1
|
Semmler L, Jeising T, Huettemeister J, Bathe-Peters M, Georgoula K, Roshanbin R, Sander P, Fu S, Bode D, Hohendanner F, Pieske B, Annibale P, Schiattarella GG, Oeing CU, Heinzel FR. Impairment of the adrenergic reserve associated with exercise intolerance in a murine model of heart failure with preserved ejection fraction. Acta Physiol (Oxf) 2024; 240:e14124. [PMID: 38436094 DOI: 10.1111/apha.14124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 12/27/2023] [Accepted: 02/20/2024] [Indexed: 03/05/2024]
Abstract
AIM Exercise intolerance is the central symptom in patients with heart failure with preserved ejection fraction. In the present study, we investigated the adrenergic reserve both in vivo and in cardiomyocytes of a murine cardiometabolic HFpEF model. METHODS 12-week-old male C57BL/6J mice were fed regular chow (control) or a high-fat diet and L-NAME (HFpEF) for 15 weeks. At 27 weeks, we performed (stress) echocardiography and exercise testing and measured the adrenergic reserve and its modulation by nitric oxide and reactive oxygen species in left ventricular cardiomyocytes. RESULTS HFpEF mice (preserved left ventricular ejection fraction, increased E/e', pulmonary congestion [wet lung weight/TL]) exhibited reduced exercise capacity and a reduction of stroke volume and cardiac output with adrenergic stress. In ventricular cardiomyocytes isolated from HFpEF mice, sarcomere shortening had a higher amplitude and faster relaxation compared to control animals. Increased shortening was caused by a shift of myofilament calcium sensitivity. With addition of isoproterenol, there were no differences in sarcomere function between HFpEF and control mice. This resulted in a reduced inotropic and lusitropic reserve in HFpEF cardiomyocytes. Preincubation with inhibitors of nitric oxide synthases or glutathione partially restored the adrenergic reserve in cardiomyocytes in HFpEF. CONCLUSION In this murine HFpEF model, the cardiac output reserve on adrenergic stimulation is impaired. In ventricular cardiomyocytes, we found a congruent loss of the adrenergic inotropic and lusitropic reserve. This was caused by increased contractility and faster relaxation at rest, partially mediated by nitro-oxidative signaling.
Collapse
Affiliation(s)
- Lukas Semmler
- Department of Internal Medicine and Cardiology, German Heart Center Charité (DHZC) - Campus Virchow-Klinikum, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
| | - Tobias Jeising
- Department of Internal Medicine and Cardiology, German Heart Center Charité (DHZC) - Campus Virchow-Klinikum, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
| | - Judith Huettemeister
- Department of Internal Medicine and Cardiology, German Heart Center Charité (DHZC) - Campus Virchow-Klinikum, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
| | - Marc Bathe-Peters
- Receptor Signalling Group, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- School of Physics and Astronomy, University of St Andrews, St Andrews, UK
| | - Konstantina Georgoula
- Receptor Signalling Group, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Rashin Roshanbin
- Department of Internal Medicine and Cardiology, German Heart Center Charité (DHZC) - Campus Virchow-Klinikum, Berlin, Germany
| | - Paulina Sander
- Department of Internal Medicine and Cardiology, German Heart Center Charité (DHZC) - Campus Virchow-Klinikum, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
| | - Shu Fu
- Department of Internal Medicine and Cardiology, German Heart Center Charité (DHZC) - Campus Virchow-Klinikum, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
| | - David Bode
- Department of Internal Medicine and Cardiology, German Heart Center Charité (DHZC) - Campus Virchow-Klinikum, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
| | - Felix Hohendanner
- Department of Internal Medicine and Cardiology, German Heart Center Charité (DHZC) - Campus Virchow-Klinikum, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
| | - Burkert Pieske
- Division of Cardiology, Department of Internal Medicine, University Medicine Rostock, Rostock, Germany
| | - Paolo Annibale
- Receptor Signalling Group, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- School of Physics and Astronomy, University of St Andrews, St Andrews, UK
| | - Gabriele G Schiattarella
- Department of Internal Medicine and Cardiology, German Heart Center Charité (DHZC) - Campus Virchow-Klinikum, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
- Translational Approaches in Heart Failure and Cardiometabolic Disease, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Christian U Oeing
- Department of Internal Medicine and Cardiology, German Heart Center Charité (DHZC) - Campus Virchow-Klinikum, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
| | - Frank R Heinzel
- Department of Internal Medicine and Cardiology, German Heart Center Charité (DHZC) - Campus Virchow-Klinikum, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
- 2. Medizinische Klinik - Kardiologie, Angiologie, Intensivmedizin, Städtisches Klinikum Dresden, Dresden, Germany
| |
Collapse
|
2
|
Ham S, Mukaida S, Sato M, Keov P, Bengtsson T, Furness S, Holliday ND, Evans BA, Summers RJ, Hutchinson DS. Role of G protein-coupled receptor kinases (GRKs) in β 2 -adrenoceptor-mediated glucose uptake. Pharmacol Res Perspect 2024; 12:e1176. [PMID: 38332691 PMCID: PMC10853676 DOI: 10.1002/prp2.1176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 12/17/2023] [Accepted: 01/22/2024] [Indexed: 02/10/2024] Open
Abstract
Truncation of the C-terminal tail of the β2 -AR, transfection of βARKct or over-expression of a kinase-dead GRK mutant reduces isoprenaline-stimulated glucose uptake, indicating that GRK is important for this response. We explored whether phosphorylation of the β2 -AR by GRK2 has a role in glucose uptake or if this response is related to the role of GRK2 as a scaffolding protein. CHO-GLUT4myc cells expressing wild-type and mutant β2 -ARs were generated and receptor affinity for [3 H]-CGP12177A and density of binding sites determined together with the affinity of isoprenaline and BRL37344. Following receptor activation by β2 -AR agonists, cAMP accumulation, GLUT4 translocation, [3 H]-2-deoxyglucose uptake, and β2 -AR internalization were measured. Bioluminescence resonance energy transfer was used to investigate interactions between β2 -AR and β-arrestin2 or between β2 -AR and GRK2. Glucose uptake after siRNA knockdown or GRK inhibitors was measured in response to β2 -AR agonists. BRL37344 was a poor partial agonist for cAMP generation but displayed similar potency and efficacy to isoprenaline for glucose uptake and GLUT4 translocation. These responses to β2 -AR agonists occurred in CHO-GLUT4myc cells expressing β2 -ARs lacking GRK or GRK/PKA phosphorylation sites as well as in cells expressing the wild-type β2 -AR. However, β2 -ARs lacking phosphorylation sites failed to recruit β-arrestin2 and did not internalize. GRK2 knock-down or GRK2 inhibitors decreased isoprenaline-stimulated glucose uptake in rat L6 skeletal muscle cells. Thus, GRK phosphorylation of the β2 -AR is not associated with isoprenaline- or BRL37344-stimulated glucose uptake. However, GRKs acting as scaffold proteins are important for glucose uptake as GRK2 knock-down or GRK2 inhibition reduces isoprenaline-stimulated glucose uptake.
Collapse
Affiliation(s)
- Seungmin Ham
- Drug Discovery BiologyMonash Institute of Pharmaceutical Sciences, Monash UniversityParkvilleVictoriaAustralia
| | - Saori Mukaida
- Drug Discovery BiologyMonash Institute of Pharmaceutical Sciences, Monash UniversityParkvilleVictoriaAustralia
| | - Masaaki Sato
- Drug Discovery BiologyMonash Institute of Pharmaceutical Sciences, Monash UniversityParkvilleVictoriaAustralia
| | - Peter Keov
- Drug Discovery BiologyMonash Institute of Pharmaceutical Sciences, Monash UniversityParkvilleVictoriaAustralia
| | - Tore Bengtsson
- Atrogi ABStockholmSweden
- Department of Molecular BiosciencesThe Wenner‐Gren Institute, Stockholm UniversityStockholmSweden
| | - Sebastian Furness
- Drug Discovery BiologyMonash Institute of Pharmaceutical Sciences, Monash UniversityParkvilleVictoriaAustralia
| | - Nicholas D. Holliday
- School of Life Sciences, The Medical School, Queen's Medical CentreUniversity of NottinghamNottinghamUK
- Excellerate Bioscience, BiocityNottinghamUK
| | - Bronwyn A. Evans
- Drug Discovery BiologyMonash Institute of Pharmaceutical Sciences, Monash UniversityParkvilleVictoriaAustralia
| | - Roger J. Summers
- Drug Discovery BiologyMonash Institute of Pharmaceutical Sciences, Monash UniversityParkvilleVictoriaAustralia
| | - Dana S. Hutchinson
- Drug Discovery BiologyMonash Institute of Pharmaceutical Sciences, Monash UniversityParkvilleVictoriaAustralia
| |
Collapse
|
3
|
Abstract
Insulin receptors are highly expressed in the heart and vasculature. Insulin signaling regulates cardiac growth, survival, substrate uptake, utilization, and mitochondrial metabolism. Insulin signaling modulates the cardiac responses to physiological and pathological stressors. Altered insulin signaling in the heart may contribute to the pathophysiology of ventricular remodeling and heart failure progression. Myocardial insulin signaling adapts rapidly to changes in the systemic metabolic milieu. What may initially represent an adaptation to protect the heart from carbotoxicity may contribute to amplifying the risk of heart failure in obesity and diabetes. This review article presents the multiple roles of insulin signaling in cardiac physiology and pathology and discusses the potential therapeutic consequences of modulating myocardial insulin signaling.
