1
|
Zhu Z, Guan Q, Xu B, Bahriz S, Shen A, West TM, Zhang Y, Deng B, Wei W, Han Y, Wang Q, Xiang YK. Inhibition of the upregulated phosphodiesterase 4D isoforms improves SERCA2a function in diabetic cardiomyopathy. Br J Pharmacol 2025; 182:1487-1507. [PMID: 39662482 DOI: 10.1111/bph.17411] [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: 12/27/2023] [Revised: 09/04/2024] [Accepted: 10/15/2024] [Indexed: 12/13/2024] Open
Abstract
BACKGROUND AND PURPOSE Sarcoplasmic reticulum Ca2+-ATPase (SERCA2a) is impaired in heart failure. Phosphodiesterases (PDEs) are implicated in the modulation of local cAMP signals and protein kinase A (PKA) activity essential for cardiac function. We characterise PDE isoforms that underlie decreased activities of SERCA2a and reduced cardiac contractile function in diabetic cardiomyopathy. EXPERIMENTAL APPROACH Wild type mice were fed with either normal chow or a high-fat diet (HFD). Cardiomyocytes were isolated for excitation-contraction coupling (ECC), fluorescence resonant energy transfer PKA biosensor and proximity ligation assays. KEY RESULTS The upregulated PDE4D3 and PDE4D9 isoforms in HFD cardiomyocytes specifically bound to SERCA2a but not ryanodine receptor 2 (RyR2) on the sarcoplasmic reticulum (SR). The increased association of PDE4D isoforms with SERCA2a in HFD cardiomyocytes led to reduced local PKA activities and phosphorylation of phospholamban (PLB) but minimally effected the PKA activities and phosphorylation of RyR2. These changes correlate with slower calcium decay tau in the SR and attenuation of ECC in HFD cardiomyocytes. Selective inhibition of PDE4D3 or PDE4D9 restored PKA activities and phosphorylation of PLB at the SERCA2a complex, recovered calcium decay tau, and increased ECC in HFD cardiomyocytes. Therapies with PDE4 inhibitor roflumilast, PDE4D inhibitor BPN14770 or genetical deletion of PDE4D restored PKA phosphorylation of PLB and cardiac contractile function. CONCLUSION AND IMPLICATIONS The current study identifies upregulation of specific PDE4D isoforms that selectively inhibit SERCA2a function in HFD-induced cardiomyopathy, indicating that this remodelling can be targeted to restore cardiac contractility in diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Zhenduo Zhu
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education; Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
- Department of Pharmacology, University of California, Davis, Davis, California, USA
| | - Qiuyun Guan
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education; Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Bing Xu
- Department of Pharmacology, University of California, Davis, Davis, California, USA
- Department of Veterans Affairs Northern California Healthcare System, Mather, California, USA
| | - Sherif Bahriz
- Department of Pharmacology, University of California, Davis, Davis, California, USA
| | - Ao Shen
- School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Toni M West
- Department of Pharmacology, University of California, Davis, Davis, California, USA
| | - Yu Zhang
- School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Bingqing Deng
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wei Wei
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education; Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Yongsheng Han
- Department of Emergency Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Qingtong Wang
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education; Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
- Department of Pharmacology, University of California, Davis, Davis, California, USA
| | - Yang K Xiang
- Department of Pharmacology, University of California, Davis, Davis, California, USA
- Department of Veterans Affairs Northern California Healthcare System, Mather, California, USA
| |
Collapse
|
2
|
Zhu Y, Yang Z, Chen Y, Qian L, Hao C, Hao L, Yang B, Duan J. Association between glyphosate exposure and cardiovascular health using "Life's Essential 8" metrics in US adults. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 290:117559. [PMID: 39693852 DOI: 10.1016/j.ecoenv.2024.117559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 12/09/2024] [Accepted: 12/13/2024] [Indexed: 12/20/2024]
Abstract
BACKGROUND Glyphosate, as one of the most widely used herbicides worldwide, has been reclassified as a potential carcinogen and linked to multiple health problems. Recent data from animal experiments have proved its potential cardiovascular toxicity. However, whether glyphosate exposure influences human cardiovascular health at the population levels remains unknown. This study aims to elucidate the correlation between glyphosate exposure and Cardiovascular Health (CVH) by utilizing comprehensive Life's Essential 8 (LE8) metrics. METHODS Data from 2842 participants, approximating 155.24 million U.S. adults, from 2013 to 2018 in National Health and Nutrition Examination Survey were analyzed. The association between natural logarithm (ln)-transformed glyphosate exposure in urine and CVH was examined using weighted linear regression and restricted cubic spline (RCS) models. Mediation analysis was used to determine potential mediators correlated with glyphosate and CVH. Further subgroup analysis and sensitive analysis were conducted to confirm the results. RESULTS In fully adjusted models, the total CVH score decreased by 1.33 points for every unit increase in continuous ln-transformed glyphosate [β = -1.33, 95 % confidence interval (CI) (-2.25, -0.41)]. There was a negative correlation between the total CVH score and the lowest quantiles of ln-transformed glyphosate (Q1), Q2, Q3, and Q4 with p for trend < 0.05. A non-linear relationship between glyphosate and total CVH emerged (p for non-linear<0.001, p for overall = 0.003), and no safe threshold of glyphosate was observed. Serum insulin was an important mediator in the adverse effects of glyphosate on CVH with an 18.73 % mediation proportion. Moreover, higher serum insulin levels and higher homeostasis model assessment of insulin resistance were associated with higher glyphosate exposure but negatively correlated with total CVH score. CONCLUSIONS Glyphosate exposure may pose a risk to cardiovascular health at the population levels, with elevated serum insulin levels acting as a crucial mediating element. Further studies are required to investigate the safe threshold and underlying mechanism of glyphosate impairment.
Collapse
Affiliation(s)
- Yuankang Zhu
- Department of Gerontology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China.
| | - Zhenlin Yang
- Shi's Center of Orthopedics and Traumatology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Yuhao Chen
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325015, China.
| | - Lingzi Qian
- Department of Pediatric Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China.
| | - Changning Hao
- Department of Gerontology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China.
| | - Liangshi Hao
- Department of Gerontology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China.
| | - Boshen Yang
- Department of Cardiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China.
| | - Junli Duan
- Department of Gerontology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China.
| |
Collapse
|
3
|
Wu X, Elsaid S, Levet F, Li W, Tee SS. Establishing Immortalized Brown and White Preadipocyte Cell Lines from Young and Aged Mice. Curr Protoc 2024; 4:e70072. [PMID: 39670655 DOI: 10.1002/cpz1.70072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2024]
Abstract
Studying adipogenesis and adipocyte biology requires the isolation of primary preadipocytes from adipose tissues. However, primary preadipocytes have a limited lifespan, can only undergo a finite number of divisions, and often lose their original biological characteristics before becoming senescent. The repeated isolation of fresh preadipocytes, particularly from young pups or aged animals, is costly and time consuming. Immortalization of these cells offers a solution by overcoming cellular senescence and maintaining proliferative capacity, allowing for long-term studies without the continuous need to isolate new cells from animals. Immortalized cell lines thus provide a consistent and reproducible experimental model, significantly reducing variability across different animals. However, successfully establishing immortalized preadipocyte cell lines presents challenges, including selecting appropriate adipose tissue depots, isolating primary preadipocytes, and choosing an effective immortalization strategy. In this article, we present optimized protocols and share first-hand experiences establishing immortalized brown and white preadipocyte cell lines from young and aging mice. These protocols offer a valuable resource for researchers studying adipogenesis and metabolism. © 2024 Wiley Periodicals LLC. Support Protocol 1: Retrovirus production Basic Protocol 1: Isolation and culture of primary brown and white preadipocytes from mouse interscapular brown adipose tissue (iBAT) and subcutaneous white adipose tissue (sWAT) in the same region Basic Protocol 2: Immortalization of mouse brown and white preadipocytes Basic Protocol 3: Selection of immortalized preadipocytes Basic Protocol 4: Selection of single-cell clones of immortalized mouse preadipocytes Basic Protocol 5: Single-cell sorting in a 96-well plate using a flow cytometer for the selection of single-cell clones of immortalized preadipocytes Support Protocol 2: Cryopreservation of immortalized mouse preadipocytes Support Protocol 3: Thawing and culture of cryopreserved immortalized mouse preadipocytes Support Protocol 4: Subculture and expansion of immortalized mouse preadipocytes Basic Protocol 6: Differentiation of immortalized mouse brown and white preadipocytes Support Protocol 5: Identification of differentiated white and brown adipocytes.
Collapse
Affiliation(s)
- Xiangdong Wu
- Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Salaheldeen Elsaid
- Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Florian Levet
- Interdisciplinary Institute for Neuroscience, University of Bordeaux, Bordeaux, France
| | - Winson Li
- Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Sui Seng Tee
- Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| |
Collapse
|
4
|
Zhu XF, Mo YT, Hu YQ, Feng YX, Liu EH. Association between single-point insulin sensitivity estimator and heart failure in older adults: A cross-sectional study. Exp Gerontol 2024; 196:112578. [PMID: 39245081 DOI: 10.1016/j.exger.2024.112578] [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: 06/17/2024] [Revised: 08/22/2024] [Accepted: 09/05/2024] [Indexed: 09/10/2024]
Abstract
BACKGROUND Heart failure (HF) is a condition caused by a malfunction of the heart's pumping function. The single-point insulin sensitivity estimator (SPISE) index is a novel indicator for assessing insulin resistance in humans. However, the connection between the SPISE index and the risk of HF in the elderly is unknown. Therefore, our study aims to evaluate the connection between the SPISE index and HF in older adults. METHODS The study was based on data collected from the 1999-2020 National Health and Nutrition Examination Survey database and included 6165 participants aged ≥60 years. The multivariable linear regression model and the smooth fitting curve model were applied to investigate the connection between the SPISE index and HF in the elderly. Furthermore, the subgroup analysis was performed to investigate the interactive factors. RESULTS In this study, the mean age of the population was 69.38 years. After adjusting for all covariates, we observed that the SPISE index was inversely related to the prevalence of HF (OR = 0.87, 95 % CI = 0.80-0.94, P < 0.001) in older adults. The interaction analysis showed that the association might be affected by diabetes mellitus and smoking status. Additionally, an inflection point between the SPISE index and HF was found among older women. CONCLUSIONS An inverse correlation was detected between the SPISE index and HF in the elderly. This could provide new insight into the prevention and management of HF in the elderly population.
