1
|
Cools JMT, Goovaerts BK, Feyen E, Van den Bogaert S, Fu Y, Civati C, Van Fraeyenhove J, Tubeeckx MRL, Ott J, Nguyen L, Wülfers EM, Van Berlo B, De Vries AAF, Vandersickel N, Pijnappels DA, Audenaert D, Roderick HL, De Winter H, De Keulenaer GW, Segers VFM. Small-molecule-induced ERBB4 activation to treat heart failure. Nat Commun 2025; 16:576. [PMID: 39794341 PMCID: PMC11724075 DOI: 10.1038/s41467-024-54908-5] [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: 03/29/2024] [Accepted: 11/25/2024] [Indexed: 01/13/2025] Open
Abstract
Heart failure is a common and deadly disease requiring new treatments. The neuregulin-1/ERBB4 pathway offers cardioprotective benefits, but using recombinant neuregulin-1 as therapy has limitations due to the need for intravenous delivery and lack of receptor specificity. We hypothesize that small-molecule activation of ERBB4 could protect against heart damage and fibrosis. To test this, we conduct a screening of 10,240 compounds and identify eight structurally similar ones (EF-1 to EF-8) that induce ERBB4 dimerization, with EF-1 being the most effective. EF-1 reduces cell death and hypertrophy in cardiomyocytes and decreases collagen production in cardiac fibroblasts in an ERBB4-dependent manner. In wild-type mice, EF-1 inhibits angiotensin-II-induced fibrosis in males and females and reduces heart damage caused by doxorubicin and myocardial infarction in females, but not in Erbb4-null mice. This study shows that small-molecule ERBB4 activation is feasible and may lead to a novel class of drugs for treating heart failure.
Collapse
Grants
- This work was supported by a Geconcerteerde onderzoeksactie grant (GOA, PID36444) of the University of Antwerp; by a Senior Clinical Investigator fellowship (to V.F.S.), a PhD fellowship (to J.MT.C. and C.C.), and research grants of the Fund for Scientific Research Flanders (Application numbers 1842219N, G021019N, G021420N, 1S49323N, and 11PBU24N); VLIR/iBOF Grant 20-VLIR-iBOF-027 (to N.V., V.F.S., H.L.R., and G.W.D.K.).
Collapse
Affiliation(s)
- Julie M T Cools
- Laboratory of PhysioPharmacology, University of Antwerp, Antwerp, Belgium
| | - Bo K Goovaerts
- Laboratory of PhysioPharmacology, University of Antwerp, Antwerp, Belgium
| | - Eline Feyen
- Laboratory of PhysioPharmacology, University of Antwerp, Antwerp, Belgium
| | | | - Yile Fu
- Laboratory of Experimental Cardiology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Céline Civati
- Laboratory of PhysioPharmacology, University of Antwerp, Antwerp, Belgium
| | | | | | - Jasper Ott
- Laboratory of Cell Biology and Histology, University of Antwerp, Antwerp, Belgium
| | - Long Nguyen
- Screening Core, VIB, Ghent, Belgium
- Centre for Bioassay Development and Screening (C-BIOS), Ghent University, Ghent, Belgium
| | - Eike M Wülfers
- Department of Physics and Astronomy, Ghent University, Ghent, Belgium
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg - Bad Krozingen, Freiburg im Breisbau, Germany and Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Benji Van Berlo
- Laboratory of PhysioPharmacology, University of Antwerp, Antwerp, Belgium
| | - Antoine A F De Vries
- Laboratory of Experimental Cardiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Nele Vandersickel
- Department of Physics and Astronomy, Ghent University, Ghent, Belgium
| | - Daniël A Pijnappels
- Laboratory of Experimental Cardiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Dominique Audenaert
- Screening Core, VIB, Ghent, Belgium
- Centre for Bioassay Development and Screening (C-BIOS), Ghent University, Ghent, Belgium
| | - H Llewelyn Roderick
- Laboratory of Experimental Cardiology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Hans De Winter
- Laboratory of Medicinal Chemistry, University of Antwerp, Antwerp, Belgium
| | - Gilles W De Keulenaer
- Laboratory of PhysioPharmacology, University of Antwerp, Antwerp, Belgium
- Department of Cardiology, ZNA Hospital, Antwerp, Belgium
| | - Vincent F M Segers
- Laboratory of PhysioPharmacology, University of Antwerp, Antwerp, Belgium.
- Department of Cardiology, University Hospital Antwerp, Antwerp, Belgium.
| |
Collapse
|
2
|
He C, Jia J, Lei Y, Hu Q, Xin Y, Chu Y, Liu C, Niu Q. The mechanism of miR-665 targeting GNB3 in aluminum-induced neuronal apoptosis. J Trace Elem Med Biol 2024; 85:127488. [PMID: 38905877 DOI: 10.1016/j.jtemb.2024.127488] [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: 03/04/2024] [Revised: 05/09/2024] [Accepted: 06/18/2024] [Indexed: 06/23/2024]
Abstract
BACKGROUND Aluminum exerts neurotoxic effects through various mechanisms, mainly manifested as impaired learning and memory function. METHODS Forty SD rats were divided into 0, 10, 20, and 40 mM maltol aluminum [Al(mal)3] groups. Cell experiments are divided into 0, 100, 200, and 400 μM Al(mal)3 dose group and control, Al(mal)3, Al(mal)3+inhibitor NC, Al(mal)3+miR-665 inhibitor intervention group. Water maze was used to detect the learning and memory function of rats, HE staining was used to observe the morphology and number of neurons in the CA1 area of the rat hippocampus, Flow cytometry was used to detect the apoptosis of PC12 cells, PCR and Western blotting were used to detect the expression of Caspase3, miR-665 and GNB3/PI3K/AKT proteins. The target binding relationship between miR-665 and GNB3 was verified by double luciferase reporter gene experiment. RESULTS In vivo experimental results showed that with the increase of Al(mal)3 concentration, the escape latency of rats was prolonged, the target quadrant dwell time was shortened, and the number of crossing platform was reduced. Moreover, the arrangement of neurons was loose and the number decreased; the expression of Caspase3 and miR-665 increased, while the expression of GNB3/PI3K/AKT proteins decreased. In vitro experiments, with the increase of Al(mal)3 concentration, apoptosis rate of PC12 cells increased, the expression of Caspase3, miR-665 and GNB3/PI3K/AKT proteins were consistent with rat results. After inhibiting miR-665 in the intervention group experiment, apoptosis rate of PC12 cells in the aluminum exposure group decreased, the expression of Caspase3 and miR-665 decreased, and the expression of GNB3/PI3K/AKT proteins increased. CONCLUSION MiR-665 plays an important role in aluminum induced neuronal apoptosis by targeting GNB3 and regulating the PI3K/AKT pathway.