Collapse
Affiliation(s)
- E Dale Abel
- Division of Endocrinology, Metabolism and Diabetes and Fraternal Order of Eagles Diabetes Research Center, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| |
Collapse
|
4
|
De Jong KA, Nikolaev VO. Multifaceted remodelling of cAMP microdomains driven by different aetiologies of heart failure. FEBS J 2021; 288:6603-6622. [DOI: 10.1111/febs.15706] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 12/22/2020] [Accepted: 01/06/2021] [Indexed: 12/14/2022]
Affiliation(s)
- Kirstie A. De Jong
- Institute of Experimental Cardiovascular Research University Medical Center Hamburg‐Eppendorf Hamburg Germany
- German Center for Cardiovascular Research (DZHK) partner site Hamburg/Kiel/Lübeck D‐20246 Hamburg Germany
| | - Viacheslav O. Nikolaev
- Institute of Experimental Cardiovascular Research University Medical Center Hamburg‐Eppendorf Hamburg Germany
- German Center for Cardiovascular Research (DZHK) partner site Hamburg/Kiel/Lübeck D‐20246 Hamburg Germany
| |
Collapse
|
5
|
The Metabolic Role of GRK2 in Insulin Resistance and Associated Conditions. Cells 2021; 10:cells10010167. [PMID: 33467677 PMCID: PMC7830135 DOI: 10.3390/cells10010167] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 01/11/2021] [Accepted: 01/12/2021] [Indexed: 02/07/2023] Open
Abstract
Insulin resistance (IRES) is a pathophysiological condition characterized by the reduced response to insulin of several tissues, including myocardial and skeletal muscle. IRES is associated with obesity, glucose intolerance, dyslipidemia, and hypertension, evolves toward type 2 diabetes, and increases the risk of developing cardiovascular diseases. Several studies designed to explore the mechanisms involved in IRES allowed the identification of a multitude of potential molecular targets. Among the most promising, G Protein Coupled Receptor Kinase type 2 (GRK2) appears to be a suitable one given its functional implications in many cellular processes. In this review, we will discuss the metabolic role of GRK2 in those conditions that are characterized by insulin resistance (diabetes, hypertension, heart failure), and the potentiality of its inhibition as a therapeutic strategy to revert both insulin resistance and its associated phenotypes.
Collapse
|
6
|
Sun X, Zhou M, Wen G, Huang Y, Wu J, Peng L, Jiang W, Yuan H, Lu Y, Cai J. Paroxetine Attenuates Cardiac Hypertrophy Via Blocking GRK2 and ADRB1 Interaction in Hypertension. J Am Heart Assoc 2020; 10:e016364. [PMID: 33372534 PMCID: PMC7955481 DOI: 10.1161/jaha.120.016364] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Background ADRB1 (adrenergic receptor beta 1) responds to neuroendocrine stimulations, which have great implications in hypertension. GRK2 (G protein‐coupled receptor kinase 2) is an essential regulator for many G protein‐coupled receptors and subsequent cell signaling cascades, but its role as a regulator of ADRB1 and associated cardiac hypertrophy in hypertension remains to be elucidated. Methods and Results In this study, we found the expressions of GRK2 and ADRB1 in peripheral blood mononuclear cells were positively associated with blood pressure levels in hypertensive patients and with their expression in heart. In vitro evidence showed a direct interaction in ADRB1 and GRK2 and genetic depletion of GRK2 blocks epinephrine‐induced upregulation of hypertrophic and fibrotic genes in cardiomyocytes. Meanwhile, we discovered a selective serotonin reuptake inhibitor paroxetine specifically blockades GRK2 and ADRB1 interaction. In vivo, paroxetine treatment ameliorates hypertension‐induced cardiac hypertrophy, dysfunction, and fibrosis in animal models. We found that paroxetine suppressed sympathetic overdrive and increased the adrenergic receptor sensitivity to catecholamines. Paroxetine treatment also blocks epinephrine‐induced upregulation of hypertrophic and fibrotic genes as well as ADRB1 internalization in cardiomyocytes. Coadministration of paroxetine further potentiates metoprolol‐induced reductions in blood pressure and heart rate, further attenuating cardiac hypertrophy in spontaneously hypertensive rats. Furthermore, in patients with hypertension accompanied with depression, we observed that cardiac remodeling was less severe in those with paroxetine treatment compared with those with other types of anti‐depressive agents. Conclusions Paroxetine promotes ADRB1 sensitivity and attenuates cardiac hypertrophy partially via blocking GRK2‐mediated ADRB1 activation and internalization in the context of hypertension.
Collapse
Affiliation(s)
- Xuejing Sun
- Department of Cardiology The Third Xiangya HospitalCentral South University Changsha China
| | - Mengli Zhou
- Department of Cardiology The Third Xiangya HospitalCentral South University Changsha China
| | - Gaiyan Wen
- Department of Pharmacy Zhejiang Hospital Hangzhou China
| | - Yun Huang
- Ningbo Medical Center Lihuili Hospital Ningbo China
| | - Junru Wu
- Department of Cardiology The Third Xiangya HospitalCentral South University Changsha China
| | - Liping Peng
- Department of Cardiology The Third Xiangya HospitalCentral South University Changsha China
| | - Weihong Jiang
- Department of Cardiology The Third Xiangya HospitalCentral South University Changsha China
| | - Hong Yuan
- The Center of Clinical Pharmacology The Third Xiangya HospitalCentral South University Changsha China
| | - Yao Lu
- The Center of Clinical Pharmacology The Third Xiangya HospitalCentral South University Changsha China
| | - Jingjing Cai
- Department of Cardiology The Third Xiangya HospitalCentral South University Changsha China.,The Center of Clinical Pharmacology The Third Xiangya HospitalCentral South University Changsha China
| |
Collapse
|
7
|
Markan KR, Boland LK, King-McAlpin AQ, Claflin KE, Leaman MP, Kemerling MK, Stonewall MM, Amendt BA, Ankrum JA, Potthoff MJ. Adipose TBX1 regulates β-adrenergic sensitivity in subcutaneous adipose tissue and thermogenic capacity in vivo. Mol Metab 2020; 36:100965. [PMID: 32240964 PMCID: PMC7115112 DOI: 10.1016/j.molmet.2020.02.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 02/07/2020] [Accepted: 02/11/2020] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVE T-box 1 (TBX1) has been identified as a genetic marker of beige adipose tissue. TBX1 is a mesodermal development transcription factor essential for tissue patterning and cell fate determination. However, whether it plays a role in the process of adipose beiging or how it functions in adipose tissue has not been reported. Here, we examined the function of TBX1 in adipose tissue as well as adipose-derived stem cells from mice and humans. METHODS Adipose-specific TBX1 transgenic (TBX1 AdipoTG) and adipose-specific TBX1 knockout (TBX1 AdipoKO) mice were generated to explore the function of TBX1 in the process of adipose beiging, metabolism and energy homeostasis in vivo. In vitro, we utilized a siRNA mediated approach to determine the function of TBX1 during adipogenesis in mouse and human stem cells. RESULTS Adipose-specific overexpression of TBX1 was not sufficient to fully induce beiging and prevent diet-induced obesity. However, adipose TBX1 expression was necessary to defend body temperature during cold through regulation of UCP1 and for maintaining β3-adrenergic sensitivity and glucose homeostasis in vivo. Loss of adipose TBX1 expression enhanced basal lipolysis and reduced the size of subcutaneous iWAT adipocytes. Reduction of TBX1 expression via siRNA significantly impaired adipogenesis of mouse stromal vascular cells but significantly enhanced adipogenesis in human adipose derived stem cells. CONCLUSIONS Adipose expression of TBX1 is necessary, but not sufficient, to defend body temperature during cold via proper UCP1 expression. Adipose TBX1 expression was also required for proper insulin signaling in subcutaneous adipose as well as for maintaining β-adrenergic sensitivity, but overexpression of TBX1 was not sufficient to induce adipocyte beiging or to prevent diet-induced obesity. TBX1 expression is enriched in adipose stem cells in which it has contrasting effects on adipogenesis in mouse versus human cells. Collectively, these data demonstrate the importance of adipose TBX1 in the regulation of beige adipocyte function, energy homeostasis, and adipocyte development.
Collapse
Affiliation(s)
- Kathleen R Markan
- Department of Neuroscience and Pharmacology, Iowa City, IA, 52242, USA; Fraternal Order of Eagles Diabetes Research Center, Iowa City, IA, 52242, USA.
| | - Lauren K Boland
- Fraternal Order of Eagles Diabetes Research Center, Iowa City, IA, 52242, USA; Roy J. Carver Department of Biomedical Engineering, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
| | - Abdul Qaadir King-McAlpin
- Department of Neuroscience and Pharmacology, Iowa City, IA, 52242, USA; Fraternal Order of Eagles Diabetes Research Center, Iowa City, IA, 52242, USA
| | - Kristin E Claflin
- Department of Neuroscience and Pharmacology, Iowa City, IA, 52242, USA; Fraternal Order of Eagles Diabetes Research Center, Iowa City, IA, 52242, USA; Iowa Neurosciences Institute, Iowa City, IA, 52242, USA
| | - Michael P Leaman
- Department of Neuroscience and Pharmacology, Iowa City, IA, 52242, USA; Fraternal Order of Eagles Diabetes Research Center, Iowa City, IA, 52242, USA
| | - Morgan K Kemerling
- Department of Neuroscience and Pharmacology, Iowa City, IA, 52242, USA; Fraternal Order of Eagles Diabetes Research Center, Iowa City, IA, 52242, USA
| | - Madison M Stonewall
- Department of Neuroscience and Pharmacology, Iowa City, IA, 52242, USA; Fraternal Order of Eagles Diabetes Research Center, Iowa City, IA, 52242, USA
| | - Brad A Amendt
- Department of Anatomy and Cell Biology and the Craniofacial Anomalies Research Center, Iowa City, IA, 52242, USA
| | - James A Ankrum
- Fraternal Order of Eagles Diabetes Research Center, Iowa City, IA, 52242, USA; Roy J. Carver Department of Biomedical Engineering, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
| | - Matthew J Potthoff
- Department of Neuroscience and Pharmacology, Iowa City, IA, 52242, USA; Fraternal Order of Eagles Diabetes Research Center, Iowa City, IA, 52242, USA; Iowa Neurosciences Institute, Iowa City, IA, 52242, USA; Department of Veterans Affairs Medical Center, Iowa City, IA, 52242, USA
| |
Collapse
|
8
|
DSCAM-AS1 mediates pro-hypertrophy role of GRK2 in cardiac hypertrophy aggravation via absorbing miR-188-5p. In Vitro Cell Dev Biol Anim 2020; 56:286-295. [PMID: 32377998 DOI: 10.1007/s11626-020-00441-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 03/11/2020] [Indexed: 12/25/2022]
Abstract
Sustained cardiac hypertrophy, as previously clarified, serves as a critical initiator of heart failure and therefore is acknowledged as an important factor for heart failure treatment. The broadly demonstrated function and participation of long non-coding RNAs (lncRNAs) in tumors are well accepted. However, the underlying mechanism implicating lncRNAs in cardiac hypertrophy is mostly unexplored and deserves to be specifically studied. The devised work was aimed to disclose the function of lncRNA DS cell adhesion molecule antisense RNA 1 (DSCAM-AS1) in angiotensin II (AngII)-induced cardiac hypertrophy. In this study, we discovered the upregulation of DSCAM-AS1 in cardiomyocytes treated with AngII by quantitative real-time polymerase chain reaction (qRT-PCR). Western blot and qRT-PCR suggested that DSCAM-AS1 silencing attenuated the highly expressed hypertrophic biomarkers including β-myosin heavy chain (β-MHC), brain natriuretic peptide (BNP), and atrial natriuretic peptide (ANP) at mRNA and protein levels. The expanded cell surface in the presence of AngII treatment area was also shrunk by DSCAM-AS1 silencing. Mechanical analysis manifested that DSCAM-AS1 sponged microRNA-188-5p to boost the pro-hypertrophy gene G protein-coupled receptor kinase 2 (GRK2) expression. Rescue experiments unveiled miR-188-5p and GRK2 managed to reverse the anti-hypertrophy impact of DSCAM-AS1 silencing. In summary, DSCAM-AS1 was identified as a positive modulator in cardiac hypertrophy through miR-188-5p decoying and GRK2 augmentation, giving rise to an enriched theoretical basis for finding a promising target in cardiac hypertrophy regulation.
Collapse
|
9
|
Xu R, Gopireddy RR, Wu Y, Wu L, Tao X, Shao J, Wang W, Li L, Jovanovic A, Xu B, Kenyon NJ, Lu Q, Xiang YK, Fu Q. Hyperinsulinemia promotes heterologous desensitization of β 2 adrenergic receptor in airway smooth muscle in obesity. FASEB J 2020; 34:3996-4008. [PMID: 31960515 DOI: 10.1096/fj.201800688rr] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 12/08/2019] [Accepted: 12/30/2019] [Indexed: 01/05/2023]
Abstract
β-Adrenergic receptor (β-AR) agonists are the most common clinical bronchodilators for asthma. Obesity influences asthma severity and may impair response to β-AR agonists. Previous studies show that in obese mice, hyperinsulinemia plays a crucial role in β-AR desensitization in the heart. We therefore investigated whether insulin promotes β-AR desensitization in airway smooth muscle (ASM) and compromises airway relaxation responsiveness to β-AR agonists. We found that human ASM cells and mouse airway tissues exposed to insulin exhibit impaired β2 AR-induced cAMP accumulation and airway relaxation. This impaired relaxation is associated with insulin-induced phosphorylation and expression of phosphodiesterase 4D (PDE4D) through transactivation of a G protein-coupled receptor kinase 2 (GRK2)-dependent β2 AR-Gi -ERK1/2 cascade. Both acute and chronic pharmacological inhibition of PDE4 effectively reversed impaired β2 AR-mediated ASM relaxation in an obesity mouse model induced by a high fat diet. Collectively, these findings reveal that cross talk between insulin and β2 AR signaling promotes ASM β2 AR desensitization in obesity through upregulation of PDE4D phosphorylation and expression. Our results identify a novel pathway of asthma pathogenesis in patients with obesity/metabolic syndrome, in which the GRK2-mediated signaling can be a potential therapeutic modality to prevent or treat β2 AR desensitization in ASM. Moreover, PDE4 inhibitors may be used as efficacious therapeutic agents for asthma in obese and diabetic subjects.
