitation to cardiac output that prevents the heart from supplying tissues with adequate oxygen and predisposes individuals to pulmonary edema. Impaired cardiac function is secondary to either decreased contractility reducing ejection (systolic failure), diminished ventricular compliance preventing filling (diastolic failure), or both. To study HF etiology, many different techniques have been developed to elicit this condition in experimental animals, with varying degrees of success. Among rats, surgically induced HF models are the most prevalent, but they bear several shortcomings, including high mortality rates and limited recapitulation of the pathophysiology, etiology, and progression of human HF. Alternatively, a number of non-invasive HF induction methods avoid many of these pitfalls, and their merits in technical simplicity, reliability, survivability, and comparability to the pathophysiologic and pathogenic characteristics of HF are reviewed herein. In particular, this review focuses on the primary pathogenic mechanisms common to genetic strains (spontaneously hypertensive and spontaneously hypertensive heart failure), pharmacological models of toxic cardiomyopathy (doxorubicin and isoproterenol), and dietary salt models, all of which have been shown to induce left ventricular HF in the rat. Additional non-invasive techniques that may potentially enable the development of new HF models are also discussed.
Collapse
Affiliation(s)
- Alex P Carll
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, 27599 USA.
| | | | | | | | | |
Collapse
|
41
|
Abstract
Pharmacological and molecular approaches have shown that an atypical β-adrenoceptor (AR), called β(3)-AR, that is distinct from β(1)-ARs and β(2)-ARs, exists in some tissues in heterogeneous populations such as β(3a)-ARs and β(3b)-ARs. β(3)-ARs belong to a superfamily of receptors linked to guanine nucleotide binding proteins (G proteins). The β(3)-AR gene contains two introns whereas the β(1)-AR and β(2)-AR genes are intronless, leading to splice variants. β(3)-ARs can couple to G(i) and G(s) and they are reported to be present in brown adipose tissue, vasculature, the heart, among other tissues. β(3)-ARs cause vasodilation of microvessels in the islets of Langerhans and may participate in the pathogenesis of cardiac failure, during which modification of β(1)-AR and β(2)-AR expression occurs. The development of β(3)-AR agonists has led to the elaboration of promising new drugs, including antiobesity and antidiabetic drugs. This article reviews the various pharmacological actions of β(3)-ARs and their clinical implications for diabetes and cardiovascular diseases.
Collapse
Affiliation(s)
| | | | - Anita A. Mehta
- Department of Pharmacology, LM College of Pharmacy, Ahmedabad, Gujarat, India
| | | |
Collapse
|
42
|
Chen Z, Miao G, Liu M, Hao G, Liu Y, Fang X, Zhang Z, Lu L, Zhang J, Zhang L. Age-related up-regulation of beta3-adrenergic receptor in heart-failure rats. J Recept Signal Transduct Res 2010; 30:227-33. [PMID: 20443655 DOI: 10.3109/10799891003801918] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Stimulation of beta1- and beta2-adrenergic receptors (ARs) in the heart results in positive inotropy. In contrast, it has been reported that the beta3-AR is also expressed in the heart and that its stimulation leads to negative inotropic effects. The aim of this study was to investigate the expression of beta3-AR in age-related heart-failure rats and its relevance to left ventricular dysfunction. Aging male Wistar rats were divided into young and aging groups according to age, and each group included sham-operation and heart-failure subgroups. Left ventricular end-diastolic pressure (LVEDP) and the ratio of left ventricular weight to body weight (LV/BW) were significantly higher for the aging heart-failure versus young heart-failure and the heart-failure versus sham-operation groups (P < 0.01, respectively). However, the left ventricular end-systolic pressure (LVESP) and the maximal rate of rise or fall of left ventricular pressure were all significantly lower for the aging heart-failure versus young heart-failure and the heart-failure versus sham-operation groups (P < 0.01, respectively). beta3-AR protein levels increased significantly when heart failure worsened in aging rats. These results suggest that beta3-AR expression in age-related heart-failure rats and left ventricular function were highly correlated.
Collapse
Affiliation(s)
- Zhe Chen
- Department of Internal Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Carll AP, Haykal-Coates N, Winsett DW, Rowan WH, Hazari MS, Ledbetter AD, Nyska A, Cascio WE, Watkinson WP, Costa DL, Farraj AK. Particulate matter inhalation exacerbates cardiopulmonary injury in a rat model of isoproterenol-induced cardiomyopathy. Inhal Toxicol 2010; 22:355-68. [PMID: 20121584 DOI: 10.3109/08958370903365692] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Ambient particulate matter (PM) exposure is linked to cardiovascular events and death, especially among individuals with heart disease. A model of toxic cardiomyopathy was developed in Spontaneously Hypertensive Heart Failure (SHHF) rats to explore potential mechanisms. Rats were infused with isoproterenol (ISO; 2.5 mg/kg/day subcutaneous [sc]), a beta-adrenergic agonist, for 28 days and subsequently exposed to PM by inhalation. ISO induced tachycardia and hypotension throughout treatment followed by postinfusion decrements in heart rate, contractility, and blood pressures (systolic, diastolic, pulse), and fibrotic cardiomyopathy. Changes in heart rate and heart rate variability (HRV) 17 days after ISO cessation indicated parasympathetic dominance with concomitantly altered ventilation. Rats were subsequently exposed to filtered air or Harvard Particle 12 (HP12) (12 mg/m(3))--a metal-rich oil combustion-derived PM--at 18 and 19 days (4 h/day) after ISO infusion via nose-only inhalation to determine if cardio-impaired rats were more responsive to the effects of PM exposure. Inhalation of PM among ISO-pretreated rats significantly increased pulmonary lactate dehydrogenase, serum high-density lipoprotein (HDL) cholesterol, and heart-to-body mass ratio. PM exposure increased the number of ISO-pretreated rats that experienced bradyarrhythmic events, which occurred concomitantly with acute alterations of HRV. PM, however, did not significantly affect mean HRV in the ISO- or saline-pretreated groups. In summary, subchronic ISO treatment elicited some pathophysiologic and histopathological features of heart failure, including cardiomyopathy. The enhanced sensitivity to PM exposure in SHHF rats with ISO-accelerated cardiomyopathy suggests that this model may be useful for elucidating the mechanisms by which PM exposure exacerbates heart disease.
Collapse
Affiliation(s)
- Alex P Carll
- Environmental Sciences and Engineering, UNC Gillings School of Global Public Health, Chapel Hill, North Carolina, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Ufer C, Germack R. Cross-regulation between beta 1- and beta 3-adrenoceptors following chronic beta-adrenergic stimulation in neonatal rat cardiomyocytes. Br J Pharmacol 2010; 158:300-13. [PMID: 19719783 DOI: 10.1111/j.1476-5381.2009.00328.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
BACKGROUND AND PURPOSE We have previously shown that beta-adrenoceptors continuously stimulated with noradrenaline induces an increase in beta(3)-adrenoceptors (G alpha(i)PCRs) and a decrease in beta(1)-adrenoceptors (G alpha(s)PCRs) at functional, genomic and protein levels. This compensatory modification induced by noradrenaline is probably one of the consequences of cardiac depression observed in heart disease. Therefore, we investigated further the interaction between beta(1)- and beta(3)-adrenoceptors in neonatal rat cardiomyocytes. EXPERIMENTAL APPROACH Functional studies were performed by cyclic adenosine monophosphate (cAMP) accumulation assays in cells untreated or treated with dobutamine and ICI 118551 (beta(1)-adrenoceptor) or CL-3162436243 (beta(3)-adrenoceptor) for 24 h in the presence or absence of protein kinase inhibitors. Beta-adrenoceptor and protein kinase expression was monitored by quantitative reverse transcription-polymerase chain reaction (RT-PCR) and by Western blotting, respectively. KEY RESULTS Chronic beta(1)- or beta(3)-adrenoceptor stimulation reduced beta(1)-adrenoceptor-mediated cAMP accumulation in association with a decrease in beta(1)-adrenoceptor mRNA and protein levels through protein kinase C (PKC), phosphoinositide 3-kinase (PI3K) and p38 mitogen-activated protein kinase (p38MAPK) activation. In contrast, both treatments induced an increase in beta(3)-adrenoceptor expression and beta(3)-adrenoceptor-inhibited forskolin response through PKC, extracellular-signal-regulated kinases 1 and 2 (ERK1/2) and p38MAPK phosphorylation, although no beta(3)-adrenoceptor response was observed in untreated cells. ERK1/2 and p38MAPK were activated by both treatments. The modulation of beta(1)- or beta(3)-adrenoceptor function did not require stress-activated protein kinase/c-Jun N-terminal kinase (SAPK/JNK) although chronic beta(1)-adrenoceptor stimulation activated SAPK/JNK. Beta(3)-adrenoceptor treatment activated Akt although PI3K was not involved in beta(3)-adrenoceptor up-regulation. CONCLUSION AND IMPLICATIONS We show for the first time that chronic beta(1)- or beta(3)-adrenoceptor stimulation leads to the modulation of beta(1)- and beta(3)-adrenoceptors by a cross-regulation involving PKC, PI3K p38MAPK and MEK/ERK1/2 pathway, and through protein kinase A when beta(1)-adrenoceptors are chronically activated.
Collapse
Affiliation(s)
- Christoph Ufer
- Institute of Biochemistry, University Medecine Berlin-Charité, Berlin, Germany; Biomedical Research Centre, School of Biomedical and Natural Sciences, Nottingham Trent University, Nottingham, UK
| | | |
Collapse
|
45
|
Cheng HJ, Grant KA, Han QH, Daunais JB, Friedman DP, Masutani S, Little WC, Cheng CP. Up-regulation and functional effect of cardiac β3-adrenoreceptors in alcoholic monkeys. Alcohol Clin Exp Res 2010; 34:1171-81. [PMID: 20477780 DOI: 10.1111/j.1530-0277.2010.01194.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
BACKGROUND Recent studies link altered cardiac beta-adrenergic receptor (AR) signaling to the pathology of alcoholic cardiomyopathy (ACM). However, the alteration and functional effect of beta(3)-AR activation in ACM are unknown. We tested the hypothesis that chronic alcohol intake causes an up-regulation of cardiac beta(3)-AR, which exacerbates myocyte dysfunction and impairs calcium regulation, thereby directly contributing to the progression of ACM. METHODS We compared myocyte beta(3)- and beta(1)-AR expression and myocyte contractile ([Ca(2+)](i)), transient ([Ca(2+)](iT)), and Ca(2+) current (I(Ca,L)) responses to beta- and beta(3)-AR stimulation in myocytes obtained from left ventricle (LV) tissue samples obtained from 10 normal control (C) and 16 monkeys with self-administered alcohol for 12 months prior to necropsy: 6 moderate (M) and 10 heavy (H) drinkers with group average alcohol intakes of 1.5 +/- 0.2 and 3.3 +/- 0.2 g/kg/d, respectively. RESULTS Compared with control myocytes (C), in alcoholic cardiomyocytes, basal cell contraction (dL/dt(max), -39%, H: 69.8 vs. C: 114.6 microm/s), relaxation (dR/dt(max), -37%, 58.2 vs. 92.9 microm/s), [Ca(2+)](iT) (-34%, 0.23 vs. 0.35), and I(Ca,L) (-25%, 4.8 vs. 6.4pA/pF) were all significantly reduced. Compared with controls, in moderate and heavy drinkers, beta(1)-AR protein levels decreased by 23% and 42%, but beta(3)-AR protein increased by 46% and 85%, respectively. These changes were associated with altered myocyte functional responses to beta-AR agonist, isoproterenol (ISO), and beta(3)-AR agonist, BRL-37344 (BRL). Compared with controls, in alcoholic myocytes, ISO (10(-8) M) produced significantly smaller increases in dL/dt(max) (H: 40% vs. C: 71%), dR/dt(max) (37% vs. 52%), [Ca(2+)](iT) (17% vs. 37%), and I(Ca,L) (17% vs. 27%), but BRL (10(-8) M) produced a significantly greater decrease in dL/dt(max) (H: -23% vs. C: -11%), [Ca(2+)](iT) (-30% vs. -11%), and I(Ca,L) (-28% vs. -17%). CONCLUSIONS Chronic alcohol consumption down-regulates cardiac beta(1)- and up-regulates beta(3)-ARs, contributing to the abnormal response to catecholamines in ACM. The up-regulation of cardiac beta(3)-AR signaling enhances inhibition of LV myocyte contraction and relaxation and exacerbates the dysfunctional [Ca(2+)](i) regulation and, thus, may precede the development of ACM.
Collapse
Affiliation(s)
- Heng-Jie Cheng
- Department of Internal Medicine-Cardiology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Moens AL, Yang R, Watts VL, Barouch LA. Beta 3-adrenoreceptor regulation of nitric oxide in the cardiovascular system. J Mol Cell Cardiol 2010; 48:1088-95. [PMID: 20184889 DOI: 10.1016/j.yjmcc.2010.02.011] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2009] [Revised: 12/08/2009] [Accepted: 02/12/2010] [Indexed: 11/18/2022]
Abstract
The presence of a third beta-adrenergic receptor (beta 3-AR) in the cardiovascular system has challenged the classical paradigm of sympathetic regulation by beta1- and beta2-adrenergic receptors. While beta 3-AR's role in the cardiovascular system remains controversial, increasing evidence suggests that it serves as a "brake" in sympathetic overstimulation - it is activated at high catecholamine concentrations, producing a negative inotropic effect that antagonizes beta1- and beta2-AR activity. The anti-adrenergic effects induced by beta 3-AR were initially linked to nitric oxide (NO) release via endothelial NO synthase (eNOS), although more recently it has been shown under some conditions to increase NO production in the cardiovascular system via the other two NOS isoforms, namely inducible NOS (iNOS) and neuronal NOS (nNOS). We summarize recent findings regarding beta 3-AR effects on the cardiovascular system and explore its prospective as a therapeutic target, particularly focusing on its emerging role as an important mediator of NO signaling in the pathogenesis of cardiovascular disorders.
Collapse
Affiliation(s)
- An L Moens
- Johns Hopkins University School of Medicine, Division of Cardiology, Baltimore, MD 21205, USA
| | | | | | | |
Collapse
|
47
|
Kohr MJ, Kaludercic N, Tocchetti CG, Dong Gao W, Kass DA, Janssen PML, Paolocci N, Ziolo MT. Nitroxyl enhances myocyte Ca2+ transients by exclusively targeting SR Ca2+-cycling. Front Biosci (Elite Ed) 2010; 2:614-26. [PMID: 20036906 DOI: 10.2741/e118] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Nitroxyl (HNO), the 1-electron reduction product of nitric oxide, improves myocardial contraction in normal and failing hearts. Here we test whether the HNO donor Angeli's salt (AS) will change myocyte action potential (AP) waveform by altering the L-type Ca2+ current (ICa) and contrast the contractile effects of HNO with that of the hydroxyl radical (.OH) and nitrite (NO2-), two potential breakdown products of AS. We confirmed the positive effect of AS/HNO on basal cardiomyocyte function, as opposed to the detrimental effect of .OH and the negligible effect of NO2-. Upon examination of the myocyte AP, we observed no change in resting membrane potential or AP duration to 20 per cent repolarization with AS/HNO, whereas AP duration to 90 per cent repolarization was slightly prolonged. However, perfusion with AS/HNO did not elicit a change in basal ICa, but did hasten ICa inactivation. Upon further examination of the SR, the AS/HNO-induced increase in cardiomyocyte Ca2+ transients was abolished with inhibition of SR Ca2+-cycling. Therefore, the HNO-induced increase in Ca2+ transients results exclusively from changes in SR Ca2+-cycling, and not from ICa.
Collapse
Affiliation(s)
- Mark J Kohr
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Dessy C, Balligand JL. Beta3-adrenergic receptors in cardiac and vascular tissues emerging concepts and therapeutic perspectives. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2010; 59:135-63. [PMID: 20933201 DOI: 10.1016/s1054-3589(10)59005-7] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Catecholamines released by the orthosympathetic system play a major role in the short- and long-term regulation of cardiovascular function. Beta1- and beta2-adrenoreceptors (ARs) have classically been considered as mediating most of their effects on cardiac contraction. After their initial cloning and pharmacologic characterization in the late 1980s, beta3-ARs have been mostly thought of as receptors mediating metabolic effects (e.g., lipolysis) in adipocytes. However, definitive evidence for their expression and functional coupling in cardiovascular tissues (including in humans) has recently initiated a re-examination of their implication in the pathophysiology of cardiovascular diseases. Distinctive pharmacodynamic properties of beta3-AR, e.g., their upregulation in disease and resistance to desensitization, suggest that they may be attractive targets for therapeutic intervention. They may substitute efficient vasodilating pathways when beta1/2-ARs are inoperative. In the heart, their contractile effects, which are functionally antipathetic to those of beta1/2-AR, may protect the myocardium against adverse effects of excessive catecholamine stimulation and perhaps mediate additional ancillary effects on key aspects of electrophysiology or remodeling. Longitudinal studies in animals and patients with different stages of heart failure are now needed to identify the optimal therapeutic scheme using specific combinations of agonists or antagonists at all three beta-ARs.
Collapse
Affiliation(s)
- Chantal Dessy
- Pole of Pharmacology and Therapeutics, Institute of Experimental and Clinical Research, Université catholique de Louvain, Brussels, Belgium
| | | |
Collapse
|
49
|
Audigane L, Kerfant BG, El Harchi A, Lorenzen-Schmidt I, Toumaniantz G, Cantereau A, Potreau D, Charpentier F, Noireaud J, Gauthier C. Rabbit, a relevant model for the study of cardiac beta 3-adrenoceptors. Exp Physiol 2009; 94:400-11. [PMID: 19151075 DOI: 10.1113/expphysiol.2008.045179] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The beta(3)-adrenoceptors (beta(3)-ARs) have been identified and characterized in the human heart. Specific beta(3)-AR stimulation, unlike beta(1)-AR or beta(2)-AR stimulation, decreases cardiac contractility, partly via the G(i)-NO pathway. However, the precise role of cardiac beta(3)-ARs is not yet completely understood. Indeed, under normal conditions, the beta(3)-AR response is present only to a very low degree in rats and mice. Therefore, we evaluated whether beta(3)-ARs were present and functional in rabbit ventricular cardiomyocytes, and whether the rabbit could serve as a relevant model for the study of cardiac beta(3)-ARs. We used RT-PCR and Western blot to measure the beta(3)-AR transcripts and protein levels in rabbit ventricular cardiomyocytes. We also analysed the effect of beta(3)-AR stimulation using isoproterenol in combination with nadolol or SR 58611A on cardiomyocyte shortening, Ca(2+) transient, L-type Ca(2+) current (I(Ca,L)), delayed rectifier potassium current (I(Ks)) and action potential duration (APD). For the first time, we show that beta(3)-ARs are expressed in rabbit ventricular cardiomyocytes. The mRNA and protein sequences present a high homology to those of rat and human beta(3)-ARs. Furthermore, beta(3)-AR stimulation decreases cardiomyocyte shortening, Ca(2+) transient and I(Ca,L) amplitudes, via a G(i)-NO pathway. Importantly, beta(3)-AR stimulation enhances I(Ks) amplitude and shortens the APD. Taken together, our results indicate that the rabbit provides a relevant model, easily used in laboratories, to study the roles of cardiac beta(3)-ARs in physiological conditions.
Collapse
Affiliation(s)
- Leslie Audigane
- INSERM, UMR 915, l'institut du thorax, Nantes, F-44035 France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Tillinger A, Myslive��ek J, Nov��kov�� M, Krizanova O, Kvet��ansk�� R. Gene Expression of Adrenoceptors in the Hearts of Cold-Acclimated Rats Exposed to a Novel Stressor. Ann N Y Acad Sci 2008; 1148:393-9. [DOI: 10.1196/annals.1410.024] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|