Collapse
Affiliation(s)
- Chanting He
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi 030001, China; Key Lab of Environmental Hazard and Health of Shanxi Province, Shanxi Medical University, Taiyuan, Shanxi 030001, China; Key Laboratory of Coal Environmental Pathogenicity and Prevention (Shanxi Medical University), Ministry of Education, China; Department of Anatomy, Shanxi Medical University, Taiyuan, Shanxi 030001, China.
| | - Jingjing Jia
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Yang Lei
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Qian Hu
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Yulu Xin
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Yafen Chu
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Congying Liu
- Department of Anatomy, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Qiao Niu
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi 030001, China; Key Lab of Environmental Hazard and Health of Shanxi Province, Shanxi Medical University, Taiyuan, Shanxi 030001, China; Key Laboratory of Coal Environmental Pathogenicity and Prevention (Shanxi Medical University), Ministry of Education, China.
| |
Collapse
|
3
|
Martin-Puig S, Menendez-Montes I. Cardiac Metabolism. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1441:365-396. [PMID: 38884721 DOI: 10.1007/978-3-031-44087-8_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
The heart is composed of a heterogeneous mixture of cellular components perfectly intermingled and able to integrate common environmental signals to ensure proper cardiac function and performance. Metabolism defines a cell context-dependent signature that plays a critical role in survival, proliferation, or differentiation, being a recognized master piece of organ biology, modulating homeostasis, disease progression, and adaptation to tissue damage. The heart is a highly demanding organ, and adult cardiomyocytes require large amount of energy to fulfill adequate contractility. However, functioning under oxidative mitochondrial metabolism is accompanied with a concomitant elevation of harmful reactive oxygen species that indeed contributes to the progression of several cardiovascular pathologies and hampers the regenerative capacity of the mammalian heart. Cardiac metabolism is dynamic along embryonic development and substantially changes as cardiomyocytes mature and differentiate within the first days after birth. During early stages of cardiogenesis, anaerobic glycolysis is the main energetic program, while a progressive switch toward oxidative phosphorylation is a hallmark of myocardium differentiation. In response to cardiac injury, different signaling pathways participate in a metabolic rewiring to reactivate embryonic bioenergetic programs or the utilization of alternative substrates, reflecting the flexibility of heart metabolism and its central role in organ adaptation to external factors. Despite the well-established metabolic pattern of fetal, neonatal, and adult cardiomyocytes, our knowledge about the bioenergetics of other cardiac populations like endothelial cells, cardiac fibroblasts, or immune cells is limited. Considering the close intercellular communication and the influence of nonautonomous cues during heart development and after cardiac damage, it will be fundamental to better understand the metabolic programs in different cardiac cells in order to develop novel interventional opportunities based on metabolic rewiring to prevent heart failure and improve the limited regenerative capacity of the mammalian heart.
Collapse
Affiliation(s)
- Silvia Martin-Puig
- Department of Metabolic and Immune Diseases, Institute for Biomedical Research "Sols-Morreale", National Spanish Research Council, CSIC, Madrid, Spain.
- Cardiac Regeneration Program, National Center for Cardiovascular Research, CNIC, Madrid, Spain.
| | - Ivan Menendez-Montes
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
4
|
Packer M. Qiliqiangxin: A multifaceted holistic treatment for heart failure or a pharmacological probe for the identification of cardioprotective mechanisms? Eur J Heart Fail 2023; 25:2130-2143. [PMID: 37877337 DOI: 10.1002/ejhf.3068] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 10/10/2023] [Accepted: 10/15/2023] [Indexed: 10/26/2023] Open
Abstract
The active ingredients in many traditional Chinese medicines are isoprene oligomers with a diterpenoid or triterpenoid structure, which exert cardiovascular effects by signalling through nutrient surplus and nutrient deprivation pathways. Qiliqiangxin (QLQX) is a commercial formulation of 11 different plant ingredients, whose active compounds include astragaloside IV, tanshione IIA, ginsenosides (Rb1, Rg1 and Re) and periplocymarin. In the QUEST trial, QLQX reduced the combined risk of cardiovascular death or heart failure hospitalization (hazard ratio 0.78, 95% confidence interval 0.68-0.90), based on 859 events in 3119 patients over a median of 18.2 months; the benefits were seen in patients taking foundational drugs except for sodium-glucose cotransporter 2 (SGLT2) inhibitors. Numerous experimental studies of QLQX in diverse cardiac injuries have yielded highly consistent findings. In marked abrupt cardiac injury, QLQX mitigated cardiac injury by upregulating nutrient surplus signalling through the PI3K/Akt/mTOR/HIF-1α/NRF2 pathway; the benefits of QLQX were abrogated by suppression of PI3K, Akt, mTOR, HIF-1α or NRF2. In contrast, in prolonged measured cardiac stress (as in chronic heart failure), QLQX ameliorated oxidative stress, maladaptive hypertrophy, cardiomyocyte apoptosis, and proinflammatory and profibrotic pathways, while enhancing mitochondrial health and promoting glucose and fatty acid oxidation and ATP production. These effects are achieved by an action of QLQX to upregulate nutrient deprivation signalling through SIRT1/AMPK/PGC-1α and enhanced autophagic flux. In particular, QLQX appears to enhance the interaction of PGC-1α with PPARα, possibly by direct binding to RXRα; silencing of SIRT1, PGC-1α and RXRα abrogated the favourable effects of QLQX in the heart. Since PGC-1α/RXRα is also a downstream effector of Akt/mTOR signalling, the actions of QLQX on PGC-1α/RXRα may explain its favourable effects in both acute and chronic stress. Intriguingly, the individual ingredients in QLQX - astragaloside IV, ginsenosides, and tanshione IIA - share QLQX's effects on PGC-1α/RXRα/PPARα signalling. QXQL also contains periplocymarin, a cardiac glycoside that inhibits Na+ -K+ -ATPase. Taken collectively, these observations support a conceptual framework for understanding the mechanism of action for QLQX in heart failure. The high likelihood of overlap in the mechanism of action of QLQX and SGLT2 inhibitors requires additional experimental studies and clinical trials.
Collapse
Affiliation(s)
- Milton Packer
- Baylor Heart and Vascular Institute, Dallas, TX, USA
- Imperial College, London, UK
| |
Collapse
|
5
|
Vaparanta K, Jokilammi A, Paatero I, Merilahti JA, Heliste J, Hemanthakumar KA, Kivelä R, Alitalo K, Taimen P, Elenius K. STAT5b is a key effector of NRG-1/ERBB4-mediated myocardial growth. EMBO Rep 2023; 24:e56689. [PMID: 37009825 PMCID: PMC10157316 DOI: 10.15252/embr.202256689] [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: 12/18/2022] [Revised: 03/07/2023] [Accepted: 03/10/2023] [Indexed: 04/04/2023] Open
Abstract
The growth factor Neuregulin-1 (NRG-1) regulates myocardial growth and is currently under clinical investigation as a treatment for heart failure. Here, we demonstrate in several in vitro and in vivo models that STAT5b mediates NRG-1/EBBB4-stimulated cardiomyocyte growth. Genetic and chemical disruption of the NRG-1/ERBB4 pathway reduces STAT5b activation and transcription of STAT5b target genes Igf1, Myc, and Cdkn1a in murine cardiomyocytes. Loss of Stat5b also ablates NRG-1-induced cardiomyocyte hypertrophy. Dynamin-2 is shown to control the cell surface localization of ERBB4 and chemical inhibition of Dynamin-2 downregulates STAT5b activation and cardiomyocyte hypertrophy. In zebrafish embryos, Stat5 is activated during NRG-1-induced hyperplastic myocardial growth, and chemical inhibition of the Nrg-1/Erbb4 pathway or Dynamin-2 leads to loss of myocardial growth and Stat5 activation. Moreover, CRISPR/Cas9-mediated knockdown of stat5b results in reduced myocardial growth and cardiac function. Finally, the NRG-1/ERBB4/STAT5b signaling pathway is differentially regulated at mRNA and protein levels in the myocardium of patients with pathological cardiac hypertrophy as compared to control human subjects, consistent with a role of the NRG-1/ERBB4/STAT5b pathway in myocardial growth.