Collapse
Affiliation(s)
- Rui Xu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan, China
| | | | - Yudi Wu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lei Wu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiang Tao
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ji Shao
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenxin Wang
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li Li
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan, China
| | | | - Bing Xu
- Department of Pharmacology, University of California at Davis, Davis, CA, USA.,VA northern California Healthcare System, Mather, CA, USA
| | - Nicolas J Kenyon
- Department of Medicine, University of California at Davis, Davis, CA, USA
| | - Quan Lu
- Department of Environmental Health, School of Public Health, Harvard University, Boston, MA, USA
| | - Yang K Xiang
- Department of Pharmacology, University of California at Davis, Davis, CA, USA.,VA northern California Healthcare System, Mather, CA, USA
| | - Qin Fu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan, China
| |
Collapse
|
10
|
West TM, Wang Q, Deng B, Zhang Y, Barbagallo F, Reddy GR, Chen D, Phan KS, Xu B, Isidori A, Xiang YK. Phosphodiesterase 5 Associates With β2 Adrenergic Receptor to Modulate Cardiac Function in Type 2 Diabetic Hearts. J Am Heart Assoc 2019; 8:e012273. [PMID: 31311394 PMCID: PMC6761630 DOI: 10.1161/jaha.119.012273] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Background In murine heart failure models and in humans with diabetic‐related heart hypertrophy, inhibition of phosphodiesterase 5 (PDE5) by sildenafil improves cardiac outcomes. However, the mechanism by which sildenafil improves cardiac function is unclear. We have observed a relationship between PDE5 and β2 adrenergic receptor (β2AR), which is characterized here as a novel mechanistic axis by which sildenafil improves symptoms of diabetic cardiomyopathy. Methods and Results Wild‐type and β2AR knockout mice fed a high fat diet (HFD) were treated with sildenafil, and echocardiogram analysis was performed. Cardiomyocytes were isolated for excitation‐contraction (E‐C) coupling, fluorescence resonant energy transfer, and proximity ligation assays; while heart tissues were implemented for biochemical and histological analyses. PDE5 selectively associates with β2AR, but not β1 adrenergic receptor, and inhibition of PDE5 with sildenafil restores the impaired response to adrenergic stimulation in HFD mice and isolated ventriculomyocytes. Sildenafil enhances β adrenergic receptor (βAR)‐stimulated cGMP and cAMP signals in HFD myocytes. Consequently, inhibition of PDE5 leads to protein kinase G–, and to a lesser extent, calcium/calmodulin‐dependent kinase II–dependent improvements in adrenergically stimulated E‐C coupling. Deletion of β2AR abolishes sildenafil's effect. Although the PDE5‐β2AR association is not altered in HFD, phosphodiesterase 3 displays an increased association with the β2AR‐PDE5 complex in HFD myocytes. Conclusions This study elucidates mechanisms by which the β2AR‐PDE5 axis can be targeted for treating diabetic cardiomyopathy. Inhibition of PDE5 enhances β2AR stimulation of cGMP and cAMP signals, as well as protein kinase G–dependent E‐C coupling in HFD myocytes.
Collapse
Affiliation(s)
- Toni M West
- Department of Pharmacology University of California Davis School of Medicine Davis CA
| | - Qingtong Wang
- Department of Pharmacology University of California Davis School of Medicine Davis CA
| | - Bingqing Deng
- Department of Pharmacology University of California Davis School of Medicine Davis CA.,Sun-Yet Sen Memorial hospital Sun-Yet Sen University Guangzhou China
| | - Yu Zhang
- Department of Pharmacology University of California Davis School of Medicine Davis CA.,College of Pharmacy Guangzhou Medical University Guangzhou China
| | - Federica Barbagallo
- Department of Pharmacology University of California Davis School of Medicine Davis CA.,Department of Experimental Medicine Sapienza University of Rome Rome Italy
| | - Gopireddy R Reddy
- Department of Pharmacology University of California Davis School of Medicine Davis CA
| | - Dana Chen
- Department of Pharmacology University of California Davis School of Medicine Davis CA
| | - Kyle S Phan
- Department of Pharmacology University of California Davis School of Medicine Davis CA
| | - Bing Xu
- Department of Pharmacology University of California Davis School of Medicine Davis CA.,College of Pharmacy Guangzhou Medical University Guangzhou China
| | - Andres Isidori
- Department of Experimental Medicine Sapienza University of Rome Rome Italy
| | - Yang K Xiang
- Department of Pharmacology University of California Davis School of Medicine Davis CA.,VA Northern California Health Care System Mather CA
| |
Collapse
|
11
|
Bencivenga L, Liccardo D, Napolitano C, Visaggi L, Rengo G, Leosco D. β-Adrenergic Receptor Signaling and Heart Failure: From Bench to Bedside. Heart Fail Clin 2019; 15:409-419. [PMID: 31079699 DOI: 10.1016/j.hfc.2019.02.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Despite improvements in management and therapeutic approach in the last decades, heart failure is still associated with high mortality rates. The sustained enhancement in the sympathetic nervous system tone, observed in patients with heart failure, causes alteration in β-adrenergic receptor signaling and function. This latter phenomenon is the result of several heart failure-related molecular abnormalities involving adrenergic receptors, G-protein-coupled receptor kinases, and β-arrestins. This article summarizes novel encouraging preclinical strategies to reactivate β-adrenergic receptor signaling in heart failure, including pharmacologic and gene therapy approaches, and attempts to translate acquired notions into the clinical setting.
Collapse
Affiliation(s)
- Leonardo Bencivenga
- Department of Translational Medical Sciences, Division of Geriatrics, Federico II University, Via Sergio Pansini, 5, Naples 80131, Italy
| | - Daniela Liccardo
- Department of Translational Medical Sciences, Division of Geriatrics, Federico II University, Via Sergio Pansini, 5, Naples 80131, Italy
| | - Carmen Napolitano
- Department of Translational Medical Sciences, Division of Geriatrics, Federico II University, Via Sergio Pansini, 5, Naples 80131, Italy
| | - Lucia Visaggi
- Department of Translational Medical Sciences, Division of Geriatrics, Federico II University, Via Sergio Pansini, 5, Naples 80131, Italy
| | - Giuseppe Rengo
- Department of Translational Medical Sciences, Division of Geriatrics, Federico II University, Via Sergio Pansini, 5, Naples 80131, Italy; Istituti Clinici Scientifici Maugeri SpA Società Benefit (ICS Maugeri SpA SB), Telese Terme, Italy
| | - Dario Leosco
- Department of Translational Medical Sciences, Division of Geriatrics, Federico II University, Via Sergio Pansini, 5, Naples 80131, Italy.
| |
Collapse
|
12
|
Cook RF, Bussey CT, Fomison‐Nurse IC, Hughes G, Bahn A, Cragg PA, Lamberts RR. β
2
‐Adrenoceptors indirectly support impaired β
1
‐adrenoceptor responsiveness in the isolated type 2 diabetic rat heart. Exp Physiol 2019; 104:808-818. [DOI: 10.1113/ep087437] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 03/18/2019] [Indexed: 12/24/2022]
Affiliation(s)
- Rosalind F. Cook
- Department of Physiology – HeartOtagoSchool of Biomedical SciencesUniversity of Otago Dunedin New Zealand
| | - Carol T. Bussey
- Department of Physiology – HeartOtagoSchool of Biomedical SciencesUniversity of Otago Dunedin New Zealand
| | - Ingrid C. Fomison‐Nurse
- Department of Physiology – HeartOtagoSchool of Biomedical SciencesUniversity of Otago Dunedin New Zealand
| | - Gillian Hughes
- Department of Physiology – HeartOtagoSchool of Biomedical SciencesUniversity of Otago Dunedin New Zealand
| | - Andrew Bahn
- Department of Physiology – HeartOtagoSchool of Biomedical SciencesUniversity of Otago Dunedin New Zealand
| | - Patricia A. Cragg
- Department of Physiology – HeartOtagoSchool of Biomedical SciencesUniversity of Otago Dunedin New Zealand
| | - Regis R. Lamberts
- Department of Physiology – HeartOtagoSchool of Biomedical SciencesUniversity of Otago Dunedin New Zealand
| |
Collapse
|
13
|
Susec M, Sencanski M, Glisic S, Veljkovic N, Pedersen C, Drinovec L, Stojan J, Nøhr J, Vrecl M. Functional characterization of β 2-adrenergic and insulin receptor heteromers. Neuropharmacology 2019; 152:78-89. [PMID: 30707913 DOI: 10.1016/j.neuropharm.2019.01.025] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 01/01/2019] [Accepted: 01/23/2019] [Indexed: 01/06/2023]
Abstract
This study aimed to functionally characterize β2-adrenergic (β2AR) and insulin receptor (IR) heteromers in regard to β-arrestin 2 (βarr2) recruitment and cAMP signaling and to examine the involvement of the cytoplasmic portion of the IR β chain in heteromerization with β2AR. Evidence for β2AR:IR:βarr2 complex formation and the specificity of the IR:βarr2 interaction was first provided by bioinfomatics analysis. Receptor-heteromer investigation technology (HIT) then provided functional evidence of β2AR:IR heterodimerization by showing isoproterenol-induced but not insulin-induced GFP2-βarr2 recruitment to the β2AR:IR complex; the IR:βarr2 interaction was found to only be constitutive. The constitutive IR:βarr2 BRET signal (BRETconst) was significantly smaller in cells coexpressing IR-RLuc8 and a GFP2-βarr2 1-185 mutant lacking the proposed IR binding domain. β2AR:IR heteromerization also influenced the pharmacological phenotype of β2AR, i.e., its efficacy in recruiting βarr2 and activating cAMP signaling. Evidence suggesting involvement of the cytoplasmic portion of the IR β chain in the interaction with β2AR was provided by BRET2 saturation and HIT assays using an IR 1-1271 stop mutant lacking the IR C-terminal tail region. For the complex consisting of IR 1-1271-RLuc8:β2AR-GFP2, saturation was not reached, most likely reflecting random collisions between IR 1-1271 and β2AR. Furthermore, in the HIT assay, no substantial agonist-induced increase in the BRET2 signal was detected that would be indicative of βarr2 recruitment to the IR 1-1271:β2AR heteromer. Complementary 3D visualization of β2AR:IR provided supporting evidence for stability of the heterotetramer complex and identified amino acid residues involved in β2AR:IR heteromerization. This article is part of the Special Issue entitled 'Receptor heteromers and their allosteric receptor-receptor interactions'.
