1
|
Lasica R, Asanin M, Vukmirovic J, Maslac L, Savic L, Zdravkovic M, Simeunovic D, Polovina M, Milosevic A, Matic D, Juricic S, Jankovic M, Marinkovic M, Djukanovic L. What Do We Know about Peripartum Cardiomyopathy? Yesterday, Today, Tomorrow. Int J Mol Sci 2024; 25:10559. [PMID: 39408885 PMCID: PMC11477285 DOI: 10.3390/ijms251910559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 09/27/2024] [Accepted: 09/27/2024] [Indexed: 10/20/2024] Open
Abstract
Peripartum cardiomyopathy is a disease that occurs during or after pregnancy and leads to a significant decline in cardiac function in previously healthy women. Peripartum cardiomyopathy has a varying prevalence among women depending on the part of the world where they live, but it is associated with a significant mortality and morbidity in this population. Therefore, timely diagnosis, treatment, and monitoring of this disease from its onset are of utmost importance. Although many risk factors are associated with the occurrence of peripartum cardiomyopathy, such as conditions of life, age of the woman, nutrient deficiencies, or multiple pregnancies, the exact cause of its onset remains unknown. Advances in research on the genetic associations with cardiomyopathies have provided a wealth of data indicating a possible association with peripartum cardiomyopathy, but due to numerous mutations and data inconsistencies, the exact connection remains unclear. Significant insights into the pathophysiological mechanisms underlying peripartum cardiomyopathy have been provided by the theory of an abnormal 16-kDa prolactin, which may be generated in an oxidative stress environment and lead to vascular and consequently myocardial damage. Recent studies supporting this disease mechanism also include research on the efficacy of bromocriptine (a prolactin synthesis inhibitor) in restoring cardiac function in affected patients. Despite significant progress in the research of this disease, there are still insufficient data on the safety of use of certain drugs treating heart failure during pregnancy and breastfeeding. Considering the metabolic changes that occur in different stages of pregnancy and the postpartum period, determining the correct dosing regimen of medications is of utmost importance not only for better treatment and survival of mothers but also for reducing the risk of toxic effects on the fetus.
Collapse
Affiliation(s)
- Ratko Lasica
- Department of Cardiology, Emergency Center, University Clinical Center of Serbia, 11000 Belgrade, Serbia;
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (M.A.); (L.S.); (M.Z.); (D.S.); (M.P.); (A.M.); (D.M.); (M.M.)
| | - Milika Asanin
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (M.A.); (L.S.); (M.Z.); (D.S.); (M.P.); (A.M.); (D.M.); (M.M.)
- Department of Cardiology, University Clinical Center of Serbia, 11000 Belgrade, Serbia; (L.M.); (S.J.); (M.J.)
| | - Jovanka Vukmirovic
- Faculty of Organizational Sciences, University of Belgrade, 11000 Belgrade, Serbia;
| | - Lidija Maslac
- Department of Cardiology, University Clinical Center of Serbia, 11000 Belgrade, Serbia; (L.M.); (S.J.); (M.J.)
| | - Lidija Savic
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (M.A.); (L.S.); (M.Z.); (D.S.); (M.P.); (A.M.); (D.M.); (M.M.)
- Department of Cardiology, University Clinical Center of Serbia, 11000 Belgrade, Serbia; (L.M.); (S.J.); (M.J.)
| | - Marija Zdravkovic
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (M.A.); (L.S.); (M.Z.); (D.S.); (M.P.); (A.M.); (D.M.); (M.M.)
- Clinical Center Bezanijska Kosa, 11000 Belgrade, Serbia
| | - Dejan Simeunovic
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (M.A.); (L.S.); (M.Z.); (D.S.); (M.P.); (A.M.); (D.M.); (M.M.)
- Department of Cardiology, University Clinical Center of Serbia, 11000 Belgrade, Serbia; (L.M.); (S.J.); (M.J.)
| | - Marija Polovina
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (M.A.); (L.S.); (M.Z.); (D.S.); (M.P.); (A.M.); (D.M.); (M.M.)
- Department of Cardiology, University Clinical Center of Serbia, 11000 Belgrade, Serbia; (L.M.); (S.J.); (M.J.)
| | - Aleksandra Milosevic
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (M.A.); (L.S.); (M.Z.); (D.S.); (M.P.); (A.M.); (D.M.); (M.M.)
- Department of Cardiology, University Clinical Center of Serbia, 11000 Belgrade, Serbia; (L.M.); (S.J.); (M.J.)
| | - Dragan Matic
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (M.A.); (L.S.); (M.Z.); (D.S.); (M.P.); (A.M.); (D.M.); (M.M.)
- Department of Cardiology, University Clinical Center of Serbia, 11000 Belgrade, Serbia; (L.M.); (S.J.); (M.J.)
| | - Stefan Juricic
- Department of Cardiology, University Clinical Center of Serbia, 11000 Belgrade, Serbia; (L.M.); (S.J.); (M.J.)
| | - Milica Jankovic
- Department of Cardiology, University Clinical Center of Serbia, 11000 Belgrade, Serbia; (L.M.); (S.J.); (M.J.)
| | - Milan Marinkovic
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (M.A.); (L.S.); (M.Z.); (D.S.); (M.P.); (A.M.); (D.M.); (M.M.)
- Department of Cardiology, University Clinical Center of Serbia, 11000 Belgrade, Serbia; (L.M.); (S.J.); (M.J.)
| | - Lazar Djukanovic
- Department of Cardiology, University Clinical Center of Serbia, 11000 Belgrade, Serbia; (L.M.); (S.J.); (M.J.)
| |
Collapse
|
2
|
Huber T, Horioka-Duplix M, Chen Y, Saca VR, Ceraudo E, Chen Y, Sakmar TP. The role of signaling pathways mediated by the GPCRs CysLTR1/2 in melanocyte proliferation and senescence. Sci Signal 2024; 17:eadp3967. [PMID: 39288219 DOI: 10.1126/scisignal.adp3967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 08/15/2024] [Indexed: 09/19/2024]
Abstract
In contrast with sun exposure-induced melanoma, rarer melanocytic tumors and neoplasms with low mutational burden present opportunities to study isolated signaling mechanisms. These include uveal melanoma and blue nevi, which are often driven by mutations within the G protein-coupled signaling cascade downstream of cysteinyl leukotriene receptor 2. Here, we review how the same mutations within this pathway drive the growth of melanocytes in one tissue but can inhibit the growth of those in another, exemplifying the role of the tissue environment in the delicate balance between uncontrolled cell growth and senescence.
Collapse
Affiliation(s)
- Thomas Huber
- Laboratory of Chemical Biology and Signal Transduction, Rockefeller University, New York, NY 10065, USA
| | - Mizuho Horioka-Duplix
- Laboratory of Chemical Biology and Signal Transduction, Rockefeller University, New York, NY 10065, USA
- Tri-Institutional PhD Program in Chemical Biology, New York, NY 10065, USA
| | - Yuanhuang Chen
- Laboratory of Chemical Biology and Signal Transduction, Rockefeller University, New York, NY 10065, USA
- Tri-Institutional PhD Program in Chemical Biology, New York, NY 10065, USA
| | - Victoria R Saca
- Laboratory of Chemical Biology and Signal Transduction, Rockefeller University, New York, NY 10065, USA
- Tri-Institutional PhD Program in Chemical Biology, New York, NY 10065, USA
| | - Emilie Ceraudo
- Laboratory of Chemical Biology and Signal Transduction, Rockefeller University, New York, NY 10065, USA
| | - Yu Chen
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Thomas P Sakmar
- Laboratory of Chemical Biology and Signal Transduction, Rockefeller University, New York, NY 10065, USA
| |
Collapse
|
3
|
Li Q, Ding J, Xia B, Liu K, Zheng K, Wu J, Huang C, Yuan X, You Q. L-theanine alleviates myocardial ischemia/reperfusion injury by suppressing oxidative stress and apoptosis through activation of the JAK2/STAT3 pathway in mice. Mol Med 2024; 30:98. [PMID: 38943069 PMCID: PMC11214244 DOI: 10.1186/s10020-024-00865-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Accepted: 06/15/2024] [Indexed: 07/01/2024] Open
Abstract
BACKGROUND L-theanine is a unique non-protein amino acid in tea that is widely used as a safe food additive. We investigated the cardioprotective effects and mechanisms of L-theanine in myocardial ischemia-reperfusion injury (MIRI). METHODS The cardioprotective effects and mechanisms of L-theanine and the role of Janus Kinase 2 (JAK2)/signal transducer and activator of transcription 3 (STAT3) signaling were investigated in MIRI mice using measures of cardiac function, oxidative stress, and apoptosis. RESULTS Administration of L-theanine (10 mg/kg, once daily) suppressed the MIRI-induced increase in infarct size and serum creatine kinase and lactate dehydrogenase levels, as well as MIRI-induced cardiac apoptosis, as evidenced by an increase in Bcl-2 expression and a decrease in Bax/caspase-3 expression. Administration of L-theanine also decreased the levels of parameters reflecting oxidative stress, such as dihydroethidium, malondialdehyde, and nitric oxide, and increased the levels of parameters reflecting anti-oxidation, such as total antioxidant capacity (T-AOC), glutathione (GSH), and superoxide dismutase (SOD) in ischemic heart tissue. Further analysis showed that L-theanine administration suppressed the MIRI-induced decrease of phospho-JAK2 and phospho-STAT3 in ischemic heart tissue. Inhibition of JAK2 by AG490 (5 mg/kg, once daily) abolished the cardioprotective effect of L-theanine, suggesting that the JAK2/STAT3 signaling pathway may play an essential role in mediating the anti-I/R effect of L-theanine. CONCLUSIONS L-theanine administration suppresses cellular apoptosis and oxidative stress in part via the JAK2/STAT3 signaling pathway, thereby attenuating MIRI-induced cardiac injury. L-theanine could be developed as a potential drug to alleviate cardiac damage in MIRI.
Collapse
Affiliation(s)
- Qi Li
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University, Nantong University, #20 Xishi Road, Nantong, 226001, Jiangsu, China
| | - Jiaqi Ding
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University, Nantong University, #20 Xishi Road, Nantong, 226001, Jiangsu, China
| | - Boyu Xia
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University, Nantong University, #20 Xishi Road, Nantong, 226001, Jiangsu, China
| | - Kun Liu
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University, Nantong University, #20 Xishi Road, Nantong, 226001, Jiangsu, China
| | - Koulong Zheng
- Department of Cardiology, The Second Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Jingjing Wu
- Department of Cardiology, Suzhou Kowloon Hospital of Shanghai Jiaotong University School of Medicine, Suzhou, Jiangsu, China
| | - Chao Huang
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Xiaomei Yuan
- Department of Cardiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China.
| | - Qingsheng You
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University, Nantong University, #20 Xishi Road, Nantong, 226001, Jiangsu, China.
| |
Collapse
|
4
|
Shi J, Wei L. ROCK1 deficiency preserves caveolar compartmentalization of signaling molecules and cell membrane integrity. FASEB Bioadv 2024; 6:85-102. [PMID: 38463696 PMCID: PMC10918988 DOI: 10.1096/fba.2024-00015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 02/05/2024] [Accepted: 02/08/2024] [Indexed: 03/12/2024] Open
Abstract
In this study, we investigated the roles of ROCK1 in regulating structural and functional features of caveolae located at the cell membrane of cardiomyocytes, adipocytes, and mouse embryonic fibroblasts (MEFs) as well as related physiopathological effects. Caveolae are small bulb-shaped cell membrane invaginations, and their roles have been associated with disease conditions. One of the unique features of caveolae is that they are physically linked to the actin cytoskeleton that is well known to be regulated by RhoA/ROCKs pathway. In cardiomyocytes, we observed that ROCK1 deficiency is coincident with an increased caveolar density, clusters, and caveolar proteins including caveolin-1 and -3. In the mouse cardiomyopathy model with transgenic overexpressing Gαq in myocardium, we demonstrated the reduced caveolar density at cell membrane and reduced caveolar protein contents. Interestingly, coexisting ROCK1 deficiency in cardiomyocytes can rescue these defects and preserve caveolar compartmentalization of β-adrenergic signaling molecules including β1-adrenergic receptor and type V/VI adenylyl cyclase. In cardiomyocytes and adipocytes, we detected that ROCK1 deficiency increased insulin signaling with increased insulin receptor activation in caveolae. In MEFs, we identified that ROCK1 deficiency increased caveolar and total levels of caveolin-1 and cell membrane repair ability after mechanical or chemical disruptions. Together, these results demonstrate that ROCK1 can regulate caveolae plasticity and multiple functions including compartmentalization of signaling molecules and cell membrane repair following membrane disruption by mechanical force and oxidative damage. These findings provide possible molecular insights into the beneficial effects of ROCK1 deletion/inhibition in cardiomyocytes, adipocytes, and MEFs under certain diseased conditions.
Collapse
Affiliation(s)
- Jianjian Shi
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, School of MedicineIndiana UniversityIndianapolisIndianaUSA
| | - Lei Wei
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, School of MedicineIndiana UniversityIndianapolisIndianaUSA
| |
Collapse
|
5
|
Grogan A, Lucero EY, Jiang H, Rockman HA. Pathophysiology and pharmacology of G protein-coupled receptors in the heart. Cardiovasc Res 2023; 119:1117-1129. [PMID: 36534965 PMCID: PMC10202650 DOI: 10.1093/cvr/cvac171] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/30/2022] [Accepted: 10/06/2022] [Indexed: 08/10/2023] Open
Abstract
G protein-coupled receptors (GPCRs), comprising the largest superfamily of cell surface receptors, serve as fundamental modulators of cardiac health and disease owing to their key roles in the regulation of heart rate, contractile dynamics, and cardiac function. Accordingly, GPCRs are heavily pursued as drug targets for a wide variety of cardiovascular diseases ranging from heart failure, cardiomyopathy, and arrhythmia to hypertension and coronary artery disease. Recent advancements in understanding the signalling mechanisms, regulation, and pharmacological properties of GPCRs have provided valuable insights that will guide the development of novel therapeutics. Herein, we review the cellular signalling mechanisms, pathophysiological roles, and pharmacological developments of the major GPCRs in the heart, highlighting the β-adrenergic, muscarinic, and angiotensin receptors as exemplar subfamilies.
Collapse
Affiliation(s)
- Alyssa Grogan
- Department of Medicine, Duke University Medical Center, DUMC 3104, 226 CARL Building, Durham, NC 27710, USA
| | - Emilio Y Lucero
- Department of Medicine, Duke University Medical Center, DUMC 3104, 226 CARL Building, Durham, NC 27710, USA
| | - Haoran Jiang
- Department of Medicine, Duke University Medical Center, DUMC 3104, 226 CARL Building, Durham, NC 27710, USA
| | - Howard A Rockman
- Department of Medicine, Duke University Medical Center, DUMC 3104, 226 CARL Building, Durham, NC 27710, USA
- Cell Biology, Duke University Medical Center, DUMC 3104, 226 CARL Building, 12 Durham, NC 27710, USA
| |
Collapse
|
6
|
Ng ML, Ang X, Yap KY, Ng JJ, Goh ECH, Khoo BBJ, Richards AM, Drum CL. Novel Oxidative Stress Biomarkers with Risk Prognosis Values in Heart Failure. Biomedicines 2023; 11:biomedicines11030917. [PMID: 36979896 PMCID: PMC10046491 DOI: 10.3390/biomedicines11030917] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/17/2023] [Accepted: 02/26/2023] [Indexed: 03/18/2023] Open
Abstract
Oxidative stress (OS) is mediated by reactive oxygen species (ROS), which in cardiovascular and other disease states, damage DNA, lipids, proteins, other cellular and extra-cellular components. OS is both initiated by, and triggers inflammation, cardiomyocyte apoptosis, matrix remodeling, myocardial fibrosis, and neurohumoral activation. These have been linked to the development of heart failure (HF). Circulating biomarkers generated by OS offer potential utility in patient management and therapeutic targeting. Novel OS-related biomarkers such as NADPH oxidases (sNox2-dp, Nrf2), advanced glycation end-products (AGE), and myeloperoxidase (MPO), are signaling molecules reflecting pathobiological changes in HF. This review aims to evaluate current OS-related biomarkers and their associations with clinical outcomes and to highlight those with greatest promise in diagnosis, risk stratification and therapeutic targeting in HF.
Collapse
Affiliation(s)
- Mei Li Ng
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
| | - Xu Ang
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
| | - Kwan Yi Yap
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
| | - Jun Jie Ng
- Vascular Surgery, Department of Cardiac, Thoracic and Vascular Surgery, National University Heart Centre, Singapore 119074, Singapore
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
| | - Eugene Chen Howe Goh
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
| | - Benjamin Bing Jie Khoo
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
| | - Arthur Mark Richards
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
- Cardiovascular Research Institute, Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, NUHS Tower Block, Level 9, NUHCS, Singapore 119228, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
| | - Chester Lee Drum
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
- Cardiovascular Research Institute, Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, NUHS Tower Block, Level 9, NUHCS, Singapore 119228, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Correspondence:
| |
Collapse
|
7
|
Altieri DI, Etzion Y, Anderson HD. Cannabinoid receptor agonist attenuates angiotensin II-induced enlargement and mitochondrial dysfunction in rat atrial cardiomyocytes. Front Pharmacol 2023; 14:1142583. [PMID: 37113758 PMCID: PMC10126395 DOI: 10.3389/fphar.2023.1142583] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 03/13/2023] [Indexed: 04/29/2023] Open
Abstract
Pathological remodeling of atrial tissue renders the atria more prone to arrhythmia upon arrival of electrical triggers. Activation of the renin-angiotensin system is an important factor that contributes to atrial remodeling, which may result in atrial hypertrophy and prolongation of P-wave duration. In addition, atrial cardiomyocytes are electrically coupled via gap junctions, and electrical remodeling of connexins may result in dysfunction of coordinated wave propagation within the atria. Currently, there is a lack of effective therapeutic strategies that target atrial remodeling. We previously proposed that cannabinoid receptors (CBR) may have cardioprotective qualities. CB13 is a dual cannabinoid receptor agonist that activates AMPK signaling in ventricular cardiomyocytes. We reported that CB13 attenuates tachypacing-induced shortening of atrial refractoriness and inhibition of AMPK signaling in the rat atria. Here, we evaluated the effects of CB13 on neonatal atrial rat cardiomyocytes (NRAM) stimulated by angiotensin II (AngII) in terms of atrial myocyte enlargement and mitochondrial function. CB13 inhibited AngII-induced enhancement of atrial myocyte surface area in an AMPK-dependent manner. CB13 also inhibited mitochondrial membrane potential deterioration in the same context. However, AngII and CB13 did not affect mitochondrial permeability transition pore opening. We further demonstrate that CB13 increased Cx43 compared to AngII-treated neonatal rat atrial myocytes. Overall, our results support the notion that CBR activation promotes atrial AMPK activation, and prevents myocyte enlargement (an indicator that suggests pathological hypertrophy), mitochondrial depolarization and Cx43 destabilization. Therefore, peripheral CBR activation should be further tested as a novel treatment strategy in the context of atrial remodeling.
Collapse
Affiliation(s)
- Danielle I. Altieri
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
- Canadian Centre for Agri-Food Research in Health and Medicine (CCARM), Albrechtsen Research Centre, St Boniface Hospital, Winnipeg, MB, Canada
| | - Yoram Etzion
- Cardiac Arrhythmia Research Laboratory, Department of Physiology and Cell Biology, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- Regenerative Medicine and Stem Cell Research Center, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Hope D. Anderson
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
- Canadian Centre for Agri-Food Research in Health and Medicine (CCARM), Albrechtsen Research Centre, St Boniface Hospital, Winnipeg, MB, Canada
- *Correspondence: Hope D. Anderson,
| |
Collapse
|
8
|
Khalilimeybodi A, Riaz M, Campbell SG, Omens JH, McCulloch AD, Qyang Y, Saucerman JJ. Signaling network model of cardiomyocyte morphological changes in familial cardiomyopathy. J Mol Cell Cardiol 2023; 174:1-14. [PMID: 36370475 PMCID: PMC10230857 DOI: 10.1016/j.yjmcc.2022.10.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 08/26/2022] [Accepted: 10/20/2022] [Indexed: 11/11/2022]
Abstract
Familial cardiomyopathy is a precursor of heart failure and sudden cardiac death. Over the past several decades, researchers have discovered numerous gene mutations primarily in sarcomeric and cytoskeletal proteins causing two different disease phenotypes: hypertrophic (HCM) and dilated (DCM) cardiomyopathies. However, molecular mechanisms linking genotype to phenotype remain unclear. Here, we employ a systems approach by integrating experimental findings from preclinical studies (e.g., murine data) into a cohesive signaling network to scrutinize genotype to phenotype mechanisms. We developed an HCM/DCM signaling network model utilizing a logic-based differential equations approach and evaluated model performance in predicting experimental data from four contexts (HCM, DCM, pressure overload, and volume overload). The model has an overall prediction accuracy of 83.8%, with higher accuracy in the HCM context (90%) than DCM (75%). Global sensitivity analysis identifies key signaling reactions, with calcium-mediated myofilament force development and calcium-calmodulin kinase signaling ranking the highest. A structural revision analysis indicates potential missing interactions that primarily control calcium regulatory proteins, increasing model prediction accuracy. Combination pharmacotherapy analysis suggests that downregulation of signaling components such as calcium, titin and its associated proteins, growth factor receptors, ERK1/2, and PI3K-AKT could inhibit myocyte growth in HCM. In experiments with patient-specific iPSC-derived cardiomyocytes (MLP-W4R;MYH7-R723C iPSC-CMs), combined inhibition of ERK1/2 and PI3K-AKT rescued the HCM phenotype, as predicted by the model. In DCM, PI3K-AKT-NFAT downregulation combined with upregulation of Ras/ERK1/2 or titin or Gq protein could ameliorate cardiomyocyte morphology. The model results suggest that HCM mutations that increase active force through elevated calcium sensitivity could increase ERK activity and decrease eccentricity through parallel growth factors, Gq-mediated, and titin pathways. Moreover, the model simulated the influence of existing medications on cardiac growth in HCM and DCM contexts. This HCM/DCM signaling model demonstrates utility in investigating genotype to phenotype mechanisms in familial cardiomyopathy.
Collapse
Affiliation(s)
- Ali Khalilimeybodi
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, United States of America
| | - Muhammad Riaz
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Stuart G Campbell
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Jeffrey H Omens
- Departments of Bioengineering and Medicine, University of California, San Diego, La Jolla, CA, United States of America
| | - Andrew D McCulloch
- Departments of Bioengineering and Medicine, University of California, San Diego, La Jolla, CA, United States of America
| | - Yibing Qyang
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA; Yale Stem Cell Center, New Haven, CT, United States of America; Department of Pathology, Yale University, New Haven, CT, United States of America; Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, United States of America
| | - Jeffrey J Saucerman
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, United States of America; Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, United States of America.
| |
Collapse
|
9
|
Jiang H, Galtes D, Wang J, Rockman HA. G protein-coupled receptor signaling: transducers and effectors. Am J Physiol Cell Physiol 2022; 323:C731-C748. [PMID: 35816644 PMCID: PMC9448338 DOI: 10.1152/ajpcell.00210.2022] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/27/2022] [Accepted: 07/10/2022] [Indexed: 01/14/2023]
Abstract
G protein-coupled receptors (GPCRs) are of considerable interest due to their importance in a wide range of physiological functions and in a large number of Food and Drug Administration (FDA)-approved drugs as therapeutic entities. With continued study of their function and mechanism of action, there is a greater understanding of how effector molecules interact with a receptor to initiate downstream effector signaling. This review aims to explore the signaling pathways, dynamic structures, and physiological relevance in the cardiovascular system of the three most important GPCR signaling effectors: heterotrimeric G proteins, GPCR kinases (GRKs), and β-arrestins. We will first summarize their prominent roles in GPCR pharmacology before transitioning into less well-explored areas. As new technologies are developed and applied to studying GPCR structure and their downstream effectors, there is increasing appreciation for the elegance of the regulatory mechanisms that mediate intracellular signaling and function.
Collapse
Affiliation(s)
- Haoran Jiang
- Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | - Daniella Galtes
- Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | - Jialu Wang
- Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | - Howard A Rockman
- Department of Medicine, Duke University Medical Center, Durham, North Carolina
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina
| |
Collapse
|
10
|
Sandroni PB, Fisher-Wellman KH, Jensen BC. Adrenergic Receptor Regulation of Mitochondrial Function in Cardiomyocytes. J Cardiovasc Pharmacol 2022; 80:364-377. [PMID: 35170492 PMCID: PMC9365878 DOI: 10.1097/fjc.0000000000001241] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 02/01/2022] [Indexed: 01/31/2023]
Abstract
ABSTRACT Adrenergic receptors (ARs) are G protein-coupled receptors that are stimulated by catecholamines to induce a wide array of physiological effects across tissue types. Both α1- and β-ARs are found on cardiomyocytes and regulate cardiac contractility and hypertrophy through diverse molecular pathways. Acute activation of cardiomyocyte β-ARs increases heart rate and contractility as an adaptive stress response. However, chronic β-AR stimulation contributes to the pathobiology of heart failure. By contrast, mounting evidence suggests that α1-ARs serve protective functions that may mitigate the deleterious effects of chronic β-AR activation. Here, we will review recent studies demonstrating that α1- and β-ARs differentially regulate mitochondrial biogenesis and dynamics, mitochondrial calcium handling, and oxidative phosphorylation in cardiomyocytes. We will identify potential mechanisms of these actions and focus on the implications of these findings for the modulation of contractile function in the uninjured and failing heart. Collectively, we hope to elucidate important physiological processes through which these well-studied and clinically relevant receptors stimulate and fuel cardiac contraction to contribute to myocardial health and disease.
Collapse
Affiliation(s)
- Peyton B. Sandroni
- University of North Carolina School of Medicine, Department of Pharmacology
- University of North Carolina School of Medicine, McAllister Heart Institute
| | - Kelsey H. Fisher-Wellman
- East Carolina University Brody School of Medicine, Department of Physiology
- East Carolina University Diabetes and Obesity Institute
| | - Brian C. Jensen
- University of North Carolina School of Medicine, Department of Pharmacology
- University of North Carolina School of Medicine, McAllister Heart Institute
- University of North Carolina School of Medicine, Department of Medicine, Division of Cardiology
| |
Collapse
|
11
|
Upregulation of Phospholipase C Gene Expression Due to Norepinephrine-Induced Hypertrophic Response. Cells 2022; 11:cells11162488. [PMID: 36010565 PMCID: PMC9406906 DOI: 10.3390/cells11162488] [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: 07/11/2022] [Revised: 08/04/2022] [Accepted: 08/09/2022] [Indexed: 11/28/2022] Open
Abstract
The activation of phospholipase C (PLC) is thought to have a key role in the cardiomyocyte response to several different hypertrophic agents such as norepinephrine, angiotensin II and endothelin-1. PLC activity results in the generation of diacylglycerol and inositol trisphosphate, which are downstream signal transducers for the expression of fetal genes, increased protein synthesis, and subsequent cardiomyocyte growth. In this article, we describe the signal transduction elements that regulate PLC gene expression. The discussion is focused on the norepinephrine- α1-adrenoceptor signaling pathway and downstream signaling processes that mediate an upregulation of PLC isozyme gene expression. Evidence is also indicated to demonstrate that PLC activities self-regulate the expression of PLC isozymes with the suggestion that PLC activities may be part of a coordinated signaling process for the perpetuation of cardiac hypertrophy. Accordingly, from the information provided, it is plausible that specific PLC isozymes could be targeted for the mitigation of cardiac hypertrophy.
Collapse
|
12
|
Tappia PS, Ramjiawan B, Dhalla NS. Role of Phospholipase C in Catecholamine-induced Increase in Myocardial Protein Synthesis. Can J Physiol Pharmacol 2022; 100:945-955. [PMID: 35767883 DOI: 10.1139/cjpp-2022-0189] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The activation of the α1-adrenoceptor-(α1-AR) by norepinephrine results in the G-protein (Gqα) mediated increase in the phosphoinositide-specific phospholipase C (PLC) activity. The byproducts of PLC hydrolytic activity, namely, 1,2-diacylglycerol and inositol-1,4,5-trisphosphate, are important downstream signal transducers for increased protein synthesis in the cardiomyocyte and the subsequent hypertrophic response. In this article, evidence is outlined to demonstrate the role of cardiomyocyte PLC isozymes in the catecholamine-induced increase in protein synthesis by using a blocker of α1-AR and an inhibitor of PLC. The discussion will be focused on the α1-AR-Gqα-PLC-mediated hypertrophic signaling pathway from the viewpoint that it may compliment the other β1-AR-Gs protein-adenylyl cyclase signal transduction mechanisms in the early stages of cardiac hypertrophy development, but may become more relevant at the late stage of cardiac hypertrophy. From the information provided here, it is suggested that some specific PLC isozymes may potentially serve as important targets for the attenuation of cardiac hypertrophy in the vulnerable patient population at-risk for heart failure.
Collapse
Affiliation(s)
- Paramjit S Tappia
- Asper Clinical Research Institute, St. Boniface Hospital, Office of Clinical Research, Winnipeg, Manitoba, Canada;
| | - Bram Ramjiawan
- University of Manitoba, Faculty of Medicine, Winnipeg, Manitoba, Canada;
| | - Naranjan S Dhalla
- St Boniface Hospital Research, 120927, Institute of Cardiovascular Sciences, Albrechtsen Research Centre, Winnipeg, Manitoba, Canada;
| |
Collapse
|
13
|
Dorn Ii GW. Neurohormonal Connections with Mitochondria in Cardiomyopathy and Other Diseases. Am J Physiol Cell Physiol 2022; 323:C461-C477. [PMID: 35759434 PMCID: PMC9363002 DOI: 10.1152/ajpcell.00167.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Neurohormonal signaling and mitochondrial dynamism are seemingly distinct processes that are almost ubiquitous among multicellular organisms. Both of these processes are regulated by GTPases, and disturbances in either can provoke disease. Here, inconspicuous pathophysiological connectivity between neurohormonal signaling and mitochondrial dynamism is reviewed in the context of cardiac and neurological syndromes. For both processes, greater understanding of basic mechanisms has evoked a reversal of conventional pathophysiological concepts. Thus, neurohormonal systems induced in, and previously thought to be critical for, cardiac functioning in heart failure are now pharmaceutically interrupted as modern standard of care. And, mitochondrial abnormalities in neuropathies that were originally attributed to an imbalance between mitochondrial fusion and fission are increasingly recognized as an interruption of axonal mitochondrial transport. The data are presented in a historical context to provided insight into how scientific thought has evolved and to foster an appreciation for how seemingly different areas of investigation can converge. Finally, some theoretical notions are presented to explain how different molecular and functional defects can evoke tissue-specific disease.
Collapse
Affiliation(s)
- Gerald W Dorn Ii
- Center for Pharmacogenomics, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
| |
Collapse
|
14
|
Abstract
Cardiovascular complications of pregnancy have risen substantially over the past decades, and now account for the majority of pregnancy-induced maternal deaths, as well as having substantial long-term consequences on maternal cardiovascular health. The causes and pathophysiology of these complications remain poorly understood, and therapeutic options are limited. Preclinical models represent a crucial tool for understanding human disease. We review here advances made in preclinical models of cardiovascular complications of pregnancy, including preeclampsia and peripartum cardiomyopathy, with a focus on pathological mechanisms elicited by the models and on relevance to human disease.
Collapse
Affiliation(s)
- Zolt Arany
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia (Z.A.)
| | - Denise Hilfiker-Kleiner
- Institute of Cardiovascular Complications in Pregnancy and in Oncologic Therapies, Philipps University Marburg, Germany (D.H.-K.)
| | - S Ananth Karumanchi
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA (S.A.K.)
| |
Collapse
|
15
|
Signaling cascades in the failing heart and emerging therapeutic strategies. Signal Transduct Target Ther 2022; 7:134. [PMID: 35461308 PMCID: PMC9035186 DOI: 10.1038/s41392-022-00972-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 03/13/2022] [Accepted: 03/20/2022] [Indexed: 12/11/2022] Open
Abstract
Chronic heart failure is the end stage of cardiac diseases. With a high prevalence and a high mortality rate worldwide, chronic heart failure is one of the heaviest health-related burdens. In addition to the standard neurohormonal blockade therapy, several medications have been developed for chronic heart failure treatment, but the population-wide improvement in chronic heart failure prognosis over time has been modest, and novel therapies are still needed. Mechanistic discovery and technical innovation are powerful driving forces for therapeutic development. On the one hand, the past decades have witnessed great progress in understanding the mechanism of chronic heart failure. It is now known that chronic heart failure is not only a matter involving cardiomyocytes. Instead, chronic heart failure involves numerous signaling pathways in noncardiomyocytes, including fibroblasts, immune cells, vascular cells, and lymphatic endothelial cells, and crosstalk among these cells. The complex regulatory network includes protein-protein, protein-RNA, and RNA-RNA interactions. These achievements in mechanistic studies provide novel insights for future therapeutic targets. On the other hand, with the development of modern biological techniques, targeting a protein pharmacologically is no longer the sole option for treating chronic heart failure. Gene therapy can directly manipulate the expression level of genes; gene editing techniques provide hope for curing hereditary cardiomyopathy; cell therapy aims to replace dysfunctional cardiomyocytes; and xenotransplantation may solve the problem of donor heart shortages. In this paper, we reviewed these two aspects in the field of failing heart signaling cascades and emerging therapeutic strategies based on modern biological techniques.
Collapse
|
16
|
Wu S, Chen L, Zhou X. Circular RNAs in the regulation of cardiac hypertrophy. MOLECULAR THERAPY. NUCLEIC ACIDS 2022; 27:484-490. [PMID: 35036059 PMCID: PMC8728521 DOI: 10.1016/j.omtn.2021.12.025] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Cardiac hypertrophy is a physiological adaptation to pressure stress that augments or preserves cardiac function. Prolonged hypertrophy can, however, eventually lead to heart failure. Although some effector molecules and signaling pathways have been associated with myocardial hypertrophy, progress has been limited, and further studies are needed to thoroughly explore the underlying mechanisms and to discover novel and effective therapeutic targets. Recently, non-coding RNAs, which are well-known physiological regulators, have attracted much attention in the field of cardiovascular research. Circular RNA, in particular, has emerged as a key player in cardiac hypertrophy, and increasing numbers of papers are now being devoted to this topic. In this review, we will give a brief introduction to circular RNA and then focus on its role as a potential therapeutic target in cardiac hypertrophy.
Collapse
Affiliation(s)
- Siyi Wu
- Department of Cardiology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Lili Chen
- Department of Cardiology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiang Zhou
- Department of Cardiology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
17
|
Kumari R, Ray AG, Mukherjee D, Chander V, Kar D, Kumar US, Bharadwaj P.V.P. D, Banerjee SK, Konar A, Bandyopadhyay A. Downregulation of PTEN Promotes Autophagy via Concurrent Reduction in Apoptosis in Cardiac Hypertrophy in PPAR α−/− Mice. Front Cardiovasc Med 2022; 9:798639. [PMID: 35224041 PMCID: PMC8881053 DOI: 10.3389/fcvm.2022.798639] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 01/14/2022] [Indexed: 01/05/2023] Open
Abstract
Cardiac hypertrophy is characterized by an increase in the size of the cardiomyocytes which is initially triggered as an adaptive response but ultimately becomes maladaptive with chronic exposure to different hypertrophic stimuli. Prolonged cardiac hypertrophy is often associated with mitochondrial dysfunctions and cardiomyocyte cell death. Peroxisome proliferator activated receptor alpha (PPAR α), which is critical for mitochondrial biogenesis and fatty acid oxidation, is down regulated in hypertrophied cardiomyocytes. Yet, the role of PPAR α in cardiomyocyte death is largely unknown. To assess the role of PPAR α in chronic hypertrophy, isoproterenol, a β-adrenergic receptor agonist was administered in PPAR α knock out (PPAR α−/−) mice for 2 weeks and hypertrophy associated changes in cardiac tissues were observed. Echocardiographic analysis ensured the development of cardiac hypertrophy and compromised hemodynamics in PPAR α−/− mice. Proteomic analysis using high resolution mass spectrometer identified about 1,200 proteins enriched in heart tissue. Proteins were classified according to biological pathway and molecular functions. We observed an unexpected down regulation of apoptotic markers, Annexin V and p53 in hypertrophied heart tissue. Further validation revealed a significant down regulation of apoptosis regulator, PTEN, along with other apoptosis markers like p53, Caspase 9 and c-PARP. The autophagy markers Atg3, Atg5, Atg7, p62, Beclin1 and LC3 A/B were up regulated in PPAR α−/− mice indicating an increase in autophagy. Similar observations were made in a high cholesterol diet fed PPAR α−/−mice. The results were further validated in vitro using NRVMs and H9C2 cell line by blocking PPAR α that resulted in enhanced autophagosome formation upon hypertrophic stimulation. The results demonstrate that in the absence of PPAR α apoptotic pathway is inhibited while autophagy is enhanced. The data suggest that PPAR α signaling might act as a molecular switch between apoptosis and autophagy thereby playing a critical role in adaptive process in cardiac hypertrophy.
Collapse
Affiliation(s)
- Ritu Kumari
- Cell Biology and Physiology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Aleepta Guha Ray
- Cell Biology and Physiology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Dibyanti Mukherjee
- Cell Biology and Physiology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Vivek Chander
- Cell Biology and Physiology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Dipak Kar
- Cell Biology and Physiology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Uppulapu Shravan Kumar
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Guwahati, India
| | - Deepak Bharadwaj P.V.P.
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Guwahati, India
| | - Sanjay K. Banerjee
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Guwahati, India
| | - Aditya Konar
- Cell Biology and Physiology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Arun Bandyopadhyay
- Cell Biology and Physiology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
- *Correspondence: Arun Bandyopadhyay ; orcid.org/0000-0002-4885-7033
| |
Collapse
|
18
|
Bourque K, Hawey C, Jiang A, Mazarura GR, Hébert TE. Biosensor-based profiling to track cellular signalling in patient-derived models of dilated cardiomyopathy. Cell Signal 2022; 91:110239. [PMID: 34990783 DOI: 10.1016/j.cellsig.2021.110239] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 12/06/2021] [Accepted: 12/29/2021] [Indexed: 12/18/2022]
Abstract
Dilated cardiomyopathies (DCM) represent a diverse group of cardiovascular diseases impacting the structure and function of the myocardium. To better treat these diseases, we need to understand the impact of such cardiomyopathies on critical signalling pathways that drive disease progression downstream of receptors we often target therapeutically. Our understanding of cellular signalling events has progressed substantially in the last few years, in large part due to the design, validation and use of biosensor-based approaches to studying such events in cells, tissues and in some cases, living animals. Another transformative development has been the use of human induced pluripotent stem cells (hiPSCs) to generate disease-relevant models from individual patients. We highlight the importance of going beyond monocellular cultures to incorporate the influence of paracrine signalling mediators. Finally, we discuss the recent coalition of these approaches in the context of DCM. We discuss recent work in generating patient-derived models of cardiomyopathies and the utility of using signalling biosensors to track disease progression and test potential therapeutic strategies that can be later used to inform treatment options in patients.
Collapse
Affiliation(s)
- Kyla Bourque
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Cara Hawey
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Alyson Jiang
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Grace R Mazarura
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Terence E Hébert
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada.
| |
Collapse
|
19
|
Tappia P, Elimban V, Dhalla N. Involvement of phospholipase C in the norepinephrine-induced hypertrophic response in Cardiomyocytes. SCRIPTA MEDICA 2022. [DOI: 10.5937/scriptamed53-36527] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
Norepinephrine (NE) is known to mediate cardiomyocyte hypertrophy through the G protein coupled a1 -adrenoceptor (a1 -AR) and the activation of the phosphoinositide-specific phospholipase C (PLC). Since the by-products of PLC activity are important downstream signal transducers for cardiac hypertrophy, the role of and the regulatory mechanisms involved in the activation of PLC isozymes in cardiac hypertrophy are highlighted in this review. The discussion is focused to underscore PLC in different experimental models of cardiac hypertrophy, as well as in isolated adult and neonatal cardiomyocytes treated with NE. Particular emphasis is laid concerning the a1 -AR-PLC-mediated hypertrophic signalling pathway. From the information provided, it is evident that the specific activation of PLC isozymes is a primary signalling event in the a1 -AR mediated response to NE as well as initiation and progression of cardiac hypertrophy. Furthermore, the possibility of PLC involvement in the perpetuation of cardiac hypertrophy is also described. It is suggested that specific PLC isozymes may serve as viable targets for the prevention of cardiac hypertrophy in patient population at-risk for the development of heart failure.
Collapse
|
20
|
Identification and characterization of an atypical Gαs-biased β 2AR agonist that fails to evoke airway smooth muscle cell tachyphylaxis. Proc Natl Acad Sci U S A 2021; 118:2026668118. [PMID: 34857633 PMCID: PMC8670521 DOI: 10.1073/pnas.2026668118] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/18/2021] [Indexed: 12/23/2022] Open
Abstract
We sought β2AR agonists for treating obstructive lung diseases such as asthma, in which this receptor relaxes airway smooth muscle (ASM) cells and opens airways. Agonists favoring Gs coupling (leads to airway relaxation) compared with activating β-arrestin (limits effectiveness due to receptor desensitization) were pursued in a 40-million-compound screening library. Of several agonists identified, one was apparently biased away from β-arrestin. Agonist–receptor–G protein modeling revealed different receptor interactions compared with other agonists. The favorable effects of the apparent biasing with this agonist were demonstrated in a physiologic system (ASM relaxation). These studies point to a different structural class of β-agonists that might be used to treat obstructive lung diseases without the adverse effects associated with tachyphylaxis. G protein–coupled receptors display multifunctional signaling, offering the potential for agonist structures to promote conformational selectivity for biased outputs. For β2-adrenergic receptors (β2AR), unbiased agonists stabilize conformation(s) that evoke coupling to Gαs (cyclic adenosine monophosphate [cAMP] production/human airway smooth muscle [HASM] cell relaxation) and β-arrestin engagement, the latter acting to quench Gαs signaling, contributing to receptor desensitization/tachyphylaxis. We screened a 40-million-compound scaffold ranking library, revealing unanticipated agonists with dihydroimidazolyl-butyl-cyclic urea scaffolds. The S-stereoisomer of compound C1 shows no detectable β-arrestin engagement/signaling by four methods. However, C1-S retained Gαs signaling—a divergence of the outputs favorable for treating asthma. Functional studies with two models confirmed the biasing: β2AR-mediated cAMP signaling underwent desensitization to the unbiased agonist albuterol but not to C1-S, and desensitization of HASM cell relaxation was observed with albuterol but not with C1-S. These HASM results indicate biologically pertinent biasing of C1-S, in the context of the relevant physiologic response, in the human cell type of interest. Thus, C1-S was apparently strongly biased away from β-arrestin, in contrast to albuterol and C5-S. C1-S structural modeling and simulations revealed binding differences compared with unbiased epinephrine at transmembrane (TM) segments 3,5,6,7 and ECL2. C1-S (R2 = cyclohexane) was repositioned in the pocket such that it lost a TM6 interaction and gained a TM7 interaction compared with the analogous unbiased C5-S (R2 = benzene group), which appears to contribute to C1-S biasing away from β-arrestin. Thus, an agnostic large chemical-space library identified agonists with receptor interactions that resulted in relevant signal splitting of β2AR actions favorable for treating obstructive lung disease.
Collapse
|
21
|
Guo Y, Yu ZY, Wu J, Gong H, Kesteven S, Iismaa SE, Chan AY, Holman S, Pinto S, Pironet A, Cox CD, Graham RM, Vennekens R, Feneley MP, Martinac B. The Ca 2+-activated cation channel TRPM4 is a positive regulator of pressure overload-induced cardiac hypertrophy. eLife 2021; 10:66582. [PMID: 34190686 PMCID: PMC8245133 DOI: 10.7554/elife.66582] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 06/20/2021] [Indexed: 01/19/2023] Open
Abstract
Pathological left ventricular hypertrophy (LVH) occurs in response to pressure overload and remains the single most important clinical predictor of cardiac mortality. The molecular pathways in the induction of pressure overload LVH are potential targets for therapeutic intervention. Current treatments aim to remove the pressure overload stimulus for LVH, but do not completely reverse adverse cardiac remodelling. Although numerous molecular signalling steps in the induction of LVH have been identified, the initial step by which mechanical stretch associated with cardiac pressure overload is converted into a chemical signal that initiates hypertrophic signalling remains unresolved. In this study, we show that selective deletion of transient receptor potential melastatin 4 (TRPM4) channels in mouse cardiomyocytes results in an approximately 50% reduction in the LVH induced by transverse aortic constriction. Our results suggest that TRPM4 channel is an important component of the mechanosensory signalling pathway that induces LVH in response to pressure overload and represents a potential novel therapeutic target for the prevention of pathological LVH.
Collapse
Affiliation(s)
- Yang Guo
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Sydney, Australia.,Cardiac Physiology and Transplantation Division, Victor Chang Cardiac Research Institute, Sydney, Australia.,St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Ze-Yan Yu
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Sydney, Australia.,Cardiac Physiology and Transplantation Division, Victor Chang Cardiac Research Institute, Sydney, Australia.,St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Jianxin Wu
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Sydney, Australia
| | - Hutao Gong
- Cardiac Physiology and Transplantation Division, Victor Chang Cardiac Research Institute, Sydney, Australia
| | - Scott Kesteven
- Cardiac Physiology and Transplantation Division, Victor Chang Cardiac Research Institute, Sydney, Australia.,St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Siiri E Iismaa
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Sydney, Australia.,St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Andrea Y Chan
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Sydney, Australia
| | - Sara Holman
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Sydney, Australia
| | - Silvia Pinto
- Laboratory of Ion Channel Research, Department of Molecular and Cellular Medicine, Katholieke Universiteit Leuven, Leuven, Belgium.,TRP Research Platform Leuven (TRPLe), Katholieke Universiteit Leuven, Leuven, Belgium
| | - Andy Pironet
- Laboratory of Ion Channel Research, Department of Molecular and Cellular Medicine, Katholieke Universiteit Leuven, Leuven, Belgium.,TRP Research Platform Leuven (TRPLe), Katholieke Universiteit Leuven, Leuven, Belgium
| | - Charles D Cox
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Sydney, Australia.,St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Robert M Graham
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Sydney, Australia.,St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Rudi Vennekens
- Laboratory of Ion Channel Research, Department of Molecular and Cellular Medicine, Katholieke Universiteit Leuven, Leuven, Belgium.,TRP Research Platform Leuven (TRPLe), Katholieke Universiteit Leuven, Leuven, Belgium
| | - Michael P Feneley
- Cardiac Physiology and Transplantation Division, Victor Chang Cardiac Research Institute, Sydney, Australia.,St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, Australia.,Department of Cardiology, St Vincent's Hospital, Sydney, Australia
| | - Boris Martinac
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Sydney, Australia.,St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, Australia
| |
Collapse
|
22
|
Kilian LS, Voran J, Frank D, Rangrez AY. RhoA: a dubious molecule in cardiac pathophysiology. J Biomed Sci 2021; 28:33. [PMID: 33906663 PMCID: PMC8080415 DOI: 10.1186/s12929-021-00730-w] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 04/23/2021] [Indexed: 02/08/2023] Open
Abstract
The Ras homolog gene family member A (RhoA) is the founding member of Rho GTPase superfamily originally studied in cancer cells where it was found to stimulate cell cycle progression and migration. RhoA acts as a master switch control of actin dynamics essential for maintaining cytoarchitecture of a cell. In the last two decades, however, RhoA has been coined and increasingly investigated as an essential molecule involved in signal transduction and regulation of gene transcription thereby affecting physiological functions such as cell division, survival, proliferation and migration. RhoA has been shown to play an important role in cardiac remodeling and cardiomyopathies; underlying mechanisms are however still poorly understood since the results derived from in vitro and in vivo experiments are still inconclusive. Interestingly its role in the development of cardiomyopathies or heart failure remains largely unclear due to anomalies in the current data available that indicate both cardioprotective and deleterious effects. In this review, we aimed to outline the molecular mechanisms of RhoA activation, to give an overview of its regulators, and the probable mechanisms of signal transduction leading to RhoA activation and induction of downstream effector pathways and corresponding cellular responses in cardiac (patho)physiology. Furthermore, we discuss the existing studies assessing the presented results and shedding light on the often-ambiguous data. Overall, we provide an update of the molecular, physiological and pathological functions of RhoA in the heart and its potential in cardiac therapeutics.
Collapse
Affiliation(s)
- Lucia Sophie Kilian
- Department of Internal Medicine III (Cardiology, Angiology, Intensive Care), University Medical Center Kiel, Rosalind-Franklin Str. 12, 24105, Kiel, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, 24105, Kiel, Germany
| | - Jakob Voran
- Department of Internal Medicine III (Cardiology, Angiology, Intensive Care), University Medical Center Kiel, Rosalind-Franklin Str. 12, 24105, Kiel, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, 24105, Kiel, Germany
| | - Derk Frank
- Department of Internal Medicine III (Cardiology, Angiology, Intensive Care), University Medical Center Kiel, Rosalind-Franklin Str. 12, 24105, Kiel, Germany. .,DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, 24105, Kiel, Germany.
| | - Ashraf Yusuf Rangrez
- Department of Internal Medicine III (Cardiology, Angiology, Intensive Care), University Medical Center Kiel, Rosalind-Franklin Str. 12, 24105, Kiel, Germany. .,DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, 24105, Kiel, Germany. .,Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany.
| |
Collapse
|
23
|
Yu ZY, Gong H, Wu J, Dai Y, Kesteven SH, Fatkin D, Martinac B, Graham RM, Feneley MP. Cardiac Gq Receptors and Calcineurin Activation Are Not Required for the Hypertrophic Response to Mechanical Left Ventricular Pressure Overload. Front Cell Dev Biol 2021; 9:639509. [PMID: 33659256 PMCID: PMC7917224 DOI: 10.3389/fcell.2021.639509] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 01/26/2021] [Indexed: 01/19/2023] Open
Abstract
Rationale Gq-coupled receptors are thought to play a critical role in the induction of left ventricular hypertrophy (LVH) secondary to pressure overload, although mechano-sensitive channel activation by a variety of mechanisms has also been proposed, and the relative importance of calcineurin- and calmodulin kinase II (CaMKII)-dependent hypertrophic pathways remains controversial. Objective To determine the mechanisms regulating the induction of LVH in response to mechanical pressure overload. Methods and Results Transgenic mice with cardiac-targeted inhibition of Gq-coupled receptors (GqI mice) and their non-transgenic littermates (NTL) were subjected to neurohumoral stimulation (continuous, subcutaneous angiotensin II (AngII) infusion for 14 days) or mechanical pressure overload (transverse aortic arch constriction (TAC) for 21 days) to induce LVH. Candidate signaling pathway activation was examined. As expected, LVH observed in NTL mice with AngII infusion was attenuated in heterozygous (GqI+/-) mice and absent in homozygous (GqI-/-) mice. In contrast, LVH due to TAC was unaltered by either heterozygous or homozygous Gq inhibition. Gene expression of atrial natriuretic peptide (ANP), B-type natriuretic peptide (BNP) and α-skeletal actin (α-SA) was increased 48 h after AngII infusion or TAC in NTL mice; in GqI mice, the increases in ANP, BNP and α-SA in response to AngII were completely absent, as expected, but all three increased after TAC. Increased nuclear translocation of nuclear factor of activated T-cells c4 (NFATc4), indicating calcineurin pathway activation, occurred in NTL mice with AngII infusion but not TAC, and was prevented in GqI mice infused with AngII. Nuclear and cytoplasmic CaMKIIδ levels increased in both NTL and GqI mice after TAC but not AngII infusion, with increased cytoplasmic phospho- and total histone deacetylase 4 (HDAC4) and increased nuclear myocyte enhancer factor 2 (MEF2) levels. Conclusion Cardiac Gq receptors and calcineurin activation are required for neurohumorally mediated LVH but not for LVH induced by mechanical pressure overload (TAC). Rather, TAC-induced LVH is associated with activation of the CaMKII-HDAC4-MEF2 pathway.
Collapse
Affiliation(s)
- Ze-Yan Yu
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia.,Cardiology Department, St Vincent's Hospital, Darlinghurst, NSW, Australia.,Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Hutao Gong
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
| | - Jianxin Wu
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
| | - Yun Dai
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
| | - Scott H Kesteven
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
| | - Diane Fatkin
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia.,Cardiology Department, St Vincent's Hospital, Darlinghurst, NSW, Australia.,Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Boris Martinac
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia.,Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Robert M Graham
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia.,Cardiology Department, St Vincent's Hospital, Darlinghurst, NSW, Australia.,Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Michael P Feneley
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia.,Cardiology Department, St Vincent's Hospital, Darlinghurst, NSW, Australia.,Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
24
|
Simões SC, Balico-Silva AL, Parreiras-E-Silva LT, Bitencourt ALB, Bouvier M, Costa-Neto CM. Signal Transduction Profiling of Angiotensin II Type 1 Receptor With Mutations Associated to Atrial Fibrillation in Humans. Front Pharmacol 2021; 11:600132. [PMID: 33424609 PMCID: PMC7786401 DOI: 10.3389/fphar.2020.600132] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 11/18/2020] [Indexed: 11/24/2022] Open
Abstract
The AT1 receptor (AT1R) has a major role in the Renin-Angiotensin System, being involved in several physiological events including blood pressure control and electrolyte balance. The AT1R is a member of the G protein coupled receptors (GPCR) family, classically known to couple Gαq and engage β-arrestin recruitment. Both G protein and arrestin signaling pathways are involved in modulation of different downstream kinases. A previous study reported that mutations in the AT1R (A244S and I103T-A244S) were positively correlated with higher risk of atrial fibrillation in men. Based on that report, we aimed to investigate if these mutations, including I103T only, could affect AT1R signal transduction profile, and consequently, implicate in atrial fibrillation outcome. To address that, we engineered an AT1R carrying the above-mentioned mutations, and functionally evaluated different signaling pathways. Phosphokinase profiler array to assess the mutations downstream effects on kinases and kinase substrates phosphorylation levels was used. Our results show that the I103T-A244S mutant receptor presents decreased β-arrestin 2 recruitment, which could lead to a harmful condition of sustained Gαq signaling. Moreover, the phosphokinase profiler array revealed that the same mutation led to downstream modulation of kinase pathways that are linked to physiological responses such as fibrous tissue formation, apoptosis and cell proliferation.
Collapse
Affiliation(s)
- Sarah C Simões
- Ribeirao Preto Medical School, Department of Biochemistry and Immunology, University of São Paulo, Ribeirao Preto, Brazil
| | - André L Balico-Silva
- Ribeirao Preto Medical School, Department of Biochemistry and Immunology, University of São Paulo, Ribeirao Preto, Brazil
| | - Lucas T Parreiras-E-Silva
- Ribeirao Preto Medical School, Department of Biochemistry and Immunology, University of São Paulo, Ribeirao Preto, Brazil
| | - André L B Bitencourt
- Ribeirao Preto Medical School, Department of Biochemistry and Immunology, University of São Paulo, Ribeirao Preto, Brazil
| | - Michel Bouvier
- Department of Biochemistry and Molecular Medicine and Institute for Research in Immunology and Cancer, University of Montréal, Montréal, QC, Canada
| | - Claudio M Costa-Neto
- Ribeirao Preto Medical School, Department of Biochemistry and Immunology, University of São Paulo, Ribeirao Preto, Brazil
| |
Collapse
|
25
|
Kaykı-Mutlu G, Papazisi O, Palmen M, Danser AHJ, Michel MC, Arioglu-Inan E. Cardiac and Vascular α 1-Adrenoceptors in Congestive Heart Failure: A Systematic Review. Cells 2020; 9:E2412. [PMID: 33158106 PMCID: PMC7694190 DOI: 10.3390/cells9112412] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/29/2020] [Accepted: 11/02/2020] [Indexed: 02/07/2023] Open
Abstract
As heart failure (HF) is a devastating health problem worldwide, a better understanding and the development of more effective therapeutic approaches are required. HF is characterized by sympathetic system activation which stimulates α- and β-adrenoceptors (ARs). The exposure of the cardiovascular system to the increased locally released and circulating levels of catecholamines leads to a well-described downregulation and desensitization of β-ARs. However, information on the role of α-AR is limited. We have performed a systematic literature review examining the role of both cardiac and vascular α1-ARs in HF using 5 databases for our search. All three α1-AR subtypes (α1A, α1B and α1D) are expressed in human and animal hearts and blood vessels in a tissue-dependent manner. We summarize the changes observed in HF regarding the density, signaling and responses of α1-ARs. Conflicting findings arise from different studies concerning the influence that HF has on α1-AR expression and function; in contrast to β-ARs there is no consistent evidence for down-regulation or desensitization of cardiac or vascular α1-ARs. Whether α1-ARs are a therapeutic target in HF remains a matter of debate.
Collapse
Affiliation(s)
- Gizem Kaykı-Mutlu
- Department of Pharmacology, Faculty of Pharmacy, Ankara University, 06560 Ankara, Turkey; (G.K.-M.); (E.A.-I.)
| | - Olga Papazisi
- Department of Cardiothoracic Surgery, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (O.P.); (M.P.)
| | - Meindert Palmen
- Department of Cardiothoracic Surgery, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (O.P.); (M.P.)
| | - A. H. Jan Danser
- Department of Internal Medicine, Division of Pharmacology, Erasmus Medical Center, 3000 CA Rotterdam, The Netherlands;
| | - Martin C. Michel
- Department of Pharmacology, Johannes Gutenberg University, 55131 Mainz, Germany
| | - Ebru Arioglu-Inan
- Department of Pharmacology, Faculty of Pharmacy, Ankara University, 06560 Ankara, Turkey; (G.K.-M.); (E.A.-I.)
| |
Collapse
|
26
|
Katan M, Cockcroft S. Phospholipase C families: Common themes and versatility in physiology and pathology. Prog Lipid Res 2020; 80:101065. [PMID: 32966869 DOI: 10.1016/j.plipres.2020.101065] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 08/14/2020] [Accepted: 09/17/2020] [Indexed: 12/20/2022]
Abstract
Phosphoinositide-specific phospholipase Cs (PLCs) are expressed in all mammalian cells and play critical roles in signal transduction. To obtain a comprehensive understanding of these enzymes in physiology and pathology, a detailed structural, biochemical, cell biological and genetic information is required. In this review, we cover all these aspects to summarize current knowledge of the entire superfamily. The families of PLCs have expanded from 13 enzymes to 16 with the identification of the atypical PLCs in the human genome. Recent structural insights highlight the common themes that cover not only the substrate catalysis but also the mechanisms of activation. This involves the release of autoinhibitory interactions that, in the absence of stimulation, maintain classical PLC enzymes in their inactive forms. Studies of individual PLCs provide a rich repertoire of PLC function in different physiologies. Furthermore, the genetic studies discovered numerous mutated and rare variants of PLC enzymes and their link to human disease development, greatly expanding our understanding of their roles in diverse pathologies. Notably, substantial evidence now supports involvement of different PLC isoforms in the development of specific cancer types, immune disorders and neurodegeneration. These advances will stimulate the generation of new drugs that target PLC enzymes, and will therefore open new possibilities for treatment of a number of diseases where current therapies remain ineffective.
Collapse
Affiliation(s)
- Matilda Katan
- Institute of Structural and Molecular Biology, Division of Biosciences, University College London, Gower Street, London WC1E 6BT, UK
| | - Shamshad Cockcroft
- Department of Neuroscience, Physiology and Pharmacology, Division of Biosciences, University College London, 21 University Street, London WC1E 6JJ, UK.
| |
Collapse
|
27
|
Chen Y, Huang L, Dong Y, Tao C, Zhang R, Shao H, Shen H. Effect of AKT1 (p. E17K) Hotspot Mutation on Malignant Tumorigenesis and Prognosis. Front Cell Dev Biol 2020; 8:573599. [PMID: 33123537 PMCID: PMC7573235 DOI: 10.3389/fcell.2020.573599] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 09/03/2020] [Indexed: 12/12/2022] Open
Abstract
The substitution of the seventeenth amino acid glutamate by lysine in the homologous structural domain of the Akt1 gene pleckstrin is a somatic cellular mutation found in breast, colorectal, and ovarian cancers, named p. Glu17Lys or E17K. In recent years, a growing number of studies have suggested that this mutation may play a unique role in the development of tumors. In this review article, we describe how AKT1(E17K) mutations stimulate downstream signals that cause cells to emerge transformed; we explore the differential regulation and function of E17K in different physiological and pathological settings; and we also describe the phenomenon that E17K impedes tumor growth by interfering with growth-promoting and chemotherapy-resistant AKT1lowQCC generation, an intriguing finding that mutants may prolong tumor patient survival by activating feedback mechanisms and disrupting transcription. This review is intended to provide a better understanding of the role of AKT1(E17K) in cancer and to inform the development of AKT1(E17K)-based antitumor strategies.
Collapse
Affiliation(s)
- Ying Chen
- Guangdong Province Key Laboratory for Biotechnology Drug Candidates, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Lan Huang
- Guangdong Province Key Laboratory for Biotechnology Drug Candidates, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yongjian Dong
- Guangdong Province Key Laboratory for Biotechnology Drug Candidates, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Changli Tao
- Guangdong Province Key Laboratory for Biotechnology Drug Candidates, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Rongxin Zhang
- Guangdong Province Key Laboratory for Biotechnology Drug Candidates, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Hongwei Shao
- Guangdong Province Key Laboratory for Biotechnology Drug Candidates, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Han Shen
- Guangdong Province Key Laboratory for Biotechnology Drug Candidates, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
28
|
Parreira RC, Gómez-Mendoza DP, de Jesus ICG, Lemos RP, Santos AK, Rezende CP, Figueiredo HCP, Pinto MCX, Kjeldsen F, Guatimosim S, Resende RR, Verano-Braga T. Cardiomyocyte Proteome Remodeling due to Isoproterenol-Induced Cardiac Hypertrophy during the Compensated Phase. Proteomics Clin Appl 2020; 14:e2000017. [PMID: 32506788 DOI: 10.1002/prca.202000017] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 04/29/2020] [Indexed: 11/09/2022]
Abstract
PURPOSE Although the pathophysiological response of cardiac tissue to pro-hypertrophic stimulus is well characterized, a comprehensive characterization of the molecular events underlying the pathological hypertrophy in cardiomyocytes during the early compensated cardiac hypertrophy is currently lacking. EXPERIMENTAL DESIGN A quantitative label-free proteomic analysis of cardiomyocytes isolated was conducted from mice treated subcutaneously with isoproterenol (ISO) during 7 days in comparison with cardiomyocytes from control animals (CT). RESULTS Canonical pathway analysis of dysregulated proteins indicated that ISO-hypertrophy drives the activation of actin cytoskeleton and integrin-linked kinase (ILK) signaling, and inhibition of the sirtuin signaling. Alteration in cardiac contractile function and calcium signaling are predicted as downstream effects of ISO-hypertrophy probably due to the upregulation of key elements such as myosin-7 (MYH7). Confocal microscopy corroborated that indeed ISO-treatment led to increased abundance of MYH7. Potential early markers for cardiac hypertrophy as APBB1, GOLGA4, HOOK1, KATNA1, KIFBP, MAN2B2, and SLC16A1 are also reported. CONCLUSIONS AND CLINICAL RELEVANCE The data consist in a complete molecular mapping of ISO-induced compensated cardiac hypertrophy model at cardiomyocyte level. Marker candidates reported may assist early diagnosis of cardiac hypertrophy and ultimately heart failure.
Collapse
Affiliation(s)
- Ricardo Cambraia Parreira
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 31270-901, Brazil.,Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 31270-901, Brazil.,Centro Universitário de Mineiros, UNIFIMES, Trindade, Golás, 75380-307, Brazil
| | - Diana Paola Gómez-Mendoza
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Itamar Couto Guedes de Jesus
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Rafael Pereira Lemos
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Anderson Kennedy Santos
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 31270-901, Brazil.,Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Cristiana Perdigão Rezende
- Departamento de Medicina Veterinária Preventiva, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Henrique César Pereira Figueiredo
- Departamento de Medicina Veterinária Preventiva, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Mauro Cunha Xavier Pinto
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia, Golás, 74968-755, Brazil
| | - Frank Kjeldsen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, 5230, Denmark
| | - Silvia Guatimosim
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Rodrigo Ribeiro Resende
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Thiago Verano-Braga
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| |
Collapse
|
29
|
Nguyen BY, Ruiz‐Velasco A, Bui T, Collins L, Wang X, Liu W. Mitochondrial function in the heart: the insight into mechanisms and therapeutic potentials. Br J Pharmacol 2019; 176:4302-4318. [PMID: 29968316 PMCID: PMC6887906 DOI: 10.1111/bph.14431] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 06/08/2018] [Accepted: 06/20/2018] [Indexed: 12/19/2022] Open
Abstract
Mitochondrial dysfunction is considered as a crucial contributory factor in cardiac pathology. This has highlighted the therapeutic potential of targeting mitochondria to prevent or treat cardiac disease. Mitochondrial dysfunction is associated with aberrant electron transport chain activity, reduced ATP production, an abnormal shift in metabolic substrates, ROS overproduction and impaired mitochondrial dynamics. This review will cover the mitochondrial functions and how they are altered in various disease conditions. Furthermore, the mechanisms that lead to mitochondrial defects and the protective mechanisms that prevent mitochondrial damage will be discussed. Finally, potential mitochondrial targets for novel therapeutic intervention will be explored. We will highlight the development of small molecules that target mitochondria from different perspectives and their current progress in clinical trials. LINKED ARTICLES: This article is part of a themed section on Mitochondrial Pharmacology: Featured Mechanisms and Approaches for Therapy Translation. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.22/issuetoc.
Collapse
Affiliation(s)
- Binh Yen Nguyen
- Faculty of Biology, Medicine and HealthThe University of ManchesterManchesterUK
| | - Andrea Ruiz‐Velasco
- Faculty of Biology, Medicine and HealthThe University of ManchesterManchesterUK
| | - Thuy Bui
- Faculty of Biology, Medicine and HealthThe University of ManchesterManchesterUK
| | - Lucy Collins
- Faculty of Biology, Medicine and HealthThe University of ManchesterManchesterUK
| | - Xin Wang
- Faculty of Biology, Medicine and HealthThe University of ManchesterManchesterUK
| | - Wei Liu
- Faculty of Biology, Medicine and HealthThe University of ManchesterManchesterUK
| |
Collapse
|
30
|
Del Re DP, Amgalan D, Linkermann A, Liu Q, Kitsis RN. Fundamental Mechanisms of Regulated Cell Death and Implications for Heart Disease. Physiol Rev 2019; 99:1765-1817. [PMID: 31364924 DOI: 10.1152/physrev.00022.2018] [Citation(s) in RCA: 570] [Impact Index Per Article: 114.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Twelve regulated cell death programs have been described. We review in detail the basic biology of nine including death receptor-mediated apoptosis, death receptor-mediated necrosis (necroptosis), mitochondrial-mediated apoptosis, mitochondrial-mediated necrosis, autophagy-dependent cell death, ferroptosis, pyroptosis, parthanatos, and immunogenic cell death. This is followed by a dissection of the roles of these cell death programs in the major cardiac syndromes: myocardial infarction and heart failure. The most important conclusion relevant to heart disease is that regulated forms of cardiomyocyte death play important roles in both myocardial infarction with reperfusion (ischemia/reperfusion) and heart failure. While a role for apoptosis in ischemia/reperfusion cannot be excluded, regulated forms of necrosis, through both death receptor and mitochondrial pathways, are critical. Ferroptosis and parthanatos are also likely important in ischemia/reperfusion, although it is unclear if these entities are functioning as independent death programs or as amplification mechanisms for necrotic cell death. Pyroptosis may also contribute to ischemia/reperfusion injury, but potentially through effects in non-cardiomyocytes. Cardiomyocyte loss through apoptosis and necrosis is also an important component in the pathogenesis of heart failure and is mediated by both death receptor and mitochondrial signaling. Roles for immunogenic cell death in cardiac disease remain to be defined but merit study in this era of immune checkpoint cancer therapy. Biology-based approaches to inhibit cell death in the various cardiac syndromes are also discussed.
Collapse
Affiliation(s)
- Dominic P Del Re
- Departments of Medicine and Cell Biology, Wilf Family Cardiovascular Research Institute, Albert Einstein Cancer Center, and Einstein-Mount Sinai Diabetes Research Center, Albert Einstein College of Medicine, Bronx, New York; Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey; Department of Internal Medicine 3, Division of Nephrology, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany; and Department of Physiology and Biophysics, University of Washington, Seattle, Washington
| | - Dulguun Amgalan
- Departments of Medicine and Cell Biology, Wilf Family Cardiovascular Research Institute, Albert Einstein Cancer Center, and Einstein-Mount Sinai Diabetes Research Center, Albert Einstein College of Medicine, Bronx, New York; Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey; Department of Internal Medicine 3, Division of Nephrology, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany; and Department of Physiology and Biophysics, University of Washington, Seattle, Washington
| | - Andreas Linkermann
- Departments of Medicine and Cell Biology, Wilf Family Cardiovascular Research Institute, Albert Einstein Cancer Center, and Einstein-Mount Sinai Diabetes Research Center, Albert Einstein College of Medicine, Bronx, New York; Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey; Department of Internal Medicine 3, Division of Nephrology, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany; and Department of Physiology and Biophysics, University of Washington, Seattle, Washington
| | - Qinghang Liu
- Departments of Medicine and Cell Biology, Wilf Family Cardiovascular Research Institute, Albert Einstein Cancer Center, and Einstein-Mount Sinai Diabetes Research Center, Albert Einstein College of Medicine, Bronx, New York; Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey; Department of Internal Medicine 3, Division of Nephrology, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany; and Department of Physiology and Biophysics, University of Washington, Seattle, Washington
| | - Richard N Kitsis
- Departments of Medicine and Cell Biology, Wilf Family Cardiovascular Research Institute, Albert Einstein Cancer Center, and Einstein-Mount Sinai Diabetes Research Center, Albert Einstein College of Medicine, Bronx, New York; Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey; Department of Internal Medicine 3, Division of Nephrology, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany; and Department of Physiology and Biophysics, University of Washington, Seattle, Washington
| |
Collapse
|
31
|
Abstract
GPCRs (G-protein [guanine nucleotide-binding protein]-coupled receptors) play a central physiological role in the regulation of cardiac function in both health and disease and thus represent one of the largest class of surface receptors targeted by drugs. Several antagonists of GPCRs, such as βARs (β-adrenergic receptors) and Ang II (angiotensin II) receptors, are now considered standard of therapy for a wide range of cardiovascular disease, such as hypertension, coronary artery disease, and heart failure. Although the mechanism of action for GPCRs was thought to be largely worked out in the 80s and 90s, recent discoveries have brought to the fore new and previously unappreciated mechanisms for GPCR activation and subsequent downstream signaling. In this review, we focus on GPCRs most relevant to the cardiovascular system and discuss traditional components of GPCR signaling and highlight evolving concepts in the field, such as ligand bias, β-arrestin-mediated signaling, and conformational heterogeneity.
Collapse
Affiliation(s)
- Jialu Wang
- From the Department of Medicine (J.W., C.G., H.A.R.)
| | | | - Howard A Rockman
- From the Department of Medicine (J.W., C.G., H.A.R.).,Department of Cell Biology (H.A.R.).,Department of Molecular Genetics and Microbiology (H.A.R.), Duke University Medical Center, Durham, NC
| |
Collapse
|
32
|
Nash CA, Wei W, Irannejad R, Smrcka AV. Golgi localized β1-adrenergic receptors stimulate Golgi PI4P hydrolysis by PLCε to regulate cardiac hypertrophy. eLife 2019; 8:48167. [PMID: 31433293 PMCID: PMC6726460 DOI: 10.7554/elife.48167] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 08/21/2019] [Indexed: 12/25/2022] Open
Abstract
Increased adrenergic tone resulting from cardiovascular stress leads to development of heart failure, in part, through chronic stimulation of β1 adrenergic receptors (βARs) on cardiac myocytes. Blocking these receptors is part of the basis for β-blocker therapy for heart failure. Recent data demonstrate that G protein-coupled receptors (GPCRs), including βARs, are activated intracellularly, although the biological significance is unclear. Here we investigated the functional role of Golgi βARs in rat cardiac myocytes and found they activate Golgi localized, prohypertrophic, phosphoinositide hydrolysis, that is not accessed by cell surface βAR stimulation. This pathway is accessed by the physiological neurotransmitter norepinephrine (NE) via an Oct3 organic cation transporter. Blockade of Oct3 or specific blockade of Golgi resident β1ARs prevents NE dependent cardiac myocyte hypertrophy. This clearly defines a pathway activated by internal GPCRs in a biologically relevant cell type and has implications for development of more efficacious β-blocker therapies.
Collapse
Affiliation(s)
- Craig A Nash
- Department of Pharmacology, University of Michigan School of Medicine, Ann Arbor, United States
| | - Wenhui Wei
- Department of Pharmacology, University of Michigan School of Medicine, Ann Arbor, United States
| | - Roshanak Irannejad
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, United States
| | - Alan V Smrcka
- Department of Pharmacology, University of Michigan School of Medicine, Ann Arbor, United States
| |
Collapse
|
33
|
Myagmar BE, Ismaili T, Swigart PM, Raghunathan A, Baker AJ, Sahdeo S, Blevitt JM, Milla ME, Simpson PC. Coupling to Gq Signaling Is Required for Cardioprotection by an Alpha-1A-Adrenergic Receptor Agonist. Circ Res 2019; 125:699-706. [PMID: 31426700 DOI: 10.1161/circresaha.118.314416] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RATIONALE Gq signaling in cardiac myocytes is classically considered toxic. Targeting Gq directly to test this is problematic, because cardiac myocytes have many Gq-coupled receptors. OBJECTIVE Test whether Gq coupling is required for the cardioprotective effects of an alpha-1A-AR (adrenergic receptor) agonist. METHODS AND RESULTS In recombinant cells, a mouse alpha-1A-AR with a 6-residue substitution in the third intracellular loop does not couple to Gq signaling. Here we studied a knockin mouse with this alpha-1A-AR mutation. Heart alpha-1A receptor levels and antagonist affinity in the knockin were identical to wild-type. In wild-type cardiac myocytes, the selective alpha-1A agonist A61603-stimulated phosphoinositide-phospholipase C and myocyte contraction. In myocytes with the alpha-1A knockin, both A61603 effects were absent, indicating that Gq coupling was absent. Surprisingly, A61603 activation of cardioprotective ERK (extracellular signal-regulated kinase) was markedly impaired in the KI mutant myocytes, and A61603 did not protect mutant myocytes from doxorubicin toxicity in vitro. Similarly, mice with the α1A KI mutation had increased mortality after transverse aortic constriction, and A61603 did not rescue cardiac function in mice with the Gq coupling-defective alpha-1A receptor. CONCLUSIONS Gq coupling is required for cardioprotection by an alpha-1A-AR agonist. Gq signaling can be adaptive.
Collapse
Affiliation(s)
- Bat-Erdene Myagmar
- From the VA Medical Center, San Francisco, CA (B.-E.M., P.M.S., A.R., A.J.B., P.C.S.).,University of California, San Francisco (B.-E.M., A.J.B., P.C.S.)
| | - Taylor Ismaili
- Janssen Research and Development, San Diego, CA (T.I., S.S., J.M.B.)
| | - Philip M Swigart
- From the VA Medical Center, San Francisco, CA (B.-E.M., P.M.S., A.R., A.J.B., P.C.S.)
| | - Anaha Raghunathan
- From the VA Medical Center, San Francisco, CA (B.-E.M., P.M.S., A.R., A.J.B., P.C.S.)
| | - Anthony J Baker
- From the VA Medical Center, San Francisco, CA (B.-E.M., P.M.S., A.R., A.J.B., P.C.S.).,University of California, San Francisco (B.-E.M., A.J.B., P.C.S.)
| | - Sunil Sahdeo
- Janssen Research and Development, San Diego, CA (T.I., S.S., J.M.B.)
| | | | | | - Paul C Simpson
- From the VA Medical Center, San Francisco, CA (B.-E.M., P.M.S., A.R., A.J.B., P.C.S.).,University of California, San Francisco (B.-E.M., A.J.B., P.C.S.)
| |
Collapse
|
34
|
Abstract
Purpose of Review Peripartum cardiomyopathy (PPCM) is an idiopathic disorder defined as heart failure occurring in women during the last month of pregnancy and up to 5 months postpartum. In this review, we outline recent reports about the disease pathogenesis and management and highlight the use of diagnosis and prognosis biomarkers. Recent Findings Novel data strengthen the implication of endothelial function in PPCM pathogenesis. The first international registry showed that patient presentations were similar globally, with heterogeneity in patient management and outcome. Summary Despite large improvement in patient management and treatment, there is still a sub-group of women who die from PPCM or who will not recover their cardiac function. Remarkable advances in the comprehension of disease incidence, pathogenesis, and prognosis could be determined with multi-center and international registries. Clinical Trials ClinicalTrials.gov Identifier: NCT02590601
Collapse
|
35
|
Navot S, Kosloff M. Structural design principles that underlie the multi-specific interactions of Gα q with dissimilar partners. Sci Rep 2019; 9:6898. [PMID: 31053791 PMCID: PMC6499889 DOI: 10.1038/s41598-019-43395-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 04/23/2019] [Indexed: 02/07/2023] Open
Abstract
Gαq is a ubiquitous molecular switch that activates the effectors phospholipase-C-β3 (PLC-β3) and Rho guanine-nucleotide exchange factors. Gαq is inactivated by regulators of G protein signaling proteins, as well as by PLC-β3. Gαq further interacts with G protein-coupled receptor kinase 2 (GRK2), although the functional role of this interaction is debated. While X-ray structures of Gαq bound to representatives of these partners have revealed details of their interactions, the mechanistic basis for differential Gαq interactions with multiple partners (i.e., Gαq multi-specificity) has not been elucidated at the individual residue resolution. Here, we map the structural determinants of Gαq multi-specificity using structure-based energy calculations. We delineate regions that specifically interact with GTPase Activating Proteins (GAPs) and residues that exclusively contribute to effector interactions, showing that only the Gαq “Switch II” region interacts with all partners. Our analysis further suggests that Gαq-GRK2 interactions are consistent with GRK2 functioning as an effector, rather than a GAP. Our multi-specificity analysis pinpoints Gαq residues that uniquely contribute to interactions with particular partners, enabling precise manipulation of these cascades. As such, we dissect the molecular basis of Gαq function as a central signaling hub, which can be used to target Gαq-mediated signaling in therapeutic interventions.
Collapse
Affiliation(s)
- Shir Navot
- The Department of Human Biology, Faculty of Natural Science, University of Haifa, Haifa, 3498838, Israel
| | - Mickey Kosloff
- The Department of Human Biology, Faculty of Natural Science, University of Haifa, Haifa, 3498838, Israel.
| |
Collapse
|
36
|
Zuo S, Kong D, Wang C, Liu J, Wang Y, Wan Q, Yan S, Zhang J, Tang J, Zhang Q, Lyu L, Li X, Shan Z, Qian L, Shen Y, Yu Y. CRTH2 promotes endoplasmic reticulum stress-induced cardiomyocyte apoptosis through m-calpain. EMBO Mol Med 2019; 10:emmm.201708237. [PMID: 29335338 PMCID: PMC5840549 DOI: 10.15252/emmm.201708237] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Apoptotic death of cardiac myocytes is associated with ischemic heart disease and chemotherapy‐induced cardiomyopathy. Chemoattractant receptor‐homologous molecule expressed on T helper type 2 cells (CRTH2) is highly expressed in the heart. However, its specific role in ischemic cardiomyopathy is not fully understood. Here, we demonstrated that CRTH2 disruption markedly improved cardiac recovery in mice postmyocardial infarction and doxorubicin challenge by suppressing cardiomyocyte apoptosis. Mechanistically, CRTH2 activation specifically facilitated endoplasmic reticulum (ER) stress‐induced cardiomyocyte apoptosis via caspase‐12‐dependent pathway. Blockage of m‐calpain prevented CRTH2‐mediated cardiomyocyte apoptosis under ER stress by suppressing caspase‐12 activity. CRTH2 was coupled with Gαq to elicit intracellular Ca2+ flux and activated m‐calpain/caspase‐12 cascade in cardiomyocytes. Knockdown of caspase‐4, an alternative to caspase‐12 in humans, markedly alleviated CRHT2 activation‐induced apoptosis in human cardiomyocyte response to anoxia. Our findings revealed an unexpected role of CRTH2 in promoting ER stress‐induced cardiomyocyte apoptosis, suggesting that CRTH2 inhibition has therapeutic potential for ischemic cardiomyopathy.
Collapse
Affiliation(s)
- Shengkai Zuo
- Department of Pharmacology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China.,Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Deping Kong
- Department of Pharmacology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Chenyao Wang
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Jiao Liu
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yuanyang Wang
- Department of Pharmacology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Qiangyou Wan
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Shuai Yan
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Jian Zhang
- Department of Pharmacology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Juan Tang
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Qianqian Zhang
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Luheng Lyu
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China.,Department of Biology, University of Miami College of Arts and Science, Miami, FL, USA
| | - Xin Li
- Department of Pharmacology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Zhixin Shan
- Medical Research Department of Guangdong General Hospital, Guangdong Cardiovascular Institute, Guangdong Academy of Medical Sciences, Guangzhou Guangdong, China
| | - Li Qian
- McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Yujun Shen
- Department of Pharmacology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Ying Yu
- Department of Pharmacology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China .,Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
37
|
Jhun BS, O-Uchi J, Adaniya SM, Cypress MW, Yoon Y. Adrenergic Regulation of Drp1-Driven Mitochondrial Fission in Cardiac Physio-Pathology. Antioxidants (Basel) 2018; 7:antiox7120195. [PMID: 30567380 PMCID: PMC6316402 DOI: 10.3390/antiox7120195] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2018] [Revised: 12/13/2018] [Accepted: 12/14/2018] [Indexed: 12/28/2022] Open
Abstract
Abnormal mitochondrial morphology, especially fragmented mitochondria, and mitochondrial dysfunction are hallmarks of a variety of human diseases including heart failure (HF). Although emerging evidence suggests a link between mitochondrial fragmentation and cardiac dysfunction, it is still not well described which cardiac signaling pathway regulates mitochondrial morphology and function under pathophysiological conditions such as HF. Mitochondria change their shape and location via the activity of mitochondrial fission and fusion proteins. This mechanism is suggested as an important modulator for mitochondrial and cellular functions including bioenergetics, reactive oxygen species (ROS) generation, spatiotemporal dynamics of Ca2+ signaling, cell growth, and death in the mammalian cell- and tissue-specific manners. Recent reports show that a mitochondrial fission protein, dynamin-like/related protein 1 (DLP1/Drp1), is post-translationally modified via cell signaling pathways, which control its subcellular localization, stability, and activity in cardiomyocytes/heart. In this review, we summarize the possible molecular mechanisms for causing post-translational modifications (PTMs) of DLP1/Drp1 in cardiomyocytes, and further discuss how these PTMs of DLP1/Drp1 mediate abnormal mitochondrial morphology and mitochondrial dysfunction under adrenergic signaling activation that contributes to the development and progression of HF.
Collapse
Affiliation(s)
- Bong Sook Jhun
- Lillehei Heart Institute, Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Jin O-Uchi
- Lillehei Heart Institute, Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Stephanie M Adaniya
- Lillehei Heart Institute, Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA.
- Cardiovascular Research Center, Rhode Island Hospital, Providence, RI 02903, USA.
- Department of Medicine, Division of Cardiology, the Alpert Medical School of Brown University, Providence, RI 02903, USA.
| | - Michael W Cypress
- Lillehei Heart Institute, Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Yisang Yoon
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA.
| |
Collapse
|
38
|
Putinski C, Abdul-Ghani M, Brunette S, Burgon PG, Megeney LA. Caspase Cleavage of Gelsolin Is an Inductive Cue for Pathologic Cardiac Hypertrophy. J Am Heart Assoc 2018; 7:e010404. [PMID: 30486716 PMCID: PMC6405540 DOI: 10.1161/jaha.118.010404] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Background Cardiac hypertrophy is an adaptive remodeling event that may improve or diminish contractile performance of the heart. Physiologic and pathologic hypertrophy yield distinct outcomes, yet both are dependent on caspase‐directed proteolysis. This suggests that each form of myocardial growth may derive from a specific caspase cleavage event(s). We examined whether caspase 3 cleavage of the actin capping/severing protein gelsolin is essential for the development of pathologic hypertrophy. Methods and Results Caspase targeting of gelsolin was established through protein analysis of hypertrophic cardiomyocytes and mass spectrometry mapping of cleavage sites. Pathologic agonists induced late‐stage caspase‐mediated cleavage of gelsolin. The requirement of caspase‐mediated gelsolin cleavage for hypertrophy induction was evaluated in primary cardiomyocytes by cell size analysis, monitoring of prohypertrophy markers, and measurement of hypertrophy‐related transcription activity. The in vivo impact of caspase‐mediated cleavage was investigated by echo‐guided intramyocardial injection of adenoviral‐expressed gelsolin. Expression of the N‐terminal gelsolin caspase cleavage fragment was necessary and sufficient to cause pathologic remodeling in isolated cardiomyocytes and the intact heart, whereas expression of a noncleavable form prevents cardiac remodeling. Alterations in myocardium structure and function were determined by echocardiography and end‐stage cardiomyocyte cell size analysis. Gelsolin secretion was also monitored for its impact on naïve cells using competitive antibody trapping, demonstrating that hypertrophic agonist stimulation of cardiomyocytes leads to gelsolin secretion, which induces hypertrophy in naïve cells. Conclusions These results suggest that cell autonomous caspase cleavage of gelsolin is essential for pathologic hypertrophy and that cardiomyocyte secretion of gelsolin may accelerate this negative remodeling response.
Collapse
Affiliation(s)
- Charis Putinski
- 1 Ottawa Hospital Research Institute Sprott Centre for Stem Cell Research Regenerative Medicine Program Ottawa Hospital Ottawa Ontario Canada.,2 Department of Cellular and Molecular Medicine Faculty of Medicine University of Ottawa Ontario Canada
| | - Mohammad Abdul-Ghani
- 1 Ottawa Hospital Research Institute Sprott Centre for Stem Cell Research Regenerative Medicine Program Ottawa Hospital Ottawa Ontario Canada.,2 Department of Cellular and Molecular Medicine Faculty of Medicine University of Ottawa Ontario Canada
| | - Steve Brunette
- 1 Ottawa Hospital Research Institute Sprott Centre for Stem Cell Research Regenerative Medicine Program Ottawa Hospital Ottawa Ontario Canada
| | - Patrick G Burgon
- 2 Department of Cellular and Molecular Medicine Faculty of Medicine University of Ottawa Ontario Canada.,3 Department of Medicine University of Ottawa Ontario Canada.,4 University of Ottawa Heart Institute Ottawa Ontario Canada
| | - Lynn A Megeney
- 1 Ottawa Hospital Research Institute Sprott Centre for Stem Cell Research Regenerative Medicine Program Ottawa Hospital Ottawa Ontario Canada.,2 Department of Cellular and Molecular Medicine Faculty of Medicine University of Ottawa Ontario Canada.,3 Department of Medicine University of Ottawa Ontario Canada
| |
Collapse
|
39
|
ERK mediated survival signaling is dependent on the Gq-G-protein coupled receptor type and subcellular localization in adult cardiac myocytes. J Mol Cell Cardiol 2018; 127:67-73. [PMID: 30528765 DOI: 10.1016/j.yjmcc.2018.11.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 11/29/2018] [Indexed: 01/05/2023]
Abstract
G protein-coupled receptors that signal through Gαq (GqPCRs), like α1-adrenergic and angiotensin receptors (α1-AR, AT-R), are traditionally thought to mediate pathologic remodeling in heart failure, including cardiac myocyte death. However, we previously demonstrated that α1- ARs are cardioprotective and identified an α1A-subtype-ERK survival-signaling pathway in adult cardiac myocytes. Recently, we demonstrated that α1-ARs localize to and signal from the nucleus, whereas AT-R localize to and signal from the sarcolemma in adult cardiac myocytes. Thus, we proposed a novel paradigm, predicated on compartmentalization of GqPCR signaling, to explain the phenotypic diversity of GqPCRs. Here, we tested the hypothesis that differential subcellular compartmentalization of α1-AR and AT-R mediated activation of ERK might explain the differential effects of these receptors on cardiac myocyte survival. Using a fluorescent ERK activity FRET-based biosensor, EKAR, to measure subcellular localization and extent of receptor-mediated ERK activation in single adult cardiac myocytes, we found that α1-ARs induced ERK activity at the nucleus and in the cytosol in 60% of cardiac myocytes, whereas AT-Rs showed no consistent ERK activation. The cell-specific α1-mediated activation of ERK in 60% of adult cardiac myocytes showed concordance with previous studies indicating that the α1A-subtype is expressed in only 60% of cardiac myocytes. Consistent with the ability to activate ERK, we found that only α1-ARs induced phosphorylation of Bcl-2 family member Bad, improved mitochondrial membrane stability, and promoted cardiac myocyte survival. In summary, our results suggest that compartmentalization of GqPCRs dictate activation of ERK and cardiac myocyte survival in adult cardiac myocytes.
Collapse
|
40
|
Ma X, Ding Y, Wang Y, Xu X. A Doxorubicin-induced Cardiomyopathy Model in Adult Zebrafish. J Vis Exp 2018. [PMID: 29939187 DOI: 10.3791/57567] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The genetically accessible adult zebrafish (Danio rerio) has been increasingly used as a vertebrate model for understanding human diseases such as cardiomyopathy. Because of its convenience and amenability to high throughput genetic manipulations, the generation of acquired cardiomyopathy models, such as the doxorubicin-induced cardiomyopathy (DIC) model in adult zebrafish, is opening the doors to new research avenues, including discovering cardiomyopathy modifiers via forward genetic screening. Different from the embryonic zebrafish DIC model, both initial acute and later chronic phases of cardiomyopathy can be determined in the adult zebrafish DIC model, enabling the study of stage-dependent signaling mechanisms and therapeutic strategies. However, variable results can be obtained with the current model, even in the hands of experienced investigators. To facilitate future implementation of the DIC model, we present a detailed protocol on how to generate this DIC model in adult zebrafish and describe two alternative ways of intraperitoneal (IP) injection. We further discuss options on how to reduce variations to obtain reliable results and provide suggestions on how to appropriately interpret the results.
Collapse
Affiliation(s)
- Xiao Ma
- Clinical and Translational Sciences Track, Mayo Clinic Graduate School of Biomedical Sciences; Department of Biochemistry and Molecular Biology, Mayo Clinic; Division of Cardiovascular Diseases, Mayo Clinic
| | - Yonghe Ding
- Department of Biochemistry and Molecular Biology, Mayo Clinic; Division of Cardiovascular Diseases, Mayo Clinic
| | - Yong Wang
- Department of Biochemistry and Molecular Biology, Mayo Clinic; Division of Cardiovascular Diseases, Mayo Clinic; Institute of Life Science, Beijing University of Chinese Medicine
| | - Xiaolei Xu
- Clinical and Translational Sciences Track, Mayo Clinic Graduate School of Biomedical Sciences; Department of Biochemistry and Molecular Biology, Mayo Clinic; Division of Cardiovascular Diseases, Mayo Clinic;
| |
Collapse
|
41
|
Compartmentalized cyclic nucleotides have opposing effects on regulation of hypertrophic phospholipase Cε signaling in cardiac myocytes. J Mol Cell Cardiol 2018; 121:51-59. [PMID: 29885334 DOI: 10.1016/j.yjmcc.2018.06.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 05/12/2018] [Accepted: 06/05/2018] [Indexed: 12/11/2022]
Abstract
In cardiac myocytes activation of an exchange factor activated by cAMP (Epac) leads to activation of phospholipase Cε (PLCε)-dependent hydrolysis of phosphatidylinositol 4-phosphate (PI4P) in the Golgi apparatus a process critical for development of cardiac hypertrophy. Here we show that β-adrenergic receptor (βAR) stimulation does not stimulate this pathway in the presence of the broad spectrum phosphodiesterase (PDE) inhibitor IBMX, but selective PDE3 inhibition revealed βAR-dependent PI4P depletion. On the other hand, selective inhibition of PDE2 or PDE9A blocked endothelin-1 (ET-1) and cAMP-dependent PI4P hydrolysis by PLCε. Direct activation of protein kinase A (PKA), protein kinase G (PKG), or the atrial natriuretic factor (ANF) receptor abolished PI4P hydrolysis in response to multiple upstream stimuli. These results reveal distinct pools of cyclic nucleotides that either inhibit PLCε at the Golgi through PKA/PKG, or activate PLCε at the Golgi through Epac. These data together reveal a new mechanism by which ANF and selective PDE inhibitors can protect against cardiac hypertrophy.
Collapse
|
42
|
Abstract
G protein-coupled receptor kinases (GRKs) are classically known for their role in regulating the activity of the largest known class of membrane receptors, which influence diverse biological processes in every cell type in the human body. As researchers have tried to uncover how this family of kinases, containing only 7 members, achieves selective and coordinated control of receptors, they have uncovered a growing number of noncanonical activities for these kinases. These activities include phosphorylation of nonreceptor targets and kinase-independent molecular interactions. In particular, GRK2, GRK3, and GRK5 are the predominant members expressed in the heart. Their canonical and noncanonical actions within cardiac and other tissues have significant implications for cardiovascular function in healthy animals and for the development and progression of disease. This review summarizes what is currently known regarding the activity of these kinases, and particularly the role of GRK2 and GRK5 in the molecular alterations that occur during heart failure. This review further highlights areas of GRK regulation that remain poorly understood and how they may represent novel targets for therapeutic development.
Collapse
|
43
|
Liu GS, Gardner G, Adly G, Jiang M, Cai WF, Lam CK, Alogaili F, Robbins N, Rubinstein J, Kranias EG. A novel human S10F-Hsp20 mutation induces lethal peripartum cardiomyopathy. J Cell Mol Med 2018; 22:3911-3919. [PMID: 29761889 PMCID: PMC6050507 DOI: 10.1111/jcmm.13665] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 03/30/2018] [Indexed: 01/20/2023] Open
Abstract
Heat shock protein 20 (Hsp20) has been shown to be a critical regulator of cardiomyocyte survival upon cardiac stress. In this study, we investigated the functional significance of a novel human Hsp20 mutation (S10F) in peripartum cardiomyopathy. Previous findings showed that cardiac-specific overexpression of this mutant were associated with reduced autophagy, left ventricular dysfunction and early death in male mice. However, this study indicates that females have normal function with no alterations in autophagy but died within a week after 1-4 pregnancies. Further examination of mutant females revealed left ventricular chamber dilation and hypertrophic remodelling. Echocardiography demonstrated increases in left ventricular end-systolic volume and left ventricular end-diastolic volume, while ejection fraction and fractional shortening were depressed following pregnancy. Subsequent studies revealed that cardiomyocyte apoptosis was elevated in mutant female hearts after the third delivery, associated with decreases in the levels of Bcl-2/Bax and Akt phosphorylation. These results indicate that the human S10F mutant is associated with dysregulation of cell survival signalling, accelerated heart failure and early death post-partum.
Collapse
Affiliation(s)
- Guan-Sheng Liu
- Department of Pharmacology & Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - George Gardner
- Department of Pharmacology & Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - George Adly
- Department of Pharmacology & Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Min Jiang
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Wen-Feng Cai
- Department of Pathology & Lab Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Chi Keung Lam
- Department of Pharmacology & Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Fawzi Alogaili
- Department of Pharmacology & Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Nathan Robbins
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Jack Rubinstein
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Evangelia G Kranias
- Department of Pharmacology & Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA.,Molecular Biology Division, Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| |
Collapse
|
44
|
Jagodzińska A, Gondek A, Pietrzak B, Cudnoch-Jędrzejewska A, Mamcarz A, Wielgoś M. Peripartum cardiomyopathy - from pathogenesis to treatment. J Perinat Med 2018; 46:237-245. [PMID: 28489560 DOI: 10.1515/jpm-2016-0247] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- Aleksandra Jagodzińska
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Banacha 1b, 02-097 Warsaw, Poland
- First Department of Obstetrics and Gynecology, Medical University of Warsaw, Pl. Starynkiewicza 1/3, 02-015 Warsaw, Poland
| | - Agata Gondek
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Banacha 1b, 02-097 Warsaw, Poland
| | - Bronisława Pietrzak
- First Department of Obstetrics and Gynecology, Medical University of Warsaw, Pl. Starynkiewicza 1/3, 02-015 Warsaw, Poland
| | - Agnieszka Cudnoch-Jędrzejewska
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Banacha 1b, 02-097 Warsaw, Poland
| | - Artur Mamcarz
- Third Department of Internal Medicine and Cardiology Medical University of Warsaw, Solec 93, Warsaw, Poland
| | - Mirosław Wielgoś
- First Department of Obstetrics and Gynecology, Medical University of Warsaw, Pl. Starynkiewicza 1/3, 02-015 Warsaw, Poland
| |
Collapse
|
45
|
Dahl EF, Wu SC, Healy CL, Harsch BA, Shearer GC, O'Connell TD. Subcellular compartmentalization of proximal Gα q-receptor signaling produces unique hypertrophic phenotypes in adult cardiac myocytes. J Biol Chem 2018; 293:8734-8749. [PMID: 29610273 DOI: 10.1074/jbc.ra118.002283] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 03/13/2018] [Indexed: 12/12/2022] Open
Abstract
G protein-coupled receptors that signal through Gαq (Gq receptors), such as α1-adrenergic receptors (α1-ARs) or angiotensin receptors, share a common proximal signaling pathway that activates phospholipase Cβ1 (PLCβ1), which cleaves phosphatidylinositol 4,5-bisphosphate (PIP2) to produce inositol 1,4,5-trisphosphate (IP3) and diacylglycerol. Despite these common proximal signaling mechanisms, Gq receptors produce distinct physiological responses, yet the mechanistic basis for this remains unclear. In the heart, Gq receptors are thought to induce myocyte hypertrophy through a mechanism termed excitation-transcription coupling, which provides a mechanistic basis for compartmentalization of calcium required for contraction versus IP3-dependent intranuclear calcium required for hypertrophy. Here, we identified subcellular compartmentalization of Gq-receptor signaling as a mechanistic basis for unique Gq receptor-induced hypertrophic phenotypes in cardiac myocytes. We show that α1-ARs co-localize with PLCβ1 and PIP2 at the nuclear membrane. Further, nuclear α1-ARs induced intranuclear PLCβ1 activity, leading to histone deacetylase 5 (HDAC5) export and a robust transcriptional response (i.e. significant up- or down-regulation of 806 genes). Conversely, we found that angiotensin receptors localize to the sarcolemma and induce sarcolemmal PLCβ1 activity, but fail to promote HDAC5 nuclear export, while producing a transcriptional response that is mostly a subset of α1-AR-induced transcription. In summary, these results link Gq-receptor compartmentalization in cardiac myocytes to unique hypertrophic transcription. They suggest a new model of excitation-transcription coupling in adult cardiac myocytes that accounts for differential Gq-receptor localization and better explains distinct physiological functions of Gq receptors.
Collapse
Affiliation(s)
| | - Steven C Wu
- Integrative Biology and Physiology, University of Minnesota, Minneapolis, Minnesota 55455 and
| | - Chastity L Healy
- Integrative Biology and Physiology, University of Minnesota, Minneapolis, Minnesota 55455 and
| | - Brian A Harsch
- the Department of Nutritional Sciences, Pennsylvania State University, University Park, Pennsylvania 16802
| | - Gregory C Shearer
- the Department of Nutritional Sciences, Pennsylvania State University, University Park, Pennsylvania 16802
| | - Timothy D O'Connell
- Integrative Biology and Physiology, University of Minnesota, Minneapolis, Minnesota 55455 and
| |
Collapse
|
46
|
Kofron CM, Kim TY, King ME, Xie A, Feng F, Park E, Qu Z, Choi BR, Mende U. G q-activated fibroblasts induce cardiomyocyte action potential prolongation and automaticity in a three-dimensional microtissue environment. Am J Physiol Heart Circ Physiol 2017; 313:H810-H827. [PMID: 28710068 DOI: 10.1152/ajpheart.00181.2017] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Revised: 06/13/2017] [Accepted: 07/03/2017] [Indexed: 11/22/2022]
Abstract
Cardiac fibroblasts (CFs) are known to regulate cardiomyocyte (CM) function in vivo and in two-dimensional in vitro cultures. This study examined the effect of CF activation on the regulation of CM electrical activity in a three-dimensional (3-D) microtissue environment. Using a scaffold-free 3-D platform with interspersed neonatal rat ventricular CMs and CFs, Gq-mediated signaling was selectively enhanced in CFs by Gαq adenoviral infection before coseeding with CMs in nonadhesive hydrogels. After 3 days, the microtissues were analyzed by signaling assay, histological staining, quantitative PCR, Western blots, optical mapping with voltage- or Ca2+-sensitive dyes, and microelectrode recordings of CF resting membrane potential (RMPCF). Enhanced Gq signaling in CFs increased microtissue size and profibrotic and prohypertrophic markers. Expression of constitutively active Gαq in CFs prolonged CM action potential duration (by 33%) and rise time (by 31%), prolonged Ca2+ transient duration (by 98%) and rise time (by 65%), and caused abnormal electrical activity based on depolarization-induced automaticity. Constitutive Gq activation in CFs also depolarized RMPCF from -33 to -20 mV and increased connexin 43 and connexin 45 expression. Computational modeling confers that elevated RMPCF and increased cell-cell coupling between CMs and CFs in a 3-D environment could lead to automaticity. In conclusion, our data demonstrate that CF activation alone is capable of altering action potential and Ca2+ transient characteristics of CMs, leading to proarrhythmic electrical activity. Our results also emphasize the importance of a 3-D environment where cell-cell interactions are prevalent, underscoring that CF activation in 3-D tissue plays a significant role in modulating CM electrophysiology and arrhythmias.NEW & NOTEWORTHY In a three-dimensional microtissue model, which lowers baseline activation of cardiac fibroblasts but enables cell-cell, paracrine, and cell-extracellular matrix interactions, we demonstrate that selective cardiac fibroblast activation by enhanced Gq signaling, a pathophysiological trigger in the diseased heart, modulates cardiomyocyte electrical activity, leading to proarrhythmogenic automaticity.
Collapse
Affiliation(s)
- C M Kofron
- Cardiovascular Research Center, Cardiovascular Institute, Rhode Island Hospital and Alpert Medical School of Brown University, Providence, Rhode Island; and
| | - T Y Kim
- Cardiovascular Research Center, Cardiovascular Institute, Rhode Island Hospital and Alpert Medical School of Brown University, Providence, Rhode Island; and
| | - M E King
- Cardiovascular Research Center, Cardiovascular Institute, Rhode Island Hospital and Alpert Medical School of Brown University, Providence, Rhode Island; and
| | - A Xie
- Cardiovascular Research Center, Cardiovascular Institute, Rhode Island Hospital and Alpert Medical School of Brown University, Providence, Rhode Island; and
| | - F Feng
- Cardiovascular Research Center, Cardiovascular Institute, Rhode Island Hospital and Alpert Medical School of Brown University, Providence, Rhode Island; and
| | - E Park
- Cardiovascular Research Center, Cardiovascular Institute, Rhode Island Hospital and Alpert Medical School of Brown University, Providence, Rhode Island; and
| | - Z Qu
- Department of Medicine, University of California, Los Angeles, California
| | - B-R Choi
- Cardiovascular Research Center, Cardiovascular Institute, Rhode Island Hospital and Alpert Medical School of Brown University, Providence, Rhode Island; and
| | - U Mende
- Cardiovascular Research Center, Cardiovascular Institute, Rhode Island Hospital and Alpert Medical School of Brown University, Providence, Rhode Island; and
| |
Collapse
|
47
|
He Z, Yang Y, Wen Z, Chen C, Xu X, Zhu Y, Wang Y, Wang DW. CYP2J2 metabolites, epoxyeicosatrienoic acids, attenuate Ang II-induced cardiac fibrotic response by targeting Gα 12/13. J Lipid Res 2017; 58:1338-1353. [PMID: 28554983 DOI: 10.1194/jlr.m074229] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 05/23/2017] [Indexed: 12/23/2022] Open
Abstract
The arachidonic acid-cytochrome P450 2J2-epoxyeicosatrienoic acid (AA-CYP2J2-EET) metabolic pathway has been identified to be protective in the cardiovascular system. This study explored the effects of the AA-CYP2J2-EET metabolic pathway on cardiac fibrosis from the perspective of cardiac fibroblasts and underlying mechanisms. In in vivo studies, 8-week-old male CYP2J2 transgenic mice (aMHC-CYP2J2-Tr) and littermates were infused with angiotensin II (Ang II) or saline for 2 weeks. Results showed that CYP2J2 overexpression increased EET production. Meanwhile, impairment of cardiac function and fibrotic response were attenuated by CYP2J2 overexpression. The effects of CYP2J2 were associated with reduced activation of the α subunits of G12 family G proteins (Gα12/13)/RhoA/Rho kinase (ROCK) cascade and elevation of the NO/cyclic guanosine monophosphate (cGMP) level in cardiac tissue. In in vitro studies, cardiac fibroblast activation, proliferation, migration, and collagen production induced by Ang II were associated with activation of the Gα12/13/RhoA/ROCK pathway, which was inhibited by exogenous 11,12-EET. Moreover, silencing of Gα12/13 or RhoA exerted similar effects as 11,12-EET. Furthermore, inhibitory effects of 11,12-EET on Gα12/13 were blocked by NO/cGMP pathway inhibitors. Our findings indicate that enhancement of the AA-CYP2J2-EET metabolic pathway by CYP2J2 overexpression attenuates Ang II-induced cardiac dysfunction and fibrosis by reducing the fibrotic response of cardiac fibroblasts by targeting the Gα12/13/RhoA/ROCK pathway via NO/cGMP signaling.
Collapse
Affiliation(s)
- Zuowen He
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Yong Yang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Zheng Wen
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Chen Chen
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Xizhen Xu
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Yanfang Zhu
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Yan Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Dao Wen Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China.
| |
Collapse
|
48
|
Pressure-overload-induced angiotensin-mediated early remodeling in mouse heart. PLoS One 2017; 12:e0176713. [PMID: 28464037 PMCID: PMC5413013 DOI: 10.1371/journal.pone.0176713] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Accepted: 04/15/2017] [Indexed: 11/19/2022] Open
Abstract
Our previous work on angiotensin II-mediated electrical-remodeling in canine left ventricle, in connection with a long history of other studies, suggested the hypothesis: increases in mechanical load induce autocrine secretion of angiotensin II (A2), which coherently regulates a coterie of membrane ion transporters in a manner that increases contractility. However, the relation between load and A2 secretion was correlative. We subsequently showed a similar or identical system was present in murine heart. To investigate whether the relation between mechanical load and A2-mediated electrical remodeling was causal, we employed transverse aortic constriction in mice to subject the left ventricle to pressure overload for short-term (1 to 2 days) or long-term (1 to 2 weeks) periods. Heart-to-body weight ratios and cell capacitance measurements were used to determine hypertrophy. Whole-cell patch clamp recordings of the predominant repolarization currents Ito,fast and IK,slow were used to assess electrical remodeling. Hearts or myocytes subjected to long-term load displayed significant hypertrophy, which was not evident in short-term load. However, short-term load induced significant reductions in Ito,fast and IK,slow. Incubation of these myocytes with the angiotensin II type 1 receptor inhibitor saralasin for 2 hours restored Ito,fast and IK,slow to control levels. The number of Ito.fast or IK,slow channels did not change with A2 or long-term load, however the hypertrophic increase in membrane area reduced the current densities for both channels. For Ito,fast but not IK,slow there was an additional reduction that was reversed by inhibition of angiotensin receptors. These results suggest increased load activates an endogenous renin angiotensin system that initially reduces Ito,fast and IK,slow prior to the onset of hypertrophic growth. However, there are functional interactions between electrical and anatomical remodeling. First, hypertrophy tends to reduce all current densities. Second, the hypertrophic program can modify signaling between the angiotensin receptor and target current.
Collapse
|
49
|
Gαq Signaling in the Regulation of Autophagy and Heart Failure. J Cardiovasc Pharmacol 2017; 69:212-214. [PMID: 28166086 DOI: 10.1097/fjc.0000000000000471] [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: 11/26/2022]
|
50
|
Yung BS, Brand CS, Xiang SY, Gray CBB, Means CK, Rosen H, Chun J, Purcell NH, Brown JH, Miyamoto S. Selective coupling of the S1P 3 receptor subtype to S1P-mediated RhoA activation and cardioprotection. J Mol Cell Cardiol 2017; 103:1-10. [PMID: 28017639 PMCID: PMC5410967 DOI: 10.1016/j.yjmcc.2016.12.008] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 12/16/2016] [Accepted: 12/19/2016] [Indexed: 01/17/2023]
Abstract
Sphingosine-1-phosphate (S1P), a bioactive lysophospholipid, is generated and released at sites of tissue injury in the heart and can act on S1P1, S1P2, and S1P3 receptor subtypes to affect cardiovascular responses. We established that S1P causes little phosphoinositide hydrolysis and does not induce hypertrophy indicating that it does not cause receptor coupling to Gq. We previously demonstrated that S1P confers cardioprotection against ischemia/reperfusion by activating RhoA and its downstream effector PKD. The S1P receptor subtypes and G proteins that regulate RhoA activation and downstream responses in the heart have not been determined. Using siRNA or pertussis toxin to inhibit different G proteins in NRVMs we established that S1P regulates RhoA activation through Gα13 but not Gα12, Gαq, or Gαi. Knockdown of the three major S1P receptors using siRNA demonstrated a requirement for S1P3 in RhoA activation and subsequent phosphorylation of PKD, and this was confirmed in studies using isolated hearts from S1P3 knockout (KO) mice. S1P treatment reduced infarct size induced by ischemia/reperfusion in Langendorff perfused wild-type (WT) hearts and this protection was abolished in the S1P3 KO mouse heart. CYM-51736, an S1P3-specific agonist, also decreased infarct size after ischemia/reperfusion to a degree similar to that achieved by S1P. The finding that S1P3 receptor- and Gα13-mediated RhoA activation is responsible for protection against ischemia/reperfusion suggests that selective targeting of S1P3 receptors could provide therapeutic benefits in ischemic heart disease.
Collapse
Affiliation(s)
- Bryan S Yung
- Department of Pharmacology, School of Medicine, University of California, San Diego, La Jolla, CA 92093, United States
| | - Cameron S Brand
- Department of Pharmacology, School of Medicine, University of California, San Diego, La Jolla, CA 92093, United States
| | - Sunny Y Xiang
- Department of Pharmacology, School of Medicine, University of California, San Diego, La Jolla, CA 92093, United States
| | - Charles B B Gray
- Department of Pharmacology, School of Medicine, University of California, San Diego, La Jolla, CA 92093, United States
| | | | - Hugh Rosen
- Department of Chemical Physiology, Scripps Research Institute, La Jolla, CA 92037, United States
| | - Jerold Chun
- Department of Molecular and Cellular Neuroscience, Dorris Neuroscience Center, Scripps Research Institute, La Jolla, CA 92037, United States
| | - Nicole H Purcell
- Department of Pharmacology, School of Medicine, University of California, San Diego, La Jolla, CA 92093, United States
| | - Joan Heller Brown
- Department of Pharmacology, School of Medicine, University of California, San Diego, La Jolla, CA 92093, United States.
| | - Shigeki Miyamoto
- Department of Pharmacology, School of Medicine, University of California, San Diego, La Jolla, CA 92093, United States.
| |
Collapse
|