Collapse
Affiliation(s)
- Katri Vaparanta
- Turku Bioscience CentreUniversity of Turku and Åbo Akademi UniversityTurkuFinland
- Medicity Research LaboratoriesUniversity of TurkuTurkuFinland
- Institute of BiomedicineUniversity of TurkuTurkuFinland
| | - Anne Jokilammi
- Turku Bioscience CentreUniversity of Turku and Åbo Akademi UniversityTurkuFinland
- Medicity Research LaboratoriesUniversity of TurkuTurkuFinland
- Institute of BiomedicineUniversity of TurkuTurkuFinland
| | - Ilkka Paatero
- Turku Bioscience CentreUniversity of Turku and Åbo Akademi UniversityTurkuFinland
| | - Johannes A Merilahti
- Turku Bioscience CentreUniversity of Turku and Åbo Akademi UniversityTurkuFinland
| | - Juho Heliste
- Turku Bioscience CentreUniversity of Turku and Åbo Akademi UniversityTurkuFinland
- Medicity Research LaboratoriesUniversity of TurkuTurkuFinland
- Institute of BiomedicineUniversity of TurkuTurkuFinland
| | - Karthik Amudhala Hemanthakumar
- Wihuri Research InstituteHelsinkiFinland
- Translational Cancer Biology Program, Research Programs Unit, Faculty of MedicineUniversity of HelsinkiHelsinkiFinland
| | - Riikka Kivelä
- Wihuri Research InstituteHelsinkiFinland
- Translational Cancer Biology Program, Research Programs Unit, Faculty of MedicineUniversity of HelsinkiHelsinkiFinland
- Faculty of Sport and Health SciencesUniversity of JyväskyläJyväskyläFinland
| | - Kari Alitalo
- Wihuri Research InstituteHelsinkiFinland
- Translational Cancer Biology Program, Research Programs Unit, Faculty of MedicineUniversity of HelsinkiHelsinkiFinland
| | - Pekka Taimen
- Institute of Biomedicine and FICAN West Cancer CentreUniversity of TurkuTurkuFinland
- Department of PathologyTurku University HospitalTurkuFinland
| | - Klaus Elenius
- Turku Bioscience CentreUniversity of Turku and Åbo Akademi UniversityTurkuFinland
- Medicity Research LaboratoriesUniversity of TurkuTurkuFinland
- Institute of BiomedicineUniversity of TurkuTurkuFinland
- Department of OncologyTurku University HospitalTurkuFinland
| |
Collapse
|
6
|
Yu M, Wu S, Gong C, Chen L. Neuregulin-1β increases glucose uptake and promotes GLUT4 translocation in palmitate-treated C2C12 myotubes by activating PI3K/AKT signaling pathway. Front Pharmacol 2023; 13:1066279. [PMID: 36703726 PMCID: PMC9871240 DOI: 10.3389/fphar.2022.1066279] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 12/26/2022] [Indexed: 01/12/2023] Open
Abstract
Insulin resistance (IR) is a feature of type 2 diabetes (T2DM) accompanied by reduced glucose uptake and glucose transporter 4 (GLUT4) translocation by skeletal muscle. Neuregulin-1β (NRG-1β) is essential for myogenesis and the regulation of skeletal muscle metabolism. Neuregulin-1β increases insulin sensitivity, promotes glucose uptake and glucose translocation in normal skeletal muscle. Here, we explored whether Neuregulin-1β increased glucose uptake and GLUT4 translocation in palmitate (PA)-treated C2C12 myotubes. After C2C12 myoblasts differentiated into myotubes, we used palmitate to induce cellular insulin resistance. Cells were incubated with or without Neuregulin-1β and glucose uptake was determined using the 2-NBDG assay. The expression level of glucose transporter 4 (GLUT4) was measured via immunofluorescence and Western blotting. MK2206, an inhibitor of AKT, was employed to reveal the important role played by AKT signaling in PA-treated C2C12 myotubes. We then established an animal model with T2DM and evaluated the effects of Neuregulin-1β on body weight and the blood glucose level. The GLUT4 level in the gastrocnemius of T2DM mice was also measured. NRG-1β not only increased glucose uptake by PA-treated myotubes but also promoted GLUT4 translocation to the plasma membrane. The effect of NRG-1β on PA-treated C2C12 myotubes was associated with AKT activation. In T2DM mice, Neuregulin-1β not only improved diabetes-induced weight loss and diabetes-induced hyperglycemia, but also promoted GLUT4 translocation in the gastrocnemius. In summary, Neuregulin-1β increased glucose uptake and promoted translocation of GLUT4 to the plasma membrane in PA-treated C2C12 myotubes by activating the PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
| | | | - Chao Gong
- *Correspondence: Chao Gong, ; Lianhua Chen,
| | | |
Collapse
|
7
|
Wang Y, Wei J, Zhang P, Zhang X, Wang Y, Chen W, Zhao Y, Cui X. Neuregulin-1, a potential therapeutic target for cardiac repair. Front Pharmacol 2022; 13:945206. [PMID: 36120374 PMCID: PMC9471952 DOI: 10.3389/fphar.2022.945206] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 08/01/2022] [Indexed: 11/13/2022] Open
Abstract
NRG1 (Neuregulin-1) is an effective cardiomyocyte proliferator, secreted and released by endothelial vascular cells, and affects the cardiovascular system. It plays a major role in heart growth, proliferation, differentiation, apoptosis, and other cardiovascular processes. Numerous experiments have shown that NRG1 can repair the heart in the pathophysiology of atherosclerosis, myocardial infarction, ischemia reperfusion, heart failure, cardiomyopathy and other cardiovascular diseases. NRG1 can connect related signaling pathways through the NRG1/ErbB pathway, which form signal cascades to improve the myocardial microenvironment, such as regulating cardiac inflammation, oxidative stress, necrotic apoptosis. Here, we summarize recent research advances on the molecular mechanisms of NRG1, elucidate the contribution of NRG1 to cardiovascular disease, discuss therapeutic approaches targeting NRG1 associated with cardiovascular disease, and highlight areas for future research.
Collapse
Affiliation(s)
- Yan Wang
- First Clinical Medical School, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Jianliang Wei
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Peng Zhang
- First Clinical Medical School, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Xin Zhang
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Yifei Wang
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Wenjing Chen
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Yanan Zhao
- First Clinical Medical School, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
- *Correspondence: Yanan Zhao, ; Xiangning Cui,
| | - Xiangning Cui
- Department of Cardiovascular, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Yanan Zhao, ; Xiangning Cui,
| |
Collapse
|
8
|
Li YY, Zhong YJ, Cheng Q, Wang YZ, Fan YY, Yang CF, Ma Z, Li YW, Li L. miR-378b Regulates Insulin Sensitivity by Targeting Insulin Receptor and p110α in Alcohol-Induced Hepatic Steatosis. Front Pharmacol 2020; 11:717. [PMID: 32508647 PMCID: PMC7251170 DOI: 10.3389/fphar.2020.00717] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 04/30/2020] [Indexed: 12/12/2022] Open
Abstract
Insulin resistance has been implicated in alcoholic liver disease. A previous study has shown that microRNAs (miRNAs) play a major role in the production, secretion, and function of insulin. MiRNAs are capable of repressing multiple target genes that in turn negatively regulate various physiological and pathological activities. However, current information on the biological function of miRNAs in insulin resistance is limited. The goal of the present study was to elucidate the role of miR-378b in alcohol-induced hepatic insulin resistance and its underlying mechanism. This study has observed that miR-378b is up-regulated in National Institute on Alcohol Abuse and Alcoholism (NIAAA) alcoholic mouse models as well as in ethanol-induced L-02 cells in vitro. Furthermore, miR-378b overexpression impaired the insulin signaling pathway, and inhibition of miR-378b improved insulin sensitivity in vivo and in vitro. A mechanistic study revealed that IR and p110α are direct targets of miR-378b. Together, these results suggest that miR-378b controls insulin sensitivity by targeting the insulin receptor (IR) as well as p110α and possibly play an inhibitory role in the development of insulin resistance, thereby providing insights into the development of novel diagnostic and treatment methods.
Collapse
Affiliation(s)
- Yuan-yuan Li
- College of Pharmacy, Guilin Medical University, Guilin, China
| | - Yu-juan Zhong
- College of Pharmacy, Guilin Medical University, Guilin, China
| | - Qi Cheng
- College of Pharmacy, Guilin Medical University, Guilin, China
| | - Ying-zhao Wang
- College of Pharmacy, Guilin Medical University, Guilin, China
| | - Yuan-yuan Fan
- College of Pharmacy, Guilin Medical University, Guilin, China
| | - Cheng-fang Yang
- College of Pharmacy, Guilin Medical University, Guilin, China
| | - Zuheng Ma
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Yong-wen Li
- College of Pharmacy, Guilin Medical University, Guilin, China
- Center for Diabetic Systems Medicine, Guangxi Key Laboratory of Excellence, Guilin, China
| | - Li Li
- College of Pharmacy, Guilin Medical University, Guilin, China
| |
Collapse
|
9
|
Shati AA, Dallak M. Acylated Ghrelin Protects the Hearts of Rats from Doxorubicin-Induced Fas/FasL Apoptosis by Stimulating SERCA2a Mediated by Activation of PKA and Akt. Cardiovasc Toxicol 2020; 19:529-547. [PMID: 31093930 DOI: 10.1007/s12012-019-09527-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
This study investigated if the cardioprotective effect of acylated ghrelin (AG) against doxorubicin (DOX)-induced cardiac toxicity in rats involves inhibition of Fas/FasL-mediated cell death. It also investigated if such an effect is mediated by restoring Ca+2 homeostasis from the aspect of stimulation of SERCA2a receptors. Adult male Wistar rats were divided into 4 groups (20 rats/each) as control, control + AG, DOX, and DOX + AG. AG was co-administered to all rats consecutively for 35 days. In addition, isolated cardiomyocytes were cultured and treated with AG in the presence or absence of DOX with or without pre-incubation with [D-Lys3]-GHRP-6 (a AG receptor antagonist), VIII (]an Akt inhibitor), or KT-5720 (a PKA inhibitor). AG increased LVSP, dp/dtmax, and dp/dtmin in both control and DOX-treated animals and improved cardiac ultrastructural changes in DOX-treated rats. It also inhibited ROS in control rats and lowered LVEDP, intracellular levels of ROS and Ca2+, and activity of calcineurin in LVs of DOX-treated rats. Concomitantly, it inhibited LV NFAT-4 nuclear translocation and downregulated their protein levels of Fas and FasL. Mechanistically, in control or DOX-treated hearts or cells, AG upregulated the levels of SERCA2a and increased the activities of PKA and Akt, leading to increase phosphorylation of phospholamban at Ser16 and Thr17. All these effects were abolished by D-Lys3-GHRP-6, VIII, or KT-5720 and were independent of food intake or GH/IGF-1. In conclusion, AG cardioprotection against DOX involves inhibition of extrinsic cell death and restoring normal Ca+2 homeostasis.
Collapse
Affiliation(s)
- Ali A Shati
- Department of Biology, College of Science, King Khalid University, Abha, Saudi Arabia.
| | - M Dallak
- Department of Physiology, College of Medicine, King Khalid University, Abha, Saudi Arabia
| |
Collapse
|
10
|
Umutlu G, Demirci N, Erdoğan AT, Acar NE, Fidanci ŞB. Neuromuscular, hormonal and cardiovascular adaptations to eight-week HIIT and continuous aerobic training combined with neuromuscular electrical stimulation. J Sports Med Phys Fitness 2020; 60:510-519. [PMID: 32043342 DOI: 10.23736/s0022-4707.19.10277-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND Whether high-or-low intensity exercise coupled with neuromuscular electrostimulation (NMES) affect IGF-1 and IGFBP-1 is unknown. The scope of this study was to test whether 8-week high-intensity interval training (HIIT) and continuous aerobic training (CA) combined with/without NMES performed at 65% and 120% of VO2max on a cycle ergometer induce different metabolic adaptations. METHODS A randomized controlled trial with a parallel groups study design was used. Thirty healthy untrained male participants (age: 21.33±1.24 years, height: 177.80±5.97 cm, weight: 73.74±7.90 kg, lean body mass: 64.29±5.11 kg, percent body fat: 12.43±5.34%) voluntarily participated in this study. Six participants were allocated to Control, six to HIIT, six to HIIT+NMES, six to CA, and six to CA+NMES. RESULTS Pre- to post-test IVO2max, blood lactate concentrations, O2 kinetics, peak torques at 60o/s and 180o/s were found statistically significant (P<0.05, P<0.001). IGF-1 pre 15 min in CA and IGF-1 post 30 min in HIIT group was found significantly higher compared to control group (16.93±8.40 vs. 6.05±4.25, P=0.024; 10.80±3.94 vs. 6.15±2.56, P=0.037), respectively. Additionally, IGFBP-1 were found significantly higher in CA+NMES group than HIIT group (0.95±0.67 vs. 1.23±0.56). Eight week post IGF-1/IGFBP-1 ratios were found higher in pre 15 min, post 30 min and post 24 h compared to baseline pre 15 min, post 30 min and post 24 h measurements in all groups (8.92±4.72 vs. 3.93±3.14; 9.41±3.72 vs. 3.99±1.76; 8.63±3.01 vs. 5.89±3.01, respectively). Also, IGFBP-1 post 30 min was significantly lower in HIIT+NMES while CA group showed significantly lower baseline and 24 h post IGFBP-1 compared to pre-test measurements (Z=-3.20, P=0.001; Z=-3.72, P=0.000; Z=-2.93, P=0.000). CONCLUSIONS HIIT and CA training induce different stimuli on IGF-1 and IGFBP-1 and NMES application combined with high-and-low intensity exercise is highly effective in improving athletic performance.
Collapse
Affiliation(s)
- Gökhan Umutlu
- School of Physical Education and Sports, Final International University, Kyrenia, Cyprus -
| | - Nevzat Demirci
- School of Physical Education and Sports, Mersin University, Mersin, Turkey
| | - Ayhan T Erdoğan
- School of Physical Education and Sports, Final International University, Kyrenia, Cyprus
| | - Nasuh E Acar
- School of Physical Education and Sports, Mersin University, Mersin, Turkey
| | - Şenay B Fidanci
- School of Medicine, Department of Medical Chemistry, Mersin University, Mersin, Turkey
| |
Collapse
|
11
|
Neuregulin-1 triggers GLUT4 translocation and enhances glucose uptake independently of insulin receptor substrate and ErbB3 in neonatal rat cardiomyocytes. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2019; 1867:118562. [PMID: 31669265 DOI: 10.1016/j.bbamcr.2019.118562] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 09/15/2019] [Accepted: 10/16/2019] [Indexed: 12/28/2022]
Abstract
During stress conditions such as pressure overload and acute ischemia, the myocardial endothelium releases neuregulin-1β (NRG-1), which acts as a cardioprotective factor and supports recovery of the heart. Recently, we demonstrated that recombinant human (rh)NRG-1 enhances glucose uptake in neonatal rat ventricular myocytes via the ErbB2/ErbB4 heterodimer and PI3Kα. The present study aimed to further elucidate the mechanism whereby rhNRG-1 activates glucose uptake in comparison to the well-established insulin and to extend the findings to adult models. Combinations of rhNRG-1 with increasing doses of insulin did not yield any additive effect on glucose uptake measured as 3H-deoxy-d-glucose incorporation, indicating that the mechanisms of the two stimuli are similar. In c-Myc-GLUT4-mCherry-transfected neonatal rat cardiomyocytes, rhNRG-1 increased sarcolemmal GLUT4 by 16-fold, similar to insulin. In contrast to insulin, rhNRG-1 did not phosphorylate IRS-1 at Tyr612, indicating that IRS-1 is not implicated in the signal transmission. Treatment of neonatal rats with rhNRG-1 induced a signaling response comparable with that observed in vitro, including increased ErbB4-pTyr1284, Akt-pThr308 and Erk1/2-pThr202/Tyr204. In contrast, in adult cardiomyocytes rhNRG-1 only increased the phosphorylation of Erk1/2 without having any significant effect on Akt and AS160 phosphorylation and glucose uptake, suggesting that rhNRG-1 function in neonatal cardiomyocytes differs from that in adult cardiomyocytes. In conclusion, our results show that similar to insulin, rhNRG-1 can induce glucose uptake by activating the PI3Kα-Akt-AS160 pathway and GLUT4 translocation. Unlike insulin, the rhNRG-1-induced effect is not mediated by IRS proteins and is observed in neonatal, but not in adult rat cardiomyocytes.
Collapse
|
12
|
Fatty acid-based monolayer culture to promote in vitro neonatal rat cardiomyocyte maturation. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2019; 1867:118561. [PMID: 31655100 DOI: 10.1016/j.bbamcr.2019.118561] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 09/19/2019] [Accepted: 09/28/2019] [Indexed: 12/20/2022]
Abstract
The development of functional and reliable in vitro cardiac models composed of fully mature cardiomyocytes is essential for improving drug screening test quality, therefore, the success of clinical trial outcomes. In their lifespan, cardiomyocytes undergo a dynamic maturation process from the fetal to adult stage, radically changing their metabolism, morphology, contractility and electrical properties. Before employing cells of human origin, in vitro models often use neonatal rat cardiomyocytes (NRCM) to obtain key proof-of-principles. Nevertheless, NRCM monolayers are prone to de-differentiate when maintained in culture. Supplementation of free fatty acids (FFA), the main energy source for mature cardiomyocytes, and co-culture with fibroblasts are each by itself known to promote the shift from fetal to adult cardiomyocytes. Using a co-culture system, our study investigates the effects of FFA on the cardiomyocyte phenotype in comparison to glucose as typical fetal energy source, and to 10% serum used as standard control condition. NRCM decreased their differentiation status and fibroblasts increased in number after 7days of culture in the control condition. On the contrary, both glucose- and FFA-supplementation better preserved protein expression of myosin-light-chain-2v, a marker of mature cardiomyocytes, and the fibroblast number at levels similar to those found in freshly isolated NRCM. Nevertheless, compared to glucose, FFA resulted in a significant increase in sarcomere striation and organization. Our findings constitute an important step forward towards the definition of the optimal culture conditions, highlighting the possible benefits of a further supplementation of specific FFA to promote CM maturation in a co-culture system with FB.
Collapse
|
13
|
De Keulenaer GW, Feyen E, Dugaucquier L, Shakeri H, Shchendrygina A, Belenkov YN, Brink M, Vermeulen Z, Segers VFM. Mechanisms of the Multitasking Endothelial Protein NRG-1 as a Compensatory Factor During Chronic Heart Failure. Circ Heart Fail 2019; 12:e006288. [DOI: 10.1161/circheartfailure.119.006288] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Heart failure is a complex syndrome whose phenotypic presentation and disease progression depends on a complex network of adaptive and maladaptive responses. One of these responses is the endothelial release of NRG (neuregulin)-1—a paracrine growth factor activating ErbB2 (erythroblastic leukemia viral oncogene homolog B2), ErbB3, and ErbB4 receptor tyrosine kinases on various targets cells. NRG-1 features a multitasking profile tuning regenerative, inflammatory, fibrotic, and metabolic processes. Here, we review the activities of NRG-1 on different cell types and organs and their implication for heart failure progression and its comorbidities. Although, in general, effects of NRG-1 in heart failure are compensatory and beneficial, translation into therapies remains unaccomplished both because of the complexity of the underlying pathways and because of the challenges in the development of therapeutics (proteins, peptides, small molecules, and RNA-based therapies) for tyrosine kinase receptors. Here, we give an overview of the complexity to be faced and how it may be tackled.
Collapse
Affiliation(s)
- Gilles W. De Keulenaer
- Laboratory of Physiopharmacology, University of Antwerp, Belgium (G.W.D.K., E.F., L.D., H.S., Z.V., V.F.M.S.)
- Department of Cardiology, ZNA Hospital, Antwerp, Belgium (G.W.D.K.)
| | - Eline Feyen
- Laboratory of Physiopharmacology, University of Antwerp, Belgium (G.W.D.K., E.F., L.D., H.S., Z.V., V.F.M.S.)
| | - Lindsey Dugaucquier
- Laboratory of Physiopharmacology, University of Antwerp, Belgium (G.W.D.K., E.F., L.D., H.S., Z.V., V.F.M.S.)
| | - Hadis Shakeri
- Laboratory of Physiopharmacology, University of Antwerp, Belgium (G.W.D.K., E.F., L.D., H.S., Z.V., V.F.M.S.)
| | - Anastasia Shchendrygina
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russian Federation (A.S., Y.N.B.)
| | - Yury N. Belenkov
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russian Federation (A.S., Y.N.B.)
| | - Marijke Brink
- Department of Biomedicine, University Hospital Basel, University of Basel, Switzerland (M.B.)
| | - Zarha Vermeulen
- Laboratory of Physiopharmacology, University of Antwerp, Belgium (G.W.D.K., E.F., L.D., H.S., Z.V., V.F.M.S.)
| | - Vincent F. M. Segers
- Laboratory of Physiopharmacology, University of Antwerp, Belgium (G.W.D.K., E.F., L.D., H.S., Z.V., V.F.M.S.)
- Department of Cardiology, University Hospital Antwerp, Edegem, Belgium (V.F.M.S.)
| |
Collapse
|
14
|
Abstract
Metabolic pathways integrate to support tissue homeostasis and to prompt changes in cell phenotype. In particular, the heart consumes relatively large amounts of substrate not only to regenerate ATP for contraction but also to sustain biosynthetic reactions for replacement of cellular building blocks. Metabolic pathways also control intracellular redox state, and metabolic intermediates and end products provide signals that prompt changes in enzymatic activity and gene expression. Mounting evidence suggests that the changes in cardiac metabolism that occur during development, exercise, and pregnancy as well as with pathological stress (eg, myocardial infarction, pressure overload) are causative in cardiac remodeling. Metabolism-mediated changes in gene expression, metabolite signaling, and the channeling of glucose-derived carbon toward anabolic pathways seem critical for physiological growth of the heart, and metabolic inefficiency and loss of coordinated anabolic activity are emerging as proximal causes of pathological remodeling. This review integrates knowledge of different forms of cardiac remodeling to develop general models of how relationships between catabolic and anabolic glucose metabolism may fortify cardiac health or promote (mal)adaptive myocardial remodeling. Adoption of conceptual frameworks based in relational biology may enable further understanding of how metabolism regulates cardiac structure and function.
Collapse
Affiliation(s)
- Andrew A Gibb
- From the Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA (A.A.G.)
| | - Bradford G Hill
- the Department of Medicine, Institute of Molecular Cardiology, Diabetes and Obesity Center, University of Louisville School of Medicine, KY (B.G.H.).
| |
Collapse
|
15
|
Chiba M, Ito Y, Nagasawa T. Phenethyl isothiocyanate stimulates glucose uptake through the Akt pathway in C2C12 myotubes. Biosci Biotechnol Biochem 2019; 83:1319-1328. [DOI: 10.1080/09168451.2019.1594675] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
ABSTRACT
Phenethyl isothiocyanate (PEITC) is an aromatic isothiocyanate present in cruciferous vegetables. Several studies have shown that isothiocyanates regulate various intracellular signaling pathways, and thereby show anti-inflammatory and detoxifying activities. However, little is known about the effects of PEITC on glucose metabolism. In this study, we examined whether PEITC promotes glucose utilization in mouse skeletal muscle cells, C2C12 myotubes. PEITC induced glucose uptake, glucose transporter 4 (Glut4) translocation to the plasma membrane, and activation of Akt and ERK in C2C12 cells. Inhibition of Akt suppressed PEITC-induced Glut4 translocation and glucose uptake, whereas ERK inhibition did not. Furthermore, PEITC increased phosphorylation of ErbB2 and ErbB3. Treatment with a pan-ErbB inhibitor reduced Akt activation and the subsequent glucose uptake induced by PEITC. These results indicate that PEITC promotes glucose utilization through the ErbB/Akt pathway in C2C12 myotubes. PEITC may therefore serve as a dietary constituent with beneficial effects on the carbohydrate metabolism.
Abbreviations: PEITC: phenethyl isothiocyanate; Glut4: glucose transporter 4; PI3K: phosphatidylinositide 3-kinase; Nrf2: erythroid−2-related factor; ARE: antioxidant response element; HO−1: heme oxygenase−1; NRG: neuregulin
Collapse
Affiliation(s)
- Maiko Chiba
- Department of Biological Chemistry and Food Science, Faculty of Agriculture, Iwate University, Morioka, Iwate, Japan
| | - Yoshiaki Ito
- Department of Biological Chemistry and Food Science, Faculty of Agriculture, Iwate University, Morioka, Iwate, Japan
| | - Takashi Nagasawa
- Department of Biological Chemistry and Food Science, Faculty of Agriculture, Iwate University, Morioka, Iwate, Japan
| |
Collapse
|
16
|
Capmany A, Gambarte Tudela J, Alonso Bivou M, Damiani MT. Akt/AS160 Signaling Pathway Inhibition Impairs Infection by Decreasing Rab14-Controlled Sphingolipids Delivery to Chlamydial Inclusions. Front Microbiol 2019; 10:666. [PMID: 31001235 PMCID: PMC6456686 DOI: 10.3389/fmicb.2019.00666] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 03/18/2019] [Indexed: 12/26/2022] Open
Abstract
Chlamydia trachomatis, an obligate intracellular bacterium, intercepts different trafficking pathways of the host cell to acquire essential lipids for its survival and replication, particularly from the Golgi apparatus via a Rab14-mediated transport. Molecular mechanisms underlying how these bacteria manipulate intracellular transport are a matter of intense study. Here, we show that C. trachomatis utilizes Akt/AS160 signaling pathway to promote sphingolipids delivery to the chlamydial inclusion through Rab14-controlled vesicular transport. C. trachomatis provokes Akt phosphorylation along its entire developmental life cycle and recruits phosphorylated Akt (pAkt) to the inclusion membrane. As a consequence, Akt Substrate of 160 kDa (AS160), also known as TBC1D4, a GTPase Activating Protein (GAP) for Rab14, is phosphorylated and therefore inactivated. Phosphorylated AS160 (pAS160) loses its ability to promote GTP hydrolysis, favoring Rab14 binding to GTP. Akt inhibition by an allosteric isoform-specific Akt inhibitor (iAkt) prevents AS160 phosphorylation and reduces Rab14 recruitment to chlamydial inclusions. iAkt further impairs sphingolipids acquisition by C. trachomatis-inclusion and provokes lipid retention at the Golgi apparatus. Consequently, treatment with iAkt decreases chlamydial inclusion size, bacterial multiplication, and infectivity in a dose-dependent manner. Similar results were found in AS160-depleted cells. By electron microscopy, we observed that iAkt generates abnormal bacterial forms as those reported after sphingolipids deprivation or Rab14 silencing. Taken together, our findings indicate that targeting the Akt/AS160/Rab14 axis could constitute a novel strategy to limit chlamydial infections, mainly for those caused by antibiotic-resistant bacteria.
Collapse
Affiliation(s)
- Anahí Capmany
- Laboratorio de Bioquímica e Inmunidad, Área de Química Biológica, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas, Mendoza, Argentina
| | - Julián Gambarte Tudela
- Laboratorio de Bioquímica e Inmunidad, Área de Química Biológica, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina.,Instituto de Medicina y Biología Experimental de Cuyo, Consejo Nacional de Investigaciones Científicas y Técnicas, Mendoza, Argentina
| | - Mariano Alonso Bivou
- Laboratorio de Bioquímica e Inmunidad, Área de Química Biológica, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina.,Instituto de Medicina y Biología Experimental de Cuyo, Consejo Nacional de Investigaciones Científicas y Técnicas, Mendoza, Argentina
| | - María T Damiani
- Laboratorio de Bioquímica e Inmunidad, Área de Química Biológica, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina.,Instituto de Medicina y Biología Experimental de Cuyo, Consejo Nacional de Investigaciones Científicas y Técnicas, Mendoza, Argentina
| |
Collapse
|
17
|
Kivelä R, Hemanthakumar KA, Vaparanta K, Robciuc M, Izumiya Y, Kidoya H, Takakura N, Peng X, Sawyer DB, Elenius K, Walsh K, Alitalo K. Endothelial Cells Regulate Physiological Cardiomyocyte Growth via VEGFR2-Mediated Paracrine Signaling. Circulation 2019; 139:2570-2584. [PMID: 30922063 PMCID: PMC6553980 DOI: 10.1161/circulationaha.118.036099] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Supplemental Digital Content is available in the text. Background: Heart failure, which is a major global health problem, is often preceded by pathological cardiac hypertrophy. The expansion of the cardiac vasculature, to maintain adequate supply of oxygen and nutrients, is a key determinant of whether the heart grows in a physiological compensated manner or a pathological decompensated manner. Bidirectional endothelial cell (EC)–cardiomyocyte (CMC) cross talk via cardiokine and angiocrine signaling plays an essential role in the regulation of cardiac growth and homeostasis. Currently, the mechanisms involved in the EC-CMC interaction are not fully understood, and very little is known about the EC-derived signals involved. Understanding how an excess of angiogenesis induces cardiac hypertrophy and how ECs regulate CMC homeostasis could provide novel therapeutic targets for heart failure. Methods: Genetic mouse models were used to delete vascular endothelial growth factor (VEGF) receptors, adeno-associated viral vectors to transduce the myocardium, and pharmacological inhibitors to block VEGF and ErbB signaling in vivo. Cell culture experiments were used for mechanistic studies, and quantitative polymerase chain reaction, microarrays, ELISA, and immunohistochemistry were used to analyze the cardiac phenotypes. Results: Both EC deletion of VEGF receptor (VEGFR)-1 and adeno-associated viral vector–mediated delivery of the VEGFR1-specific ligands VEGF-B or placental growth factor into the myocardium increased the coronary vasculature and induced CMC hypertrophy in adult mice. The resulting cardiac hypertrophy was physiological, as indicated by preserved cardiac function and exercise capacity and lack of pathological gene activation. These changes were mediated by increased VEGF signaling via endothelial VEGFR2, because the effects of VEGF-B and placental growth factor on both angiogenesis and CMC growth were fully inhibited by treatment with antibodies blocking VEGFR2 or by endothelial deletion of VEGFR2. To identify activated pathways downstream of VEGFR2, whole-genome transcriptomics and secretome analyses were performed, and the Notch and ErbB pathways were shown to be involved in transducing signals for EC-CMC cross talk in response to angiogenesis. Pharmacological or genetic blocking of ErbB signaling also inhibited part of the VEGF-B–induced effects in the heart. Conclusions: This study reveals that cross talk between the EC VEGFR2 and CMC ErbB signaling pathways coordinates CMC hypertrophy with angiogenesis, contributing to physiological cardiac growth.
Collapse
Affiliation(s)
- Riikka Kivelä
- Wihuri Research Institute, Helsinki, Finland and Translational Cancer Biology Program, Research Programs Unit, Faculty of Medicine, University of Helsinki, Finland (R.K., K.A.H., M.R., K.A.)
| | - Karthik Amudhala Hemanthakumar
- Wihuri Research Institute, Helsinki, Finland and Translational Cancer Biology Program, Research Programs Unit, Faculty of Medicine, University of Helsinki, Finland (R.K., K.A.H., M.R., K.A.)
| | - Katri Vaparanta
- MediCity Research Laboratories and Institute of Biomedicine, Faculty of Medicine, University of Turku, Finland (K.V., K.E.).,Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Finland (K.V., K.E.)
| | - Marius Robciuc
- Wihuri Research Institute, Helsinki, Finland and Translational Cancer Biology Program, Research Programs Unit, Faculty of Medicine, University of Helsinki, Finland (R.K., K.A.H., M.R., K.A.)
| | - Yasuhiro Izumiya
- Department of Cardiovascular Medicine, Osaka City University Graduate School of Medicine, Japan (Y.I.)
| | - Hiroyasu Kidoya
- Department of Signal Transduction, Research Institute for Microbial Diseases, Osaka University, Japan (H.K., N.T.)
| | - Nobuyuki Takakura
- Department of Signal Transduction, Research Institute for Microbial Diseases, Osaka University, Japan (H.K., N.T.)
| | - Xuyang Peng
- Maine Medical Center, Portland (X.P., D.B.S.)
| | | | - Klaus Elenius
- MediCity Research Laboratories and Institute of Biomedicine, Faculty of Medicine, University of Turku, Finland (K.V., K.E.).,Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Finland (K.V., K.E.).,Department of Oncology and Radiotherapy, University of Turku and Turku University Hospital, Finland (K.E.)
| | - Kenneth Walsh
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville (K.W.)
| | - Kari Alitalo
- Wihuri Research Institute, Helsinki, Finland and Translational Cancer Biology Program, Research Programs Unit, Faculty of Medicine, University of Helsinki, Finland (R.K., K.A.H., M.R., K.A.)
| |
Collapse
|
18
|
Catestatin Induces Glucose Uptake and GLUT4 Trafficking in Adult Rat Cardiomyocytes. BIOMED RESEARCH INTERNATIONAL 2018; 2018:2086109. [PMID: 30370303 PMCID: PMC6189662 DOI: 10.1155/2018/2086109] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 09/10/2018] [Accepted: 09/13/2018] [Indexed: 12/26/2022]
Abstract
Catestatin is a cationic and hydrophobic peptide derived from the enzymatic cleavage of the prohormone Chromogranin A. Initially identified as a potent endogenous nicotinic-cholinergic antagonist, Catestatin has recently been shown to act as a novel regulator of cardiac function and blood pressure and as a cardioprotective agent in both pre- and postconditioning through AKT-dependent mechanisms. The aim of this study is to investigate the potential role of Catestatin also on cardiac metabolism modulation, particularly on cardiomyocytes glucose uptake. Experiments were performed on isolated adult rat cardiomyocytes. Glucose uptake was assessed by fluorescent glucose incubation and confocal microscope analysis. Glut4 plasma membrane translocation was studied by immunofluorescence experiments and evaluation of the ratio peripheral vs internal Glut4 staining. Furthermore, we performed immunoblot experiments to investigate the involvement of the intracellular pathway AKT/AS160 in the Catestatin dependent Glut4 trafficking. Our results show that 10 nM Catestatin induces a significant increase in the fluorescent glucose uptake, comparable to that exerted by 100 nM Insulin. Moreover, Catestatin stimulates Glut4 translocation to plasma membrane and both AKT and AS160 phosphorylation. All these effects were inhibited by Wortmannin. On the whole, we show for the first time that Catestatin is able to modulate cardiac glucose metabolism, by inducing an increase in glucose uptake through Glut4 translocation to the plasma membrane and that this mechanism is mediated by the AKT/AS160 intracellular pathway.
Collapse
|
19
|
Ferreira R, Nogueira-Ferreira R, Trindade F, Vitorino R, Powers SK, Moreira-Gonçalves D. Sugar or fat: The metabolic choice of the trained heart. Metabolism 2018; 87:98-104. [PMID: 30077622 DOI: 10.1016/j.metabol.2018.07.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 06/13/2018] [Accepted: 07/19/2018] [Indexed: 12/13/2022]
Abstract
Mammals respond to muscular exercise by increasing cardiac output to meet the increased demand for oxygen in the working muscles and it is well-established that regular bouts of exercise results in myocardial remodeling. Depending on exercise type, intensity and duration, these cardiac adaptations lead to changes in the energetic substrates required to sustain cardiac contractility. In contrast to the failing heart, fatty acids are the preferred substrate in the trained heart, though glucose metabolism is also enhanced to support oxidative phosphorylation. The participation of AMPK/eNOS and PPARα/PGC-1α pathways in the regulation of cardiac metabolism is well known but other players also contribute including sirtuins and integrins-mediated outside-in activation of FAK and other kinases. These regulatory players act by up-regulating fatty acid uptake, transport to mitochondria and oxidation, and glucose uptake via GLUT4. This exercise-induced increase in mitochondria metabolic flexibility is important to sustain the energetic demand associated with cardiomyocyte hypertrophy and hyperplasia promoted by IGF-1 and neuregulin-1-induced PI3K/Akt signaling. So, the timeless advice of Hippocrates "walking is the best medicine" seems to be justified by the promotion of mitochondrial health and, consequently, the beneficial metabolic remodeling of the heart.
Collapse
Affiliation(s)
- Rita Ferreira
- QOPNA, Department of Chemistry, University of Aveiro, Aveiro, Portugal.
| | - Rita Nogueira-Ferreira
- Unidade de Investigação Cardiovascular, Departamento de Cirurgia e Fisiologia, Faculdade de Medicina, Universidade do Porto, Porto, Portugal
| | - Fábio Trindade
- Unidade de Investigação Cardiovascular, Departamento de Cirurgia e Fisiologia, Faculdade de Medicina, Universidade do Porto, Porto, Portugal; iBiMED, Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| | - Rui Vitorino
- Unidade de Investigação Cardiovascular, Departamento de Cirurgia e Fisiologia, Faculdade de Medicina, Universidade do Porto, Porto, Portugal; iBiMED, Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| | - Scott K Powers
- Department of Applied Physiology and Kinesiology, University of Florida, United States
| | - Daniel Moreira-Gonçalves
- Unidade de Investigação Cardiovascular, Departamento de Cirurgia e Fisiologia, Faculdade de Medicina, Universidade do Porto, Porto, Portugal; CIAFEL, Faculty of Sport, University of Porto, Porto, Portugal.
| |
Collapse
|
20
|
Fulghum K, Hill BG. Metabolic Mechanisms of Exercise-Induced Cardiac Remodeling. Front Cardiovasc Med 2018; 5:127. [PMID: 30255026 PMCID: PMC6141631 DOI: 10.3389/fcvm.2018.00127] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 08/23/2018] [Indexed: 12/13/2022] Open
Abstract
Exercise has a myriad of physiological benefits that derive in part from its ability to improve cardiometabolic health. The periodic metabolic stress imposed by regular exercise appears fundamental in driving cardiovascular tissue adaptation. However, different types, intensities, or durations of exercise elicit different levels of metabolic stress and may promote distinct types of tissue remodeling. In this review, we discuss how exercise affects cardiac structure and function and how exercise-induced changes in metabolism regulate cardiac adaptation. Current evidence suggests that exercise typically elicits an adaptive, beneficial form of cardiac remodeling that involves cardiomyocyte growth and proliferation; however, chronic levels of extreme exercise may increase the risk for pathological cardiac remodeling or sudden cardiac death. An emerging theme underpinning acute as well as chronic cardiac adaptations to exercise is metabolic periodicity, which appears important for regulating mitochondrial quality and function, for stimulating metabolism-mediated exercise gene programs and hypertrophic kinase activity, and for coordinating biosynthetic pathway activity. In addition, circulating metabolites liberated during exercise trigger physiological cardiac growth. Further understanding of how exercise-mediated changes in metabolism orchestrate cell signaling and gene expression could facilitate therapeutic strategies to maximize the benefits of exercise and improve cardiac health.
Collapse
Affiliation(s)
- Kyle Fulghum
- Department of Medicine, Envirome Institute, Institute of Molecular Cardiology, Diabetes and Obesity Center, Louisville, KY, United States
- Department of Physiology, University of Louisville, Louisville, KY, United States
| | - Bradford G. Hill
- Department of Medicine, Envirome Institute, Institute of Molecular Cardiology, Diabetes and Obesity Center, Louisville, KY, United States
| |
Collapse
|
21
|
Du X, Li X, Chen L, Zhang M, Lei L, Gao W, Shi Z, Dong Y, Wang Z, Li X, Liu G. Hepatic miR-125b inhibits insulin signaling pathway by targeting PIK3CD. J Cell Physiol 2018; 233:6052-6066. [PMID: 29319168 DOI: 10.1002/jcp.26442] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 01/05/2018] [Indexed: 12/26/2022]
Abstract
Insulin resistance is often characterized as the most critical factor contributing to the development of (T2D) type 2 diabetes. MicroRNAs (miRNAs) are endogenous non-coding short single-stranded RNAs that function as negative regulators in many physiological and pathological processes. The objective of this study was to evaluate the roles of miR-125b in the regulation of insulin sensitivity in hepatocytes. We found that hepatic miR-125b levels were significantly increased in the patients with type 2 diabetes, high fat diet (HFD) mice, ob/ob and db/db mice. In vitro, miR-125b was also significantly up-regulated in tumor necrosis factor-alpha- (TNF-α) and glucosamine-induced insulin resistance conditions. Furthermore, miR-125b overexpression impaired the insulin signaling pathway in HepG2 cells, L02c cells, and primary hepatocytes. Inhibition of miR-125b improved insulin sensitivity, especially in insulin-resistant cells induced by either TNF-α or glucosamine. We demonstrated that miR-125b targeted the 3'-untranslated region (3'-UTR) of phosphoinositide 3-kinase catalytic subunit delta (PIK3CD) mRNA. The hepatic PIK3CD protein levels were markedly decreased in patients with type 2 diabetes, HFD, ob/ob, and db/db mice. Inhibition of PIK3CD markedly attenuated the improvement of insulin sensitivity induced by miR-125b inhibitors. More importantly, overexpressing miR-125b in mice causes insulin resistance and impairs glucose homeostasis. Together, these findings indicate that miR-125b inhibits insulin sensitivity by targeting PIK3CD in hepatocytes, supporting hepatic miR-125b, or PIK3CD are potential therapeutic target of insulin resistance.
Collapse
Affiliation(s)
- Xiliang Du
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Xiaobing Li
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Liang Chen
- Heilongjiang Institute of Veterinary Science, Qiqihar, Heilongjiang, China
| | - Min Zhang
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Lin Lei
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Wenwen Gao
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Zhen Shi
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Yuhao Dong
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Zhe Wang
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Xinwei Li
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Guowen Liu
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| |
Collapse
|
22
|
Ryzhov S, Robich MP, Roberts DJ, Favreau-Lessard AJ, Peterson SM, Jachimowicz E, Rath R, Vary CPH, Quinn R, Kramer RS, Sawyer DB. ErbB2 promotes endothelial phenotype of human left ventricular epicardial highly proliferative cells (eHiPC). J Mol Cell Cardiol 2018; 115:39-50. [PMID: 29291395 PMCID: PMC5926239 DOI: 10.1016/j.yjmcc.2017.12.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 12/20/2017] [Accepted: 12/28/2017] [Indexed: 12/13/2022]
Abstract
The adult human heart contains a subpopulation of highly proliferative cells. The role of ErbB receptors in these cells has not been studied. From human left ventricular (LV) epicardial biopsies, we isolated highly proliferative cells (eHiPC) to characterize the cell surface expression and function of ErbB receptors in the regulation of cell proliferation and phenotype. We found that human LV eHiPC express all four ErbB receptor subtypes. However, the expression of ErbB receptors varied widely among eHiPC isolated from different subjects. eHiPC with higher cell surface expression of ErbB2 reproduced the phenotype of endothelial cells and were characterized by endothelial cell-like functional properties. We also found that EGF/ErbB1 induces VEGFR2 expression, while ligands for both ErbB1 and ErbB3/4 induce expression of Tie2. The number of CD31posCD45neg endothelial cells is higher in LV biopsies from subjects with high ErbB2 (ErbB2high) eHiPC compared to low ErbB2 (ErbB2low) eHiPC. These findings have important implications for potential strategies to increase the efficacy of cell-based revascularization of the injured heart, through promotion of an endothelial phenotype in cardiac highly proliferative cells.
Collapse
Affiliation(s)
- Sergey Ryzhov
- Maine Medical Center Research Institute, Scarborough, ME, United States
| | - Michael P Robich
- Maine Medical Center Research Institute, Scarborough, ME, United States; Maine Medical Center, Portland, ME, United States
| | - Daniel J Roberts
- Maine Medical Center Research Institute, Scarborough, ME, United States; Maine Medical Center, Portland, ME, United States
| | | | - Sarah M Peterson
- Maine Medical Center Research Institute, Scarborough, ME, United States
| | | | - Rutwik Rath
- Maine Medical Center Research Institute, Scarborough, ME, United States
| | - Calvin P H Vary
- Maine Medical Center Research Institute, Scarborough, ME, United States
| | - Reed Quinn
- Maine Medical Center, Portland, ME, United States
| | | | - Douglas B Sawyer
- Maine Medical Center Research Institute, Scarborough, ME, United States; Maine Medical Center, Portland, ME, United States.
| |
Collapse
|
23
|
Gardner JD. Neuregulin-1β as a potential therapeutic for targeting fibroblasts in heart disease. J Mol Cell Cardiol 2017; 112:132-133. [PMID: 28689005 DOI: 10.1016/j.yjmcc.2017.07.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 06/30/2017] [Accepted: 07/03/2017] [Indexed: 01/19/2023]
Affiliation(s)
- Jason D Gardner
- Department of Physiology, School of Medicine, Louisiana State University Health Sciences Center, MEB, 1901 Perdido St., New Orleans, LA, United States.
| |
Collapse
|
24
|
Neuregulin 1 improves complex 2-mediated mitochondrial respiration in skeletal muscle of healthy and diabetic mice. Sci Rep 2017; 7:1742. [PMID: 28496106 PMCID: PMC5431817 DOI: 10.1038/s41598-017-02029-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 04/04/2017] [Indexed: 12/12/2022] Open
Abstract
It has been reported that neuregulin1 (NRG1) improves glucose tolerance in healthy and diabetic rodents. In vitro studies also suggest that NRG1 regulates myocyte oxidative capacity. To confirm this observation in vivo, we evaluated the effect on mitochondrial function of an 8-week treatment with NRG1 in db/db diabetic mice and C57BL/6JRJ healthy controls. NRG1 treatment improved complex 2-mediated mitochondrial respiration in the gastrocnemius of both control and diabetic mice and increased mitochondrial complex 2 subunit content by 2-fold. This effect was not associated with an increase in mitochondrial biogenesis markers. Enhanced ERBB4 phosphorylation could mediate NRG1 effects on mitochondrial function through signalling pathways, independently of ERK1/2, AKT or AMPK.
Collapse
|
25
|
Wu G, Liu XX, Lu NN, Liu QB, Tian Y, Ye WF, Jiang GJ, