1
|
Koopmans T, van Rooij E. Molecular gatekeepers of endogenous adult mammalian cardiomyocyte proliferation. Nat Rev Cardiol 2025:10.1038/s41569-025-01145-y. [PMID: 40195566 DOI: 10.1038/s41569-025-01145-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/10/2025] [Indexed: 04/09/2025]
Abstract
Irreversible cardiac fibrosis, cardiomyocyte death and chronic cardiac dysfunction after myocardial infarction pose a substantial global health-care challenge, with no curative treatments available. To regenerate the injured heart, cardiomyocytes must proliferate to replace lost myocardial tissue - a capability that adult mammals have largely forfeited to adapt to the demanding conditions of life. Using various preclinical models, our understanding of cardiomyocyte proliferation has progressed remarkably, leading to the successful reactivation of cell cycle induction in adult animals, with functional recovery after cardiac injury. Central to this success is the targeting of key pathways and structures that drive cardiomyocyte maturation after birth - nucleation and ploidy, sarcomere structure, developmental signalling, chromatin and epigenetic regulation, the microenvironment and metabolic maturation - forming a complex regulatory framework that allows efficient cellular contraction but restricts cardiomyocyte proliferation. In this Review, we explore the molecular pathways underlying these core mechanisms and how their manipulation can reactivate the cell cycle in cardiomyocytes, potentially contributing to cardiac repair.
Collapse
Affiliation(s)
- Tim Koopmans
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, Netherlands
| | - Eva van Rooij
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, Netherlands.
- Department of Cardiology, University Medical Center Utrecht, Utrecht, Netherlands.
| |
Collapse
|
2
|
Sim G, Ok SH, Lee SH, Park KE, Park S, Sohn JT. Lipid Emulsion Mitigates the Cardiotoxic Effects of Labetalol in Rat Cardiomyoblasts. Cells 2025; 14:187. [PMID: 39936979 PMCID: PMC11816916 DOI: 10.3390/cells14030187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 01/21/2025] [Accepted: 01/23/2025] [Indexed: 02/13/2025] Open
Abstract
Lipid emulsion has recently emerged as an effective agent for improving the cardiotoxicity of highly lipophilic drugs. However, its effect on cardiotoxicity induced by labetalol, a nonselective beta-blocker, remains unknown. In this study, we investigated the effects of lipid emulsion on the cardiotoxicity of labetalol in rat cardiomyoblasts and tried to decipher the underlying mechanisms. The effects of lipid emulsion on labetalol-induced changes in cell viability, expression of Bax/Bcl-2, cleaved caspase-3, and cleaved caspase-9, and phosphorylation of GSK-3β, Akt, and PI3K were examined. Lipid emulsion inhibited labetalol-induced decrease in cell viability, whereas LY294002, MK2206, and SB216763, the inhibitors of phosphoinositide 3-kinase (PI3K), Akt, glycogen synthase kinase-3β (GSK-3β), respectively, partially attenuated this restoration of cell viability. Lipid emulsion reversed the increase in expression of cleaved caspase-3, cleaved caspase-9, and Bax/Bcl-2 and decrease in the phosphorylation of GSK-3β, Akt, and PI3K by labetalol. Lipid emulsion and cyclosporin, a mitochondrial permeability transition pore (MPTP) inhibitor, reduced the labetalol-induced increase in the number of TUNEL-positive cells and promoted late-stage apoptosis. Overall, lipid emulsion inhibited apoptotic cell death caused by labetalol toxicity via the inhibition of intrinsic apoptotic pathway and MPTP in rat cardiomyoblasts, which appears to involve PI3K, Akt, and GSK-3β signaling pathways.
Collapse
Affiliation(s)
- Gyujin Sim
- Department of Anesthesiology and Pain Medicine, Gyeongsang National University Hospital, Jinju-si 52727, Republic of Korea
- Institute of Medical Science, Gyeongsang National University, Jinju-si 52727, Republic of Korea
| | - Seong-Ho Ok
- Institute of Medical Science, Gyeongsang National University, Jinju-si 52727, Republic of Korea
- Department of Anesthesiology and Pain Medicine, Gyeongsang National University Changwon Hospital, Changwon-si 51472, Republic of Korea
- Department of Anesthesiology and Pain Medicine, Gyeongsang National University College of Medicine, Jinju-si 52727, Republic of Korea
| | - Soo Hee Lee
- Institute of Medical Science, Gyeongsang National University, Jinju-si 52727, Republic of Korea
- Department of Anesthesiology and Pain Medicine, Gyeongsang National University Changwon Hospital, Changwon-si 51472, Republic of Korea
- Department of Anesthesiology and Pain Medicine, Gyeongsang National University College of Medicine, Jinju-si 52727, Republic of Korea
| | - Kyeong-Eon Park
- Department of Anesthesiology and Pain Medicine, Gyeongsang National University College of Medicine, Gyeongsang National University Hospital, Jinju-si 52727, Republic of Korea
| | - Seunghyeon Park
- Department of Anesthesiology and Pain Medicine, Gyeongsang National University Hospital, Jinju-si 52727, Republic of Korea
| | - Ju-Tae Sohn
- Institute of Medical Science, Gyeongsang National University, Jinju-si 52727, Republic of Korea
- Department of Anesthesiology and Pain Medicine, Gyeongsang National University College of Medicine, Gyeongsang National University Hospital, Jinju-si 52727, Republic of Korea
| |
Collapse
|
3
|
Mousavi SM, Jalali-Zefrei F, Shourmij M, Tabaghi S, Davari A, Khalili SB, Farzipour S, Salari A. Targeting Wnt Pathways with Small Molecules as New Approach in Cardiovascular Disease. Curr Cardiol Rev 2025; 21:108-122. [PMID: 39482911 PMCID: PMC12060913 DOI: 10.2174/011573403x333038241023153349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 10/01/2024] [Accepted: 10/04/2024] [Indexed: 11/03/2024] Open
Abstract
The increasing incidences of morbidity and mortality associated with cardiovascular diseases represent significant difficulties for clinical treatment and have a major impact on patient health. Wnt signaling pathways are highly conserved and are well known for their regulatory roles in embryonic development, tissue regeneration, and adult tissue homeostasis. Wnt signaling is classified into two distinct pathways: canonical Wnt/β-catenin signaling and noncanonical pathways, including planar cell polarity and Wnt/Ca2+ pathways. A growing body of experimental evidence suggests the involvement of both canonical and non-canonical Wnt signaling pathways in the development of cardiovascular diseases, including myocardial hypertrophy, arrhythmias, diabetic cardiomyopathy, arrhythmogenic cardiomyopathy, and myocardial infarction. Thus, to enhance patient quality of life, diagnosing and treating cardiac illnesses may require a thorough understanding of the molecular functions played by the Wnt pathway in these disorders. Many small-molecule inhibitors specifically target various components within the Wnt signaling pathways, such as Frizzled, Disheveled, Porcupine, and Tankyrase. This study aims to present an overview of the latest findings regarding the functions of Wnt signaling in human cardiac disorders and possible inhibitors of Wnt, which could lead to novel approaches for treating cardiac ailments.
Collapse
Affiliation(s)
- Seyed Mehdi Mousavi
- Cardiovascular Diseases Research Center, Department of Cardiology, School of Medicine, Heshmat Hospital, Guilan University of Medical Sciences, Rasht, Iran
| | - Fatemeh Jalali-Zefrei
- Department of radiology, Faculty of Medicine, Guilan University of Medical Science, Rasht, Iran
| | - Mohammad Shourmij
- Razi Herbal Medicines Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Shiva Tabaghi
- Cardiovascular Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amirhesam Davari
- Cardiovascular Diseases Research Center, Department of Cardiology, School of Medicine, Heshmat Hospital, Guilan University of Medical Sciences, Rasht, Iran
| | - Saeed Bahador Khalili
- Department of Electronic Engineering, Universitat Rovira i Virgili, Tarragona, 43007, Spain
| | - Soghra Farzipour
- Cardiovascular Diseases Research Center, Department of Cardiology, School of Medicine, Heshmat Hospital, Guilan University of Medical Sciences, Rasht, Iran
| | - Arsalan Salari
- Cardiovascular Diseases Research Center, Department of Cardiology, School of Medicine, Heshmat Hospital, Guilan University of Medical Sciences, Rasht, Iran
| |
Collapse
|
4
|
Alotaiq N, Khalifa AS, Youssef A, El-Nagar EG, Elwali NE, Habib HM, AlZaim I, Eid AH, Bakkar NMZ, El-Yazbi AF. Targeting GSK-3β for adipose dysfunction and cardiovascular complications of metabolic disease: An entangled WNT/β-catenin question. FASEB J 2024; 38:e70273. [PMID: 39726401 DOI: 10.1096/fj.202402470r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 12/10/2024] [Accepted: 12/17/2024] [Indexed: 12/28/2024]
Abstract
Individuals with metabolic syndrome have a high risk of developing cardiovascular disorders that is closely tied to visceral adipose tissue dysfunction, as well as an altered interaction between adipose tissue and the cardiovascular system. In metabolic syndrome, adipose tissue dysfunction is associated with increased hypertrophy, reduced vascularization, and hypoxia of adipocytes, leading to a pro-oxidative and pro-inflammatory environment. Among the pathways regulating adipose tissue homeostasis is the wingless-type mammary tumor virus integration site family (Wnt) signaling pathway, with both its canonical and non-canonical arms. Various modulators of the Wnt signaling have been identified to contribute to the development of metabolic diseases and their cardiovascular complications, with a particularly significant role played by Glycogen Synthase Kinase-3β (GSK-3β). GSK-3β levels and activities have various and often contrasting roles in obesity and related metabolic disorders, as well as their cardiovascular sequelae. Here, we explore the possibility that altered Wnt signaling and GSK-3β activities could serve as a connection between adipose tissue dysfunction and the development of cardiovascular disease in individuals with metabolic syndrome. We attempt to define a context-specific approach for intervention, which could possibly serve as a novel disease modifying therapy for the mitigation of such complications.
Collapse
Affiliation(s)
- Nasser Alotaiq
- Health Sciences Research Center, Imam Muhammad Ibn Saud Islamic University (IMISIU), Riyadh, Kingdom of Saudi Arabia
| | - Ahmed S Khalifa
- Faculty of Pharmacy, Alamein International University, Alamein, Egypt
| | - Amr Youssef
- Faculty of Pharmacy, Alamein International University, Alamein, Egypt
| | - Esraa G El-Nagar
- Faculty of Pharmacy, Alamein International University, Alamein, Egypt
| | - Nasr Eldin Elwali
- Deanship of Scientific Research, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Kingdom of Saudi Arabia
| | - Hosam M Habib
- Research & Innovation Hub, Alamein International University, Alamein, Egypt
| | - Ibrahim AlZaim
- Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Ali H Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | | | - Ahmed F El-Yazbi
- Faculty of Pharmacy, Alamein International University, Alamein, Egypt
- Research & Innovation Hub, Alamein International University, Alamein, Egypt
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| |
Collapse
|
5
|
Bai Y, Li R, Hao JF, Chen LW, Liu ST, Zhang XL, Lip GYH, Yang JK, Zou YX, Wang H. Accumulated β-catenin is associated with human atrial fibrosis and atrial fibrillation. J Transl Med 2024; 22:734. [PMID: 39103891 PMCID: PMC11302159 DOI: 10.1186/s12967-024-05558-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 07/29/2024] [Indexed: 08/07/2024] Open
Abstract
BACKGROUND Atrial fibrillation (AF) is associated with increased risk of stroke and mortality. It has been reported that the process of atrial fibrosis was regulated by β-catenin in rats with AF. However, pathophysiological mechanisms of this process in human with AF remain unclear. This study aims to investigate the possible mechanisms of β-catenin in participating in the atrial fibrosis using human right atrial appendage (hRAA) tissues . METHODS We compared the difference of β-catenin expression in hRAA tissues between the patients with AF and sinus rhythm (SR). The possible function of β-catenin in the development of AF was also explored in mice and primary cells. RESULTS Firstly, the space between the membrane of the gap junctions of cardiomyocytes was wider in the AF group. Secondly, the expression of the gap junction function related proteins, Connexin40 and Connexin43, was decreased, while the expression of β-catenin and its binding partner E-cadherin was increased in hRAA and cardiomyocytes of the AF group. Thirdly, β-catenin colocalized with E-cadherin on the plasma membrane of cardiomyocytes in the SR group, while they were dissociated and accumulated intracellularly in the AF group. Furthermore, the expression of glycogen synthase kinase 3β (GSK-3β) and Adenomatous Polyposis Coli (APC), which participated in the degradation of β-catenin, was decreased in hRAA tissues and cardiomyocytes of the AF group. Finally, the development of atrial fibrosis and AF were proved to be prevented after inhibiting β-catenin expression in the AF model mice. CONCLUSIONS Based on human atrial pathological and molecular analyses, our findings provided evidence that β-catenin was associated with atrial fibrosis and AF progression.
Collapse
Affiliation(s)
- Ying Bai
- Cardiovascular Center, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Rui Li
- Department of Pathology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Jun-Feng Hao
- Core Facility for Protein Research, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Lian-Wan Chen
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Si-Tong Liu
- Cardiovascular Center, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Xi-Lin Zhang
- Cardiovascular Center, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Gregory Y H Lip
- Liverpool Centre for Cardiovascular Science at University of Liverpool, Liverpool John Moores University and Liverpool Heart & Chest Hospital, Liverpool, UK
- Danish Center for Health Services Research, Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Jin-Kui Yang
- Department of Endocrinology, Beijing Key Laboratory of Diabetes Research and Care, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Yi-Xi Zou
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China.
| | - Hao Wang
- Beijing Key Laboratory of Diabetes Research and Care, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China.
| |
Collapse
|
6
|
Alkhatib B, Ciarelli J, Ghnenis A, Pallas B, Olivier N, Padmanabhan V, Vyas AK. Early- to mid-gestational testosterone excess leads to adverse cardiac outcomes in postpartum sheep. Am J Physiol Heart Circ Physiol 2024; 327:H315-H330. [PMID: 38819385 PMCID: PMC11687963 DOI: 10.1152/ajpheart.00763.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 05/13/2024] [Accepted: 05/22/2024] [Indexed: 06/01/2024]
Abstract
Cardiovascular dysfunctions complicate 10-20% of pregnancies, increasing the risk for postpartum mortality. Various gestational insults, including preeclampsia are reported to be associated with adverse maternal cardiovascular outcomes. One such insult, gestational hyperandrogenism increases the risk for preeclampsia and other gestational morbidities but its impact on postpartum maternal health is not well known. We hypothesize that gestational hyperandrogenism such as testosterone (T) excess will adversely impact the maternal heart in the postpartum period. Pregnant ewes were injected with T propionate from day 30 to day 90 of gestation (term 147 days). Three months postpartum, echocardiograms, plasma cytokine profiles, cardiac morphometric, and molecular analysis were conducted [control (C) n = 6, T-treated (T) n = 7 number of animals]. Data were analyzed by two-tailed Student's t test and Cohen's effect size (d) analysis. There was a nonsignificant large magnitude decrease in cardiac output (7.64 ± 1.27 L/min vs. 10.19 ± 1.40, P = 0.22, d = 0.81) and fractional shortening in the T ewes compared with C (35.83 ± 2.33% vs. 41.50 ± 2.84, P = 0.15, d = 0.89). T treatment significantly increased 1) left ventricle (LV) weight-to-body weight ratio (2.82 ± 0.14 g/kg vs. 2.46 ± 0.08) and LV thickness (14.56 ± 0.52 mm vs. 12.50 ± 0.75), 2) proinflammatory marker [tumor necrosis factor-alpha (TNF-α)] in LV (1.66 ± 0.35 vs. 1.06 ± 0.18), 3) LV collagen (Masson's Trichrome stain: 3.38 ± 0.35 vs. 1.49 ± 0.15 and Picrosirius red stain: 5.50 ± 0.32 vs. 3.01 ± 0.23), 4) markers of LV apoptosis, including TUNEL (8.3 ± 1.1 vs. 0.9 ± 0.18), bcl-2-associated X protein (Bax)+-to-b-cell lymphoma 2 (Bcl2)+ ratio (0.68 ± 0.30 vs. 0.13 ± 0.02), and cleaved caspase 3 (15.4 ± 1.7 vs. 4.4 ± 0.38). These findings suggest that gestational testosterone excess adversely programs the maternal LV, leading to adverse structural and functional consequences in the postpartum period.NEW & NOTEWORTHY Using a sheep model of human translational relevance, this study provides evidence that excess gestational testosterone exposure such as that seen in hyperandrogenic disorders adversely impacts postpartum maternal hearts.