Collapse
Affiliation(s)
- Xiao-Feng Zhu
- Department of Clinical Medicine, The Nanshan School of Guangzhou Medical University, Guangzhou, 511436, China.
| | - Ye-Tong Mo
- Department of Clinical Medicine, The Third Clinical School of Guangzhou Medical University, Guangzhou, 511436, China
| | - Yu-Qi Hu
- Department of Clinical Medicine, The Third Clinical School of Guangzhou Medical University, Guangzhou, 511436, China
| | - Yu-Xue Feng
- Department of Clinical Medicine, The First Clinical School of Guangzhou Medical University, Guangzhou, 511436, China
| | - En-Hui Liu
- Department of Pediatrics, Pediatrics School, Guangzhou Medical University, Guangzhou, 511436, China
| |
Collapse
|
5
|
Achter JS, Vega ET, Sorrentino A, Kahnert K, Galsgaard KD, Hernandez-Varas P, Wierer M, Holst JJ, Wojtaszewski JFP, Mills RW, Kjøbsted R, Lundby A. In-depth phosphoproteomic profiling of the insulin signaling response in heart tissue and cardiomyocytes unveils canonical and specialized regulation. Cardiovasc Diabetol 2024; 23:258. [PMID: 39026321 PMCID: PMC11264841 DOI: 10.1186/s12933-024-02338-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 06/26/2024] [Indexed: 07/20/2024] Open
Abstract
BACKGROUND Insulin signaling regulates cardiac substrate utilization and is implicated in physiological adaptations of the heart. Alterations in the signaling response within the heart are believed to contribute to pathological conditions such as type-2 diabetes and heart failure. While extensively investigated in several metabolic organs using phosphoproteomic strategies, the signaling response elicited in cardiac tissue in general, and specifically in the specialized cardiomyocytes, has not yet been investigated to the same extent. METHODS Insulin or vehicle was administered to male C57BL6/JRj mice via intravenous injection into the vena cava. Ventricular tissue was extracted and subjected to quantitative phosphoproteomics analysis to evaluate the insulin signaling response. To delineate the cardiomyocyte-specific response and investigate the role of Tbc1d4 in insulin signal transduction, cardiomyocytes from the hearts of cardiac and skeletal muscle-specific Tbc1d4 knockout mice, as well as from wildtype littermates, were studied. The phosphoproteomic studies involved isobaric peptide labeling with Tandem Mass Tags (TMT), enrichment for phosphorylated peptides, fractionation via micro-flow reversed-phase liquid chromatography, and high-resolution mass spectrometry measurements. RESULTS We quantified 10,399 phosphorylated peptides from ventricular tissue and 12,739 from isolated cardiomyocytes, localizing to 3,232 and 3,128 unique proteins, respectively. In cardiac tissue, we identified 84 insulin-regulated phosphorylation events, including sites on the Insulin Receptor (InsrY1351, Y1175, Y1179, Y1180) itself as well as the Insulin receptor substrate protein 1 (Irs1S522, S526). Predicted kinases with increased activity in response to insulin stimulation included Rps6kb1, Akt1 and Mtor. Tbc1d4 emerged as a major phosphorylation target in cardiomyocytes. Despite limited impact on the global phosphorylation landscape, Tbc1d4 deficiency in cardiomyocytes attenuated insulin-induced Glut4 translocation and induced protein remodeling. We observed 15 proteins significantly regulated upon knockout of Tbc1d4. While Glut4 exhibited decreased protein abundance consequent to Tbc1d4-deficiency, Txnip levels were notably increased. Stimulation of wildtype cardiomyocytes with insulin led to the regulation of 262 significant phosphorylation events, predicted to be regulated by kinases such as Akt1, Mtor, Akt2, and Insr. In cardiomyocytes, the canonical insulin signaling response is elicited in addition to regulation on specialized cardiomyocyte proteins, such as Kcnj11Y12 and DspS2597. Details of all phosphorylation sites are provided. CONCLUSION We present a first global outline of the insulin-induced phosphorylation signaling response in heart tissue and in isolated adult cardiomyocytes, detailing the specific residues with changed phosphorylation abundances. Our study marks an important step towards understanding the role of insulin signaling in cardiac diseases linked to insulin resistance.
Collapse
Affiliation(s)
- Jonathan Samuel Achter
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Estefania Torres Vega
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Andrea Sorrentino
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Konstantin Kahnert
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Katrine Douglas Galsgaard
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Pablo Hernandez-Varas
- Core Facility for Integrated Microscopy, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Michael Wierer
- Proteomics Research Infrastructure, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jens Juul Holst
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jørgen Frank Pind Wojtaszewski
- The August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Robert William Mills
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Rasmus Kjøbsted
- The August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Alicia Lundby
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
6
|
Darwish R, Chen E, Minear S, Sheffield C. Resolution of insulin resistance, lactic acidosis, and decrease in mechanical support requirements in patients post orthotopic heart transplant with the use of long-acting insulin glargine. J Cardiothorac Surg 2024; 19:99. [PMID: 38365663 PMCID: PMC10873932 DOI: 10.1186/s13019-024-02543-y] [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: 07/13/2023] [Accepted: 01/28/2024] [Indexed: 02/18/2024] Open
Abstract
OBJECTIVE This study investigates the efficacy of using a long-acting insulin analog, along with the infusion of regular insulin, in achieving appropriate glycemic control and correcting lactic acidosis in patients post orthotopic heart transplant who demonstrate severe lactic acidosis and insulin resistance. METHODS This was a retrospective study of two cohorts (IRB FLA 20-003) of patients post orthotopic heart transplant with severe lactic acidosis and insulin resistance who were admitted to a tertiary intensive care unit and treated with (group 1) or without long-acting insulin analog (group 2) within the first 24 h of admission to the intensive care unit. Insulin resistance is defined as the requirement for intravenous regular insulin infusion of more than 20 units/h without the ability to achieve appropriate serum glucose level (120-180 mg /dL). Severe lactic acidosis is defined as arterial lactic acid of more than 10 mmol/L. The following parameters were investigated: time to correct lactic acidosis, duration of postoperative mechanical ventilation, the need for periprocedural mechanical circulatory support, and 28-day mortality. RESULTS The 28-day mortality was zero in both groups. Two patients required periprocedural mechanical support in group one, and ten patients required mechanical support in group two (RR = 0.224, 95%, confidence interval 0.052-0.95, Z = 2.029, p = 0.042). Three patients required tracheostomy in group one, and four patients required tracheostomy in group two (RR 0.84, 95 confidence interval 0.20-3.48, Z = 0.23, P = 0.81). Wilcoxon rank-sum test was used to compare time to correct lactic acidosis, with lactic acid resolution being faster in group one ([Formula: see text]1 = 19.7 h, SD ± 12.6 h [Formula: see text]2 = 29.3 h, SD ± 19.6 h, Z-value - 2.02, p-value 0.043). The duration of mechanical ventilation was less in group one ([Formula: see text]1 = 29 h, SD ± 12.7 h, [Formula: see text]2 = 55.1 h, SD ± 44.5 h, Z-value: - 1.92, p-value 0.05). CONCLUSION Administration of low-dose long-acting insulin glargine led to the resolution of the lactic acidosis, insulin resistance, and decreased requirements for pressor and inotropic support, which led to decreased need for mechanical circulatory support.
Collapse
Affiliation(s)
- Ribal Darwish
- Anesthesiology Institute, Surgical Critical Care Division, Cleveland Clinic Florida, Weston, USA.
| | - Eva Chen
- Anesthesiology Institute, Surgical Critical Care Division, Cleveland Clinic Florida, Weston, USA
| | - Steven Minear
- Anesthesiology Institute, Surgical Critical Care Division, Cleveland Clinic Florida, Weston, USA
| | - Cedric Sheffield
- Department of Thoracic and Cardiovascular Surgery, Cleveland Clinic Florida, Weston, USA
| |
Collapse
|
7
|
Smith A, Franklin O, McCallion N, Breathnach F, El-Khuffash A. Assessment of Myocardial Function in Infants of Mothers with Gestational Diabetes Mellitus Using Deformation Imaging over the First Year of Age. J Pediatr 2023; 263:113645. [PMID: 37517648 DOI: 10.1016/j.jpeds.2023.113645] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 07/19/2023] [Accepted: 07/25/2023] [Indexed: 08/01/2023]
Abstract
OBJECTIVE To assess serial myocardial performance and pulmonary vascular resistance (PVR) in infants of mothers with gestational diabetes mellitus (GDM) over the first year of life. STUDY DESIGN This was a prospective, observational study. Echocardiography was performed at birth, 6 months, and 1 year of age. Pulmonary artery acceleration time and left ventricular (LV) eccentricity index provided surrogate measurements of PVR. Biventricular function was assessed by tissue Doppler imaging and deformation analysis. RESULTS Fifty infants of mothers with GDM were compared with 50 controls with no difference in gestation (38.9 ± 0.8 weeks vs 39.3 ± 0.9 weeks; P = .05) or birthweight (3.55 ± 0.49 kg vs 3.56 ± 0.41 kg; P = .95). At 1 year of age, the pulmonary artery acceleration time was lower (70 ± 11 vs 79 ± 10; P = .01) in the GDM group. LV global longitudinal strain (24.7 ± 1.9 vs 28.8 ± 1.8 %; P < .01), LV systolic strain rate (1.8 ± 0.2 vs 2.1 ± 0.3 1/s; P < .01), and RV free wall strain (31.1 ± 4.8 vs 34.6 ± 3.9 %; P < .01) were lower in the GDM cohort at 1 year of age (all P values adjusted for gestation, mode of delivery, and maternal body mass index). CONCLUSIONS Our findings demonstrate higher indices of PVR and lower biventricular function in infants of mothers with GDM compared with controls at each time point assessed in this study over the first year of life.
Collapse
Affiliation(s)
- Aisling Smith
- Department of Neonatology, The Rotunda Hospital, Dublin, Ireland.
| | - Orla Franklin
- Department of Pediatric Cardiology, Our Lady's Children's Hospital Crumlin, Dublin, Ireland
| | - Naomi McCallion
- Department of Neonatology, The Rotunda Hospital, Dublin, Ireland; Department of Pediatrics, The Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Fionnuala Breathnach
- Department of Obstetrics & Gynaecology, The Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Afif El-Khuffash
- Department of Neonatology, The Rotunda Hospital, Dublin, Ireland; Department of Pediatrics, The Royal College of Surgeons in Ireland, Dublin, Ireland
| |
Collapse
|
8
|
Xu J, Xiao H, He K, Zhang Y. Crosstalk between adrenergic receptors and catalytic receptors. CURRENT OPINION IN PHYSIOLOGY 2023; 36:100718. [DOI: 10.1016/j.cophys.2023.100718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
9
|
Jovanovic A, Xu B, Zhu C, Ren D, Wang H, Krause-Hauch M, Abel ED, Li J, Xiang YK. Characterizing Adrenergic Regulation of Glucose Transporter 4-Mediated Glucose Uptake and Metabolism in the Heart. JACC Basic Transl Sci 2023. [DOI: 10.1016/j.jacbts.2022.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
|
10
|
Wang Y, Song X, Wang Y, Wang N. Specific interaction of insulin receptor and GLP-1 receptor mediates crosstalk between their signaling. Biochem Biophys Res Commun 2022; 636:31-39. [DOI: 10.1016/j.bbrc.2022.10.094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 10/27/2022] [Indexed: 11/02/2022]
|
11
|
Xu B, Wang Y, Bahriz SMFM, Zhao M, Zhu C, Xiang YK. Probing spatiotemporal PKA activity at the ryanodine receptor and SERCA2a nanodomains in cardomyocytes. Cell Commun Signal 2022; 20:143. [PMID: 36104752 PMCID: PMC9472443 DOI: 10.1186/s12964-022-00947-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 07/23/2022] [Indexed: 11/10/2022] Open
Abstract
AbstractSpatiotemporal regulation of subcellular protein kinase A (PKA) activity for precise substrate phosphorylation is essential for cellular responses to hormonal stimulation. Ryanodine receptor 2 (RyR2) and (sarco)endoplasmic reticulum calcium ATPase 2a (SERCA2a) represent two critical targets of β adrenoceptor (βAR) signaling on the sarcoplasmic reticulum membrane for cardiac excitation and contraction coupling. Using novel biosensors, we show that cardiac β1AR signals to both RyR2 and SERCA2a nanodomains in cardiomyocytes from mice, rats, and rabbits, whereas the β2AR signaling is restricted from these nanodomains. Phosphodiesterase 4 (PDE4) and PDE3 control the baseline PKA activity and prevent β2AR signaling from reaching the RyR2 and SERCA2a nanodomains. Moreover, blocking inhibitory G protein allows β2AR signaling to the RyR2 but not the SERCA2a nanodomains. This study provides evidence for the differential roles of inhibitory G protein and PDEs in controlling the adrenergic subtype signaling at the RyR2 and SERCA2a nanodomains in cardiomyocytes.
Collapse
|
12
|
Tadinada SM, Grzesik WJ, Kutschke W, Weiss RM, Abel ED. Acute effects of euglycemic-hyperinsulinemia on myocardial contractility in male mice. Physiol Rep 2022; 10:e15388. [PMID: 36073057 PMCID: PMC9453172 DOI: 10.14814/phy2.15388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 06/19/2022] [Accepted: 06/24/2022] [Indexed: 06/15/2023] Open
Abstract
Type 2 diabetes and obesity are associated with increased risk of cardiovascular disease, including heart failure. A hallmark of these dysmetabolic states is hyperinsulinemia and decreased cardiac reserve. However, the direct effects of hyperinsulinemia on myocardial function are incompletely understood. In this study, using invasive hemodynamics in mice, we studied the effects of short-term euglycemic hyperinsulinemia on basal myocardial function and subsequent responses of the myocardium to β-adrenergic stimulation. We found that cardiac function as measured by left ventricular (LV) invasive hemodynamics is not influenced by acute exposure to hyperinsulinemia, induced by an intravenous insulin injection with concurrent inotropic stimulation induced by β-adrenergic stimulation secondary to isoproterenol administration. When animals were exposed to 120-min of hyperinsulinemia by euglycemic-hyperinsulinemic clamps, there was a significant decrease in LV developed pressure, perhaps secondary to the systemic vasodilatory effects of insulin. Despite the baseline reduction, the contractile response to β-adrenergic stimulation remained intact in animals subject to euglycemic hyperinsulinemic clamps. β-adrenergic activation of phospholamban phosphorylation was not impaired by hyperinsulinemia. These results suggest that short-term hyperinsulinemia does not impair cardiac inotropic response to β-adrenergic stimulation in vivo.
Collapse
Affiliation(s)
- Satya Murthy Tadinada
- Department of Neuroscience and Pharmacology, Carver College of MedicineUniversity of IowaIowa CityIowaUSA
- Fraternal Order of Eagles Diabetes Research Center, Carver College of MedicineUniversity of IowaIowa CityIowaUSA
| | - Wojciech J. Grzesik
- Fraternal Order of Eagles Diabetes Research Center, Carver College of MedicineUniversity of IowaIowa CityIowaUSA
| | - William Kutschke
- Abboud Cardiovascular Research Center, Carver College of MedicineUniversity of IowaIowa CityIowaUSA
| | - Robert M. Weiss
- Abboud Cardiovascular Research Center, Carver College of MedicineUniversity of IowaIowa CityIowaUSA
- Division of Cardiology, Department of Internal Medicine, Carver College of MedicineUniversity of IowaIowa CityIowaUSA
| | - E. Dale Abel
- Department of Neuroscience and Pharmacology, Carver College of MedicineUniversity of IowaIowa CityIowaUSA
- Fraternal Order of Eagles Diabetes Research Center, Carver College of MedicineUniversity of IowaIowa CityIowaUSA
- Abboud Cardiovascular Research Center, Carver College of MedicineUniversity of IowaIowa CityIowaUSA
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Carver College of MedicineUniversity of IowaIowa CityIowaUSA
- Department of MedicineUniversity of California Los AngelesLos AngelesCaliforniaUSA
| |
Collapse
|
13
|
Xu R, Fu J, Hu Y, Yang X, Tao X, Chen L, Huang K, Fu Q. Roflumilast-Mediated Phosphodiesterase 4D Inhibition Reverses Diabetes-Associated Cardiac Dysfunction and Remodeling: Effects Beyond Glucose Lowering. Diabetes 2022; 71:1660-1678. [PMID: 35594380 DOI: 10.2337/db21-0898] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 05/02/2022] [Indexed: 11/13/2022]
Abstract
Patients with type 2 diabetes have a substantial risk of developing cardiovascular disease. Phosphodiesterase 4 (PDE4) dysregulation is of pathophysiological importance in metabolic disorders. For determination of the role of PDE4 in diabetic cardiac dysfunction, mice fed with a high-fat diet (HFD) were treated by pharmacological inhibition of PDE4 or cardiac specific knocking down of PDE4D. Mice on HFD developed diabetes and cardiac dysfunction with increased cardiac PDE4D5 expression. PDE4 inhibitor roflumilast can reverse hyperglycemia and cardiac dysfunction, accompanied by the decrease of PDE4D expression and increase of muscle specific miRNA miR-1 level in hearts. Either cardiac specific PDE4D knockdown or miR-1 overexpression significantly reversed cardiac dysfunction in HFD mice, despite persistence of hyperglycemia. Findings of gain- and loss-of-function studies of PDE4D in cardiomyocytes indicated that inhibition of insulin-induced PDE4D protected cardiac hypertrophy by preserving miR-1 expression in cardiomyocytes through promoting cAMP-CREB-Sirt1 signaling-induced SERCA2a expression. We further revealed that insulin also induced PDE4D expression in cardiac fibroblasts, which causes cardiac fibrosis through TGF-β1 signaling-mediated miR-1 reduction. Importantly, the expression of PDE4D5 was increased in human failing hearts of individuals with diabetes. These studies elucidate a novel mechanism by which hyperinsulinemia-induced cardiac PDE4D expression contributes to diabetic cardiac remodeling through reducing the expression of miR-1 and upregulation of miR-1 target hypertrophy and fibrosis-associated genes. Our study suggests a therapeutic potential of PDE4 inhibitor roflumilast in preventing or treating cardiac dysfunction in diabetes in addition to lowering glucose.
Collapse
Affiliation(s)
- Rui Xu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory for Drug Target Research and Pharmacodynamic Evaluation of Hubei Province, Wuhan, China
| | - Jing Fu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- 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
| | - Xiaoyan Yang
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory for Drug Target Research and Pharmacodynamic Evaluation of Hubei Province, Wuhan, China
| | - Xiang Tao
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Clinical Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Long Chen
- Clinical Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kai Huang
- Clinical Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qin Fu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory for Drug Target Research and Pharmacodynamic Evaluation of Hubei Province, Wuhan, China
| |
Collapse
|
14
|
Ren H, Chen Y, Ao Z, Cheng Q, Yang X, Tao H, Zhao L, Shen A, Li P, Fu Q. PDE4D binds and interacts with YAP to cooperatively promote HCC progression. Cancer Lett 2022; 541:215749. [PMID: 35597479 DOI: 10.1016/j.canlet.2022.215749] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 04/27/2022] [Accepted: 05/16/2022] [Indexed: 11/02/2022]
Abstract
The role of cAMP in the development of hepatocellular carcinoma (HCC) is controversial and the biological function of cAMP-hydrolysing enzyme phosphodiesterase 4D (PDE4D) in HCC remains unclear. In this study, we observed markedly higher PDE4D expression in HCC patients with poor survival. PDE4D bound to yes-associated protein (YAP), and PDE4D expression positively correlated with YAP expression in HCC. Overexpression of PDE4D increased YAP dephosphorylation and activity and promoted HCC cell growth in vitro and in vivo, which was attenuated by the YAP inhibitor verteporfin. In contrast, silencing PDE4D reduced YAP expression and HCC cell growth. Notably, forced expression of YAP promoted PDE4D and YAP target gene expression and cell growth, which were abrogated by the PDE4D inhibitor roflumilast. Mechanistically, silencing of YAP caused PDE4D downregulation and HCC cell apoptosis via extracellular signal-regulated kinase (ERK) activation. Roflumilast activated cAMP-PKA signaling and induced cAMP-PKA-dependent YAP phosphorylation at serine 127, resulting in YAP degradation and suppression of HCC growth, which were reversed by the PKA inhibitor PKI. Additionally, transfection of the YAP-S127A mutant reversed roflumilast-mediated suppression of YAP and cell growth. Taken together, our findings indicate that PDE4D binds to and interacts with YAP to promote HCC progression. Targeting the PDE4D-YAP interaction with roflumilast may be an effective strategy for HCC treatment.
Collapse
Affiliation(s)
- Huili Ren
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yingxiang Chen
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhou Ao
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qi Cheng
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoyan Yang
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Key Laboratory for Drug Target Research and Pharmacodynamic Evaluation of Hubei Province, Wuhan, China
| | - Hua Tao
- Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Lixin Zhao
- Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Ao Shen
- Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Peiyuan Li
- Department of Gastroenterology, Wenchang People's Hospital, Hainan, China; Division of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Qin Fu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Key Laboratory for Drug Target Research and Pharmacodynamic Evaluation of Hubei Province, Wuhan, China.
| |
Collapse
|
15
|
Insulin Receptors and Insulin Action in the Heart: The Effects of Left Ventricular Assist Devices. Biomolecules 2022; 12:biom12040578. [PMID: 35454166 PMCID: PMC9024449 DOI: 10.3390/biom12040578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 04/11/2022] [Accepted: 04/12/2022] [Indexed: 02/01/2023] Open
Abstract
This year, 2022, marks the 100th anniversary of the isolation of human insulin and its administration to patients suffering from diabetes mellitus (DM). Insulin exerts many effects on the human body, including the cardiac tissue. The pathways implicated include the PKB/Akt signaling pathway, the Janus kinase, and the mitogen-activated protein kinase pathway and lead to normal cardiac growth, vascular smooth muscle regulation, and cardiac contractility. This review aims to summarize the existing knowledge and provide new insights on insulin pathways of cardiac tissue, along with the role of left ventricular assist devices on insulin regulation and cardiac function.
Collapse
|
16
|
Limberg JK, Soares RN, Padilla J. Role of the Autonomic Nervous System in the Hemodynamic Response to Hyperinsulinemia-Implications for Obesity and Insulin Resistance. Curr Diab Rep 2022; 22:169-175. [PMID: 35247145 PMCID: PMC9012695 DOI: 10.1007/s11892-022-01456-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/30/2021] [Indexed: 11/29/2022]
Abstract
PURPOSE OF REVIEW Herein, we summarize recent advances which provide new insights into the role of the autonomic nervous system in the control of blood flow and blood pressure during hyperinsulinemia. We also highlight remaining gaps in knowledge as it pertains to the translation of findings to relevant human chronic conditions such as obesity, insulin resistance, and type 2 diabetes. RECENT FINDINGS Our findings in insulin-sensitive adults show that increases in muscle sympathetic nerve activity with hyperinsulinemia do not result in greater sympathetically mediated vasoconstriction in the peripheral circulation. Both an attenuation of α-adrenergic-receptor vasoconstriction and augmented β-adrenergic vasodilation in the setting of high insulin likely explain these findings. In the absence of an increase in sympathetically mediated restraint of peripheral vasodilation during hyperinsulinemia, blood pressure is supported by increases in cardiac output in insulin-sensitive individuals. We highlight a dynamic interplay between central and peripheral mechanisms during hyperinsulinemia to increase sympathetic nervous system activity and maintain blood pressure in insulin-sensitive adults. Whether these results translate to the insulin-resistant condition and implications for long-term cardiovascular regulation warrants further exploration.
Collapse
Affiliation(s)
- Jacqueline K Limberg
- Department of Nutrition and Exercise Physiology, University of Missouri, 204 Gwynn Hall, Columbia, MO, 65211, USA.
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA.
| | - Rogerio N Soares
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
| | - Jaume Padilla
- Department of Nutrition and Exercise Physiology, University of Missouri, 204 Gwynn Hall, Columbia, MO, 65211, USA
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
| |
Collapse
|
17
|
Shen X, He S, Wang J, Qian X, Wang H, Zhang B, Chen Y, Li H, Li G. Phenotype of higher post-load insulin response as a predictor of all-cause mortality and cardiovascular mortality in the Chinese non-diabetic population. Diabetol Metab Syndr 2022; 14:19. [PMID: 35090539 PMCID: PMC8796343 DOI: 10.1186/s13098-022-00786-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 01/06/2022] [Indexed: 11/10/2022] Open
Abstract
AIM This study aimed to assess whether a higher insulin response increased the long-term risk of mortality in a non-diabetic population. METHODS A total of 446 people with normal glucose tolerance (NGT) or impaired glucose tolerance (IGT) who participated in the Da Qing Diabetes Study, were stratified into quartiles subgroups according to their baseline insulin area under the curve (AUC) during oral glucose tolerance test, defined as Q1, Q2, Q3 and Q4. The participants were followed from 1986 to 2016 to assess the risk of death in association with the magnitude of post-load insulin response. RESULTS Over 30 years, the rates of all cause death were 9.94, 14.81, 15.02, and 17.58 per 1000 person-years across the four groups respectively. The rate for cardiovascular disease (CVD) death was 5.14, 6.50, 6.80 and 10.47 per 1000 person-years. Compared with Q1, the risk of all-cause death was significantly higher in participants in Q4 (HR = 2.14, 95% CI 1.34-3.42), Q3 (HR = 1.94, 95% CI 1.20-3.14), and Q2 group (HR = 1.70, 95% CI 1.06-2.74). In the Fine-Gray model with non-CVD death as competing risk, the increased insulin AUC were also significantly associated with the CVD death (Q4 vs Q1, HR = 2.04, 95% CI 1.10-3.79). In the fractional polynomial regression analysis, a nonlinear association between insulin AUC and all-cause and CVD death was demonstrated. In addition, insulin AUC was associated with a progressively higher risk of all-cause death and CVD death (fractional power 3, P < 0.001). CONCLUSION A higher post-load insulin response was significantly associated with a long-term increased risk of all-cause and CVD deaths in the Chinese non-diabetic population. It suggests that people featured by this phenotype is a potential important target for further intervention.
Collapse
Affiliation(s)
- Xiaoxia Shen
- Center of Endocrinology, Fuwai Hospital, Chinese Academy of Medical Sciences, No. 167 North Lishi Road, Xicheng District, Beijing, 100037, China
| | - Siyao He
- Center of Endocrinology, Fuwai Hospital, Chinese Academy of Medical Sciences, No. 167 North Lishi Road, Xicheng District, Beijing, 100037, China
| | - Jinping Wang
- Department of Cardiology, Da Qing First Hospital, No. 9 Zhongkang Street, Saltu District, Daqing, 163411, Heilongjiang, China
| | - Xin Qian
- Center of Endocrinology, Fuwai Hospital, Chinese Academy of Medical Sciences, No. 167 North Lishi Road, Xicheng District, Beijing, 100037, China
| | - Hui Wang
- Center of Endocrinology, Fuwai Hospital, Chinese Academy of Medical Sciences, No. 167 North Lishi Road, Xicheng District, Beijing, 100037, China
| | - Bo Zhang
- Department of Endocrinology, China-Japan Friendship Hospital, No 2, East Yinghua Road, Chaoyang District, Beijing, 100029, China
| | - Yanyan Chen
- Center of Endocrinology, Fuwai Hospital, Chinese Academy of Medical Sciences, No. 167 North Lishi Road, Xicheng District, Beijing, 100037, China
| | - Hui Li
- Department of Cardiology, Da Qing First Hospital, No. 9 Zhongkang Street, Saltu District, Daqing, 163411, Heilongjiang, China
| | - Guangwei Li
- Center of Endocrinology, Fuwai Hospital, Chinese Academy of Medical Sciences, No. 167 North Lishi Road, Xicheng District, Beijing, 100037, China.
- Department of Endocrinology, China-Japan Friendship Hospital, No 2, East Yinghua Road, Chaoyang District, Beijing, 100029, China.
| |
Collapse
|
18
|
Carruth ED, Fielden SW, Nevius CD, Fornwalt BK, Haggerty CM. 3D-Encoded DENSE MRI with Zonal Excitation for Quantifying Biventricular Myocardial Strain During a Breath-Hold. Cardiovasc Eng Technol 2021; 12:589-597. [PMID: 34244904 DOI: 10.1007/s13239-021-00561-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 06/25/2021] [Indexed: 11/28/2022]
Abstract
PURPOSE Right ventricular (RV) function is increasingly recognized for its prognostic value in many disease states. As with the left ventricle (LV), strain-based measurements may have better prognostic value than typical chamber volumes or ejection fraction. Complete functional characterization of the RV requires high-resolution, 3D displacement tracking methods, which have been prohibitively challenging to implement. Zonal excitation during Displacement ENcoding with Stimulated Echoes (DENSE) magnetic resonance imaging (MRI) has helped reduce scan time for 2D LV strain quantification. We hypothesized that zonal excitation could alternatively be used to reproducibly acquire higher resolution, 3D-encoded DENSE images for quantification of bi-ventricular strain within a single breath-hold. METHODS We modified sequence parameters for a 3D zonal excitation DENSE sequence to achieve in-plane resolution < 2 mm and acquired two sets of images in eight healthy adult male volunteers with median (IQR) age 32.5 (32.0-33.8) years. We assessed the inter-test reproducibility of this technique, and compared computed strains and torsion with previously published data. RESULTS Data for one subject was excluded based on image artifacts. Reproducibility for LV (CoV: 6.1-9.0%) and RV normal strains (CoV: 6.3-8.2%) and LV torsion (CoV = 7.1%) were all very good. Reproducibility of RV torsion was lower (CoV = 16.7%), but still within acceptable limits. Computed global strains and torsion were within reasonable agreement with published data, but further studies in larger cohorts are needed to confirm. CONCLUSION Reproducible acquisition of 3D-encoded biventricular myocardial strain data in a breath-hold is feasible using DENSE with zonal excitation.
Collapse
Affiliation(s)
- Eric D Carruth
- Department of Translational Data Science and Informatics, Geisinger, Danville, PA, USA
| | - Samuel W Fielden
- Department of Translational Data Science and Informatics, Geisinger, Danville, PA, USA.,Medical and Health Physics, Geisinger, Danville, PA, USA
| | - Christopher D Nevius
- Department of Translational Data Science and Informatics, Geisinger, Danville, PA, USA
| | - Brandon K Fornwalt
- Department of Translational Data Science and Informatics, Geisinger, Danville, PA, USA.,The Heart Institute, Geisinger, Danville, PA, USA.,Department of Radiology, Geisinger, Danville, PA, USA
| | - Christopher M Haggerty
- Department of Translational Data Science and Informatics, Geisinger, Danville, PA, USA. .,The Heart Institute, Geisinger, Danville, PA, USA.
| |
Collapse
|
19
|
Abstract
Alterations in cardiac energy metabolism contribute to the severity of heart failure. However, the energy metabolic changes that occur in heart failure are complex and are dependent not only on the severity and type of heart failure present but also on the co-existence of common comorbidities such as obesity and type 2 diabetes. The failing heart faces an energy deficit, primarily because of a decrease in mitochondrial oxidative capacity. This is partly compensated for by an increase in ATP production from glycolysis. The relative contribution of the different fuels for mitochondrial ATP production also changes, including a decrease in glucose and amino acid oxidation, and an increase in ketone oxidation. The oxidation of fatty acids by the heart increases or decreases, depending on the type of heart failure. For instance, in heart failure associated with diabetes and obesity, myocardial fatty acid oxidation increases, while in heart failure associated with hypertension or ischemia, myocardial fatty acid oxidation decreases. Combined, these energy metabolic changes result in the failing heart becoming less efficient (ie, a decrease in cardiac work/O2 consumed). The alterations in both glycolysis and mitochondrial oxidative metabolism in the failing heart are due to both transcriptional changes in key enzymes involved in these metabolic pathways, as well as alterations in NAD redox state (NAD+ and nicotinamide adenine dinucleotide levels) and metabolite signaling that contribute to posttranslational epigenetic changes in the control of expression of genes encoding energy metabolic enzymes. Alterations in the fate of glucose, beyond flux through glycolysis or glucose oxidation, also contribute to the pathology of heart failure. Of importance, pharmacological targeting of the energy metabolic pathways has emerged as a novel therapeutic approach to improving cardiac efficiency, decreasing the energy deficit and improving cardiac function in the failing heart.
Collapse
Affiliation(s)
- Gary D Lopaschuk
- Cardiovascular Research Centre, University of Alberta, Edmonton, Canada (G.D.L., Q.G.K.)
| | - Qutuba G Karwi
- Cardiovascular Research Centre, University of Alberta, Edmonton, Canada (G.D.L., Q.G.K.)
| | - Rong Tian
- Mitochondria and Metabolism Center, University of Washington, Seattle (R.T.)
| | - Adam R Wende
- Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham (A.R.W.)
| | - E Dale Abel
- Division of Endocrinology and Metabolism, University of Iowa Carver College of Medicine, Iowa City (E.D.A.).,Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City (E.D.A.)
| |
Collapse
|
20
|
Limberg JK, Soares RN, Power G, Harper JL, Smith JA, Shariffi B, Jacob DW, Manrique-Acevedo C, Padilla J. Hyperinsulinemia blunts sympathetic vasoconstriction: a possible role of β-adrenergic activation. Am J Physiol Regul Integr Comp Physiol 2021; 320:R771-R779. [PMID: 33851554 DOI: 10.1152/ajpregu.00018.2021] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Herein we report in a sample of healthy young men (n = 14) and women (n = 12) that hyperinsulinemia induces time-dependent decreases in total peripheral resistance and its contribution to the maintenance of blood pressure. In the same participants, we observe profound vasodilatory effects of insulin in the lower limb despite concomitant activation of the sympathetic nervous system. We hypothesized that this prominent peripheral vasodilation is possibly due to the ability of the leg vasculature to escape sympathetic vasoconstriction during systemic insulin stimulation. Consistent with this notion, we demonstrate in a subset of healthy men (n = 9) and women (n = 7) that systemic infusion of insulin blunts sympathetically mediated leg vasoconstriction evoked by a cold pressor test, a well-established sympathoexcitatory stimulus. Further substantiating this observation, we show in mouse aortic rings that insulin exposure suppresses epinephrine and norepinephrine-induced vasoconstriction. Notably, we found that such insulin-suppressing effects on catecholamine-induced constriction are diminished following β-adrenergic receptor blockade. In accordance, we also reveal that insulin augments β-adrenergic-mediated vasorelaxation in isolated arteries. Collectively, these findings support the idea that sympathetic vasoconstriction can be attenuated during systemic hyperinsulinemia in the leg vasculature of both men and women and that this phenomenon may be in part mediated by potentiation of β-adrenergic vasodilation neutralizing α-adrenergic vasoconstriction.
Collapse
Affiliation(s)
- Jacqueline K Limberg
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
| | - Rogerio N Soares
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| | - Gavin Power
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
| | - Jennifer L Harper
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
| | - James A Smith
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
| | - Brian Shariffi
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
| | - Dain W Jacob
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
| | - Camila Manrique-Acevedo
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri.,Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Missouri, Columbia, Missouri.,Research Services, Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri
| | - Jaume Padilla
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri.,Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| |
Collapse
|
21
|
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: 0.8] [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
|
22
|
|
23
|
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: 3.0] [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
|
24
|
Smith A, Franklin O, McCallion N, Breatnach F, El-Khuffash A. Effect of Gestational Diabetes Mellitus on Neonatal Myocardial Function. Neonatology 2021; 118:64-72. [PMID: 33596570 DOI: 10.1159/000513041] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 11/13/2020] [Indexed: 11/19/2022]
Abstract
BACKGROUND AND AIMS Infants born to mothers with gestational diabetes mellitus (GDM) have impaired myocardial performance and are at risk of pulmonary hypertension. We aimed to assess myocardial deformation and left ventricular (LV) rotational mechanics in this population. METHODS We studied 40 infants of mothers with GDM and 40 control infants. Three echocardiograms were carried out over the first 3 days after birth. RESULTS GDM infants had a lower gestation at birth and a thicker septal wall, a higher LV eccentricity index (indicating septal bowing), and a lower PAATi (indicating higher pulmonary vascular resistance) (all p < 0.05). GDM infants had lower LV strain, systolic and early diastolic strain rates, lower right ventricular (RV) strain, and early diastolic strain rates over the study period (all p < 0.05). By day 3, GDM infants had higher twist, torsion, and higher LV twist and untwist rates (all p < 0.05). GDM status was an independent predictor of LV and RV function and pulmonary vascular resistance (p < 0.01). CONCLUSION Infants of mothers with GDM demonstrate important changes in myocardial function in addition to pulmonary vascular resistance that do not resolve by hospital discharge. The observed LV twist increase in GDM infants may be a compensatory mechanism for the lower longitudinal function in this cohort.
Collapse
Affiliation(s)
- Aisling Smith
- Department of Neonatology, The Rotunda Hospital, Dublin, Ireland
| | - Orla Franklin
- Department of Paediatric Cardiology, Our Lady's Children's Hospital Crumlin, Dublin, Ireland
| | - Naomi McCallion
- Department of Neonatology, The Rotunda Hospital, Dublin, Ireland.,Department of Paediatrics, The Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Fionnouala Breatnach
- Department of Obstetrics & Gynaecology, The Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Afif El-Khuffash
- Department of Neonatology, The Rotunda Hospital, Dublin, Ireland, .,Department of Paediatrics, The Royal College of Surgeons in Ireland, Dublin, Ireland,
| |
Collapse
|
25
|
Pei J, Xiao Z, Guo Z, Pei Y, Wei S, Wu H, Wang D. Sustained Stimulation of β 2AR Inhibits Insulin Signaling in H9C2 Cardiomyoblast Cells Through the PKA-Dependent Signaling Pathway. Diabetes Metab Syndr Obes 2020; 13:3887-3898. [PMID: 33116735 PMCID: PMC7585860 DOI: 10.2147/dmso.s268028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 09/05/2020] [Indexed: 12/16/2022] Open
Abstract
INTRODUCTION This study aimed to investigate the role of β2 adrenergic receptor (β2AR) in insulin signaling transduction in H9C2 cardiomyoblast cells to understand the formation of the β2AR-insulin receptor (IR) protein complex and its role in insulin-induced Glut4 expression. METHODS H9C2 cells were treated with various protein inhibitors (CGP, β1AR inhibitor CGP20712; ICI, β2AR inhibitor ICI 118,551; PKI, PKA inhibitor myristoylated PKI; PD 0325901, MEK inhibitor; SP600125, JNK inhibitor) with or without insulin or isoproterenol (ISO) before RNA-sequencing (RNA-Seq) and quantitative-PCR (Q-PCR). Yeast two-hybrid, co-immunoprecipitation and His-tag pull-down assay were carried out to investigate the formation of the β2AR-IR protein complex. The intracellular concentrations of cAMP in H9C2 cells were tested by high performance liquid chromatography (HPLC) and the phosphorylation of JNK was tested by Western blot. RESULTS Gene Ontology (GO) analysis revealed that the most significantly enriched processes in the domain of molecular function (MF) were catalytic activity and binding, whereas in the domain of biological processes (BP) were metabolic process and cellular process. Furthermore, the enriched processes in the domain of cellular components (CC) were cell and cell parts. The Kyoto encyclopedia of genes and genomes (KEGG) pathway analysis showed that the most significant pathways that have been altered included the PI3K-Akt and MAPK signaling pathways. Q-PCR, which was performed to verify the gene expression levels exhibited consistent results. In evaluating the signaling pathways, the sustained stimulation of β2AR by ISO inhibited insulin signalling, and the effect was primarily through the cAMP-PKA-JNK pathway and MEK/JNK signaling pathway. Yeast two-hybrid, co-immunoprecipitation and His-tag pull-down assay revealed that β2AR, IR, insulin receptor substrate 1 (IRS1), Grb2-associated binding protein 1 (GAB1) and Grb2 existed in the same protein complex. CONCLUSION The sustained stimulation of β2AR might inhibit insulin signaling transduction through the cAMP-PKA-JNK and MEK/JNK pathways in H9C2 cells.
Collapse
Affiliation(s)
- Jinli Pei
- Key Laboratory of Ministry of Education for Tropical Bioresources, Hainan University, Haikou, Hainan570228, People's Republic of China
- Laboratory of Biotechnology and Molecular Pharmacology, School of Life and Pharmaceutical Sciences, Hainan University, Haikou, Hainan570228, People's Republic of China
| | - Zhengpan Xiao
- Key Laboratory of Ministry of Education for Tropical Bioresources, Hainan University, Haikou, Hainan570228, People's Republic of China
- Laboratory of Biotechnology and Molecular Pharmacology, School of Life and Pharmaceutical Sciences, Hainan University, Haikou, Hainan570228, People's Republic of China
| | - Ziyi Guo
- Key Laboratory of Ministry of Education for Tropical Bioresources, Hainan University, Haikou, Hainan570228, People's Republic of China
- Laboratory of Biotechnology and Molecular Pharmacology, School of Life and Pharmaceutical Sciences, Hainan University, Haikou, Hainan570228, People's Republic of China
| | - Yechun Pei
- Key Laboratory of Ministry of Education for Tropical Bioresources, Hainan University, Haikou, Hainan570228, People's Republic of China
- Laboratory of Biotechnology and Molecular Pharmacology, School of Life and Pharmaceutical Sciences, Hainan University, Haikou, Hainan570228, People's Republic of China
| | - Shuangshuang Wei
- Key Laboratory of Ministry of Education for Tropical Bioresources, Hainan University, Haikou, Hainan570228, People's Republic of China
- Laboratory of Biotechnology and Molecular Pharmacology, School of Life and Pharmaceutical Sciences, Hainan University, Haikou, Hainan570228, People's Republic of China
| | - Hao Wu
- Key Laboratory of Ministry of Education for Tropical Bioresources, Hainan University, Haikou, Hainan570228, People's Republic of China
- Laboratory of Biotechnology and Molecular Pharmacology, School of Life and Pharmaceutical Sciences, Hainan University, Haikou, Hainan570228, People's Republic of China
| | - Dayong Wang
- Key Laboratory of Ministry of Education for Tropical Bioresources, Hainan University, Haikou, Hainan570228, People's Republic of China
- Laboratory of Biotechnology and Molecular Pharmacology, School of Life and Pharmaceutical Sciences, Hainan University, Haikou, Hainan570228, People's Republic of China
| |
Collapse
|
26
|
Abstract
Diabetes mellitus predisposes affected individuals to a significant spectrum of cardiovascular complications, one of the most debilitating in terms of prognosis is heart failure. Indeed, the increasing global prevalence of diabetes mellitus and an aging population has given rise to an epidemic of diabetes mellitus-induced heart failure. Despite the significant research attention this phenomenon, termed diabetic cardiomyopathy, has received over several decades, understanding of the full spectrum of potential contributing mechanisms, and their relative contribution to this heart failure phenotype in the specific context of diabetes mellitus, has not yet been fully resolved. Key recent preclinical discoveries that comprise the current state-of-the-art understanding of the basic mechanisms of the complex phenotype, that is, the diabetic heart, form the basis of this review. Abnormalities in each of cardiac metabolism, physiological and pathophysiological signaling, and the mitochondrial compartment, in addition to oxidative stress, inflammation, myocardial cell death pathways, and neurohumoral mechanisms, are addressed. Further, the interactions between each of these contributing mechanisms and how they align to the functional, morphological, and structural impairments that characterize the diabetic heart are considered in light of the clinical context: from the disease burden, its current management in the clinic, and where the knowledge gaps remain. The need for continued interrogation of these mechanisms (both known and those yet to be identified) is essential to not only decipher the how and why of diabetes mellitus-induced heart failure but also to facilitate improved inroads into the clinical management of this pervasive clinical challenge.
Collapse
Affiliation(s)
- Rebecca H. Ritchie
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University (Parkville campus), Parkville, Victoria 3052, Australia
| | - E. Dale Abel
- Division of Endocrinology and Metabolism, University of Iowa Carver College of Medicine, Iowa City, IA 52242, United States
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, IA 52242, United States
| |
Collapse
|
27
|
Vetter L, Cortassa S, O'Rourke B, Armoundas AA, Bedja D, Jende JME, Bendszus M, Paolocci N, Sollot SJ, Aon MA, Kurz FT. Diabetes Increases the Vulnerability of the Cardiac Mitochondrial Network to Criticality. Front Physiol 2020; 11:175. [PMID: 32210835 PMCID: PMC7077512 DOI: 10.3389/fphys.2020.00175] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 02/14/2020] [Indexed: 02/06/2023] Open
Abstract
Mitochondrial criticality describes a state in which the mitochondrial cardiac network under intense oxidative stress becomes very sensitive to small perturbations, leading from local to cell-wide depolarization and synchronized oscillations that may escalate to the myocardial syncytium generating arrhythmias. Herein, we describe the occurrence of mitochondrial criticality in the chronic setting of a metabolic disorder, type 1 diabetes (T1DM), using a streptozotocin (STZ)-treated guinea pig (GP) animal model. Using wavelet analysis of mitochondrial networks from two-photon microscopy imaging of cardiac myocytes loaded with a fluorescent probe of the mitochondrial membrane potential, we show that cardiomyocytes from T1DM GPs are closer to criticality, making them more vulnerable to cell-wide mitochondrial oscillations as can be judged by the latency period to trigger oscillations after a laser flash perturbation, and their propensity to oscillate. Insulin treatment of T1DM GPs rescued cardiac myocytes to sham control levels of susceptibility, a protective condition that could also be attained with interventions leading to improvement of the cellular redox environment such as preincubation of diabetic cardiac myocytes with the lipid palmitate or a cell-permeable form of glutathione, in the presence of glucose.
Collapse
Affiliation(s)
- Larissa Vetter
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States.,Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Sonia Cortassa
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
| | - Brian O'Rourke
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Antonis A Armoundas
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States.,Institute for Medical Engineering and Science, Massachusetts Institute of Technology Cambridge, MA, United States
| | - Djahida Bedja
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Johann M E Jende
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Martin Bendszus
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Nazareno Paolocci
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD, United States.,Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Steven J Sollot
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
| | - Miguel A Aon
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
| | - Felix T Kurz
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany.,Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States
| |
Collapse
|
28
|
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: 2.0] [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
|
29
|
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: 36] [Impact Index Per Article: 6.0] [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
|
30
|
Yang HQ, Wang LP, Gong YY, Fan XX, Zhu SY, Wang XT, Wang YP, Li LL, Xing X, Liu XX, Ji GS, Hou T, Zhang Y, Xiao RP, Wang SQ. β
2
-Adrenergic Stimulation Compartmentalizes β
1
Signaling Into Nanoscale Local Domains by Targeting the C-Terminus of β
1
-Adrenoceptors. Circ Res 2019; 124:1350-1359. [DOI: 10.1161/circresaha.118.314322] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Hua-Qian Yang
- From the State Key Lab of Membrane Biology, College of Life Sciences and Institute of Molecular Medicine, Peking University, Beijing, China (H.-Q.Y., L.-P.W., X.-X.F., S.-Y.Z., X.-T.W., Y.-P.W., L.-L.L., X.X., X.-X.L., G.-S.J., T.T.H., Y.Z., R.-P.X., S.-Q.W.)
| | - Li-Peng Wang
- From the State Key Lab of Membrane Biology, College of Life Sciences and Institute of Molecular Medicine, Peking University, Beijing, China (H.-Q.Y., L.-P.W., X.-X.F., S.-Y.Z., X.-T.W., Y.-P.W., L.-L.L., X.X., X.-X.L., G.-S.J., T.T.H., Y.Z., R.-P.X., S.-Q.W.)
| | - Yun-Yun Gong
- Beijing Advanced Innovation Center for Biomedical Engineering, and School of Biological Science and Medical Engineering, Beihang University, Beijing, China (Y.-Y.G)
| | - Xue-Xin Fan
- From the State Key Lab of Membrane Biology, College of Life Sciences and Institute of Molecular Medicine, Peking University, Beijing, China (H.-Q.Y., L.-P.W., X.-X.F., S.-Y.Z., X.-T.W., Y.-P.W., L.-L.L., X.X., X.-X.L., G.-S.J., T.T.H., Y.Z., R.-P.X., S.-Q.W.)
| | - Si-Yu Zhu
- From the State Key Lab of Membrane Biology, College of Life Sciences and Institute of Molecular Medicine, Peking University, Beijing, China (H.-Q.Y., L.-P.W., X.-X.F., S.-Y.Z., X.-T.W., Y.-P.W., L.-L.L., X.X., X.-X.L., G.-S.J., T.T.H., Y.Z., R.-P.X., S.-Q.W.)
| | - Xiao-Ting Wang
- From the State Key Lab of Membrane Biology, College of Life Sciences and Institute of Molecular Medicine, Peking University, Beijing, China (H.-Q.Y., L.-P.W., X.-X.F., S.-Y.Z., X.-T.W., Y.-P.W., L.-L.L., X.X., X.-X.L., G.-S.J., T.T.H., Y.Z., R.-P.X., S.-Q.W.)
| | - Yu-Pu Wang
- From the State Key Lab of Membrane Biology, College of Life Sciences and Institute of Molecular Medicine, Peking University, Beijing, China (H.-Q.Y., L.-P.W., X.-X.F., S.-Y.Z., X.-T.W., Y.-P.W., L.-L.L., X.X., X.-X.L., G.-S.J., T.T.H., Y.Z., R.-P.X., S.-Q.W.)
| | - Lin-Lin Li
- From the State Key Lab of Membrane Biology, College of Life Sciences and Institute of Molecular Medicine, Peking University, Beijing, China (H.-Q.Y., L.-P.W., X.-X.F., S.-Y.Z., X.-T.W., Y.-P.W., L.-L.L., X.X., X.-X.L., G.-S.J., T.T.H., Y.Z., R.-P.X., S.-Q.W.)
| | - Xin Xing
- From the State Key Lab of Membrane Biology, College of Life Sciences and Institute of Molecular Medicine, Peking University, Beijing, China (H.-Q.Y., L.-P.W., X.-X.F., S.-Y.Z., X.-T.W., Y.-P.W., L.-L.L., X.X., X.-X.L., G.-S.J., T.T.H., Y.Z., R.-P.X., S.-Q.W.)
| | - Xiao-Xiao Liu
- From the State Key Lab of Membrane Biology, College of Life Sciences and Institute of Molecular Medicine, Peking University, Beijing, China (H.-Q.Y., L.-P.W., X.-X.F., S.-Y.Z., X.-T.W., Y.-P.W., L.-L.L., X.X., X.-X.L., G.-S.J., T.T.H., Y.Z., R.-P.X., S.-Q.W.)
| | - Guang-Shen Ji
- From the State Key Lab of Membrane Biology, College of Life Sciences and Institute of Molecular Medicine, Peking University, Beijing, China (H.-Q.Y., L.-P.W., X.-X.F., S.-Y.Z., X.-T.W., Y.-P.W., L.-L.L., X.X., X.-X.L., G.-S.J., T.T.H., Y.Z., R.-P.X., S.-Q.W.)
| | - TingTing Hou
- From the State Key Lab of Membrane Biology, College of Life Sciences and Institute of Molecular Medicine, Peking University, Beijing, China (H.-Q.Y., L.-P.W., X.-X.F., S.-Y.Z., X.-T.W., Y.-P.W., L.-L.L., X.X., X.-X.L., G.-S.J., T.T.H., Y.Z., R.-P.X., S.-Q.W.)
| | - Yan Zhang
- From the State Key Lab of Membrane Biology, College of Life Sciences and Institute of Molecular Medicine, Peking University, Beijing, China (H.-Q.Y., L.-P.W., X.-X.F., S.-Y.Z., X.-T.W., Y.-P.W., L.-L.L., X.X., X.-X.L., G.-S.J., T.T.H., Y.Z., R.-P.X., S.-Q.W.)
| | - Rui-Ping Xiao
- From the State Key Lab of Membrane Biology, College of Life Sciences and Institute of Molecular Medicine, Peking University, Beijing, China (H.-Q.Y., L.-P.W., X.-X.F., S.-Y.Z., X.-T.W., Y.-P.W., L.-L.L., X.X., X.-X.L., G.-S.J., T.T.H., Y.Z., R.-P.X., S.-Q.W.)
| | - Shi-Qiang Wang
- From the State Key Lab of Membrane Biology, College of Life Sciences and Institute of Molecular Medicine, Peking University, Beijing, China (H.-Q.Y., L.-P.W., X.-X.F., S.-Y.Z., X.-T.W., Y.-P.W., L.-L.L., X.X., X.-X.L., G.-S.J., T.T.H., Y.Z., R.-P.X., S.-Q.W.)
| |
Collapse
|
31
|
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: 1.0] [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
|
32
|
Murga C, Arcones AC, Cruces-Sande M, Briones AM, Salaices M, Mayor F. G Protein-Coupled Receptor Kinase 2 (GRK2) as a Potential Therapeutic Target in Cardiovascular and Metabolic Diseases. Front Pharmacol 2019; 10:112. [PMID: 30837878 PMCID: PMC6390810 DOI: 10.3389/fphar.2019.00112] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 01/28/2019] [Indexed: 12/20/2022] Open
Abstract
G protein-coupled receptor kinase 2 (GRK2) is a central signaling node involved in the modulation of many G protein-coupled receptors (GPCRs) and also displaying regulatory functions in other cell signaling routes. GRK2 levels and activity have been reported to be enhanced in patients or in preclinical models of several relevant pathological situations, such as heart failure, cardiac hypertrophy, hypertension, obesity and insulin resistance conditions, or non-alcoholic fatty liver disease (NAFLD), and to contribute to disease progression by a variety of mechanisms related to its multifunctional roles. Therefore, targeting GRK2 by different strategies emerges as a potentially relevant approach to treat cardiovascular disease, obesity, type 2 diabetes, or NAFLD, pathological conditions which are frequently interconnected and present as co-morbidities.
Collapse
Affiliation(s)
- Cristina Murga
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa (UAM-CSIC), Universidad Autónoma de Madrid, Madrid, Spain.,CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain.,Instituto de Investigación Sanitaria La Princesa, Madrid, Spain
| | - Alba C Arcones
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa (UAM-CSIC), Universidad Autónoma de Madrid, Madrid, Spain.,CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain.,Instituto de Investigación Sanitaria La Princesa, Madrid, Spain
| | - Marta Cruces-Sande
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa (UAM-CSIC), Universidad Autónoma de Madrid, Madrid, Spain.,CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain.,Instituto de Investigación Sanitaria La Princesa, Madrid, Spain
| | - Ana M Briones
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain.,Departamento de Farmacología, Universidad Autónoma de Madrid (UAM), Madrid, Spain.,Instituto de Investigación Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
| | - Mercedes Salaices
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain.,Departamento de Farmacología, Universidad Autónoma de Madrid (UAM), Madrid, Spain.,Instituto de Investigación Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
| | - Federico Mayor
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa (UAM-CSIC), Universidad Autónoma de Madrid, Madrid, Spain.,CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain.,Instituto de Investigación Sanitaria La Princesa, Madrid, Spain
| |
Collapse
|
33
|
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: 0.8] [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
|
34
|
Wu L, Tai Y, Hu S, Zhang M, Wang R, Zhou W, Tao J, Han Y, Wang Q, Wei W. Bidirectional Role of β2-Adrenergic Receptor in Autoimmune Diseases. Front Pharmacol 2018; 9:1313. [PMID: 30538630 PMCID: PMC6277539 DOI: 10.3389/fphar.2018.01313] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 10/29/2018] [Indexed: 12/20/2022] Open
Abstract
Disorder of the sympathetic nervous system (SNS) is closely related to the pathogenesis of various autoimmune diseases (ADs). Catecholamine triggered beta2-adrenergic receptor (β2-AR) signaling is important in creating a bidirectional response in the progression of ADs due to factors including diverse expression patterns, single nucleotide polymorphisms (SNPs), biased signals, and desensitization of β2-AR, as well as different subtypes of Gα binding to β2-AR. In this review, we summarize the actions of β2-AR signaling in regulating the functions of immunocytes and in the pathogenesis of ADs, and the application of β2-AR agonists or antagonists in treating major types of ADs is also discussed. We suggest that restoring the immune balance via a soft regulation of the expression or activation of β2-AR is one of the promising therapeutic strategies for systematic ADs.
Collapse
Affiliation(s)
- Li Wu
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Yu Tai
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Shanshan Hu
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Mei Zhang
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Rui Wang
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Weijie Zhou
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Juan Tao
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Yongsheng Han
- Department of Emergency Medicine, The First Affiliated Hospital, University of Science and Technology of China, Hefei, China
| | - Qingtong Wang
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Wei Wei
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| |
Collapse
|
35
|
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: 147] [Impact Index Per Article: 21.0] [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
|
36
|
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
|
37
|
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: 199] [Impact Index Per Article: 28.4] [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
|
38
|
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
|
39
|
G protein-coupled receptor kinase 2 (GRK2) as an integrative signalling node in the regulation of cardiovascular function and metabolic homeostasis. Cell Signal 2018; 41:25-32. [DOI: 10.1016/j.cellsig.2017.04.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 03/22/2017] [Accepted: 04/03/2017] [Indexed: 12/23/2022]
|
40
|
Steinhorn B, Sartoretto JL, Sorrentino A, Romero N, Kalwa H, Abel ED, Michel T. Insulin-dependent metabolic and inotropic responses in the heart are modulated by hydrogen peroxide from NADPH-oxidase isoforms NOX2 and NOX4. Free Radic Biol Med 2017; 113:16-25. [PMID: 28917508 PMCID: PMC5699944 DOI: 10.1016/j.freeradbiomed.2017.09.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 08/22/2017] [Accepted: 09/09/2017] [Indexed: 01/08/2023]
Abstract
RATIONALE Hydrogen peroxide (H2O2) is a stable reactive oxygen species (ROS) that has long been implicated in insulin signal transduction in adipocytes. However, H2O2's role in mediating insulin's effects on the heart are unknown. OBJECTIVE We investigated the role of H2O2 in activating insulin-dependent changes in cardiac myocyte metabolic and inotropic pathways. The sources of insulin-dependent H2O2 generation were also studied. METHODS AND RESULTS In addition to the canonical role of insulin in modulating cardiac metabolic pathways, we found that insulin also inhibited beta adrenergic-induced increases in cardiac contractility. Catalase and NADPH oxidase (NOX) inhibitors blunted activation of insulin-responsive kinases Akt and mTOR and attenuated beta adrenergic receptor-mediated responses. These insulin responses were lost in a mouse model of type 2 diabetes, suggesting a role for these H2O2-dependent pathways in the diabetic heart. The H2O2-sensitive fluorescent biosensor HyPer revealed rapid increases in cytosolic and caveolar H2O2 concentrations in response to insulin treatment, which were blocked by NOX inhibitors and attenuated in NOX2 KO and NOX4 KO mice. In NOX2 KO cardiac myocytes, insulin-mediated phosphorylation of Akt and mTOR was blocked, while these responses were unaffected in cardiac myocytes from NOX4 KO mice. In contrast, insulin's effects on contractility were lost in cardiac myocytes from NOX4 KO animals but were retained in NOX2 KO mice. CONCLUSIONS These studies identify a proximal point of bifurcation in cardiac insulin signaling through the simultaneous activation of both NOX2 and NOX4. Each NOX isoform generates H2O2 in cardiac myocytes with distinct time courses, with H2O2 derived from NOX2 augmenting Akt-dependent metabolic effects of insulin, while H2O2 from NOX4 blocks beta adrenergic increases in inotropy. These findings suggest that insulin resistance in the diabetic heart may lead to potentially deleterious potentiation of beta adrenergic responses.
Collapse
Affiliation(s)
- Benjamin Steinhorn
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, United States
| | - Juliano L Sartoretto
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, United States
| | - Andrea Sorrentino
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, United States
| | - Natalia Romero
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, United States
| | - Hermann Kalwa
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, United States
| | - E Dale Abel
- University of Iowa School of Medicine, United States
| | - Thomas Michel
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, United States.
| |
Collapse
|
41
|
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: 0.9] [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
|
42
|
Jing L, Pulenthiran A, Nevius CD, Mejia-Spiegeler A, Suever JD, Wehner GJ, Kirchner HL, Haggerty CM, Fornwalt BK. Impaired right ventricular contractile function in childhood obesity and its association with right and left ventricular changes: a cine DENSE cardiac magnetic resonance study. J Cardiovasc Magn Reson 2017; 19:49. [PMID: 28659144 PMCID: PMC5490166 DOI: 10.1186/s12968-017-0363-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 05/17/2017] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Pediatric obesity is a growing public health problem, which is associated with increased risk of cardiovascular disease and premature death. Left ventricular (LV) remodeling (increased myocardial mass and thickness) and contractile dysfunction (impaired longitudinal strain) have been documented in obese children, but little attention has been paid to the right ventricle (RV). We hypothesized that obese/overweight children would have evidence of RV remodeling and contractile dysfunction. METHODS One hundred and three children, ages 8-18 years, were prospectively recruited and underwent cardiovascular magnetic resonance (CMR), including both standard cine imaging and displacement encoding with stimulated echoes (DENSE) imaging, which allowed for quantification of RV geometry and function/mechanics. RV free wall longitudinal strain was quantified from the end-systolic four-chamber DENSE image. Linear regression was used to quantify correlations of RV strain with LV strain and measurements of body composition (adjusted for sex and height). Analysis of variance was used to study the relationship between RV strain and LV remodeling types (concentric remodeling, eccentric/concentric hypertrophy). RESULTS The RV was sufficiently visualized with DENSE in 70 (68%) subjects, comprising 36 healthy weight (13.6 ± 2.7 years) and 34 (12.1 ± 2.9 years) obese/overweight children. Obese/overweight children had a 22% larger RV mass index (8.2 ± 0.9 vs 6.7 ± 1.1 g/m2.7, p < 0.001) compared to healthy controls. RV free wall longitudinal strain was impaired in obese/overweight children (-16 ± 4% vs -19 ± 5%, p = 0.02). Ten (14%) out of 70 children had LV concentric hypertrophy, and these children had the most impaired RV longitudinal strain compared to those with normal LV geometry (-13 ± 4% vs -19 ± 5%, p = 0.002). RV longitudinal strain was correlated with LV longitudinal strain (r = 0.34, p = 0.004), systolic blood pressure (r = 0.33, p = 0.006), as well as BMI z-score (r = 0.28, p = 0.02), waist (r = 0.31, p = 0.01), hip (r = 0.40, p = 0.004) and abdominal (r = 0.38, p = 0.002) circumference, height and sex adjusted. CONCLUSIONS Obese/overweight children have evidence of RV remodeling (increased RV mass) and RV contractile dysfunction (impaired free wall longitudinal strain). Moreover, RV longitudinal strain correlates with LV longitudinal strain, and children with LV concentric hypertrophy show the most impaired RV function. These results suggest there may be a common mechanism underlying both remodeling and dysfunction of the left and right ventricles in obese/overweight children.
Collapse
MESH Headings
- Adolescent
- Child
- Female
- Humans
- Hypertrophy, Left Ventricular/diagnostic imaging
- Hypertrophy, Left Ventricular/etiology
- Hypertrophy, Left Ventricular/physiopathology
- Image Interpretation, Computer-Assisted
- Kentucky
- Linear Models
- Magnetic Resonance Imaging, Cine
- Male
- Myocardial Contraction
- Observer Variation
- Pediatric Obesity/complications
- Pediatric Obesity/diagnosis
- Pediatric Obesity/physiopathology
- Pennsylvania
- Predictive Value of Tests
- Prospective Studies
- Reproducibility of Results
- Ventricular Dysfunction, Left/diagnostic imaging
- Ventricular Dysfunction, Left/etiology
- Ventricular Dysfunction, Left/physiopathology
- Ventricular Dysfunction, Right/diagnostic imaging
- Ventricular Dysfunction, Right/etiology
- Ventricular Dysfunction, Right/physiopathology
- Ventricular Function, Left
- Ventricular Function, Right
- Ventricular Remodeling
Collapse
Affiliation(s)
- Linyuan Jing
- Department of Imaging Science and Innovation, Geisinger Health System, 100 North Academy Avenue, Danville, 17822-4400 PA USA
- Biomedical and Translational Informatics Institute, Geisinger Health System, Danville, PA USA
| | - Arichanah Pulenthiran
- Department of Imaging Science and Innovation, Geisinger Health System, 100 North Academy Avenue, Danville, 17822-4400 PA USA
- Biomedical and Translational Informatics Institute, Geisinger Health System, Danville, PA USA
| | - Christopher D. Nevius
- Department of Imaging Science and Innovation, Geisinger Health System, 100 North Academy Avenue, Danville, 17822-4400 PA USA
- Biomedical and Translational Informatics Institute, Geisinger Health System, Danville, PA USA
| | - Abba Mejia-Spiegeler
- Department of Imaging Science and Innovation, Geisinger Health System, 100 North Academy Avenue, Danville, 17822-4400 PA USA
- Biomedical and Translational Informatics Institute, Geisinger Health System, Danville, PA USA
| | - Jonathan D. Suever
- Department of Imaging Science and Innovation, Geisinger Health System, 100 North Academy Avenue, Danville, 17822-4400 PA USA
- Biomedical and Translational Informatics Institute, Geisinger Health System, Danville, PA USA
| | - Gregory J. Wehner
- Department of Biomedical Engineering, University of Kentucky, Lexington, KY USA
| | - H. Lester Kirchner
- Biomedical and Translational Informatics Institute, Geisinger Health System, Danville, PA USA
| | - Christopher M. Haggerty
- Department of Imaging Science and Innovation, Geisinger Health System, 100 North Academy Avenue, Danville, 17822-4400 PA USA
- Biomedical and Translational Informatics Institute, Geisinger Health System, Danville, PA USA
| | - Brandon K. Fornwalt
- Department of Imaging Science and Innovation, Geisinger Health System, 100 North Academy Avenue, Danville, 17822-4400 PA USA
- Biomedical and Translational Informatics Institute, Geisinger Health System, Danville, PA USA
- Department of Radiology, Geisinger Health System, Danville, PA USA
| |
Collapse
|
43
|
Catabolic and anabolic faces of insulin resistance and their disorders: a new insight into circadian control of metabolic disorders leading to diabetes. Future Sci OA 2017; 3:FSO201. [PMID: 28884000 PMCID: PMC5583684 DOI: 10.4155/fsoa-2017-0015] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 03/23/2017] [Indexed: 12/18/2022] Open
Abstract
Maintenance of glucose homeostasis during circadian behavioral cycles is critical. The processes controlling the switch between predominant lipolysis/fatty oxidation during fasting and predominant lipid storage/glucose oxidation following feeding are determined principally by insulin. Chronic elevated threshold of insulin resistance (IR) is a key pathological feature of obesity, Type 2 diabetes, sepsis and cancer cachexia; however, a temporal reduced threshold of IR is widely met in fasting/hibernation, pregnancy, antibacterial immunity, exercise and stress. Paradoxically, some of these cases are associated with catabolic metabolism, whereas others are related to anabolic pathways. This article considers the possible causes of circadian disorders in glucose and lipid metabolism that act as a driving force for obesity-promoted development of Type 2 diabetes. This is intended to provide improved insight into the pathogenesis of chronic circadian disorders that increase the risk of diabetes, and consider new targets for its metabolic and drug correction. Insulin resistance (IR) is a common adaptive mechanism, acting under opposite anabolic and catabolic conditions. However, chronic IR is a key pathological feature of obesity, Type 2 diabetes and cancer cachexia, whereas a temporal IR is widely seen in fasting, pregnancy, exercise and stress. Therefore, it is important to understand when this transient IR-mechanism shifts to chronic IR-associated diseases. What factors result in the switch between the anabolic and catabolic conditions and what defect(s) in this switch is associated with chronic IR induction? The present opinion article aimed to address these questions to the metabolic changes typical for circadian regulation in lean, obese and diabetic patients. Graphical abstract: Early circadian IR disorders caused by overweight and obesity are associated with increased risk for diabetes via formation of a vicious cycle between lipid anabolic and catabolic programs thus distorting insulin and lipid levels in day/night period.
Collapse
|
44
|
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: 3.6] [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
|
45
|
Guo CA, Guo S. Insulin receptor substrate signaling controls cardiac energy metabolism and heart failure. J Endocrinol 2017; 233:R131-R143. [PMID: 28381504 PMCID: PMC9675292 DOI: 10.1530/joe-16-0679] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 04/05/2017] [Indexed: 12/11/2022]
Abstract
The heart is an insulin-dependent and energy-consuming organ in which insulin and nutritional signaling integrates to the regulation of cardiac metabolism, growth and survival. Heart failure is highly associated with insulin resistance, and heart failure patients suffer from the cardiac energy deficiency and structural and functional dysfunction. Chronic pathological conditions, such as obesity and type 2 diabetes mellitus, involve various mechanisms in promoting heart failure by remodeling metabolic pathways, modulating cardiac energetics and impairing cardiac contractility. Recent studies demonstrated that insulin receptor substrates 1 and 2 (IRS-1,-2) are major mediators of both insulin and insulin-like growth factor-1 (IGF-1) signaling responsible for myocardial energetics, structure, function and organismal survival. Importantly, the insulin receptor substrates (IRS) play an important role in the activation of the phosphatidylinositide-3-dependent kinase (PI-3K) that controls Akt and Foxo1 signaling cascade, regulating the mitochondrial function, cardiac energy metabolism and the renin-angiotensin system. Dysregulation of this branch in signaling cascades by insulin resistance in the heart through the endocrine system promotes heart failure, providing a novel mechanism for diabetic cardiomyopathy. Therefore, targeting this branch of IRS→PI-3K→Foxo1 signaling cascade and associated pathways may provide a fundamental strategy for the therapeutic and nutritional development in control of metabolic and cardiovascular diseases. In this review, we focus on insulin signaling and resistance in the heart and the role energetics play in cardiac metabolism, structure and function.
Collapse
Affiliation(s)
- Cathy A Guo
- Department of Nutrition and Food ScienceCollege of Agriculture and Life Sciences, Texas A&M University, College Station, Texas, USA
| | - Shaodong Guo
- Department of Nutrition and Food ScienceCollege of Agriculture and Life Sciences, Texas A&M University, College Station, Texas, USA
| |
Collapse
|
46
|
Shi Q, Li M, Mika D, Fu Q, Kim S, Phan J, Shen A, Vandecasteele G, Xiang YK. Heterologous desensitization of cardiac β-adrenergic signal via hormone-induced βAR/arrestin/PDE4 complexes. Cardiovasc Res 2017; 113:656-670. [PMID: 28339772 PMCID: PMC5852637 DOI: 10.1093/cvr/cvx036] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 10/20/2017] [Accepted: 02/17/2017] [Indexed: 12/22/2022] Open
Abstract
AIMS Cardiac β-adrenergic receptor (βAR) signalling is susceptible to heterologous desensitization by different neurohormonal stimuli in clinical conditions associated with heart failure. We aim to examine the underlying mechanism of cross talk between βARs and a set of G-protein coupled receptors (GPCRs) activated by hormones/agonists. METHODS AND RESULTS Rat ventricular cardiomyocytes were used to determine heterologous phosphorylation of βARs under a series of GPCR agonists. Activation of Gs-coupled dopamine receptor, adenosine receptor, relaxin receptor and prostaglandin E2 receptor, and Gq-coupled α1 adrenergic receptor and angiotensin II type 1 receptor promotes phosphorylation of β1AR and β2AR at putative protein kinase A (PKA) phosphorylation sites; but activation of Gi-coupled α2 adrenergic receptor and activation of protease-activated receptor does not. The GPCR agonists that promote β2AR phosphorylation effectively inhibit βAR agonist isoproterenol-induced PKA phosphorylation of phospholamban and contractile function in ventricular cardiomyocytes. Heterologous GPCR stimuli have minimal to small effect on isoproterenol-induced β2AR activation and G-protein coupling for cyclic adenosine monophosphate (cAMP) production. However, these GPCR stimuli significantly promote phosphorylation of phosphodiesterase 4D (PDE4D), and recruit PDE4D to the phosphorylated β2AR in a β-arrestin 2 dependent manner without promoting β2AR endocytosis. The increased binding between β2AR and PDE4D effectively hydrolyzes cAMP signal generated by subsequent stimulation with isoproterenol. Mutation of PKA phosphorylation sites in β2AR, inhibition of PDE4, or genetic ablation of PDE4D or β-arrestin 2 abolishes this heterologous inhibitory effect. Ablation of β-arrestin 2 or PDE4D gene also rescues β-adrenergic stimuli-induced myocyte contractile function. CONCLUSIONS These data reveal essential roles of β-arrestin 2 and PDE4D in a common mechanism for heterologous desensitization of cardiac βARs under hormonal stimulation, which is associated with impaired cardiac function during the development of pathophysiological conditions.
Collapse
MESH Headings
- Animals
- Cells, Cultured
- Cyclic AMP-Dependent Protein Kinases/metabolism
- Cyclic Nucleotide Phosphodiesterases, Type 4/genetics
- Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism
- Hormones/pharmacology
- Male
- Mice, Knockout
- Myocardial Contraction/drug effects
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Phosphorylation
- Protein Kinase C/metabolism
- Rats
- Receptor Cross-Talk
- Receptors, Adrenergic, beta-1/drug effects
- Receptors, Adrenergic, beta-1/genetics
- Receptors, Adrenergic, beta-1/metabolism
- Receptors, Adrenergic, beta-2/drug effects
- Receptors, Adrenergic, beta-2/genetics
- Receptors, Adrenergic, beta-2/metabolism
- Signal Transduction/drug effects
- Time Factors
- beta-Arrestin 1/genetics
- beta-Arrestin 1/metabolism
- beta-Arrestin 2/genetics
- beta-Arrestin 2/metabolism
Collapse
Affiliation(s)
- Qian Shi
- Department of Pharmacology, University of California at Davis, Davis, CA 95616, USA
| | - Minghui Li
- Department of Pharmacology, University of California at Davis, Davis, CA 95616, USA
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210001, China
| | - Delphine Mika
- INSERM UMR-S 1180, Univ. Paris-Sud, Université Paris-Saclay, Châtenay-Malabry, France
| | - Qin Fu
- Department of Pharmacology, Tongji Medical College, Huazhong University of Technology and Sciences, Wuhan 430030, China
| | - Sungjin Kim
- Department of Pharmacology, University of California at Davis, Davis, CA 95616, USA
| | - Jason Phan
- Department of Pharmacology, University of California at Davis, Davis, CA 95616, USA
| | - Ao Shen
- Department of Pharmacology, University of California at Davis, Davis, CA 95616, USA
| | | | - Yang K. Xiang
- Department of Pharmacology, University of California at Davis, Davis, CA 95616, USA
- VA Northern California Health care system, Mather, CA 95655, USA
| |
Collapse
|
47
|
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: 0.8] [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
|
48
|
Bologna Z, Teoh JP, Bayoumi AS, Tang Y, Kim IM. Biased G Protein-Coupled Receptor Signaling: New Player in Modulating Physiology and Pathology. Biomol Ther (Seoul) 2017; 25:12-25. [PMID: 28035079 PMCID: PMC5207460 DOI: 10.4062/biomolther.2016.165] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2016] [Revised: 08/19/2016] [Accepted: 08/23/2016] [Indexed: 01/03/2023] Open
Abstract
G protein-coupled receptors (GPCRs) are a family of cell-surface proteins that play critical roles in regulating a variety of pathophysiological processes and thus are targeted by almost a third of currently available therapeutics. It was originally thought that GPCRs convert extracellular stimuli into intracellular signals through activating G proteins, whereas β-arrestins have important roles in internalization and desensitization of the receptor. Over the past decade, several novel functional aspects of β-arrestins in regulating GPCR signaling have been discovered. These previously unanticipated roles of β-arrestins to act as signal transducers and mediators of G protein-independent signaling have led to the concept of biased agonism. Biased GPCR ligands are able to engage with their target receptors in a manner that preferentially activates only G protein- or β-arrestin-mediated downstream signaling. This offers the potential for next generation drugs with high selectivity to therapeutically relevant GPCR signaling pathways. In this review, we provide a summary of the recent studies highlighting G protein- or β-arrestin-biased GPCR signaling and the effects of biased ligands on disease pathogenesis and regulation.
Collapse
Affiliation(s)
- Zuzana Bologna
- Vascular Biology Center, Medical College of Georgia, Augusta University, GA 30912, USA
| | - Jian-Peng Teoh
- Vascular Biology Center, Medical College of Georgia, Augusta University, GA 30912, USA
| | - Ahmed S Bayoumi
- Vascular Biology Center, Medical College of Georgia, Augusta University, GA 30912, USA
| | - Yaoliang Tang
- Vascular Biology Center, Medical College of Georgia, Augusta University, GA 30912, USA
| | - Il-Man Kim
- Vascular Biology Center, Medical College of Georgia, Augusta University, GA 30912, USA.,Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, GA 30912, USA
| |
Collapse
|
49
|
Llano-Diez M, Sinclair J, Yamada T, Zong M, Fauconnier J, Zhang SJ, Katz A, Jardemark K, Westerblad H, Andersson DC, Lanner JT. The Role of Reactive Oxygen Species in β-Adrenergic Signaling in Cardiomyocytes from Mice with the Metabolic Syndrome. PLoS One 2016; 11:e0167090. [PMID: 27907040 PMCID: PMC5131978 DOI: 10.1371/journal.pone.0167090] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 11/07/2016] [Indexed: 12/20/2022] Open
Abstract
The metabolic syndrome is associated with prolonged stress and hyperactivity of the sympathetic nervous system and afflicted subjects are prone to develop cardiovascular disease. Under normal conditions, the cardiomyocyte response to acute β-adrenergic stimulation partly depends on increased production of reactive oxygen species (ROS). Here we investigated the interplay between beta-adrenergic signaling, ROS and cardiac contractility using freshly isolated cardiomyocytes and whole hearts from two mouse models with the metabolic syndrome (high-fat diet and ob/ob mice). We hypothesized that cardiomyocytes of mice with the metabolic syndrome would experience excessive ROS levels that trigger cellular dysfunctions. Fluorescent dyes and confocal microscopy were used to assess mitochondrial ROS production, cellular Ca2+ handling and contractile function in freshly isolated adult cardiomyocytes. Immunofluorescence, western blot and enzyme assay were used to study protein biochemistry. Unexpectedly, our results point towards decreased cardiac ROS signaling in a stable, chronic phase of the metabolic syndrome because: β-adrenergic-induced increases in the amplitude of intracellular Ca2+ signals were insensitive to antioxidant treatment; mitochondrial ROS production showed decreased basal rate and smaller response to β-adrenergic stimulation. Moreover, control hearts and hearts with the metabolic syndrome showed similar basal levels of ROS-mediated protein modification, but only control hearts showed increases after β-adrenergic stimulation. In conclusion, in contrast to the situation in control hearts, the cardiomyocyte response to acute β-adrenergic stimulation does not involve increased mitochondrial ROS production in a stable, chronic phase of the metabolic syndrome. This can be seen as a beneficial adaptation to prevent excessive ROS levels.
Collapse
Affiliation(s)
- Monica Llano-Diez
- Karolinska Institutet, Department of Physiology & Pharmacology, Stockholm, Sweden
| | - Jon Sinclair
- Karolinska Institutet, Department of Physiology & Pharmacology, Stockholm, Sweden
| | - Takashi Yamada
- Karolinska Institutet, Department of Physiology & Pharmacology, Stockholm, Sweden
| | - Mei Zong
- Karolinska University Hospital, Rheumatology unit, CMM, Stockholm Sweden
| | - Jeremy Fauconnier
- Karolinska Institutet, Department of Physiology & Pharmacology, Stockholm, Sweden
| | - Shi-Jin Zhang
- Karolinska Institutet, Department of Physiology & Pharmacology, Stockholm, Sweden
| | - Abram Katz
- Karolinska Institutet, Department of Physiology & Pharmacology, Stockholm, Sweden
| | - Kent Jardemark
- Karolinska Institutet, Department of Physiology & Pharmacology, Stockholm, Sweden
| | - Håkan Westerblad
- Karolinska Institutet, Department of Physiology & Pharmacology, Stockholm, Sweden
| | | | - Johanna T. Lanner
- Karolinska Institutet, Department of Physiology & Pharmacology, Stockholm, Sweden
- * E-mail:
| |
Collapse
|
50
|
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: 96] [Impact Index Per Article: 10.7] [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
|