Collapse
Affiliation(s)
- Maja Susec
- Institute of Preclinical Sciences, Veterinary Faculty, University of Ljubljana, Slovenia
| | - Milan Sencanski
- Center for Multidisciplinary Research, Institute of Nuclear Sciences VINCA, University of Belgrade, Belgrade, Serbia
| | - Sanja Glisic
- Center for Multidisciplinary Research, Institute of Nuclear Sciences VINCA, University of Belgrade, Belgrade, Serbia
| | - Nevena Veljkovic
- Center for Multidisciplinary Research, Institute of Nuclear Sciences VINCA, University of Belgrade, Belgrade, Serbia
| | - Christina Pedersen
- Department of Incretin & Islet Biology, Novo Nordisk A/S, Måløv, Denmark
| | - Luka Drinovec
- Department of Condensed Matter Physics, Jožef Stefan Institute, Slovenia
| | - Jurij Stojan
- Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Jane Nøhr
- Department of Incretin & Islet Biology, Novo Nordisk A/S, Måløv, Denmark
| | - Milka Vrecl
- Institute of Preclinical Sciences, Veterinary Faculty, University of Ljubljana, Slovenia.
| |
Collapse
|
14
|
Riehle C, Bauersachs J. Of mice and men: models and mechanisms of diabetic cardiomyopathy. Basic Res Cardiol 2018; 114:2. [PMID: 30443826 PMCID: PMC6244639 DOI: 10.1007/s00395-018-0711-0] [Citation(s) in RCA: 133] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 11/09/2018] [Indexed: 02/07/2023]
Abstract
Diabetes mellitus increases the risk of heart failure independent of co-existing hypertension and coronary artery disease. Although several molecular mechanisms for the development of diabetic cardiomyopathy have been identified, they are incompletely understood. The pathomechanisms are multifactorial and as a consequence, no causative treatment exists at this time to modulate or reverse the molecular changes contributing to accelerated cardiac dysfunction in diabetic patients. Numerous animal models have been generated, which serve as powerful tools to study the impact of type 1 and type 2 diabetes on the heart. Despite specific limitations of the models generated, they mimic various perturbations observed in the diabetic myocardium and continue to provide important mechanistic insight into the pathogenesis underlying diabetic cardiomyopathy. This article reviews recent studies in both diabetic patients and in these animal models, and discusses novel hypotheses to delineate the increased incidence of heart failure in diabetic patients.
Collapse
Affiliation(s)
- Christian Riehle
- Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Str. 1, Hannover, 30625, Germany.
| | - Johann Bauersachs
- Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Str. 1, Hannover, 30625, Germany
| |
Collapse
|
15
|
Abstract
Heart failure (HF) has become increasingly common within the elderly population, decreasing their survival and overall quality of life. In fact, despite the improvements in treatment, many elderly people suffer from cardiac dysfunction (HF, valvular diseases, arrhythmias or hypertension-induced cardiac hypertrophy) that are much more common in an older fragile heart. Since β-adrenergic receptor (β-AR) signaling is abnormal in failing as well as aged hearts, this pathway is an effective diagnostic and therapeutic target. Both HF and aging are characterized by activation/hyperactivity of various neurohormonal pathways, the most important of which is the sympathetic nervous system (SNS). SNS hyperactivity is initially a compensatory mechanism to stimulate contractility and maintain cardiac output. Unfortunately, this chronic stimulation becomes detrimental and causes decreased cardiac function as well as reduced inotropic reserve due to a decrease in cardiac β-ARs responsiveness. Therapies which (e.g., β-blockers and physical activity) restore β-ARs responsiveness can ameliorate cardiac performance and outcomes during HF, particularly in older patients. In this review, we will discuss physiological β-adrenergic signaling and its alterations in both HF and aging as well as the potential clinical application of targeting β-adrenergic signaling in these disease processes.
Collapse
|
16
|
de Lucia C, Eguchi A, Koch WJ. New Insights in Cardiac β-Adrenergic Signaling During Heart Failure and Aging. Front Pharmacol 2018; 9:904. [PMID: 30147654 PMCID: PMC6095970 DOI: 10.3389/fphar.2018.00904] [Citation(s) in RCA: 184] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Accepted: 07/24/2018] [Indexed: 12/13/2022] Open
Abstract
Heart failure (HF) has become increasingly common within the elderly population, decreasing their survival and overall quality of life. In fact, despite the improvements in treatment, many elderly people suffer from cardiac dysfunction (HF, valvular diseases, arrhythmias or hypertension-induced cardiac hypertrophy) that are much more common in an older fragile heart. Since β-adrenergic receptor (β-AR) signaling is abnormal in failing as well as aged hearts, this pathway is an effective diagnostic and therapeutic target. Both HF and aging are characterized by activation/hyperactivity of various neurohormonal pathways, the most important of which is the sympathetic nervous system (SNS). SNS hyperactivity is initially a compensatory mechanism to stimulate contractility and maintain cardiac output. Unfortunately, this chronic stimulation becomes detrimental and causes decreased cardiac function as well as reduced inotropic reserve due to a decrease in cardiac β-ARs responsiveness. Therapies which (e.g., β-blockers and physical activity) restore β-ARs responsiveness can ameliorate cardiac performance and outcomes during HF, particularly in older patients. In this review, we will discuss physiological β-adrenergic signaling and its alterations in both HF and aging as well as the potential clinical application of targeting β-adrenergic signaling in these disease processes.
Collapse
Affiliation(s)
| | | | - Walter J. Koch
- Department of Pharmacology – Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| |
Collapse
|
17
|
Abstract
Diabetes is a major risk factor for the development of heart failure. One of the hallmarks of diabetes is insulin resistance associated with hyperinsulinemia. The literature shows that insulin and adrenergic signaling is intimately linked to each other; however, whether and how insulin may modulate cardiac adrenergic signaling and cardiac function remains unknown. Notably, recent studies have revealed that insulin receptor and β2 adrenergic receptor (β2AR) forms a membrane complex in animal hearts, bringing together the direct contact between 2 receptor signaling systems, and forming an integrated and dynamic network. Moreover, insulin can drive cardiac adrenergic desensitization via protein kinase A and G protein-receptor kinases phosphorylation of the β2AR, which compromises adrenergic regulation of cardiac contractile function. In this review, we will explore the current state of knowledge linking insulin and G protein-coupled receptor signaling, especially β-adrenergic receptor signaling in the heart, with emphasis on molecular insights regarding its role in diabetic cardiomyopathy.
Collapse
|
18
|
Abstract
G protein-coupled receptors (GPCRs) transduce pleiotropic intracellular signals in a broad range of physiological responses and disease states. Activated GPCRs can undergo agonist-induced phosphorylation by G protein receptor kinases (GRKs) and second messenger-dependent protein kinases such as protein kinase A (PKA). Here, we characterize spatially segregated subpopulations of β2-adrenergic receptor (β2AR) undergoing selective phosphorylation by GRKs or PKA in a single cell. GRKs primarily label monomeric β2ARs that undergo endocytosis, whereas PKA modifies dimeric β2ARs that remain at the cell surface. In hippocampal neurons, PKA-phosphorylated β2ARs are enriched in dendrites, whereas GRK-phosphorylated β2ARs accumulate in soma, being excluded from dendrites in a neuron maturation-dependent manner. Moreover, we show that PKA-phosphorylated β2ARs are necessary to augment the activity of L-type calcium channel. Collectively, these findings provide evidence that functionally distinct subpopulations of this prototypical GPCR exist in a single cell. β2-adrenergic receptor (β2AR) can be phosphorylated by G protein receptor kinases and second messenger-dependent kinases. Here, the authors demonstrate that these phosphorylation events are specific to functionally distinct and spatially segregated subpopulations of β2AR that co-exist in a single cell.
Collapse
|
19
|
Blurring Boundaries: Receptor Tyrosine Kinases as functional G Protein-Coupled Receptors. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2018; 339:1-40. [DOI: 10.1016/bs.ircmb.2018.02.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
20
|
Guillory AN, Clayton RP, Prasai A, El Ayadi A, Herndon DN, Finnerty CC. Biventricular differences in β-adrenergic receptor signaling following burn injury. PLoS One 2017; 12:e0189527. [PMID: 29232706 PMCID: PMC5726759 DOI: 10.1371/journal.pone.0189527] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 11/26/2017] [Indexed: 12/17/2022] Open
Abstract
Burn injury detrimentally affects the myocardium, primarily due to over-activation of β-adrenergic receptors (β-AR). Autopsy reports from our institution reveal that patients often suffer from right ventricle (RV) failure. Since burn injury affects β-AR signaling in the left ventricle (LV), we proposed that β-AR signaling may also be altered in the RV. A rodent model with a scald burn of 60% of the total body surface area was used to test this hypothesis. Ventricles were isolated 7 days post-burn. We examined the expression of β-ARs via Western blotting and the mRNA expression of downstream signaling proteins via qRT-PCR. Cyclic adenosine monophosphate (cAMP) production and protein kinase A (PKA) activity were measured in membrane and cytosolic fractions, respectively, using enzyme immunoassay kits. β1-AR protein expression was significantly increased in the RV following burn injury compared to non-burned RV but not in the LV (p = 0.0022). In contrast, β2-AR expression was unaltered among the groups while Gαi expression was significantly higher in the LV post-burn (p = 0.023). B-arrestin-1 and G-protein coupled receptor kinase-2 mRNA expression were significantly increased in the left ventricle post-burn (p = 0.001, p<0.0001, respectively). cAMP production and PKA activity were significantly lower in the LV post-burn (p = 0.0063, 0.0042, respectively). These data indicate that burn injury affects the β-AR signaling pathway in the RV independently of the LV. Additionally, non-canonical β-AR signaling may be activated in the RV as cAMP production and PKA activity were unchanged despite changes in β1-AR protein expression.
Collapse
Affiliation(s)
- Ashley N. Guillory
- Department of Surgery, University of Texas Medical Branch, Galveston, Texas, United States of America
- Shriners Hospitals for Children—Galveston, Galveston, Texas, United States of America
- Institute for Translational Sciences, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Robert P. Clayton
- Institute for Translational Sciences, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Anesh Prasai
- Department of Surgery, University of Texas Medical Branch, Galveston, Texas, United States of America
- Shriners Hospitals for Children—Galveston, Galveston, Texas, United States of America
| | - Amina El Ayadi
- Department of Surgery, University of Texas Medical Branch, Galveston, Texas, United States of America
- Shriners Hospitals for Children—Galveston, Galveston, Texas, United States of America
- Institute for Translational Sciences, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - David N. Herndon
- Department of Surgery, University of Texas Medical Branch, Galveston, Texas, United States of America
- Shriners Hospitals for Children—Galveston, Galveston, Texas, United States of America
- Institute for Translational Sciences, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Celeste C. Finnerty
- Department of Surgery, University of Texas Medical Branch, Galveston, Texas, United States of America
- Shriners Hospitals for Children—Galveston, Galveston, Texas, United States of America
- Institute for Translational Sciences, University of Texas Medical Branch, Galveston, Texas, United States of America
- * E-mail:
| |
Collapse
|
21
|
Molecular mechanisms of cardiac pathology in diabetes - Experimental insights. Biochim Biophys Acta Mol Basis Dis 2017; 1864:1949-1959. [PMID: 29109032 DOI: 10.1016/j.bbadis.2017.10.035] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 10/09/2017] [Accepted: 10/27/2017] [Indexed: 12/11/2022]
Abstract
Diabetic cardiomyopathy is a distinct pathology independent of co-morbidities such as coronary artery disease and hypertension. Diminished glucose uptake due to impaired insulin signaling and decreased expression of glucose transporters is associated with a shift towards increased reliance on fatty acid oxidation and reduced cardiac efficiency in diabetic hearts. The cardiac metabolic profile in diabetes is influenced by disturbances in circulating glucose, insulin and fatty acids, and alterations in cardiomyocyte signaling. In this review, we focus on recent preclinical advances in understanding the molecular mechanisms of diabetic cardiomyopathy. Genetic manipulation of cardiomyocyte insulin signaling intermediates has demonstrated that partial cardiac functional rescue can be achieved by upregulation of the insulin signaling pathway in diabetic hearts. Inconsistent findings have been reported relating to the role of cardiac AMPK and β-adrenergic signaling in diabetes, and systemic administration of agents targeting these pathways appear to elicit some cardiac benefit, but whether these effects are related to direct cardiac actions is uncertain. Overload of cardiomyocyte fuel storage is evident in the diabetic heart, with accumulation of glycogen and lipid droplets. Cardiac metabolic dysregulation in diabetes has been linked with oxidative stress and autophagy disturbance, which may lead to cell death induction, fibrotic 'backfill' and cardiac dysfunction. This review examines the weight of evidence relating to the molecular mechanisms of diabetic cardiomyopathy, with a particular focus on metabolic and signaling pathways. Areas of uncertainty in the field are highlighted and important knowledge gaps for further investigation are identified. This article is part of a Special issue entitled Cardiac adaptations to obesity, diabetes and insulin resistance, edited by Professors Jan F.C. Glatz, Jason R.B. Dyck and Christine Des Rosiers.
Collapse
|
22
|
Hu X, Xiao RP. MG53 and disordered metabolism in striated muscle. Biochim Biophys Acta Mol Basis Dis 2017; 1864:1984-1990. [PMID: 29017896 DOI: 10.1016/j.bbadis.2017.10.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 10/06/2017] [Accepted: 10/06/2017] [Indexed: 12/25/2022]
Abstract
MG53 is a member of tripartite motif family (TRIM) that expressed most abundantly in striated muscle. Using rodent models, many studies have demonstrated the MG53 not only facilitates membrane repair after ischemia reperfusion injury, but also contributes to the protective effects of both pre- and post-conditioning. Recently, however, it has been shown that MG53 participates in the regulation of many metabolic processes, especially insulin signaling pathway. Thus, sustained overexpression of MG53 may contribute to the development of various metabolic disorders in striated muscle. In this review, using cardiac muscle as an example, we will discuss muscle metabolic disturbances associated with diabetes and the current understanding of the underlying molecular mechanisms; in particular, the pathogenesis of diabetic cardiomyopathy. We will focus on the pathways that MG53 regulates and how the dysregulation of MG53 leads to metabolic disorders, thereby establishing a causal relationship between sustained upregulation of MG53 and the development of muscle insulin resistance and metabolic disorders. This article is part of a Special issue entitled Cardiac adaptations to obesity, diabetes and insulin resistance, edited by Professors Jan F.C. Glatz, Jason R.B. Dyck and Christine Des Rosiers.
Collapse
Affiliation(s)
- Xinli Hu
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, Peking University, Beijing 100871, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China; Beijing City Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing 100871, China
| | - Rui-Ping Xiao
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, Peking University, Beijing 100871, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China; Beijing City Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing 100871, China.
| |
Collapse
|
23
|
Fu Q, Wang Q, Xiang YK. Insulin and β Adrenergic Receptor Signaling: Crosstalk in Heart. Trends Endocrinol Metab 2017; 28:416-427. [PMID: 28256297 PMCID: PMC5535765 DOI: 10.1016/j.tem.2017.02.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Revised: 01/29/2017] [Accepted: 02/01/2017] [Indexed: 02/03/2023]
Abstract
Recent advances show that insulin may affect β adrenergic receptor (βAR) signaling in the heart to modulate cardiac function in clinically relevant states, such as diabetes mellitus (DM) and heart failure (HF). Conversely, activation of βAR regulates cardiac glucose uptake and promotes insulin resistance (IR) in HF. Here, we discuss the recent characterization of the interaction between the cardiac insulin receptor (InsR) and βAR in the myocardium, in which insulin stimulation crosstalks with cardiac βAR via InsR substrate (IRS)-dependent and G-protein receptor kinase 2 (GRK2)-mediated phosphorylation of β2AR. The insulin-induced phosphorylation promotes β2AR coupling to Gi and expression of phosphodiesterase 4D, which both inhibit cardiac adrenergic signaling and compromise cardiac contractile function. These recent developments could support new approaches for the effective prevention or treatment of obesity- or DM-related HF.
Collapse
Affiliation(s)
- Qin Fu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Hubei Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Huazhong University of Science and Technology, Wuhan, China.
| | - Qingtong Wang
- Institute of Clinical Pharmacology, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Anhui Medical University, Hefei, China.
| | - Yang K Xiang
- Department of Pharmacology, University of California, Davis, CA, USA; VA Northern California Health Care System, Mather, CA, USA.
| |
Collapse
|
24
|
Fu Q, Hu Y, Wang Q, Liu Y, Li N, Xu B, Kim S, Chiamvimonvat N, Xiang YK. High-fat diet induces protein kinase A and G-protein receptor kinase phosphorylation of β 2 -adrenergic receptor and impairs cardiac adrenergic reserve in animal hearts. J Physiol 2017; 595:1973-1986. [PMID: 27983752 PMCID: PMC5350441 DOI: 10.1113/jp273314] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 11/23/2016] [Indexed: 11/08/2022] Open
Abstract
Key points Patients with diabetes show a blunted cardiac inotropic response to β‐adrenergic stimulation despite normal cardiac contractile reserve. Acute insulin stimulation impairs β‐adrenergically induced contractile function in isolated cardiomyocytes and Langendorff‐perfused hearts. In this study, we aimed to examine the potential effects of hyperinsulinaemia associated with high‐fat diet (HFD) feeding on the cardiac β2‐adrenergic receptor signalling and the impacts on cardiac contractile function. We showed that 8 weeks of HFD feeding leads to reductions in cardiac functional reserve in response to β‐adrenergic stimulation without significant alteration of cardiac structure and function, which is associated with significant changes in β2‐adrenergic receptor phosphorylation at protein kinase A and G‐protein receptor kinase sites in the myocardium. The results suggest that clinical intervention might be applied to subjects in early diabetes without cardiac symptoms to prevent further cardiac complications.
Abstract Patients with diabetes display reduced exercise capability and impaired cardiac contractile reserve in response to adrenergic stimulation. We have recently uncovered an insulin receptor and adrenergic receptor signal network in the heart. The aim of this study was to understand the impacts of high‐fat diet (HFD) on the insulin–adrenergic receptor signal network in hearts. After 8 weeks of HFD feeding, mice exhibited diabetes, with elevated insulin and glucose concentrations associated with body weight gain. Mice fed an HFD had normal cardiac structure and function. However, the HFD‐fed mice displayed a significant elevation of phosphorylation of the β2‐adrenergic receptor (β2AR) at both the protein kinase A site serine 261/262 and the G‐protein‐coupled receptor kinase site serine 355/356 and impaired adrenergic reserve when compared with mice fed on normal chow. Isolated myocytes from HFD‐fed mice also displayed a reduced contractile response to adrenergic stimulation when compared with those of control mice fed normal chow. Genetic deletion of the β2AR led to a normalized adrenergic response and preserved cardiac contractile reserve in HFD‐fed mice. Together, these data indicate that HFD promotes phosphorylation of the β2AR, contributing to impairment of cardiac contractile reserve before cardiac structural and functional remodelling, suggesting that early intervention in the insulin–adrenergic signalling network might be effective in prevention of cardiac complications in diabetes. Patients with diabetes show a blunted cardiac inotropic response to β‐adrenergic stimulation despite normal cardiac contractile reserve. Acute insulin stimulation impairs β‐adrenergically induced contractile function in isolated cardiomyocytes and Langendorff‐perfused hearts. In this study, we aimed to examine the potential effects of hyperinsulinaemia associated with high‐fat diet (HFD) feeding on the cardiac β2‐adrenergic receptor signalling and the impacts on cardiac contractile function. We showed that 8 weeks of HFD feeding leads to reductions in cardiac functional reserve in response to β‐adrenergic stimulation without significant alteration of cardiac structure and function, which is associated with significant changes in β2‐adrenergic receptor phosphorylation at protein kinase A and G‐protein receptor kinase sites in the myocardium. The results suggest that clinical intervention might be applied to subjects in early diabetes without cardiac symptoms to prevent further cardiac complications.
Collapse
Affiliation(s)
- Qin Fu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,The Key Laboratory for Drug Target Research and Pharmacodynamic Evaluation of Hubei Province, Wuhan, China
| | - Yuting Hu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,The Key Laboratory for Drug Target Research and Pharmacodynamic Evaluation of Hubei Province, Wuhan, China
| | - Qingtong Wang
- Department of Pharmacology, University of California, Davis, CA, USA.,Institute of Clinical Pharmacology, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Centre of Anti-inflammatory and Immune Medicine, Anhui Medical University, Hefei, China
| | - Yongming Liu
- Department of Pharmacology, University of California, Davis, CA, USA.,Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ning Li
- Division of Cardiovascular Medicine, Department of Medicine, University of California, Davis, CA, USA
| | - Bing Xu
- Department of Pharmacology, University of California, Davis, CA, USA
| | - Sungjin Kim
- Department of Pharmacology, University of California, Davis, CA, USA
| | - Nipavan Chiamvimonvat
- Division of Cardiovascular Medicine, Department of Medicine, University of California, Davis, CA, USA.,VA Northern California Healthcare System, Mather, CA, USA
| | - Yang K Xiang
- Department of Pharmacology, University of California, Davis, CA, USA.,Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,VA Northern California Healthcare System, Mather, CA, USA
| |
Collapse
|
25
|
Song L, Wang F, Dong Z, Hua X, Xia Q. Label-free quantitative phosphoproteomic profiling of cellular response induced by an insect cytokine paralytic peptide. J Proteomics 2016; 154:49-58. [PMID: 27903465 DOI: 10.1016/j.jprot.2016.11.018] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2016] [Revised: 11/18/2016] [Accepted: 11/21/2016] [Indexed: 12/18/2022]
Abstract
Paralytic peptide (PP) participates in diverse physiological processes as an insect cytokine, such as immunity control, paralysis induction, regulation of cell morphology and proliferation. To investigate the molecular mechanism underlying those physiological activities, we systematically investigated the global phosphorylation events in fat body of silkworm larvae induced by PP through label-free quantitative phosphoproteomics. 2534 phosphosites were finally identified, of which the phosphorylation level of 620 phosphosites on 244 proteins was significantly up-regulated and 67 phosphosites on 43 proteins was down-regulated. Among those proteins, 13 were protein kinases (PKs), 13 were transcription factors (TFs) across 10 families and 17 were metabolism related enzymes. Meanwhile, Motif-X analysis of the phosphorylation sites showed that 16 motifs are significantly enriched, including 8 novel phosphorylation motifs. In addition, KEGG and functional interacting network analysis revealed that phosphorylation cascades play the crucial regulation roles in PP-dependent signaling pathways, and highlighted the potential central position of the mitogen-activated protein kinases (MAPKs) in them. These analyses provide direct insights into the molecule mechanisms of cellular response induced by PP. SIGNIFICANCE PP as an insect cytokine participated in diverse functions including immunity control paralysis induction, regulation of cell morphology and proliferation. In this study, we performed firstly a label-free quantitative phosphoproteomics analysis. We found some new phosphorylation targets of PP-stimulation. Meanwhile, Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis and functional networks revealed that phosphorylation cascades play the crucial regulation roles in PP-dependent signaling pathways. In addition, the potential central position of the mitogen-activated protein kinases (MAPKs) was highlighted in PP-dependent signaling pathways. We think our findings may help us gain a systematic understanding of the cytokine-dependent response regulation in insects.
Collapse
Affiliation(s)
- Liang Song
- State Key Laboratory of Silkworm Genome Biology, Southwest University, 216, Tiansheng Road, Beibei, Chongqing 400716, China.
| | - Fei Wang
- State Key Laboratory of Silkworm Genome Biology, Southwest University, 216, Tiansheng Road, Beibei, Chongqing 400716, China.
| | - Zhaoming Dong
- State Key Laboratory of Silkworm Genome Biology, Southwest University, 216, Tiansheng Road, Beibei, Chongqing 400716, China.
| | - Xiaoting Hua
- State Key Laboratory of Silkworm Genome Biology, Southwest University, 216, Tiansheng Road, Beibei, Chongqing 400716, China.
| | - Qingyou Xia
- State Key Laboratory of Silkworm Genome Biology, Southwest University, 216, Tiansheng Road, Beibei, Chongqing 400716, China.
| |
Collapse
|
26
|
Wang Q, Liu Y, Fu Q, Xu B, Zhang Y, Kim S, Tan R, Barbagallo F, West T, Anderson E, Wei W, Abel ED, Xiang YK. Inhibiting Insulin-Mediated β2-Adrenergic Receptor Activation Prevents Diabetes-Associated Cardiac Dysfunction. Circulation 2016; 135:73-88. [PMID: 27815373 DOI: 10.1161/circulationaha.116.022281] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 10/13/2016] [Indexed: 12/31/2022]
Abstract
BACKGROUND Type 2 diabetes mellitus (DM) and obesity independently increase the risk of heart failure by incompletely understood mechanisms. We propose that hyperinsulinemia might promote adverse consequences in the hearts of subjects with type-2 DM and obesity. METHODS High-fat diet feeding was used to induce obesity and DM in wild-type mice or mice lacking β2-adrenergic receptor (β2AR) or β-arrestin2. Wild-type mice fed with high-fat diet were treated with a β-blocker carvedilol or a GRK2 (G-protein-coupled receptor kinase 2) inhibitor. We examined signaling and cardiac contractile function. RESULTS High-fat diet feeding selectively increases the expression of phosphodiesterase 4D (PDE4D) in mouse hearts, in concert with reduced protein kinase A phosphorylation of phospholamban, which contributes to systolic and diastolic dysfunction. The expression of PDE4D is also elevated in human hearts with DM. The induction of PDE4D expression is mediated by an insulin receptor, insulin receptor substrate, and GRK2 and β-arrestin2-dependent transactivation of a β2AR-extracellular regulated protein kinase signaling cascade. Thus, pharmacological inhibition of β2AR or GRK2, or genetic deletion of β2AR or β-arrestin2, all significantly attenuate insulin-induced phosphorylation of extracellular regulated protein kinase and PDE4D induction to prevent DM-related contractile dysfunction. CONCLUSIONS These studies elucidate a novel mechanism by which hyperinsulinemia contributes to heart failure by increasing PDE4D expression and identify β2AR or GRK2 as plausible therapeutic targets for preventing or treating heart failure in subjects with type 2 DM.
Collapse
MESH Headings
- Animals
- Carbazoles/pharmacology
- Carvedilol
- Cells, Cultured
- Cyclic Nucleotide Phosphodiesterases, Type 4/genetics
- Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism
- Diabetes Mellitus, Type 2/complications
- Diet, High-Fat
- Extracellular Signal-Regulated MAP Kinases/metabolism
- G-Protein-Coupled Receptor Kinase 2/antagonists & inhibitors
- G-Protein-Coupled Receptor Kinase 2/metabolism
- Heart Failure/etiology
- Humans
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Myocardial Contraction/drug effects
- Myocardium/metabolism
- Myocytes, Cardiac/cytology
- Myocytes, Cardiac/metabolism
- Obesity/complications
- Propanolamines/pharmacology
- Receptors, Adrenergic, beta-2/deficiency
- Receptors, Adrenergic, beta-2/genetics
- Signal Transduction
- Vasodilator Agents/pharmacology
- beta-Arrestin 2/deficiency
- beta-Arrestin 2/genetics
Collapse
Affiliation(s)
- Qingtong Wang
- Department of Pharmacology, University of California at Davis, CA 95616, USA
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, 230032, China
| | - Yongming Liu
- Department of Pharmacology, University of California at Davis, CA 95616, USA
- Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200000, China
| | - Qin Fu
- Department of Pharmacology, University of California at Davis, CA 95616, USA
- Department of Pharmacology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Bing Xu
- Department of Pharmacology, University of California at Davis, CA 95616, USA
| | - Yuan Zhang
- Department of Medicine, Division of Endocrinology and Metabolism and Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA 52242, USA
| | - Sungjin Kim
- Department of Pharmacology, University of California at Davis, CA 95616, USA
| | - Ruensern Tan
- Department of Pharmacology, University of California at Davis, CA 95616, USA
| | - Federica Barbagallo
- Department of Pharmacology, University of California at Davis, CA 95616, USA
| | - Toni West
- Department of Pharmacology, University of California at Davis, CA 95616, USA
| | - Ethan Anderson
- Department of Pharmacology and Toxicology, East Carolina University, Greenville, NC 27834, USA
| | - Wei Wei
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, 230032, China
| | - E Dale Abel
- Department of Medicine, Division of Endocrinology and Metabolism and Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA 52242, USA
| | - Yang K Xiang
- Department of Pharmacology, University of California at Davis, CA 95616, USA
- VA Northern California Health Care System, Mather, CA 95655, USA
| |
Collapse
|
27
|
Cornacchione M, Pellegrini M, Fassina L, Mognaschi ME, Di Siena S, Gimmelli R, Ambrosino P, Soldovieri MV, Taglialatela M, Gianfrilli D, Isidori AM, Lenzi A, Naro F. β-Adrenergic response is counteracted by extremely-low-frequency pulsed electromagnetic fields in beating cardiomyocytes. J Mol Cell Cardiol 2016; 98:146-58. [DOI: 10.1016/j.yjmcc.2016.07.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 07/08/2016] [Indexed: 12/18/2022]
|
28
|
Cardiovascular Dysfunction Following Burn Injury: What We Have Learned from Rat and Mouse Models. Int J Mol Sci 2016; 17:ijms17010053. [PMID: 26729111 PMCID: PMC4730298 DOI: 10.3390/ijms17010053] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Revised: 12/14/2015] [Accepted: 12/23/2015] [Indexed: 12/11/2022] Open
Abstract
Severe burn profoundly affects organs both proximal and distal to the actual burn site. Cardiovascular dysfunction is a well-documented phenomenon that increases morbidity and mortality following a massive thermal trauma. Beginning immediately post-burn, during the ebb phase, cardiac function is severely depressed. By 48 h post-injury, cardiac function rebounds and the post-burn myocardium becomes tachycardic and hyperinflammatory. While current clinical trials are investigating a variety of drugs targeted at reducing aspects of the post-burn hypermetabolic response such as heart rate and cardiac work, there is still a paucity of knowledge regarding the underlying mechanisms that induce cardiac dysfunction in the severely burned. There are many animal models of burn injury, from rodents, to sheep or swine, but the majority of burn related cardiovascular investigations have occurred in rat and mouse models. This literature review consolidates the data supporting the prevalent role that β-adrenergic receptors play in mediating post-burn cardiac dysfunction and the idea that pharmacological modulation of this receptor family is a viable therapeutic target for resolving burn-induced cardiac deficits.
Collapse
|
29
|
“Barcode” and Differential Effects of GPCR Phosphorylation by Different GRKs. METHODS IN PHARMACOLOGY AND TOXICOLOGY 2016. [DOI: 10.1007/978-1-4939-3798-1_5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
30
|
Trafficking of β-Adrenergic Receptors: Implications in Intracellular Receptor Signaling. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2015; 132:151-88. [PMID: 26055058 DOI: 10.1016/bs.pmbts.2015.03.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
β-Adrenergic receptors (βARs), prototypical G-protein-coupled receptors, play a pivotal role in regulating neuronal and cardiovascular responses to catecholamines during stress. Agonist-induced receptor endocytosis is traditionally considered as a primary mechanism to turn off the receptor signaling (or receptor desensitization). However, recent progress suggests that intracellular trafficking of βAR presents a mean to translocate receptor signaling machinery to intracellular organelles/compartments while terminating the signaling at the cell surface. Moreover, the apparent multidimensionality of ligand efficacy in space and time in a cell has forecasted exciting pathophysiological implications, which are just beginning to be explored. As we begin to understand how these pathways impact downstream cellular programs, this will have significant implications for a number of pathophysiological conditions in heart and other systems, that in turn open up new therapeutic opportunities.
Collapse
|