Collapse
Affiliation(s)
- Bashar Alkhatib
- Department of Pediatrics, Washington University, St Louis, Missouri, United States
| | - Joseph Ciarelli
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan, United States
| | - Adel Ghnenis
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan, United States
| | - Brooke Pallas
- Unit for Laboratory Animal Medicine, University of Michigan, Ann Arbor, Michigan, United States
| | - Nicholas Olivier
- Department of Veterinary Medicine, Michigan State University, Lansing, Michigan, United States
| | - Vasantha Padmanabhan
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan, United States
| | - Arpita Kalla Vyas
- Department of Pediatrics, Washington University, St Louis, Missouri, United States
| |
Collapse
|
7
|
Krishnan V, Atanasova N, Aujla PK, Hupka D, Owen CA, Kassiri Z. Loss of ADAM15 in female mice does not worsen pressure overload cardiomyopathy, independent of ovarian hormones. Am J Physiol Heart Circ Physiol 2024; 327:H409-H416. [PMID: 38607341 DOI: 10.1152/ajpheart.00116.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 04/03/2024] [Accepted: 04/04/2024] [Indexed: 04/13/2024]
Abstract
Cardiac hypertrophy is a common feature in several cardiomyopathies. We previously reported that loss of ADAM15 (disintegrin and metalloproteinase 15) worsened cardiac hypertrophy and dilated cardiomyopathy following cardiac pressure overload. Here, we investigated the impact of ADAM15 loss in female mice following cardiac pressure overload induced by transverse aortic constriction (TAC). Female Adam15-/- mice developed the same degree of cardiac hypertrophy, dilation, and dysfunction as the parallel female wild-type (WT) mice at 6 wk post-TAC. To determine if this is due to the protective effects of estrogen, which could mask the negative impact of Adam15 loss, WT and Adam15-/- mice underwent ovariectomy (OVx) 2 wk before TAC. Cardiac structure and function analyses were performed at 6 wk post-TAC. OVx similarly impacted females of both genotypes post-TAC. Calcineurin (Cn) activity was increased post-OVx-TAC, and more in Adam15-/- mice; however, this increase was not reflected in the total-to-phospho-NFAT levels. Integrin-α7 expression, which was upstream of Cn activation in male Adam15-/- -TAC mice, remained unchanged in female mice. However, activation of the mitogen-activated protein kinases (ERK, JNK, P38) was greater in Adam15-/--OVx-TAC than in WT-OVx-TAC mice. In addition, ADAM15 protein levels were significantly increased post-TAC in male but not in female WT mice. Myocardial fibrosis was comparable in non-OVx WT-TAC and Adam15-/- -TAC mice. OVx increased the perivascular fibrosis more in Adam15-/- compared with WT mice post-TAC. Our data demonstrate that loss of ovarian hormones did not fully replicate the male phenotype in the female Adam15-/- mice post-TAC. As ADAM15 levels were increased in males but not in females post-TAC, it is plausible that ADAM15 does not play a prominent role in post-TAC events in female mice. Our findings highlight the significance of factors other than sex hormones in mediating cardiomyopathies in females, which require a more thorough understanding.NEW & NOTEWORTHY Loss of ADAM15 in female mice, unlike the male mice, does not worsen the cardiomyopathy following cardiac pressure overload. Ovariectomy does not worsen the post-TAC cardiomyopathy in female Adam15-/- mice compared with female WT mice. Lack of deleterious impact of Adam15 deficiency in female mice is not because of the protective effects of ovarian hormones but could be due to a less prominent role of ADAM15 in cardiac response to post-TAC remodeling in female mice.
Collapse
Affiliation(s)
- Vidhya Krishnan
- Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Nikki Atanasova
- Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Preetinder K Aujla
- Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Devon Hupka
- Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Caroline A Owen
- Brigham and Women's Hospital, Boston, Massachusetts, United States
- Harvard Medical School, Boston, Massachusetts, United States
| | - Zamaneh Kassiri
- Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
8
|
Boengler K, Eickelmann C, Kleinbongard P. Mitochondrial Kinase Signaling for Cardioprotection. Int J Mol Sci 2024; 25:4491. [PMID: 38674076 PMCID: PMC11049936 DOI: 10.3390/ijms25084491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 04/15/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
Myocardial ischemia/reperfusion injury is reduced by cardioprotective adaptations such as local or remote ischemic conditioning. The cardioprotective stimuli activate signaling cascades, which converge on mitochondria and maintain the function of the organelles, which is critical for cell survival. The signaling cascades include not only extracellular molecules that activate sarcolemmal receptor-dependent or -independent protein kinases that signal at the plasma membrane or in the cytosol, but also involve kinases, which are located to or within mitochondria, phosphorylate mitochondrial target proteins, and thereby modify, e.g., respiration, the generation of reactive oxygen species, calcium handling, mitochondrial dynamics, mitophagy, or apoptosis. In the present review, we give a personal and opinionated overview of selected protein kinases, localized to/within myocardial mitochondria, and summarize the available data on their role in myocardial ischemia/reperfusion injury and protection from it. We highlight the regulation of mitochondrial function by these mitochondrial protein kinases.
Collapse
Affiliation(s)
- Kerstin Boengler
- Institute of Physiology, Justus-Liebig University, 35392 Giessen, Germany
| | - Chantal Eickelmann
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, 45147 Essen, Germany; (C.E.); (P.K.)
| | - Petra Kleinbongard
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, 45147 Essen, Germany; (C.E.); (P.K.)
| |
Collapse
|
9
|
Zhan X, Yang Y, Li Q, He F. The role of deubiquitinases in cardiac disease. Expert Rev Mol Med 2024; 26:e3. [PMID: 38525836 PMCID: PMC11062144 DOI: 10.1017/erm.2024.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 12/10/2023] [Accepted: 12/28/2023] [Indexed: 03/26/2024]
Abstract
Deubiquitinases are a group of proteins that identify and digest monoubiquitin chains or polyubiquitin chains attached to substrate proteins, preventing the substrate protein from being degraded by the ubiquitin-proteasome system. Deubiquitinases regulate cellular autophagy, metabolism and oxidative stress by acting on different substrate proteins. Recent studies have revealed that deubiquitinases act as a critical regulator in various cardiac diseases, and control the onset and progression of cardiac disease through a board range of mechanism. This review summarizes the function of different deubiquitinases in cardiac disease, including cardiac hypertrophy, myocardial infarction and diabetes mellitus-related cardiac disease. Besides, this review briefly recapitulates the role of deubiquitinases modulators in cardiac disease, providing the potential therapeutic targets in the future.
Collapse
Affiliation(s)
- Xiaona Zhan
- Department of Nephrology, Tongji Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Yi Yang
- Department of Nephrology, Tongji Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Qing Li
- Department of Nephrology, Tongji Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Fan He
- Department of Nephrology, Tongji Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| |
Collapse
|
10
|
Shi YJ, Yang CG, Qiao WB, Liu YC, Liu SY, Dong GJ. Sacubitril/valsartan attenuates myocardial inflammation, hypertrophy, and fibrosis in rats with heart failure with preserved ejection fraction. Eur J Pharmacol 2023; 961:176170. [PMID: 37939991 DOI: 10.1016/j.ejphar.2023.176170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 10/16/2023] [Accepted: 10/26/2023] [Indexed: 11/10/2023]
Abstract
Heart failure with preserved ejection fraction (HFpEF) represents a multifaceted syndrome related to complex pathologic mechanisms. Sacubitril/valsartan (Sac/val) has demonstrated therapeutic efficacy in HFpEF treatment. However, additional research is required to elucidate its pharmacological mechanisms. Accordingly, this study aimed to explore the potential therapeutic effects of Sac/val in HFpEF rats and the underlying molecular mechanisms. In this study, rats with HFpEF were induced by subjecting spontaneously hypertensive rats to a diet rich in fats, salts, and sugars, along with administering streptozotocin. Subsequently, they were administered Sac/val at a daily dosage of 18 mg/kg. Finally, cardiac structure and function were assessed using echocardiography; Hematoxylin and eosin staining and Masson's trichrome staining were employed to evaluate the pathological changes; Quantitative real-time polymerase chain reaction and Western blot analysis were conducted to determine the expression of pertinent mRNA and proteins. Sac/val treatment attenuated left ventricular (LV) remodeling and diastolic dysfunction in HFpEF rats, possibly related to its anti-inflammatory, anti-hypertrophic, and anti-fibrotic efficacy. Mechanistically, Sac/val might inhibit inflammation by down-regulating cell adhesion molecule (intercellular adhesion molecule-1 (ICAM-1) and vascular endothelial cell adhesion molecule-1 (VCAM-1)) expression. Additionally, it blocked the phosphorylation of glycogen synthase kinase 3β (GSK-3β) to prevent cardiomyocyte hypertrophy. Furthermore, it effectively suppressed myocardial fibrosis by inhibiting the transforming growth factor-beta1 (TGF-β1)/Smads pathway. Our findings suggest that Sac/val improved LV remodeling and diastolic dysfunction, potentially attributed to its anti-inflammatory, anti-hypertrophic, and anti-fibrotic effects. These results provide a sound theoretical rationale for the clinical application of Sac/val in patients with HFpEF.
Collapse
Affiliation(s)
- Yu Jiao Shi
- Department of Cardiovascular Internal Medicine, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Chen Guang Yang
- Department of Cardiovascular Internal Medicine, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Wen Bo Qiao
- Department of Cardiovascular Internal Medicine, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Yong Cheng Liu
- Department of Cardiovascular Internal Medicine, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Si Yu Liu
- Department of Cardiovascular Internal Medicine, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Guo Ju Dong
- Department of Cardiovascular Internal Medicine, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China; National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China.
| |
Collapse
|
11
|
Umbarkar P, Ruiz Ramirez SY, Toro Cora A, Tousif S, Lal H. GSK-3 at the heart of cardiometabolic diseases: Isoform-specific targeting is critical to therapeutic benefit. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166724. [PMID: 37094727 PMCID: PMC10247467 DOI: 10.1016/j.bbadis.2023.166724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 04/14/2023] [Accepted: 04/18/2023] [Indexed: 04/26/2023]
Abstract
Glycogen synthase kinase-3 (GSK-3) is a family of serine/threonine kinases. The GSK-3 family has 2 isoforms, GSK-3α and GSK-3β. The GSK-3 isoforms have been shown to play overlapping as well as isoform-specific-unique roles in both, organ homeostasis and the pathogenesis of multiple diseases. In the present review, we will particularly focus on expanding the isoform-specific role of GSK-3 in the pathophysiology of cardiometabolic disorders. We will highlight recent data from our lab that demonstrated the critical role of cardiac fibroblast (CF) GSK-3α in promoting injury-induced myofibroblast transformation, adverse fibrotic remodeling, and deterioration of cardiac function. We will also discuss studies that found the exact opposite role of CF-GSK-3β in cardiac fibrosis. We will review emerging studies with inducible cardiomyocyte (CM)-specific as well as global isoform-specific GSK-3 KOs that demonstrated inhibition of both GSK-3 isoforms provides benefits against obesity-associated cardiometabolic pathologies. The underlying molecular interactions and crosstalk among GSK-3 and other signaling pathways will be discussed. We will briefly review the specificity and limitations of the available small molecule inhibitors targeting GSK-3 and their potential applications to treat metabolic disorders. Finally, we will summarize these findings and offer our perspective on envisioning GSK-3 as a therapeutic target for the management of cardiometabolic diseases.
Collapse
Affiliation(s)
- Prachi Umbarkar
- Division of Cardiovascular Disease, The University of Alabama at Birmingham, Birmingham, AL, USA.
| | - Sulivette Y Ruiz Ramirez
- Division of Cardiovascular Disease, The University of Alabama at Birmingham, Birmingham, AL, USA.
| | - Angelica Toro Cora
- Division of Cardiovascular Disease, The University of Alabama at Birmingham, Birmingham, AL, USA.
| | - Sultan Tousif
- Division of Cardiovascular Disease, The University of Alabama at Birmingham, Birmingham, AL, USA.
| | - Hind Lal
- Division of Cardiovascular Disease, The University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
12
|
Shi Y, Liu C, Yang C, Qiao W, Liu Y, Liu S, Dong G. A rat model of metabolic syndrome-related heart failure with preserved ejection fraction phenotype: pathological alterations and possible molecular mechanisms. Front Cardiovasc Med 2023; 10:1208370. [PMID: 37469482 PMCID: PMC10352810 DOI: 10.3389/fcvm.2023.1208370] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 06/13/2023] [Indexed: 07/21/2023] Open
Abstract
Background Heart failure with preserved ejection fraction (HFpEF) represents a syndrome involving multiple pathophysiologic disorders and clinical phenotypes. This complexity makes it challenging to develop a comprehensive preclinical model, which presents an obstacle to elucidating disease mechanisms and developing new drugs. Metabolic syndrome (MetS) is a major phenotype of HFpEF. Thus, we produced a rat model of the MetS-related HFpEF phenotype and explored the molecular mechanisms underpinning the observed pathological changes. Methods A rat model of the MetS-related HFpEF phenotype was created by feeding spontaneously hypertensive rats a high-fat-salt-sugar diet and administering streptozotocin solution intraperitoneally. Subsequently, pathological changes in the rat heart and their possible molecular mechanisms were explored. Results The HFpEF rats demonstrated primary features of MetS, such as hypertension, hyperglycemia, hyperlipidemia, insulin resistance, and cardiac anomalies, such as left ventricular (LV) remodeling and diastolic impairment, and left atrial dilation. Additionally, inflammation, myocardial hypertrophy, and fibrosis were observed in LV myocardial tissue, which may be associated with diverse cellular and molecular signaling cascades. First, the inflammatory response might be related to the overexpression of inflammatory regulators (growth differentiation factor 15 (GDF-15), intercellular adhesion molecule-1 (ICAM-1), and vascular endothelial cell adhesion molecule-1 (VCAM-1)). Secondly, phosphorylated glycogen synthase kinase 3β (GSK-3β) may stimulate cardiac hypertrophy, which was regulated by activated -RAC-alpha serine/threonine-protein kinase (AKT). Finally, the transforming growth factor-β1 (TGF-β1)/Smads pathway might regulate collagen production and fibroblast activation, promoting myocardial fibrosis. Conclusion The HFpEF rat replicates the pathology and clinical presentation of human HFpEF with MetS and may be a reliable preclinical model that helps elucidate HFpEF pathogenesis and develop effective treatment strategies.
Collapse
Affiliation(s)
- Yujiao Shi
- Department of Cardiovascular Internal Medicine, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing, China
| | - Chunqiu Liu
- Department of Cardiovascular Internal Medicine, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing, China
| | - Chenguang Yang
- Department of Cardiovascular Internal Medicine, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing, China
| | - Wenbo Qiao
- Department of Cardiovascular Internal Medicine, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing, China
| | - Yongcheng Liu
- Department of Cardiovascular Internal Medicine, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing, China
| | - Siyu Liu
- Department of Cardiovascular Internal Medicine, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing, China
| | - GuoJu Dong
- Department of Cardiovascular Internal Medicine, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing, China
| |
Collapse
|
13
|
Matsuno M, Yokoe S, Nagatsuka T, Morihara H, Moriwaki K, Asahi M. O-GlcNAcylation-induced GSK-3β activation deteriorates pressure overload-induced heart failure via lack of compensatory cardiac hypertrophy in mice. Front Endocrinol (Lausanne) 2023; 14:1122125. [PMID: 37033243 PMCID: PMC10073727 DOI: 10.3389/fendo.2023.1122125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 02/27/2023] [Indexed: 04/11/2023] Open
Abstract
O-GlcNAc transferase (OGT) modulates many functions of proteins via O-GlcNAcylation that adds O-linked β-N-acetylglucosamine (O-GlcNAc) to the serine/threonine residues of proteins. However, the role of O-GlcNAcylation in cardiac remodeling and function is not fully understood. To examine the effect of O-GlcNAcylation on pressure overload-induced cardiac hypertrophy and subsequent heart failure, transverse aortic constriction (TAC) surgery was performed in wild type (WT) and Ogt transgenic (Ogt-Tg) mice. Four weeks after TAC (TAC4W), the heart function of Ogt-Tg mice was significantly lower than that of WT mice (reduced fractional shortening and increased ANP levels). The myocardium of left ventricle (LV) in Ogt-Tg mice became much thinner than that in WT mice. Moreover, compared to the heart tissues of WT mice, O-GlcNAcylation of GSK-3β at Ser9 was increased and phosphorylation of GSK-3β at Ser9 was reduced in the heart tissues of Ogt-Tg mice, resulting in its activation and subsequent inactivation of nuclear factor of activated T cell (NFAT) activity. Finally, the thinned LV wall and reduced cardiac function induced by TAC4W in Ogt-Tg mice was reversed by the treatment of a GSK-3β inhibitor, TDZD-8. These results imply that augmented O-GlcNAcylation exacerbates pressure overload-induced heart failure due to a lack of compensatory cardiac hypertrophy via O-GlcNAcylation of GSK-3β, which deprives the phosphorylation site of GSK-3β to constantly inactivate NFAT activity to prevent cardiac hypertrophy. Our findings may provide a new therapeutic strategy for cardiac hypertrophy and subsequent heart failure.
Collapse
Affiliation(s)
- Mahito Matsuno
- Department of Pharmacology, Faculty of Medicine, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Shunichi Yokoe
- Department of Pharmacology, Faculty of Medicine, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Takehiro Nagatsuka
- Center for Medical Research & Development, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Hirofumi Morihara
- Department of Pharmacology, Faculty of Medicine, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Kazumasa Moriwaki
- Department of Pharmacology, Faculty of Medicine, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Michio Asahi
- Department of Pharmacology, Faculty of Medicine, Osaka Medical and Pharmaceutical University, Osaka, Japan
- *Correspondence: Michio Asahi,
| |
Collapse
|
14
|
Tarasov KV, Chakir K, Riordon DR, Lyashkov AE, Ahmet I, Perino MG, Silvester AJ, Zhang J, Wang M, Lukyanenko YO, Qu JH, Barrera MCR, Juhaszova M, Tarasova YS, Ziman B, Telljohann R, Kumar V, Ranek M, Lammons J, Bychkov R, de Cabo R, Jun S, Keceli G, Gupta A, Yang D, Aon MA, Adamo L, Morrell CH, Otu W, Carroll C, Chambers S, Paolocci N, Huynh T, Pacak K, Weiss R, Field L, Sollott SJ, Lakatta EG. A remarkable adaptive paradigm of heart performance and protection emerges in response to marked cardiac-specific overexpression of ADCY8. eLife 2022; 11:e80949. [PMID: 36515265 PMCID: PMC9822292 DOI: 10.7554/elife.80949] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 12/08/2022] [Indexed: 12/15/2022] Open
Abstract
Adult (3 month) mice with cardiac-specific overexpression of adenylyl cyclase (AC) type VIII (TGAC8) adapt to an increased cAMP-induced cardiac workload (~30% increases in heart rate, ejection fraction and cardiac output) for up to a year without signs of heart failure or excessive mortality. Here, we show classical cardiac hypertrophy markers were absent in TGAC8, and that total left ventricular (LV) mass was not increased: a reduced LV cavity volume in TGAC8 was encased by thicker LV walls harboring an increased number of small cardiac myocytes, and a network of small interstitial proliferative non-cardiac myocytes compared to wild type (WT) littermates; Protein synthesis, proteosome activity, and autophagy were enhanced in TGAC8 vs WT, and Nrf-2, Hsp90α, and ACC2 protein levels were increased. Despite increased energy demands in vivo LV ATP and phosphocreatine levels in TGAC8 did not differ from WT. Unbiased omics analyses identified more than 2,000 transcripts and proteins, comprising a broad array of biological processes across multiple cellular compartments, which differed by genotype; compared to WT, in TGAC8 there was a shift from fatty acid oxidation to aerobic glycolysis in the context of increased utilization of the pentose phosphate shunt and nucleotide synthesis. Thus, marked overexpression of AC8 engages complex, coordinate adaptation "circuity" that has evolved in mammalian cells to defend against stress that threatens health or life (elements of which have already been shown to be central to cardiac ischemic pre-conditioning and exercise endurance cardiac conditioning) that may be of biological significance to allow for proper healing in disease states such as infarction or failure of the heart.
Collapse
Affiliation(s)
- Kirill V Tarasov
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of HealthBaltimoreUnited States
| | - Khalid Chakir
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of HealthBaltimoreUnited States
| | - Daniel R Riordon
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of HealthBaltimoreUnited States
| | - Alexey E Lyashkov
- Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of HealthBaltimoreUnited States
| | - Ismayil Ahmet
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of HealthBaltimoreUnited States
| | - Maria Grazia Perino
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of HealthBaltimoreUnited States
| | - Allwin Jennifa Silvester
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of HealthBaltimoreUnited States
| | - Jing Zhang
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of HealthBaltimoreUnited States
| | - Mingyi Wang
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of HealthBaltimoreUnited States
| | - Yevgeniya O Lukyanenko
- Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of HealthBaltimoreUnited States
| | - Jia-Hua Qu
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of HealthBaltimoreUnited States
| | - Miguel Calvo-Rubio Barrera
- Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of HealthBaltimoreUnited States
| | - Magdalena Juhaszova
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of HealthBaltimoreUnited States
| | - Yelena S Tarasova
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of HealthBaltimoreUnited States
| | - Bruce Ziman
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of HealthBaltimoreUnited States
| | - Richard Telljohann
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of HealthBaltimoreUnited States
| | - Vikas Kumar
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of HealthBaltimoreUnited States
| | - Mark Ranek
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - John Lammons
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of HealthBaltimoreUnited States
| | - Rostislav Bychkov
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of HealthBaltimoreUnited States
| | - Rafael de Cabo
- Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of HealthBaltimoreUnited States
| | - Seungho Jun
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Gizem Keceli
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Ashish Gupta
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Dongmei Yang
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of HealthBaltimoreUnited States
| | - Miguel A Aon
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of HealthBaltimoreUnited States
| | - Luigi Adamo
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Christopher H Morrell
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of HealthBaltimoreUnited States
| | - Walter Otu
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of HealthBaltimoreUnited States
| | - Cameron Carroll
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of HealthBaltimoreUnited States
| | - Shane Chambers
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of HealthBaltimoreUnited States
| | - Nazareno Paolocci
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Thanh Huynh
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of HealthBethesdaUnited States
| | - Karel Pacak
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of HealthBethesdaUnited States
| | - Robert Weiss
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Loren Field
- Kraennert Institute of Cardiology, Indiana University School of MedicineIdianapolisUnited States
| | - Steven J Sollott
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of HealthBaltimoreUnited States
| | - Edward G Lakatta
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of HealthBaltimoreUnited States
| |
Collapse
|
15
|
Lunde IG, Skrbic B, Sjaastad I, Christensen G, Carlson CR, Tønnessen T. Calcineurin-NFAT dynamics correspond to cardiac remodeling during aortic banding and debanding, mimicking aortic valve replacement. FRONTIERS IN MOLECULAR MEDICINE 2022; 2:980717. [PMID: 39086965 PMCID: PMC11285616 DOI: 10.3389/fmmed.2022.980717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 10/06/2022] [Indexed: 08/02/2024]
Abstract
Aortic valve stenosis (AS) is a major health problem. Extensive myocardial remodeling increases operative risk and might lead to incomplete reverse remodeling with persistent symptoms after aortic valve replacement (AVR); this makes the optimal timing of AVR a clinical challenge. The pathogenesis behind incomplete reverse remodeling is unclear. Central among signaling pathways in the remodeling heart is the pro-hypertrophic Ca2+-activated calcineurin and its downstream nuclear factor of activated T-cell (NFATc1-c4) transcription factors. We investigated calcineurin-NFATc dynamics in patient and mouse hearts during remodeling and reverse remodeling. Myocardial biopsies were obtained from AS patients during AVR and left ventricles harvested from mice subjected to aortic banding (AB) and debanding (DB). The transcript and protein of the NFATc-responsive gene regulator of calcineurin 1-4 (RCAN1-4) and luciferase activity in NFAT-luciferase mice were used as read-outs for calcineurin-NFATc activity. Calcineurin-NFATc activation was sustained through AB 24 h to 18 weeks and elevated in AS patients. All four NFATc isoforms were elevated in AS, while NFATc4 was persistently elevated during chronic remodeling after AB in mice. NFAT activation remained reversible when 1 week's AB was followed by 1 week's DB and accompanied functional improvement. However, when DB for 1 week followed AB for 4 weeks, NFAT activation was not reversed. In conclusion, calcineurin-NFAT dynamics correspond with cardiac remodeling and reverse remodeling during experimental AB and DB. Our data suggest that calcineurin-NFATc attenuation is important for reverse remodeling and outcomes after AVR for AS.
Collapse
Affiliation(s)
- Ida G. Lunde
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- KG Jebsen Cardiac Research Center, University of Oslo, Oslo, Norway
- Division of Diagnostics and Technology, Akershus University Hospital, Lørenskog, Norway
| | - Biljana Skrbic
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- KG Jebsen Cardiac Research Center, University of Oslo, Oslo, Norway
- Department of Cardiothoracic Surgery, Oslo University Hospital Ullevaal, Oslo, Norway
| | - Ivar Sjaastad
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- KG Jebsen Cardiac Research Center, University of Oslo, Oslo, Norway
| | - Geir Christensen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- KG Jebsen Cardiac Research Center, University of Oslo, Oslo, Norway
| | - Cathrine R. Carlson
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- KG Jebsen Cardiac Research Center, University of Oslo, Oslo, Norway
| | - Theis Tønnessen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- KG Jebsen Cardiac Research Center, University of Oslo, Oslo, Norway
- Department of Cardiothoracic Surgery, Oslo University Hospital Ullevaal, Oslo, Norway
| |
Collapse
|
16
|
Kim HK, Kim M, Marquez JC, Jeong SH, Ko TH, Noh YH, Kha PT, Choi HM, Kim DH, Kim JT, Yang YI, Ko KS, Rhee BD, Shubina LK, Makarieva TN, Yashunsky DY, Gerbst AG, Nifantiev NE, Stonik VA, Han J. Novel GSK-3β Inhibitor Neopetroside A Protects Against Murine Myocardial Ischemia/Reperfusion Injury. JACC Basic Transl Sci 2022; 7:1102-1116. [PMID: 36687267 PMCID: PMC9849271 DOI: 10.1016/j.jacbts.2022.05.004] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 03/31/2022] [Accepted: 05/09/2022] [Indexed: 02/01/2023]
Abstract
Recent trends suggest novel natural compounds as promising treatments for cardiovascular disease. The authors examined how neopetroside A, a natural pyridine nucleoside containing an α-glycoside bond, regulates mitochondrial metabolism and heart function and investigated its cardioprotective role against ischemia/reperfusion injury. Neopetroside A treatment maintained cardiac hemodynamic status and mitochondrial respiration capacity and significantly prevented cardiac fibrosis in murine models. These effects can be attributed to preserved cellular and mitochondrial function caused by the inhibition of glycogen synthase kinase-3 beta, which regulates the ratio of nicotinamide adenine dinucleotide to nicotinamide adenine dinucleotide, reduced, through activation of the nuclear factor erythroid 2-related factor 2/NAD(P)H quinone oxidoreductase 1 axis in a phosphorylation-independent manner.
Collapse
Key Words
- ATP, adenosine triphosphate
- GSK-3, glycogen synthase kinase–3
- GSK-3β inhibition
- I/R, ischemia/reperfusion
- MI, myocardial infarction
- NAD+, nicotinamide adenine dinucleotide
- NADH, nicotinamide adenine dinucleotide, reduced
- NPS A
- NPS A, neopetroside A
- Nqo1, NAD(P)H:quinone oxidoreductase 1
- Nrf2, nuclear factor erythroid 2–related factor 2
- OCR, oxygen consumption rate
- ischemia/reperfusion injury
- mPTP, mitochondrial permeability transition pore
- mTOR, mammalian target of rapamycin
- marine pyridine α-nucleoside
- mitochondria
Collapse
Affiliation(s)
- Hyoung Kyu Kim
- Cardiovascular and Metabolic Disease Center, Inje University, Busan, South Korea,Department of Health Sciences and Technology, Graduate School, Inje University, Busan, South Korea
| | - Min Kim
- Cardiovascular and Metabolic Disease Center, Inje University, Busan, South Korea,Department of Physiology, BK Plus Project Team, College of Medicine, Inje University, Busan, South Korea
| | - Jubert C. Marquez
- Cardiovascular and Metabolic Disease Center, Inje University, Busan, South Korea,Department of Health Sciences and Technology, Graduate School, Inje University, Busan, South Korea
| | - Seung Hun Jeong
- Cardiovascular and Metabolic Disease Center, Inje University, Busan, South Korea,Department of Physiology, BK Plus Project Team, College of Medicine, Inje University, Busan, South Korea
| | - Tae Hee Ko
- Cardiovascular and Metabolic Disease Center, Inje University, Busan, South Korea,Department of Physiology, BK Plus Project Team, College of Medicine, Inje University, Busan, South Korea
| | - Yeon Hee Noh
- Cardiovascular and Metabolic Disease Center, Inje University, Busan, South Korea,Department of Physiology, BK Plus Project Team, College of Medicine, Inje University, Busan, South Korea
| | - Pham Trong Kha
- Cardiovascular and Metabolic Disease Center, Inje University, Busan, South Korea,Department of Physiology, BK Plus Project Team, College of Medicine, Inje University, Busan, South Korea
| | - Ha Min Choi
- Cardiovascular and Metabolic Disease Center, Inje University, Busan, South Korea,Department of Physiology, BK Plus Project Team, College of Medicine, Inje University, Busan, South Korea
| | - Dong Hyun Kim
- Department of Pharmacology and Pharmaco-Genomics Research Center, College of Medicine, Inje University, Busan, South Korea
| | - Jong Tae Kim
- Paik Institute for Clinical Research, Inje University College of Medicine, Busan, South Korea
| | - Young Il Yang
- Paik Institute for Clinical Research, Inje University College of Medicine, Busan, South Korea
| | - Kyung Soo Ko
- Cardiovascular and Metabolic Disease Center, Inje University, Busan, South Korea,Department of Health Sciences and Technology, Graduate School, Inje University, Busan, South Korea
| | - Byoung Doo Rhee
- Cardiovascular and Metabolic Disease Center, Inje University, Busan, South Korea,Department of Health Sciences and Technology, Graduate School, Inje University, Busan, South Korea
| | - Larisa K. Shubina
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far-Eastern Branch of the Russian Academy of Science, Vladivostok, Russia
| | - Tatyana N. Makarieva
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far-Eastern Branch of the Russian Academy of Science, Vladivostok, Russia
| | - Dmitry Y. Yashunsky
- Laboratory of Glycoconjugate Chemistry, N.D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Alexey G. Gerbst
- Laboratory of Glycoconjugate Chemistry, N.D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Nikolay E. Nifantiev
- Laboratory of Glycoconjugate Chemistry, N.D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Valentin A. Stonik
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far-Eastern Branch of the Russian Academy of Science, Vladivostok, Russia
| | - Jin Han
- Cardiovascular and Metabolic Disease Center, Inje University, Busan, South Korea,Department of Health Sciences and Technology, Graduate School, Inje University, Busan, South Korea,Department of Physiology, BK Plus Project Team, College of Medicine, Inje University, Busan, South Korea,Address for correspondence: Dr Jin Han, National Research Laboratory for Mitochondrial Signaling, Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan 47393, South Korea.
| |
Collapse
|
17
|
Liu C, Gu L, Deng W, Meng Q, Li N, Dai G, Yu S, Fang H. N6-Methyladenosine RNA Methylation in Cardiovascular Diseases. Front Cardiovasc Med 2022; 9:887838. [PMID: 35571209 PMCID: PMC9098837 DOI: 10.3389/fcvm.2022.887838] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 04/12/2022] [Indexed: 12/18/2022] Open
Abstract
N6-methyladenosine (m6A) modification is the most universal and abundant post-transcriptional modification of eukaryotic RNA and occurs mainly at the consensus motif RR (m6A) CH (R = A or G, H = A, C, or U) in long internal exons, near stop codons, or in the 3' untranslated region (UTR). "Writers," "erasers," and "readers" are responsible for the occurrence, removal, and recognition of m6A modification, respectively. Substantial evidence has shown that m6A RNA modification can exert important functions in physiological and pathological processes. Cardiovascular diseases (CVDs) are a wide array of disorders affecting heart or vessels, including atherosclerosis (AS), hypertension (HT), ischemia/reperfusion (I/R) injury, myocardial infarction (MI), stroke, cardiac hypertrophy, heart failure (HF), and so on. Despite the advances in lipid-lowering drugs, antihypertensives, antiplatelet agents, and anticoagulation therapy, CVDs are still the leading cause of death worldwide. Recent studies have suggested that m6A modification of RNA may contribute to the pathogenesis of CVDs, providing a novel research insight for CVDs. Herein, we provide an up-of-date summarization of the molecular mechanism of m6A and the roles of m6A in different types of CVDs. At last, we propose that m6A might be a potiential biomarker or therapeutic target for CVDs.
Collapse
Affiliation(s)
- Chi Liu
- Department of Cardiology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
- Department of Geriatrics Center, National Clinical Research Center for Aging and Medicine, Jing’an District Central Hospital of Shanghai, Fudan University, Shanghai, China
| | - Lei Gu
- Department of Internal Medicine, Shanghai Shende Hospital, Shanghai, China
| | - Wenjuan Deng
- Department of Geriatrics Center, National Clinical Research Center for Aging and Medicine, Jing’an District Central Hospital of Shanghai, Fudan University, Shanghai, China
| | - Qianchao Meng
- Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Nan Li
- Department of Geriatrics Center, National Clinical Research Center for Aging and Medicine, Jing’an District Central Hospital of Shanghai, Fudan University, Shanghai, China
| | - Guifeng Dai
- Department of Geriatrics Center, National Clinical Research Center for Aging and Medicine, Jing’an District Central Hospital of Shanghai, Fudan University, Shanghai, China
| | - Suli Yu
- Department of Hand and Upper Extremity Surgery and Limb Function Reconstruction Center, Jing’an District Central Hospital, Shanghai, China
| | - Hong Fang
- Department of Cardiology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
18
|
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: 36] [Impact Index Per Article: 12.0] [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
|
19
|
Hartmann N, Preuß L, Mohamed BA, Schnelle M, Renner A, Hasenfuß G, Toischer K. Different activation of MAPKs and Akt/GSK3β after preload vs. afterload elevation. ESC Heart Fail 2022; 9:1823-1831. [PMID: 35315235 PMCID: PMC9065823 DOI: 10.1002/ehf2.13877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 02/07/2022] [Accepted: 02/28/2022] [Indexed: 11/28/2022] Open
Abstract
Aims Pressure overload (PO) and volume overload (VO) lead to concentric or eccentric hypertrophy. Previously, we could show that activation of signalling cascades differ in in vivo mouse models. Activation of these signal cascades could either be induced by intrinsic load sensing or neuro‐endocrine substances like catecholamines or the renin‐angiotensin‐aldosterone system. Methods and results We therefore analysed the activation of classical cardiac signal pathways [mitogen‐activated protein kinases (MAPKs) (ERK, p38, and JNK) and Akt‐GSK3β] in in vitro of mechanical overload (ejecting heart model, rabbit and human isolated muscle strips). Selective elevation of preload in vitro increased AKT and GSK3β phosphorylation after 15 min in isolated rabbit muscles strips (AKT 49%, GSK3β 26%, P < 0.05) and in mouse ejecting hearts (AKT 51%, GSK49%, P < 0.05), whereas phosphorylation of MAPKs was not influenced by increased preload. Selective elevation of afterload revealed an increase in ERK phosphorylation in the ejecting heart (43%, P < 0.05), but not in AKT, GSK3β, and the other MAPKs. Elevation of preload and afterload in the ejecting heart induced a significant phosphorylation of ERK (95%, P < 0.001) and showed a moderate increased AKT (P = 0.14) and GSK3β (P = 0.21) phosphorylation, which did not reach significance. Preload and afterload elevation in muscles strips from human failing hearts showed neither AKT nor ERK phosphorylation changes. Conclusions Our data show that preload activates the AKT–GSK3β and afterload the ERK pathway in vitro, indicating an intrinsic mechanism independent of endocrine signalling.
Collapse
Affiliation(s)
- Nico Hartmann
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Robert-Koch-Str. 40, Göttingen, 37075, Germany
| | - Lena Preuß
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Robert-Koch-Str. 40, Göttingen, 37075, Germany
| | - Belal A Mohamed
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Robert-Koch-Str. 40, Göttingen, 37075, Germany.,DZHK, German Centre for Cardiovascular Research, Göttingen, Germany
| | - Moritz Schnelle
- Institute for Clinical Chemistry, University Medical Center Göttingen, Göttingen, Germany.,DZHK, German Centre for Cardiovascular Research, Göttingen, Germany
| | - Andre Renner
- Department of Thoracic, Cardiac and Vascular Surgery (Heart and Diabetes Center), North Rhine Westphalia, Bad Oeynhausen, Germany
| | - Gerd Hasenfuß
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Robert-Koch-Str. 40, Göttingen, 37075, Germany.,DZHK, German Centre for Cardiovascular Research, Göttingen, Germany
| | - Karl Toischer
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Robert-Koch-Str. 40, Göttingen, 37075, Germany.,DZHK, German Centre for Cardiovascular Research, Göttingen, Germany
| |
Collapse
|
20
|
Stachowski-Doll MJ, Papadaki M, Martin TG, Ma W, Gong HM, Shao S, Shen S, Muntu NA, Kumar M, Perez E, Martin JL, Moravec CS, Sadayappan S, Campbell SG, Irving T, Kirk JA. GSK-3β Localizes to the Cardiac Z-Disc to Maintain Length Dependent Activation. Circ Res 2022; 130:871-886. [PMID: 35168370 PMCID: PMC8930626 DOI: 10.1161/circresaha.121.319491] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 02/07/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND Altered kinase localization is gaining appreciation as a mechanism of cardiovascular disease. Previous work suggests GSK-3β (glycogen synthase kinase 3β) localizes to and regulates contractile function of the myofilament. We aimed to discover GSK-3β's in vivo role in regulating myofilament function, the mechanisms involved, and the translational relevance. METHODS Inducible cardiomyocyte-specific GSK-3β knockout mice and left ventricular myocardium from nonfailing and failing human hearts were studied. RESULTS Skinned cardiomyocytes from knockout mice failed to exhibit calcium sensitization with stretch indicating a loss of length-dependent activation (LDA), the mechanism underlying the Frank-Starling Law. Titin acts as a length sensor for LDA, and knockout mice had decreased titin stiffness compared with control mice, explaining the lack of LDA. Knockout mice exhibited no changes in titin isoforms, titin phosphorylation, or other thin filament phosphorylation sites known to affect passive tension or LDA. Mass spectrometry identified several z-disc proteins as myofilament phospho-substrates of GSK-3β. Agreeing with the localization of its targets, GSK-3β that is phosphorylated at Y216 binds to the z-disc. We showed pY216 was necessary and sufficient for z-disc binding using adenoviruses for wild-type, Y216F, and Y216E GSK-3β in neonatal rat ventricular cardiomyocytes. One of GSK-3β's z-disc targets, abLIM-1 (actin-binding LIM protein 1), binds to the z-disc domains of titin that are important for maintaining passive tension. Genetic knockdown of abLIM-1 via siRNA in human engineered heart tissues resulted in enhancement of LDA, indicating abLIM-1 may act as a negative regulator that is modulated by GSK-3β. Last, GSK-3β myofilament localization was reduced in left ventricular myocardium from failing human hearts, which correlated with depressed LDA. CONCLUSIONS We identified a novel mechanism by which GSK-3β localizes to the myofilament to modulate LDA. Importantly, z-disc GSK-3β levels were reduced in patients with heart failure, indicating z-disc localized GSK-3β is a possible therapeutic target to restore the Frank-Starling mechanism in patients with heart failure.
Collapse
Affiliation(s)
- Marisa J Stachowski-Doll
- Department of Cell and Molecular Physiology, Loyola University Stritch School of Medicine, Maywood, IL (M.J.S.-D., M.P., T.G.M., N.A.M., E.P., J.A.K.)
| | - Maria Papadaki
- Department of Cell and Molecular Physiology, Loyola University Stritch School of Medicine, Maywood, IL (M.J.S.-D., M.P., T.G.M., N.A.M., E.P., J.A.K.)
| | - Thomas G Martin
- Department of Cell and Molecular Physiology, Loyola University Stritch School of Medicine, Maywood, IL (M.J.S.-D., M.P., T.G.M., N.A.M., E.P., J.A.K.)
| | - Weikang Ma
- Center for Synchrotron Radiation Research and Instrumentation and Department of Biological Sciences, Illinois Institute of Technology, Chicago (W.M., H.M.G., T.I.)
| | - Henry M Gong
- Center for Synchrotron Radiation Research and Instrumentation and Department of Biological Sciences, Illinois Institute of Technology, Chicago (W.M., H.M.G., T.I.)
| | - Stephanie Shao
- Department of Bioengineering, Yale University, New Haven, CT (S. Shao, S. Shen, S.G.C.)
| | - Shi Shen
- Department of Bioengineering, Yale University, New Haven, CT (S. Shao, S. Shen, S.G.C.)
| | - Nitha Aima Muntu
- Department of Cell and Molecular Physiology, Loyola University Stritch School of Medicine, Maywood, IL (M.J.S.-D., M.P., T.G.M., N.A.M., E.P., J.A.K.)
| | - Mohit Kumar
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, Heart, Lung, and Vascular Institute, University of Cincinnati, OH (M.K., S. Sadayappan)
| | - Edith Perez
- Department of Cell and Molecular Physiology, Loyola University Stritch School of Medicine, Maywood, IL (M.J.S.-D., M.P., T.G.M., N.A.M., E.P., J.A.K.)
| | - Jody L Martin
- Department of Pharmacology, Cardiovascular Research Institute, UC Davis School of Medicine, CA (J.L.M.)
| | - Christine S Moravec
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic, OH (C.S.M.)
| | - Sakthivel Sadayappan
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, Heart, Lung, and Vascular Institute, University of Cincinnati, OH (M.K., S. Sadayappan)
| | - Stuart G Campbell
- Department of Bioengineering, Yale University, New Haven, CT (S. Shao, S. Shen, S.G.C.)
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT (S.G.C.)
| | - Thomas Irving
- Center for Synchrotron Radiation Research and Instrumentation and Department of Biological Sciences, Illinois Institute of Technology, Chicago (W.M., H.M.G., T.I.)
| | - Jonathan A Kirk
- Department of Cell and Molecular Physiology, Loyola University Stritch School of Medicine, Maywood, IL (M.J.S.-D., M.P., T.G.M., N.A.M., E.P., J.A.K.)
| |
Collapse
|
21
|
Lavin KM, Coen PM, Baptista LC, Bell MB, Drummer D, Harper SA, Lixandrão ME, McAdam JS, O’Bryan SM, Ramos S, Roberts LM, Vega RB, Goodpaster BH, Bamman MM, Buford TW. State of Knowledge on Molecular Adaptations to Exercise in Humans: Historical Perspectives and Future Directions. Compr Physiol 2022; 12:3193-3279. [PMID: 35578962 PMCID: PMC9186317 DOI: 10.1002/cphy.c200033] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
For centuries, regular exercise has been acknowledged as a potent stimulus to promote, maintain, and restore healthy functioning of nearly every physiological system of the human body. With advancing understanding of the complexity of human physiology, continually evolving methodological possibilities, and an increasingly dire public health situation, the study of exercise as a preventative or therapeutic treatment has never been more interdisciplinary, or more impactful. During the early stages of the NIH Common Fund Molecular Transducers of Physical Activity Consortium (MoTrPAC) Initiative, the field is well-positioned to build substantially upon the existing understanding of the mechanisms underlying benefits associated with exercise. Thus, we present a comprehensive body of the knowledge detailing the current literature basis surrounding the molecular adaptations to exercise in humans to provide a view of the state of the field at this critical juncture, as well as a resource for scientists bringing external expertise to the field of exercise physiology. In reviewing current literature related to molecular and cellular processes underlying exercise-induced benefits and adaptations, we also draw attention to existing knowledge gaps warranting continued research effort. © 2021 American Physiological Society. Compr Physiol 12:3193-3279, 2022.
Collapse
Affiliation(s)
- Kaleen M. Lavin
- Center for Exercise Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Cell, Developmental, and Integrative Biology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
- Center for Human Health, Resilience, and Performance, Institute for Human and Machine Cognition, Pensacola, Florida, USA
| | - Paul M. Coen
- Translational Research Institute for Metabolism and Diabetes, Advent Health, Orlando, Florida, USA
- Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida, USA
| | - Liliana C. Baptista
- Center for Exercise Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Medicine, Division of Gerontology, Geriatrics and Palliative Care, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Margaret B. Bell
- Center for Exercise Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Cell, Developmental, and Integrative Biology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Devin Drummer
- Center for Exercise Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Cell, Developmental, and Integrative Biology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Sara A. Harper
- Center for Exercise Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Medicine, Division of Gerontology, Geriatrics and Palliative Care, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Manoel E. Lixandrão
- Center for Exercise Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Cell, Developmental, and Integrative Biology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Jeremy S. McAdam
- Center for Exercise Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Cell, Developmental, and Integrative Biology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Samia M. O’Bryan
- Center for Exercise Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Cell, Developmental, and Integrative Biology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Sofhia Ramos
- Translational Research Institute for Metabolism and Diabetes, Advent Health, Orlando, Florida, USA
- Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida, USA
| | - Lisa M. Roberts
- Center for Exercise Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Medicine, Division of Gerontology, Geriatrics and Palliative Care, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Rick B. Vega
- Translational Research Institute for Metabolism and Diabetes, Advent Health, Orlando, Florida, USA
- Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida, USA
| | - Bret H. Goodpaster
- Translational Research Institute for Metabolism and Diabetes, Advent Health, Orlando, Florida, USA
- Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida, USA
| | - Marcas M. Bamman
- Center for Exercise Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Cell, Developmental, and Integrative Biology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
- Center for Human Health, Resilience, and Performance, Institute for Human and Machine Cognition, Pensacola, Florida, USA
| | - Thomas W. Buford
- Center for Exercise Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Medicine, Division of Gerontology, Geriatrics and Palliative Care, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
22
|
Ritchie JA, Ng JQ, Kemi OJ. When one says yes and the other says no; does calcineurin participate in physiologic cardiac hypertrophy? ADVANCES IN PHYSIOLOGY EDUCATION 2022; 46:84-95. [PMID: 34762541 DOI: 10.1152/advan.00104.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 11/08/2021] [Indexed: 06/13/2023]
Abstract
Developing engaging activities that build skills for understanding and appreciating research is important for undergraduate and postgraduate science students. Comparing and contrasting opposing research studies does this, and more: it also appropriately for these cohorts challenges higher level cognitive processing. Here, we present and discuss one such scenario, that of calcineurin in the heart and its response to exercise training. This scenario is further accentuated by the existence of only two studies. The background is that regular aerobic endurance exercise training stimulates the heart to physiologically adapt to chronically increase its ability to produce a greater cardiac output to meet the increased demand for oxygenated blood in working muscles, and this happens by two main mechanisms: 1) increased cardiac contractile function and 2) physiologic hypertrophy. The major underlying mechanisms have been delineated over the last decades, but one aspect has not been resolved: the potential role of calcineurin in modulating physiologic hypertrophy. This is partly because the existing research has provided opposing and contrasting findings, one line showing that exercise training does activate cardiac calcineurin in conjunction with myocardial hypertrophy, but another line showing that exercise training does not activate cardiac calcineurin even if myocardial hypertrophy is blatantly occurring. Here, we review and present the current evidence in the field and discuss reasons for this controversy. We present real-life examples from physiology research and discuss how this may enhance student engagement and participation, widen the scope of learning, and thereby also further facilitate higher level cognitive processing.
Collapse
Affiliation(s)
- Jonathan A Ritchie
- School of Medicine, Dentistry and Nursing, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Jun Q Ng
- School of Life Sciences, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Ole J Kemi
- School of Life Sciences, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
23
|
Vainio L, Taponen S, Kinnunen SM, Halmetoja E, Szabo Z, Alakoski T, Ulvila J, Junttila J, Lakkisto P, Magga J, Kerkelä R. GSK3β Serine 389 Phosphorylation Modulates Cardiomyocyte Hypertrophy and Ischemic Injury. Int J Mol Sci 2021; 22:13586. [PMID: 34948382 PMCID: PMC8707850 DOI: 10.3390/ijms222413586] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 12/03/2021] [Accepted: 12/15/2021] [Indexed: 11/17/2022] Open
Abstract
Prior studies show that glycogen synthase kinase 3β (GSK3β) contributes to cardiac ischemic injury and cardiac hypertrophy. GSK3β is constitutionally active and phosphorylation of GSK3β at serine 9 (S9) inactivates the kinase and promotes cellular growth. GSK3β is also phosphorylated at serine 389 (S389), but the significance of this phosphorylation in the heart is not known. We analyzed GSK3β S389 phosphorylation in diseased hearts and utilized overexpression of GSK3β carrying ser→ala mutations at S9 (S9A) and S389 (S389A) to study the biological function of constitutively active GSK3β in primary cardiomyocytes. We found that phosphorylation of GSK3β at S389 was increased in left ventricular samples from patients with dilated cardiomyopathy and ischemic cardiomyopathy, and in hearts of mice subjected to thoracic aortic constriction. Overexpression of either GSK3β S9A or S389A reduced the viability of cardiomyocytes subjected to hypoxia-reoxygenation. Overexpression of double GSK3β mutant (S9A/S389A) further reduced cardiomyocyte viability. Determination of protein synthesis showed that overexpression of GSK3β S389A or GSK3β S9A/S389A increased both basal and agonist-induced cardiomyocyte growth. Mechanistically, GSK3β S389A mutation was associated with activation of mTOR complex 1 signaling. In conclusion, our data suggest that phosphorylation of GSK3β at S389 enhances cardiomyocyte survival and protects from cardiomyocyte hypertrophy.
Collapse
Affiliation(s)
- Laura Vainio
- Research Unit of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu 90220, Finland; (L.V.); (S.T.); (S.M.K.); (E.H.); (Z.S.); (T.A.); (J.U.); (J.M.)
- Biocenter Oulu, University of Oulu, Oulu 90220, Finland;
| | - Saija Taponen
- Research Unit of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu 90220, Finland; (L.V.); (S.T.); (S.M.K.); (E.H.); (Z.S.); (T.A.); (J.U.); (J.M.)
- Biocenter Oulu, University of Oulu, Oulu 90220, Finland;
| | - Sini M. Kinnunen
- Research Unit of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu 90220, Finland; (L.V.); (S.T.); (S.M.K.); (E.H.); (Z.S.); (T.A.); (J.U.); (J.M.)
- Drug Research Program, Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki 00014, Finland
| | - Eveliina Halmetoja
- Research Unit of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu 90220, Finland; (L.V.); (S.T.); (S.M.K.); (E.H.); (Z.S.); (T.A.); (J.U.); (J.M.)
- Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu 90220, Finland
| | - Zoltan Szabo
- Research Unit of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu 90220, Finland; (L.V.); (S.T.); (S.M.K.); (E.H.); (Z.S.); (T.A.); (J.U.); (J.M.)
| | - Tarja Alakoski
- Research Unit of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu 90220, Finland; (L.V.); (S.T.); (S.M.K.); (E.H.); (Z.S.); (T.A.); (J.U.); (J.M.)
- Biocenter Oulu, University of Oulu, Oulu 90220, Finland;
| | - Johanna Ulvila
- Research Unit of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu 90220, Finland; (L.V.); (S.T.); (S.M.K.); (E.H.); (Z.S.); (T.A.); (J.U.); (J.M.)
| | - Juhani Junttila
- Biocenter Oulu, University of Oulu, Oulu 90220, Finland;
- Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu 90220, Finland
- Research Unit of Internal Medicine, Division of Cardiology, Oulu University Hospital and University of Oulu, Oulu 90220, Finland
| | - Päivi Lakkisto
- Unit of Cardiovascular Research, Minerva Institute for Medical Research, Helsinki 00014, Finland;
- Department of Clinical Chemistry and Hematology, University of Helsinki and Helsinki University Hospital, Helsinki 00014, Finland
| | - Johanna Magga
- Research Unit of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu 90220, Finland; (L.V.); (S.T.); (S.M.K.); (E.H.); (Z.S.); (T.A.); (J.U.); (J.M.)
- Biocenter Oulu, University of Oulu, Oulu 90220, Finland;
| | - Risto Kerkelä
- Research Unit of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu 90220, Finland; (L.V.); (S.T.); (S.M.K.); (E.H.); (Z.S.); (T.A.); (J.U.); (J.M.)
- Biocenter Oulu, University of Oulu, Oulu 90220, Finland;
- Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu 90220, Finland
| |
Collapse
|
24
|
Taneja A, Ravi V, Hong JY, Lin H, Sundaresan NR. Emerging roles of Sirtuin 2 in cardiovascular diseases. FASEB J 2021; 35:e21841. [PMID: 34582046 DOI: 10.1096/fj.202100490r] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 07/07/2021] [Accepted: 07/23/2021] [Indexed: 11/11/2022]
Abstract
Sirtuins are a family of NAD+ -dependent deacetylases implicated in a wide variety of age-associated pathologies, including cardiovascular disorders. Among the seven mammalian sirtuins, SIRT2 modulates various cellular processes through the deacetylation or deacylation of their target proteins. Notably, the levels of SIRT2 in the heart decline with age and other pathological conditions, leading to cardiovascular dysfunction. In the present review, we discuss the emerging roles of SIRT2 in cardiovascular dysfunction and heart failure associated with factors like age, hypertension, oxidative stress, and diabetes. We also discuss the potential of using inhibitors to study the unexplored role of SIRT2 in the heart. While SIRT2 undoubtedly plays a crucial role in the cardiovascular system, its functions are only beginning to be understood, making it an attractive candidate for further research in the field.
Collapse
Affiliation(s)
- Arushi Taneja
- Department of Microbiology and Cell Biology, Cardiovascular and Muscle Research Laboratory, Indian Institute of Science, Bengaluru, India
| | - Venkatraman Ravi
- Department of Microbiology and Cell Biology, Cardiovascular and Muscle Research Laboratory, Indian Institute of Science, Bengaluru, India
| | - Jun Young Hong
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, USA
| | - Hening Lin
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, USA.,Howard Hughes Medical Institute, Cornell University, Ithaca, NY, USA
| | - Nagalingam Ravi Sundaresan
- Department of Microbiology and Cell Biology, Cardiovascular and Muscle Research Laboratory, Indian Institute of Science, Bengaluru, India
| |
Collapse
|
25
|
Kumari R, Ranjan P, Suleiman ZG, Goswami SK, Li J, Prasad R, Verma SK. mRNA modifications in cardiovascular biology and disease: with a focus on m6A modification. Cardiovasc Res 2021; 118:1680-1692. [PMID: 33956076 DOI: 10.1093/cvr/cvab160] [Citation(s) in RCA: 126] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 05/04/2021] [Indexed: 12/23/2022] Open
Abstract
Among several known RNA modifications, N6-methyladenosine (m6A) is the most studied RNA epitranscriptomic modification and controls multiple cellular functions during development, differentiation, and disease. Current research advancements have made it possible to examine the regulatory mechanisms associated with RNA methylation and reveal its functional consequences in the pathobiology of many diseases, including heart failure. m6A methylation has been described both on coding (mRNA) and non-coding RNA species including rRNA, tRNA, small nuclear RNA and circular RNAs. The protein components which catalyze the m6A methylation are termed methyltransferase or "m6A writers." The family of proteins that recognize this methylation are termed "m6A readers" and finally the enzymes involved in the removal of a methyl group from RNA are known as demethylases or "m6A erasers." At the cellular level, different components of methylation machinery are tightly regulated by many factors to maintain the m6A methylation dynamics. The m6A methylation process impacts different stages of mRNA metabolism and the biogenesis of long non-coding RNA and miRNA. Although, mRNA methylation was initially described in the 1970s, its regulatory roles in various diseases, including cardiovascular diseases are broadly unexplored. Recent investigations suggest the important role of m6A mRNA methylation in both hypertrophic and ischemic heart diseases. In the present review, we evaluate the significance of m6A methylation in the cardiovascular system, in cardiac homeostasis and disease, all of which may help to improve therapeutic intervention for the treatment of heart failure.
Collapse
Affiliation(s)
- Rajesh Kumari
- Division of Cardiovascular Disease, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Prabhat Ranjan
- Division of Cardiovascular Disease, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Zainab Gbongbo Suleiman
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Sumanta Kumar Goswami
- Division of Cardiovascular Disease, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Jing Li
- Division of Cardiovascular Disease, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Ram Prasad
- Department of Ophthalmology and Visual Sciences, School of Medicine, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Suresh Kumar Verma
- Division of Cardiovascular Disease, The University of Alabama at Birmingham, Birmingham, AL, United States.,Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
26
|
Francois A, Canella A, Marcho LM, Stratton MS. Protein acetylation in cardiac aging. J Mol Cell Cardiol 2021; 157:90-97. [PMID: 33915138 DOI: 10.1016/j.yjmcc.2021.04.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 03/14/2021] [Accepted: 04/21/2021] [Indexed: 11/15/2022]
Abstract
Biological aging is attributed to progressive dysfunction in systems governing genetic and metabolic integrity. At the cellular level, aging is evident by accumulated DNA damage and mutation, reactive oxygen species, alternate lipid and protein modifications, alternate gene expression programs, and mitochondrial dysfunction. These effects sum to drive altered tissue morphology and organ dysfunction. Protein-acylation has emerged as a critical mediator of age-dependent changes in these processes. Despite decades of research focus from academia and industry, heart failure remains a leading cause of death in the United States while the 5 year mortality rate for heart failure remains over 40%. Over 90% of heart failure deaths occur in patients over the age of 65 and heart failure is the leading cause of hospitalization in Medicare beneficiaries. In 1931, Cole and Koch discovered age-dependent accumulation of phosphates in skeletal muscle. These and similar findings provided supporting evidence for, now well accepted, theories linking metabolism and aging. Nearly two decades later, age-associated alterations in biochemical molecules were described in the heart. From these small beginnings, the field has grown substantially in recent years. This growing research focus on cardiac aging has, in part, been driven by advances on multiple public health fronts that allow population level clinical presentation of aging related disorders. It is estimated that by 2030, 25% of the worldwide population will be over the age of 65. This review provides an overview of acetylation-dependent regulation of biological processes related to cardiac aging and introduces emerging non-acetyl, acyl-lysine modifications in cardiac function and aging.
Collapse
Affiliation(s)
- Ashley Francois
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Alessandro Canella
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Lynn M Marcho
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Matthew S Stratton
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
| |
Collapse
|
27
|
Budhram-Mahadeo VS, Solomons MR, Mahadeo-Heads EAO. Linking metabolic dysfunction with cardiovascular diseases: Brn-3b/POU4F2 transcription factor in cardiometabolic tissues in health and disease. Cell Death Dis 2021; 12:267. [PMID: 33712567 PMCID: PMC7955040 DOI: 10.1038/s41419-021-03551-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 02/15/2021] [Accepted: 02/16/2021] [Indexed: 12/26/2022]
Abstract
Metabolic and cardiovascular diseases are highly prevalent and chronic conditions that are closely linked by complex molecular and pathological changes. Such adverse effects often arise from changes in the expression of genes that control essential cellular functions, but the factors that drive such effects are not fully understood. Since tissue-specific transcription factors control the expression of multiple genes, which affect cell fate under different conditions, then identifying such regulators can provide valuable insight into the molecular basis of such diseases. This review explores emerging evidence that supports novel and important roles for the POU4F2/Brn-3b transcription factor (TF) in controlling cellular genes that regulate cardiometabolic function. Brn-3b is expressed in insulin-responsive metabolic tissues (e.g. skeletal muscle and adipose tissue) and is important for normal function because constitutive Brn-3b-knockout (KO) mice develop profound metabolic dysfunction (hyperglycaemia; insulin resistance). Brn-3b is highly expressed in the developing hearts, with lower levels in adult hearts. However, Brn-3b is re-expressed in adult cardiomyocytes following haemodynamic stress or injury and is necessary for adaptive cardiac responses, particularly in male hearts, because male Brn-3b KO mice develop adverse remodelling and reduced cardiac function. As a TF, Brn-3b regulates the expression of multiple target genes, including GLUT4, GSK3β, sonic hedgehog (SHH), cyclin D1 and CDK4, which have known functions in controlling metabolic processes but also participate in cardiac responses to stress or injury. Therefore, loss of Brn-3b and the resultant alterations in the expression of such genes could potentially provide the link between metabolic dysfunctions with adverse cardiovascular responses, which is seen in Brn-3b KO mutants. Since the loss of Brn-3b is associated with obesity, type II diabetes (T2DM) and altered cardiac responses to stress, this regulator may provide a new and important link for understanding how pathological changes arise in such endemic diseases.
Collapse
Affiliation(s)
- Vishwanie S Budhram-Mahadeo
- Molecular Biology Development and Disease, Institute of Cardiovascular Science, University College London, London, UK.
| | - Matthew R Solomons
- Molecular Biology Development and Disease, Institute of Cardiovascular Science, University College London, London, UK
| | - Eeshan A O Mahadeo-Heads
- Molecular Biology Development and Disease, Institute of Cardiovascular Science, University College London, London, UK.,College of Medicine and Health, University of Exeter Medical School, St Luke's Campus, Exeter, UK
| |
Collapse
|
28
|
Rubio B, Mora C, Pintado C, Mazuecos L, Fernández A, López V, Andrés A, Gallardo N. The nutrient sensing pathways FoxO1/3 and mTOR in the heart are coordinately regulated by central leptin through PPARβ/δ. Implications in cardiac remodeling. Metabolism 2021; 115:154453. [PMID: 33249043 DOI: 10.1016/j.metabol.2020.154453] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 11/11/2020] [Accepted: 11/23/2020] [Indexed: 10/22/2022]
Abstract
BACKGROUND Cardiovascular disease in obese individuals with type 2 diabetes is often associated with hyperleptinemia and leptin resistance, while other studies support that leptin has cardioprotective effects. Besides, the role of leptin in regulating cardiac atrophy or hypertrophy remains to be clearly defined. In fact, in rats with normal leptin sensitivity, the molecular underpinnings of the effects of central leptin regulating cardiac structural pathways remain poorly understood. OBJECTIVE Hence, we assessed the effects of intracerebroventricular (icv) leptin infusion on cardiac remodeling analyzing FOXO1/3 and mTORC1 pathways, focusing special attention to PPARβ/δ as mediator of central leptin's effects on cardiac metabolism. METHODS Male 3-months-old Wistar rats, infused with icv leptin (0.2 μg/day) for 7 days, were daily co-treated intraperitoneally with the specific PPARβ/δ antagonist GSK0660, at 1 mg/kg per day along leptin treatment. RESULTS Central leptin regulated dynamically, in an opposite manner, the network between FOXOs and mTORC1 and induced an atrophy-related gene program in cardiac tissue. Leptin activated the anti-hypertrophic kinase GSK3β and increased the protein levels of muscle-specific ubiquitin ligases, muscle RING finger 1 (MuRF1) and muscle atrophy F-box (MAFbx)/Atrogin-1 involved in limiting cardiac hypertrophy. FOXO1 activity and the expression of their target genes, Sod2 and Lpl, were also increased in the heart upon central leptin infusion. Besides, Beclin-1 and LC3B-II, gene products of the autophagic pathway response, were upregulated, while the content and expression levels of phenotypic markers of cardiac hypertrophy as ANP and β-myosin heavy chain, gene product of Myh7 were significantly decreased. On the other hand, mTORC1 activity and OXPHOS protein levels were decreased suggesting a key role of central leptin preventing cardiac oxidative stress. In fact, the content of carbonylated proteins, TBARS and ROS/RSN were not increased in cardiac tissue in response to central leptin infusion. Finally, the pharmacological inhibition of PPARβ/δ, via in vivo administration of the selective antagonist GSK0660, blunted the induction of FOXO1/3, Atrogin-1, MuRF1 and GSK3β in the heart mediated by icv leptin infusion. CONCLUSIONS Our results demonstrate that, in lean rats with normal leptin sensitivity, central leptin regulates nutrient sensing pathways in heart contributing to balance cardiac remodeling through the anti- and pro-hypertrophic programs, and in this process is involved PPARβ/δ.
Collapse
Affiliation(s)
- Blanca Rubio
- Universidad de Castilla-La Mancha, Regional Centre for Biomedical Research, Spain; Universidad de Castilla-La Mancha, Biochemistry Section, Faculty of Science and Chemical Technologies, Avda Camilo José Cela 10, 13071 Ciudad Real, Spain
| | - Cristina Mora
- Universidad de Castilla-La Mancha, Regional Centre for Biomedical Research, Spain; Universidad de Castilla-La Mancha, Biochemistry Section, Faculty of Science and Chemical Technologies, Avda Camilo José Cela 10, 13071 Ciudad Real, Spain
| | - Cristina Pintado
- Universidad de Castilla-La Mancha, Regional Centre for Biomedical Research, Spain; Universidad de Castilla-La Mancha, Biochemistry Section, Faculty of Environmental Sciences and Biochemistry, Avda. Carlos III s/n, 45071 Toledo, Spain
| | - Lorena Mazuecos
- Universidad de Castilla-La Mancha, Regional Centre for Biomedical Research, Spain; Universidad de Castilla-La Mancha, Biochemistry Section, Faculty of Science and Chemical Technologies, Avda Camilo José Cela 10, 13071 Ciudad Real, Spain
| | - Alejandro Fernández
- Universidad de Castilla-La Mancha, Regional Centre for Biomedical Research, Spain; Universidad de Castilla-La Mancha, Biochemistry Section, Faculty of Science and Chemical Technologies, Avda Camilo José Cela 10, 13071 Ciudad Real, Spain
| | - Virginia López
- Universidad de Castilla-La Mancha, Regional Centre for Biomedical Research, Spain; Universidad de Castilla-La Mancha, Biochemistry Section, Faculty of Science and Chemical Technologies, Avda Camilo José Cela 10, 13071 Ciudad Real, Spain
| | - Antonio Andrés
- Universidad de Castilla-La Mancha, Regional Centre for Biomedical Research, Spain; Universidad de Castilla-La Mancha, Biochemistry Section, Faculty of Science and Chemical Technologies, Avda Camilo José Cela 10, 13071 Ciudad Real, Spain.
| | - Nilda Gallardo
- Universidad de Castilla-La Mancha, Regional Centre for Biomedical Research, Spain; Universidad de Castilla-La Mancha, Biochemistry Section, Faculty of Science and Chemical Technologies, Avda Camilo José Cela 10, 13071 Ciudad Real, Spain.
| |
Collapse
|
29
|
Liu Y, Chen J, Fontes SK, Bautista EN, Cheng Z. Physiological And Pathological Roles Of Protein Kinase A In The Heart. Cardiovasc Res 2021; 118:386-398. [PMID: 33483740 DOI: 10.1093/cvr/cvab008] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 11/30/2020] [Accepted: 01/08/2021] [Indexed: 12/21/2022] Open
Abstract
Protein kinase A (PKA) is a central regulator of cardiac performance and morphology. Myocardial PKA activation is induced by a variety of hormones, neurotransmitters and stress signals, most notably catecholamines secreted by the sympathetic nervous system. Catecholamines bind β-adrenergic receptors to stimulate cAMP-dependent PKA activation in cardiomyocytes. Elevated PKA activity enhances Ca2+ cycling and increases cardiac muscle contractility. Dynamic control of PKA is essential for cardiac homeostasis, as dysregulation of PKA signaling is associated with a broad range of heart diseases. Specifically, abnormal PKA activation or inactivation contributes to the pathogenesis of myocardial ischemia, hypertrophy, heart failure, as well as diabetic, takotsubo, or anthracycline cardiomyopathies. PKA may also determine sex-dependent differences in contractile function and heart disease predisposition. Here, we describe the recent advances regarding the roles of PKA in cardiac physiology and pathology, highlighting previous study limitations and future research directions. Moreover, we discuss the therapeutic strategies and molecular mechanisms associated with cardiac PKA biology. In summary, PKA could serve as a promising drug target for cardioprotection. Depending on disease types and mechanisms, therapeutic intervention may require either inhibition or activation of PKA. Therefore, specific PKA inhibitors or activators may represent valuable drug candidates for the treatment of heart diseases.
Collapse
Affiliation(s)
- Yuening Liu
- Department of Pharmaceutical Sciences, Washington State University, PBS 423, 412 E. Spokane Falls Blvd, ., Spokane, WA, 99202-2131, USA
| | - Jingrui Chen
- Department of Pharmaceutical Sciences, Washington State University, PBS 423, 412 E. Spokane Falls Blvd, ., Spokane, WA, 99202-2131, USA
| | - Shayne K Fontes
- Department of Pharmaceutical Sciences, Washington State University, PBS 423, 412 E. Spokane Falls Blvd, ., Spokane, WA, 99202-2131, USA
| | - Erika N Bautista
- Department of Pharmaceutical Sciences, Washington State University, PBS 423, 412 E. Spokane Falls Blvd, ., Spokane, WA, 99202-2131, USA
| | - Zhaokang Cheng
- Department of Pharmaceutical Sciences, Washington State University, PBS 423, 412 E. Spokane Falls Blvd, ., Spokane, WA, 99202-2131, USA
| |
Collapse
|
30
|
The piRNA CHAPIR regulates cardiac hypertrophy by controlling METTL3-dependent N 6-methyladenosine methylation of Parp10 mRNA. Nat Cell Biol 2020; 22:1319-1331. [PMID: 33020597 DOI: 10.1038/s41556-020-0576-y] [Citation(s) in RCA: 128] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 08/11/2020] [Indexed: 12/21/2022]
Abstract
PIWI-interacting RNAs (piRNAs) are abundantly expressed during cardiac hypertrophy. However, their functions and molecular mechanisms remain unknown. Here, we identified a cardiac-hypertrophy-associated piRNA (CHAPIR) that promotes pathological hypertrophy and cardiac remodelling by targeting METTL3-mediated N6-methyladenosine (m6A) methylation of Parp10 mRNA transcripts. CHAPIR deletion markedly attenuates cardiac hypertrophy and restores heart function, while administration of a CHAPIR mimic enhances the pathological hypertrophic response in pressure-overloaded mice. Mechanistically, CHAPIR-PIWIL4 complexes directly interact with METTL3 and block the m6A methylation of Parp10 mRNA transcripts, which upregulates PARP10 expression. The CHAPIR-dependent increase in PARP10 promotes the mono-ADP-ribosylation of GSK3β and inhibits its kinase activity, which results in the accumulation of nuclear NFATC4 and the progression of pathological hypertrophy. Hence, our findings reveal that a piRNA-mediated RNA epigenetic mechanism is involved in the regulation of cardiac hypertrophy and that the CHAPIR-METTL3-PARP10-NFATC4 signalling axis could be therapeutically targeted for treating pathological hypertrophy and maladaptive cardiac remodelling.
Collapse
|
31
|
Zhang L, Cao M, Yu Z, Xu S, Ju L, Qian J, Li T, Xu J, Qian W, Zhou J, Li Z. The restoration of Wnt/β-catenin signalling activity by a tuna backbone-derived peptide ameliorates hypoxia-induced cardiomyocyte injury. Am J Transl Res 2020; 12:5221-5236. [PMID: 33042415 PMCID: PMC7540155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Accepted: 08/20/2020] [Indexed: 06/11/2023]
Abstract
Myocardial infarction (MI) is a serious disease with high morbidity and mortality worldwide. Reducing myocardial reperfusion injury in MI patients remains a challenge. The generation of excessive reactive oxygen species (ROS) during reperfusion is known to be responsible for injury. A peptide from tuna backbone protein (APTBP) captured our attention due to its strong antioxidant activity. Here, we aimed to assess the function of APTBP in protecting against myocardial ischaemia-reperfusion (I/R) injury and to clarify the associated mechanism. Two in vitro models generated by hypoxia and cobalt chloride treatment were used to determine the effect of APTBP on cardiomyocytes under hypoxic stress. In vivo, a rat model of I/R was generated to evaluate APTBP functions. As a result, APTBP attenuated hypoxia- or cobalt chloride-induced injury to H9C2 cells and primary cardiomyocytes. Moreover, hypoxia-induced apoptosis, ROS generation and impaired mitochondrial function were also suppressed by APTBP administration. In vivo, tail vein injection of APTBP ameliorated pathological damage and mildly restored cardiac function. To clarify the mechanism, RNA-seq was performed and revealed that the Wnt signalling pathway may be associated with this mechanism. Rescue analysis showed that β-catenin knockdown diminished the protective effect of APTBP and that the expression of an ROS generator abolished the restoration of Wnt/β-catenin signalling induced by APTBP. Collectively, our findings suggest that APTBP reduces cardiomyocyte apoptosis and protects against myocardial ischaemia-reperfusion injury by scavenging ROS and subsequently restoring Wnt/β-catenin signalling.
Collapse
Affiliation(s)
- Le Zhang
- Department of Neonatology, The Affiliated Wuxi Children’s Hospital of Nanjing Medical UniversityWuxi 214023, Jiangsu, China
| | - Minkai Cao
- Department of Obstetrics, The Affiliated Wuxi Maternity and Child Health Care Hospital of Nanjing Medical UniversityWuxi 214002, Jiangsu, China
| | - Zhiwei Yu
- Department of Paediatrics, The Affiliated Wuxi Children’s Hospital of Nanjing Medical UniversityWuxi 214023, Jiangsu, China
| | - Siliang Xu
- State Key Laboratory of Reproductive Medicine, Clinical Centre of Reproductive Medicine, First Affiliated Hospital, Nanjing Medical UniversityNanjing 210029, China
| | - Liang Ju
- Department of Paediatrics, The Affiliated Wuxi Children’s Hospital of Nanjing Medical UniversityWuxi 214023, Jiangsu, China
| | - Jun Qian
- Department of Paediatrics, The Affiliated Wuxi Children’s Hospital of Nanjing Medical UniversityWuxi 214023, Jiangsu, China
| | - Tianyu Li
- Department of Paediatrics, The Affiliated Wuxi Children’s Hospital of Nanjing Medical UniversityWuxi 214023, Jiangsu, China
| | - Jianjuan Xu
- Department of Obstetrics, The Affiliated Wuxi Maternity and Child Health Care Hospital of Nanjing Medical UniversityWuxi 214002, Jiangsu, China
| | - Wei Qian
- Department of Paediatrics, The Affiliated Wuxi Children’s Hospital of Nanjing Medical UniversityWuxi 214023, Jiangsu, China
| | - Jian Zhou
- Department of Paediatric Laboratory, The Affiliated Wuxi Children’s Hospital of Nanjing Medical UniversityWuxi 214023, Jiangsu, China
| | - Zhengying Li
- Department of Neonatology, The Affiliated Wuxi Children’s Hospital of Nanjing Medical UniversityWuxi 214023, Jiangsu, China
| |
Collapse
|
32
|
Ni M, Zhou H, Zhang J, Jin D, Lu T, Busuttil RW, Kupiec-Weglinski JW, Wang X, Zhai Y. Isoform- and Cell Type-Specific Roles of Glycogen Synthase Kinase 3 N-Terminal Serine Phosphorylation in Liver Ischemia Reperfusion Injury. THE JOURNAL OF IMMUNOLOGY 2020; 205:1147-1156. [PMID: 32680958 DOI: 10.4049/jimmunol.2000397] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 06/21/2020] [Indexed: 12/31/2022]
Abstract
Glycogen synthase kinase 3 (Gsk3) α and β are both constitutively active and inhibited upon stimulation by N-terminal serine phosphorylation. Although roles of active Gsk3 in liver ischemia reperfusion injury (IRI) have been well appreciated, whether Gsk3 N-terminal serine phosphorylation has any functional significance in the disease process remains unclear. In a murine liver partial warm ischemia model, we studied Gsk3 N-terminal serine mutant knock-in (KI) mice and showed that liver IRI was decreased in Gsk3αS21A but increased in Gsk3βS9A mutant KI mice. Bone marrow chimeric experiments revealed that the Gsk3α, but not β, mutation in liver parenchyma protected from IRI, and both mutations in bone marrow-derived cells exacerbated liver injuries. Mechanistically, mutant Gsk3α protected hepatocytes from inflammatory (TNF-α) cell death by the activation of HIV-1 TAT-interactive protein 60 (TIP60)-mediated autophagy pathway. The pharmacological inhibition of TIP60 or autophagy diminished the protection of the Gsk3α mutant hepatocytes from inflammatory cell death in vitro and the Gsk3α mutant KI mice from liver IRI in vivo. Thus, Gsk3 N-terminal serine phosphorylation inhibits liver innate immune activation but suppresses hepatocyte autophagy in response to inflammation. Gsk3 αS21, but not βS9, mutation is sufficient to sustain Gsk4 activities in hepatocytes and protect livers from IRI via TIP60 activation.
Collapse
Affiliation(s)
- Ming Ni
- Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095.,Department of Liver Surgery, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210008, China
| | - Haoming Zhou
- Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095.,Department of Liver Surgery, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210008, China
| | - Jing Zhang
- Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095
| | - Dan Jin
- Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095.,Department of Obstetrics and Gynecology, Shanghai Jiaotong University, Shanghai 200025, China; and
| | - Tianfei Lu
- Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095.,Liver Surgery, Renji Hospital, Shanghai Jiaotong University, Shanghai 200025, China
| | - Ronald W Busuttil
- Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095
| | - Jerzy W Kupiec-Weglinski
- Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095
| | - Xuehao Wang
- Department of Liver Surgery, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210008, China;
| | - Yuan Zhai
- Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095;
| |
Collapse
|
33
|
Nair VD, Ge Y, Li S, Pincas H, Jain N, Seenarine N, Amper MAS, Goodpaster BH, Walsh MJ, Coen PM, Sealfon SC. Sedentary and Trained Older Men Have Distinct Circulating Exosomal microRNA Profiles at Baseline and in Response to Acute Exercise. Front Physiol 2020; 11:605. [PMID: 32587527 PMCID: PMC7298138 DOI: 10.3389/fphys.2020.00605] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 05/14/2020] [Indexed: 12/20/2022] Open
Abstract
Exercise has multi-systemic benefits and attenuates the physiological impairments associated with aging. Emerging evidence suggests that circulating exosomes mediate some of the beneficial effects of exercise via the transfer of microRNAs between tissues. However, the impact of regular exercise and acute exercise on circulating exosomal microRNAs (exomiRs) in older populations remains unknown. In the present study, we analyzed circulating exomiR expression in endurance-trained elderly men (n = 5) and age-matched sedentary males (n = 5) at baseline (Pre), immediately after a forty minute bout of aerobic exercise on a cycle ergometer (Post), and three hours after this acute exercise (3hPost). Following the isolation and enrichment of exosomes from plasma, exosome-enriched preparations were characterized and exomiR levels were determined by sequencing. The effect of regular exercise on circulating exomiRs was assessed by comparing the baseline expression levels in the trained and sedentary groups. The effect of acute exercise was determined by comparing baseline and post-training expression levels in each group. Regular exercise resulted in significantly increased baseline expression of three exomiRs (miR-486-5p, miR-215-5p, miR-941) and decreased expression of one exomiR (miR-151b). Acute exercise altered circulating exomiR expression in both groups. However, exomiRs regulated by acute exercise in the trained group (7 miRNAs at Post and 8 at 3hPost) were distinct from those in the sedentary group (9 at Post and 4 at 3hPost). Pathway analysis prediction and reported target validation experiments revealed that the majority of exercise-regulated exomiRs are targeting genes that are related to IGF-1 signaling, a pathway involved in exercise-induced muscle and cardiac hypertrophy. The immediately post-acute exercise exomiR signature in the trained group correlates with activation of IGF-1 signaling, whereas in the sedentary group it is associated with inhibition of IGF-1 signaling. While further validation is needed, including measurements of IGF-1/IGF-1 signaling in blood or skeletal muscle, our results suggest that training status may counteract age-related anabolic resistance by modulating circulating exomiR profiles both at baseline and in response to acute exercise.
Collapse
Affiliation(s)
- Venugopalan D. Nair
- Department of Neurology, Center for Advanced Research on Diagnostic Assays, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Yongchao Ge
- Department of Neurology, Center for Advanced Research on Diagnostic Assays, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Side Li
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Hanna Pincas
- Department of Neurology, Center for Advanced Research on Diagnostic Assays, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Nimisha Jain
- Department of Neurology, Center for Advanced Research on Diagnostic Assays, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Nitish Seenarine
- Department of Neurology, Center for Advanced Research on Diagnostic Assays, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Mary Anne S. Amper
- Department of Neurology, Center for Advanced Research on Diagnostic Assays, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Bret H. Goodpaster
- Translational Research Institute, AdventHealth, Orlando, FL, United States
| | - Martin J. Walsh
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Paul M. Coen
- Translational Research Institute, AdventHealth, Orlando, FL, United States
| | - Stuart C. Sealfon
- Department of Neurology, Center for Advanced Research on Diagnostic Assays, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
34
|
Aryan L, Medzikovic L, Umar S, Eghbali M. Pregnancy-associated cardiac dysfunction and the regulatory role of microRNAs. Biol Sex Differ 2020; 11:14. [PMID: 32252821 PMCID: PMC7137306 DOI: 10.1186/s13293-020-00292-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 03/18/2020] [Indexed: 12/14/2022] Open
Abstract
Many crucial cardiovascular adaptations occur in the body during pregnancy to ensure successful gestation. Maladaptation of the cardiovascular system during pregnancy can lead to complications that promote cardiac dysfunction and may lead to heart failure (HF). About 12% of pregnancy-related deaths in the USA have been attributed to HF and the detrimental effects of cardiovascular complications on the heart can be long-lasting, pre-disposing the mother to HF later in life. Indeed, cardiovascular complications such as gestational diabetes mellitus, preeclampsia, gestational hypertension, and peripartum cardiomyopathy have been shown to induce cardiac metabolic dysfunction, oxidative stress, fibrosis, apoptosis, and diastolic and systolic dysfunction in the hearts of pregnant women, all of which are hallmarks of HF. The exact etiology and cardiac pathophysiology of pregnancy-related complications is not yet fully deciphered. Furthermore, diagnosis of cardiac dysfunction in pregnancy is often made only after clinical symptoms are already present, thus necessitating the need for novel diagnostic and prognostic biomarkers. Mounting data demonstrates an altered expression of maternal circulating miRNAs during pregnancy affected by cardiovascular complications. Throughout the past decade, miRNAs have become of growing interest as modulators and biomarkers of pathophysiology, diagnosis, and prognosis in cardiac dysfunction. While the association between pregnancy-related cardiovascular complications and cardiac dysfunction or HF is becoming increasingly evident, the roles of miRNA-mediated regulation herein remain poorly understood. Therefore, this review will summarize current reports on pregnancy-related cardiovascular complications that may lead to cardiac dysfunction and HF during and after pregnancy in previously healthy women, with a focus on the pathophysiological role of miRNAs.
Collapse
Affiliation(s)
- Laila Aryan
- Department of Anesthesiology, Division of Molecular Medicine, David Geffen School of Medicine at University of California, Los Angeles, BH-550 CHS, Los Angeles, CA, 90095-7115, USA
| | - Lejla Medzikovic
- Department of Anesthesiology, Division of Molecular Medicine, David Geffen School of Medicine at University of California, Los Angeles, BH-550 CHS, Los Angeles, CA, 90095-7115, USA
| | - Soban Umar
- Department of Anesthesiology, Division of Molecular Medicine, David Geffen School of Medicine at University of California, Los Angeles, BH-550 CHS, Los Angeles, CA, 90095-7115, USA
| | - Mansoureh Eghbali
- Department of Anesthesiology, Division of Molecular Medicine, David Geffen School of Medicine at University of California, Los Angeles, BH-550 CHS, Los Angeles, CA, 90095-7115, USA.
| |
Collapse
|
35
|
Cell-to-cell lactate shuttle operates in heart and is important in age-related heart failure. Aging (Albany NY) 2020; 12:3388-3406. [PMID: 32035422 PMCID: PMC7066931 DOI: 10.18632/aging.102818] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 01/27/2020] [Indexed: 01/02/2023]
Abstract
Recent studies have revealed a resemblance of a HIF-regulated heart and brain glycolytic profiles prompting the hypothesis that the classical cell-to-cell lactate shuttle observed between astrocytes and neurons operates also in heart - between cardiac fibroblasts and cardiomyocytes. Here, we demonstrate that co-culturing of cardiomyocytes with cardiac fibroblasts leads to orchestrated changes in expression and/or localization pattern of glucose metabolism enzymes and lactate transport proteins in both cell types. These changes are regulated by paracrine signaling using microvesicle-packed and soluble factors released to the culture medium and, taken together, they concur with the cardiac lactate shuttle hypothesis. The results presented here show that similarity of heart and brain proteomes demonstrated earlier extend to physiological level and provide a theoretical rationale for designing novel therapeutic strategies for treatment of cardiomyopathies resulting from disruption of the maturation of cardiac metabolic pathways, and of heart failure associated with metabolic complications and age-related heart failure linked with extracellular matrix deposition and hypoxia.
Collapse
|
36
|
Simpson LJ, Reader JS, Tzima E. Mechanical Regulation of Protein Translation in the Cardiovascular System. Front Cell Dev Biol 2020; 8:34. [PMID: 32083081 PMCID: PMC7006472 DOI: 10.3389/fcell.2020.00034] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 01/15/2020] [Indexed: 12/12/2022] Open
Abstract
The cardiovascular system can sense and adapt to changes in mechanical stimuli by remodeling the physical properties of the heart and blood vessels in order to maintain homeostasis. Imbalances in mechanical forces and/or impaired sensing are now not only implicated but are, in some cases, considered to be drivers for the development and progression of cardiovascular disease. There is now growing evidence to highlight the role of mechanical forces in the regulation of protein translation pathways. The canonical mechanism of protein synthesis typically involves transcription and translation. Protein translation occurs globally throughout the cell to maintain general function but localized protein synthesis allows for precise spatiotemporal control of protein translation. This Review will cover studies on the role of biomechanical stress -induced translational control in the heart (often in the context of physiological and pathological hypertrophy). We will also discuss the much less studied effects of mechanical forces in regulating protein translation in the vasculature. Understanding how the mechanical environment influences protein translational mechanisms in the cardiovascular system, will help to inform disease pathogenesis and potential areas of therapeutic intervention.
Collapse
Affiliation(s)
- Lisa J Simpson
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - John S Reader
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Ellie Tzima
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
37
|
Sodium Selenate Ameliorates Cardiac Injury Developed from High-Fat Diet in Mice through Regulation of Autophagy Activity. Sci Rep 2019; 9:18752. [PMID: 31822702 PMCID: PMC6904559 DOI: 10.1038/s41598-019-54985-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 11/19/2019] [Indexed: 02/06/2023] Open
Abstract
Obesity is often accompanied by dyslipidemia, high blood glucose, hypertension, atherosclerosis, and myocardial dysfunction. Selenate is a vital antioxidant in the cardiovascular system. The beneficial effects of selenate on obesity-associated cardiac dysfunction and potential molecular mechanism were identified in both H9C2 cells and C57BL/6J mice hearts. The cardiac histological preformation in C57BL/6J mice were evaluated by cross-sectional area (CSA) of cardiomyocytes and percent area of fibrosis in the left ventricles. The cardiac autophagy flux in H9C2 cells and C57BL/6J mice hearts was analyzed by Western blots and the number of autophagosomes and autolysosome in H9C2 cells. In the present study, we found that lipid overload caused increases in serum lipid, CSA, and percent area of fibrosis. We further found that lipid-induced accumulation of autophagosomes was due to depressed autophagy degradation, which was not restored in the pretreatment with 3-methyladenine and chloroquine, whereas, it was improved by rapamycin. Moreover, we demonstrated that increased levels of serum lipid, CSA, percent area of fibrosis and mRNA expression related to cardiomyocytes hypertrophy and fibrosis were significantly reduced after selenate treatments of mice. We also found selenate treatment significantly down-regulated activity of the Akt pathway, which was activated in response to lipid-overload. Furthermore, selenate dramatically improved cardiac autophagic degradation which was suppressed after exposure to lipid-overload in both H9C2 cells and C57BL/6J mice hearts. Taken together, selenate offers therapeutic intervention in lipid-related metabolic disorders, and protection against cardiac remodeling, likely through regulation of the activity of autophagic degradation and Akt pathway.
Collapse
|
38
|
2,5-Dimethylcelecoxib prevents isoprenaline-induced cardiomyocyte hypertrophy and cardiac fibroblast activation by inhibiting Akt-mediated GSK-3 phosphorylation. Biochem Pharmacol 2019; 168:82-90. [DOI: 10.1016/j.bcp.2019.06.018] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 06/18/2019] [Indexed: 11/22/2022]
|
39
|
Oláh A, Mátyás C, Kellermayer D, Ruppert M, Barta BA, Sayour AA, Török M, Koncsos G, Giricz Z, Ferdinandy P, Merkely B, Radovits T. Sex Differences in Morphological and Functional Aspects of Exercise-Induced Cardiac Hypertrophy in a Rat Model. Front Physiol 2019; 10:889. [PMID: 31354526 PMCID: PMC6639783 DOI: 10.3389/fphys.2019.00889] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 06/26/2019] [Indexed: 01/20/2023] Open
Abstract
Background: Recent evidences suggest that sex hormones may be involved in the regulation of exercise-induced left ventricular (LV) hypertrophy. However, the sex-specific functional consequences of exercise-induced myocardial hypertrophy is still not investigated in detail. We aimed at understanding the sex-specific functional and morphological alterations in the LV and the underlying molecular changes in a rat model of athlete’s heart. Methods: We divided our young, adult male and female rats into control and exercised groups. Athlete’s heart was induced by a 12-week long swim training. Following the training period, we assessed LV hypertrophy with echocardiography, while pressure-volume analysis was performed to investigate in vivo LV function. After in vivo experiments, molecular biological studies and histological investigations were performed. Results: Echocardiography and post-mortem measured heart weight data indicated LV hypertrophy in both genders, nevertheless it was more pronounced in females. Despite the more significant relative hypertrophy in females, characteristic functional parameters did not show notable differences between the genders. LV pressure-volume analysis showed increased stroke volume, improved contractility and stroke work and unaltered LV stiffness in both male and female exercised rats, while active relaxation was ameliorated solely in male animals. The induction of Akt signaling was more significant in females compared to males. There was also a characteristic difference in the mitogen-activated protein kinase pathway as suppressed phosphorylation of p44/42 MAPK (Erk) and mTOR was observed in female exercised rats, but not in male ones. Myosin heavy chain α (MHC)/β-MHC ratio did not differ in males, but increased markedly in females. Conclusion: Our results confirm that there is a more pronounced exercise-induced LV hypertrophy in females as compared to the males, however, there are only minor differences regarding LV function. There are characteristic molecular differences between male and female animals, that can explain different degrees of LV hypertrophy.
Collapse
Affiliation(s)
- Attila Oláh
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Csaba Mátyás
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | | | - Mihály Ruppert
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | | | - Alex Ali Sayour
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Marianna Török
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Gábor Koncsos
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Zoltáng Giricz
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary.,Pharmahungary Group, Szeged, Hungary
| | - Béla Merkely
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Tamás Radovits
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| |
Collapse
|
40
|
Shang L, Pin L, Zhu S, Zhong X, Zhang Y, Shun M, Liu Y, Hou M. Plantamajoside attenuates isoproterenol-induced cardiac hypertrophy associated with the HDAC2 and AKT/ GSK-3β signaling pathway. Chem Biol Interact 2019; 307:21-28. [DOI: 10.1016/j.cbi.2019.04.024] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 04/05/2019] [Accepted: 04/17/2019] [Indexed: 01/01/2023]
|
41
|
Badimon L, Casaní L, Camino-Lopez S, Juan-Babot O, Borrell-Pages M. GSK3β inhibition and canonical Wnt signaling in mice hearts after myocardial ischemic damage. PLoS One 2019; 14:e0218098. [PMID: 31220102 PMCID: PMC6586285 DOI: 10.1371/journal.pone.0218098] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 05/25/2019] [Indexed: 12/22/2022] Open
Abstract
Aims Myocardial infarction induces myocardial injury and tissue damage. During myocardial infarction strong cellular response is initiated to salvage the damaged tissues. This response is associated with the induction of different signaling pathways. Of these, the canonical Wnt signaling is increasingly important for its prosurvival cellular role, making it a good candidate for the search of new molecular targets to develop therapies to prevent heart failure in infarcted patients. Methods Herein we report that GSK3β regulates the canonical Wnt signaling in C57Bl6 mice hearts. GSK3β is a canonical Wnt pathway inhibitor. Using GSK3β inhibitors and inducing myocardial injury (MI) in Lrp5-/- mice model we show that GSK3β phosphorylation levels regulate downstream canonical Wnt pathway genes in the ischemic heart. In the setting of MI, myocardial damage assessment usually correlates with functional and clinical outcomes. Therefore, we measured myocardial injury size in Wt and Lrp5-/- mice in the presence and absence of two different GSK3 inhibitors prior to MI. Myocardial injury was independent of GSK3 inhibitor treatments and GSK3β expression levels. Results These studies support a central role for GSK3β in the activation of the canonical Wnt pathway in the Wt heart. Although LRP5 is protective against myocardial injury, GSK3β expression levels do not regulate heart damage.
Collapse
Affiliation(s)
- Lina Badimon
- Cardiovascular Program ICCC, Institut de Recerca de l’-Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- CIBER-CV, Instituto de Salud Carlos III, Madrid, Spain
- Cardiovascular Research Chair, UAB, Barcelona, Spain
| | - Laura Casaní
- Cardiovascular Program ICCC, Institut de Recerca de l’-Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Sandra Camino-Lopez
- Cardiovascular Program ICCC, Institut de Recerca de l’-Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Oriol Juan-Babot
- Cardiovascular Program ICCC, Institut de Recerca de l’-Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Maria Borrell-Pages
- Cardiovascular Program ICCC, Institut de Recerca de l’-Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- CIBER-CV, Instituto de Salud Carlos III, Madrid, Spain
- * E-mail:
| |
Collapse
|
42
|
Narasimhan G, Henderson J, Luong HT, Rajasekaran NS, Qin G, Zhang J, Krishnamurthy P. OBG-like ATPase 1 inhibition attenuates angiotensin II-induced hypertrophic response in human ventricular myocytes via GSK-3beta/beta-catenin signalling. Clin Exp Pharmacol Physiol 2019; 46:743-751. [PMID: 31063653 DOI: 10.1111/1440-1681.13101] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 04/23/2019] [Accepted: 04/30/2019] [Indexed: 11/28/2022]
Abstract
Obg-like ATPase 1 (OLA1) that possesses both GTP and ATP hydrolyzing activities has been shown to be involved in translational regulation of cancer cell growth and survival. Also, GSK3β signalling has been implicated in cardiac development and disease. However, the role of OLA1 in pathological cardiac hypertrophy is unknown. We sought to understand the mechanism by which OLA1 regulates GSK3β-β-Catenin signalling and its functional significance in angiotensin-II (ANG II)-induced cardiac hypertrophic response. OLA1 function and its endogenous interaction with GSK3β/β-catenin signalling in cultured human ventricular cardiomyocytes (AC16 cells) and mouse hearts (in vivo) was evaluated with/without ANG II-stimulated hypertrophic response. ANG II administration in mice increases myocardial OLA1 protein expression with a corresponding increase in GSK3β phosphorylation and decrease in β-Catenin phosphorylation. Cultured cardiomyocytes treated with ANG II show endogenous interaction between OLA1 and GSK3β, nuclear accumulation of β-Catenin and significant increase in cell size and expression of hypertrophic marker genes such as atrial natriuretic factor (ANF; NPPA) and β-myosin heavy chain (MYH7). Intriguingly, OLA1 inhibition attenuates the above hypertrophic response in cardiomyocytes. Taken together, our data suggest that OLA1 plays a detrimental role in hypertrophic response via GSK3β/β-catenin signalling. Translation strategies to target OLA1 might potentially limit the underlying molecular derangements leading to left ventricular dysfunction in patients with maladaptive cardiac hypertrophy.
Collapse
Affiliation(s)
- Gayathri Narasimhan
- Department of Biomedical Engineering, Schools of Medicine and Engineering, University of Alabama at Birmingham, Birmingham, Alabama
| | - John Henderson
- Department of Biomedical Engineering, Schools of Medicine and Engineering, University of Alabama at Birmingham, Birmingham, Alabama
| | - Hien T Luong
- Department of Biomedical Engineering, Schools of Medicine and Engineering, University of Alabama at Birmingham, Birmingham, Alabama
| | - Namakkal Soorapan Rajasekaran
- Division of Molecular & Cellular Pathology, Department of Pathology, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Gangjian Qin
- Department of Biomedical Engineering, Schools of Medicine and Engineering, University of Alabama at Birmingham, Birmingham, Alabama
| | - Jianyi Zhang
- Department of Biomedical Engineering, Schools of Medicine and Engineering, University of Alabama at Birmingham, Birmingham, Alabama
| | - Prasanna Krishnamurthy
- Department of Biomedical Engineering, Schools of Medicine and Engineering, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
43
|
Mohajeri M, Behnam B, Barreto GE, Sahebkar A. Carbon nanomaterials and amyloid-beta interactions: potentials for the detection and treatment of Alzheimer's disease? Pharmacol Res 2019; 143:186-203. [DOI: 10.1016/j.phrs.2019.03.023] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Revised: 03/26/2019] [Accepted: 03/26/2019] [Indexed: 01/24/2023]
|
44
|
Gizak A, Duda P, Wisniewski J, Rakus D. Fructose-1,6-bisphosphatase: From a glucose metabolism enzyme to multifaceted regulator of a cell fate. Adv Biol Regul 2019; 72:41-50. [PMID: 30871972 DOI: 10.1016/j.jbior.2019.03.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 03/04/2019] [Accepted: 03/05/2019] [Indexed: 06/09/2023]
Abstract
Fructose-1,6-bisphosphatase (FBPase) is one of the ancient, evolutionarily conserved enzymes of carbohydrate metabolism. It has been described for a first time in 1943, however, for the next half a century mostly kinetic and structural parameters of animal FBPases have been studied. Discovery of ubiquitous expression of the muscle isozyme of FBPase, thus far considered to merely regulate glycogen synthesis from carbohydrate precursors, and its nuclear localisation in several cell types has risen new interest in the protein, resulting in numerous publications revealing complex functions/properties of FBPase. This review summarises the current knowledge of FBPase in animal cells providing evidence that the enzyme merits the name of moonlighting protein.
Collapse
Affiliation(s)
- Agnieszka Gizak
- Department of Molecular Physiology and Neurobiology, Wroclaw University, Wroclaw, Poland
| | - Przemyslaw Duda
- Department of Molecular Physiology and Neurobiology, Wroclaw University, Wroclaw, Poland
| | - Janusz Wisniewski
- Department of Molecular Physiology and Neurobiology, Wroclaw University, Wroclaw, Poland
| | - Dariusz Rakus
- Department of Molecular Physiology and Neurobiology, Wroclaw University, Wroclaw, Poland.
| |
Collapse
|
45
|
Ye J, Zheng Q, Jia S, Qiao X, Cao Y, Xu C, Weng L, Zhao L, Chen Y, Liu J, Wang T, Cheng H, Zheng M. Programmed Cell Death 5 Provides Negative Feedback on Cardiac Hypertrophy Through the Stabilization of Sarco/Endoplasmic Reticulum Ca 2+-ATPase 2a Protein. Hypertension 2019; 72:889-901. [PMID: 30354711 DOI: 10.1161/hypertensionaha.118.11357] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
PDCD5 (programmed cell death 5) is ubiquitously expressed in tissues, including the heart; however, the mechanism underlying the cardiac function of PDCD5 has not been understood. We investigated the mechanisms of PDCD5 in the pathogenesis of cardiac hypertrophy. Cardiac-specific PDCD5 knockout mice developed severe cardiac hypertrophy and impaired cardiac function, whereas PDCD5 protein was significantly increased in transverse aortic constriction mouse hearts and phenylephrine-stimulated cardiomyocytes. Overexpression of PDCD5 inhibited phenylephrine-induced cardiomyocyte hypertrophy, and knockdown of PDCD5 induced cardiomyocyte hypertrophy and aggravated phenylephrine-induced hypertrophy. The expression of PDCD5 protein was regulated by NFATc2 (nuclear factor of activated T cells c2) during hypertrophy. SERCA2a (sarco/endoplasmic reticulum Ca2+-ATPase 2a) expression was decreased in PDCD5-deficient mouse hearts because of increased ubiquitination. PDCD5-deficient cardiomyocytes displayed decreased calcium uptake rate, slowed decay of Ca2+ transients, decreased calcium stores, and diastolic dysfunction. Moreover, reintroduction of PDCD5 in PDCD5-deficient mouse hearts reserved SERCA2a protein, suppressed NFATc2 protein, and rescued the hypertrophy and cardiac dysfunction. Our results revealed that PDCD5 is a novel target of NFATc2 in the hypertrophic heart and provides negative feedback to protect the heart against excessive hypertrophy via the stabilization of SERCA2a protein.
Collapse
Affiliation(s)
- Jingjing Ye
- From the Department of Physiology and Pathophysiology, School of Basic Medical Sciences (J.Y., S.J., X.Q., Y.C., C.X., L.W., L.Z., M.Z.), Peking University Health Science Center, Beijing, China
| | - Qiaoxia Zheng
- Institute of Molecular Medicine, Peking University, Beijing, P.R. China (Q.Z., H.C.)
| | - Shi Jia
- From the Department of Physiology and Pathophysiology, School of Basic Medical Sciences (J.Y., S.J., X.Q., Y.C., C.X., L.W., L.Z., M.Z.), Peking University Health Science Center, Beijing, China
| | - Xue Qiao
- From the Department of Physiology and Pathophysiology, School of Basic Medical Sciences (J.Y., S.J., X.Q., Y.C., C.X., L.W., L.Z., M.Z.), Peking University Health Science Center, Beijing, China
| | - Yangpo Cao
- From the Department of Physiology and Pathophysiology, School of Basic Medical Sciences (J.Y., S.J., X.Q., Y.C., C.X., L.W., L.Z., M.Z.), Peking University Health Science Center, Beijing, China
| | - Chunling Xu
- From the Department of Physiology and Pathophysiology, School of Basic Medical Sciences (J.Y., S.J., X.Q., Y.C., C.X., L.W., L.Z., M.Z.), Peking University Health Science Center, Beijing, China
| | - Lin Weng
- From the Department of Physiology and Pathophysiology, School of Basic Medical Sciences (J.Y., S.J., X.Q., Y.C., C.X., L.W., L.Z., M.Z.), Peking University Health Science Center, Beijing, China
| | - Lifang Zhao
- From the Department of Physiology and Pathophysiology, School of Basic Medical Sciences (J.Y., S.J., X.Q., Y.C., C.X., L.W., L.Z., M.Z.), Peking University Health Science Center, Beijing, China
| | - Yingyu Chen
- Key Laboratory of Medical Immunology, Ministry of Health (Y.C.), Peking University Health Science Center, Beijing, China
| | - Jian Liu
- Departments of Cardiology (J.L.), Peking University People's Hospital, Beijing, China
| | - Tianbing Wang
- Trauma and Orthopedics (T.W.), Peking University People's Hospital, Beijing, China
| | - Heping Cheng
- Institute of Molecular Medicine, Peking University, Beijing, P.R. China (Q.Z., H.C.)
| | - Ming Zheng
- From the Department of Physiology and Pathophysiology, School of Basic Medical Sciences (J.Y., S.J., X.Q., Y.C., C.X., L.W., L.Z., M.Z.), Peking University Health Science Center, Beijing, China
| |
Collapse
|
46
|
Ahmad F, Singh AP, Tomar D, Rahmani M, Zhang Q, Woodgett JR, Tilley DG, Lal H, Force T. Cardiomyocyte-GSK-3α promotes mPTP opening and heart failure in mice with chronic pressure overload. J Mol Cell Cardiol 2019; 130:65-75. [PMID: 30928428 DOI: 10.1016/j.yjmcc.2019.03.020] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 02/09/2019] [Accepted: 03/25/2019] [Indexed: 01/19/2023]
Abstract
Chronic pressure-overload (PO)- induced cardiomyopathy is one of the leading causes of left ventricular (LV) remodeling and heart failure. The role of the α isoform of glycogen synthase kinase-3 (GSK-3α) in PO-induced cardiac remodeling is unclear and its downstream molecular targets are largely unknown. To investigate the potential roles of GSK-3α, cardiomyocyte-specific GSK-3α conditional knockout (cKO) and control mice underwent trans-aortic constriction (TAC) or sham surgeries. Cardiac function in the cKOs and littermate controls declined equally up to 2 weeks of TAC. At 4 week, cKO animals retained concentric LV remodeling and showed significantly less decline in contractile function both at systole and diastole, vs. controls which remained same until the end of the study (6 wk). Histological analysis confirmed preservation of LV chamber and protection against TAC-induced cellular hypertrophy in the cKO. Consistent with attenuated hypertrophy, significantly lower level of cardiomyocyte apoptosis was observed in the cKO. Mechanistically, GSK-3α was found to regulate mitochondrial permeability transition pore (mPTP) opening and GSK-3α-deficient mitochondria showed delayed mPTP opening in response to Ca2+ overload. Consistently, overexpression of GSK-3α in cardiomyocytes resulted in elevated Bax expression, increased apoptosis, as well as a reduction of maximum respiration capacity and cell viability. Taken together, we show for the first time that GSK-3α regulates mPTP opening under pathological conditions, likely through Bax overexpression. Genetic ablation of cardiomyocyte GSK-3α protects against chronic PO-induced cardiomyopathy and adverse LV remodeling, and preserves contractile function. Selective inhibition of GSK-3α using isoform-specific inhibitors could be a viable therapeutic strategy to limit PO-induced heart failure.
Collapse
Affiliation(s)
- Firdos Ahmad
- College of Medicine and Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates.
| | - Anand P Singh
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Dhanendra Tomar
- Center for Translational Medicine, School of Medicine, Temple University, Philadelphia, PA, USA
| | - Mohamed Rahmani
- College of Medicine and Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
| | - Qinkun Zhang
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - James R Woodgett
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Douglas G Tilley
- Center for Translational Medicine, School of Medicine, Temple University, Philadelphia, PA, USA
| | - Hind Lal
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Thomas Force
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
47
|
Abstract
Glycogen synthase kinase-3 (GSK-3) is a cytoplasmic serine/threonine protein kinase which is known to regulate a variety of cellular processes through a number of signaling pathways important for cell proliferation, stem cell renewal, apoptosis and development. Although GSK-3 exists in a variety of tissues, this kinase plays very important roles in the heart to control its development through the formation of heart and cardiomyocyte proliferation. GSK-3 is also recognized as one of the main molecules that control cardiac hypertrophy and fibrosis. Therefore, GSK-3 could be an attractive target for the development of new drugs to cure cardiac diseases. The present review summarizes the roles of GSK-3 in the signaling pathways and the heart, and discusses the possibility of new drug development targeting this kinase.
Collapse
|
48
|
Miranda-Silva D, Gonçalves-Rodrigues P, Almeida-Coelho J, Hamdani N, Lima T, Conceição G, Sousa-Mendes C, Cláudia-Moura, González A, Díez J, Linke WA, Leite-Moreira A, Falcão-Pires I. Characterization of biventricular alterations in myocardial (reverse) remodelling in aortic banding-induced chronic pressure overload. Sci Rep 2019; 9:2956. [PMID: 30814653 PMCID: PMC6393473 DOI: 10.1038/s41598-019-39581-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 01/18/2019] [Indexed: 01/03/2023] Open
Abstract
Aortic Stenosis (AS) is the most frequent valvulopathy in the western world. Traditionally aortic valve replacement (AVR) has been recommended immediately after the onset of heart failure (HF) symptoms. However, recent evidence suggests that AVR outcome can be improved if performed earlier. After AVR, the process of left ventricle (LV) reverse remodelling (RR) is variable and frequently incomplete. In this study, we aimed at detecting mechanism underlying the process of LV RR regarding myocardial structural, functional and molecular changes before the onset of HF symptoms. Wistar-Han rats were subjected to 7-weeks of ascending aortic-banding followed by a 2-week period of debanding to resemble AS-induced LV remodelling and the early events of AVR-induced RR, respectively. This resulted in 3 groups: Sham (n = 10), Banding (Ba, n = 15) and Debanding (Deb, n = 10). Concentric hypertrophy and diastolic dysfunction (DD) were patent in the Ba group. Aortic-debanding induced RR, which promoted LV functional recovery, while cardiac structure did not normalise. Cardiac parameters of RV dysfunction, assessed by echocardiography and at the cardiomyocyte level prevailed altered after debanding. After debanding, these alterations were accompanied by persistent changes in pathways associated to myocardial hypertrophy, fibrosis and LV inflammation. Aortic banding induced pulmonary arterial wall thickness to increase and correlates negatively with effort intolerance and positively with E/e′ and left atrial area. We described dysregulated pathways in LV and RV remodelling and RR after AVR. Importantly we showed important RV-side effects of aortic constriction, highlighting the impact that LV-reverse remodelling has on both ventricles.
Collapse
Affiliation(s)
| | | | | | - Nazha Hamdani
- Department of Systems Physiology, Ruhr University, Bochum, Germany
| | - Tânia Lima
- Department of Surgery and Physiology, University of Porto, Porto, Portugal
| | - Glória Conceição
- Department of Surgery and Physiology, University of Porto, Porto, Portugal
| | | | - Cláudia-Moura
- Department of Surgery and Physiology, University of Porto, Porto, Portugal
| | - Arantxa González
- Program of Cardiovascular Diseases, Centre for Applied Medical Research, University of Navarra and CIBERCV, Pamplona, Spain.,Department of Cardiology and Cardiac Surgery and Department of Nephrology, University of Navarra Clinic, Pamplona, Spain
| | - Javier Díez
- Program of Cardiovascular Diseases, Centre for Applied Medical Research, University of Navarra and CIBERCV, Pamplona, Spain.,Department of Cardiology and Cardiac Surgery and Department of Nephrology, University of Navarra Clinic, Pamplona, Spain
| | - Wolfgang A Linke
- Institute of Physiology II, University of Muenster, Muenster, Germany
| | | | - Inês Falcão-Pires
- Department of Surgery and Physiology, University of Porto, Porto, Portugal.
| |
Collapse
|
49
|
Iqubal A, Iqubal MK, Sharma S, Ansari MA, Najmi AK, Ali SM, Ali J, Haque SE. Molecular mechanism involved in cyclophosphamide-induced cardiotoxicity: Old drug with a new vision. Life Sci 2018; 218:112-131. [PMID: 30552952 DOI: 10.1016/j.lfs.2018.12.018] [Citation(s) in RCA: 185] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 11/08/2018] [Accepted: 12/10/2018] [Indexed: 12/20/2022]
Abstract
Cyclophosphamide (CP) is an important anticancer drug which belongs to the class of alkylating agent. Cyclophosphamide is mostly used in bone marrow transplantation, rheumatoid arthritis, lupus erythematosus, multiple sclerosis, neuroblastoma and other types of cancer. Dose-related cardiotoxicity is a limiting factor for its use. CP-induced cardiotoxicity ranges from 7 to 28% and mortality ranges from 11 to 43% at the therapeutic dose of 170-180 mg/kg, i.v. CP undergoes hepatic metabolism that results in the production of aldophosphamide. Aldophosphamide decomposes into phosphoramide mustard & acrolein. Phosphoramide is an active neoplastic agent, and acrolein is a toxic metabolite which acts on the myocardium and endothelial cells. This is the first review article that talks about cyclophosphamide-induced cardiotoxicity and the different signaling pathways involved in its pathogenicity. Based on the available literature, CP is accountable for cardiomyocytes energy pool alteration by affecting the heart fatty acid binding proteins (H-FABP). CP has been found associated with cardiomyocytes apoptosis, inflammation, endothelial dysfunction, calcium dysregulation, endoplasmic reticulum damage, and mitochondrial damage. Molecular mechanism of cardiotoxicity has been discussed in detail through crosstalk of Nrf2/ARE, Akt/GSK-3β/NFAT/calcineurin, p53/p38MAPK, NF-kB/TLR-4, and Phospholamban/SERCA-2a signaling pathway. Based on the available literature we support the fact that metabolites of CP are responsible for cardiotoxicity due to depletion of antioxidants/ATP level, altered contractility, damaged endothelium and enhanced pro-inflammatory/pro-apoptotic activities resulting into cardiomyopathy, myocardial infarction, and heart failure. Dose adjustment, elimination/excretion of acrolein and maintenance of endogenous antioxidant pool could be the therapeutic approach to mitigate the toxicities.
Collapse
Affiliation(s)
- Ashif Iqubal
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Mohammad Kashif Iqubal
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Sumit Sharma
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Mohd Asif Ansari
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Abul Kalam Najmi
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Syed Mansoor Ali
- Department of Biosciences, Jamia Millia Islamia,110025 New Delhi, India
| | - Javed Ali
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Syed Ehtaishamul Haque
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India.
| |
Collapse
|
50
|
Kirchhefer U, Hammer E, Heinick A, Herpertz T, Isensee G, Müller FU, Neumann J, Schulte K, Seidl MD, Boknik P, Schulte JS. Chronic β-adrenergic stimulation reverses depressed Ca handling in mice overexpressing inhibitor-2 of protein phosphatase 1. J Mol Cell Cardiol 2018; 125:195-204. [PMID: 30389400 DOI: 10.1016/j.yjmcc.2018.10.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 10/26/2018] [Indexed: 11/16/2022]
Abstract
RATIONALE A higher expression/activity of type 1 serine/threonine protein phosphatase 1 (PP1) may contribute to dephosphorylation of cardiac regulatory proteins triggering the development of heart failure. OBJECTIVE Here, we tested the putatively protective effects of PP1 inhibitor-2 (I2) overexpression using a heart failure model induced by chronic β-adrenergic stimulation. METHODS AND RESULTS Transgenic (TG) and wild-type (WT) mice were subjected to isoprenaline (ISO) or isotonic NaCl solution supplied via osmotic minipumps for 7 days. I2 overexpression was associated with a depressed PP1 activity. Basal contractility was unchanged in catheterized mice and isolated cardiomyocytes between TGNaCl and WTNaCl. TGISO mice exhibited more fibrosis and a higher expression of hypertrophy marker proteins as compared to WTISO. After acute administration of ISO, the contractile response was accompanied by a higher sensitivity in TGISO as compared to WTISO. In contrast to basal contractility, the peak amplitude of [Ca]i and SR Ca load were reduced in TGNaCl as compared to WTNaCl. These effects were normalized to WT levels after chronic ISO stimulation. Cardiomyocyte relaxation and [Ca]i decay kinetics were hastened in TGISO as compared to WTISO, which can be explained by a higher phospholamban phosphorylation at Ser16. Chronic catecholamine stimulation was followed by an enhanced expression of GSK3β, whereas the phosphorylation at Ser9 was lower in TG as compared to the corresponding WT group. This resulted in a higher I2 phosphorylation that may reactivate PP1. CONCLUSION Our findings suggest that the basal desensitization of β-adrenergic signaling and the depressed Ca handling in TG by inhibition of PP1 is restored by a GSK3β-dependent phosphorylation of I2.
Collapse
Affiliation(s)
- Uwe Kirchhefer
- Institut für Pharmakologie und Toxikologie, Westfälische Wilhelms-Universität, Münster, Germany.
| | - Elke Hammer
- Interfakultäres Institut für Genetik und Funktionelle Genomforschung, Universitätsmedizin Greifswald, Germany
| | - Alexander Heinick
- Institut für Pharmakologie und Toxikologie, Westfälische Wilhelms-Universität, Münster, Germany
| | - Thomas Herpertz
- Institut für Pharmakologie und Toxikologie, Westfälische Wilhelms-Universität, Münster, Germany
| | - Gunnar Isensee
- Institut für Pharmakologie und Toxikologie, Westfälische Wilhelms-Universität, Münster, Germany
| | - Frank U Müller
- Institut für Pharmakologie und Toxikologie, Westfälische Wilhelms-Universität, Münster, Germany
| | - Joachim Neumann
- Institute of Pharmacology and Toxicology, Faculty of Medicine, Martin-Luther-University, Halle, Germany
| | - Kirsten Schulte
- Institut für Pharmakologie und Toxikologie, Westfälische Wilhelms-Universität, Münster, Germany
| | - Matthias D Seidl
- Institut für Pharmakologie und Toxikologie, Westfälische Wilhelms-Universität, Münster, Germany
| | - Peter Boknik
- Institut für Pharmakologie und Toxikologie, Westfälische Wilhelms-Universität, Münster, Germany
| | - Jan S Schulte
- Institut für Pharmakologie und Toxikologie, Westfälische Wilhelms-Universität, Münster, Germany
| |
Collapse
|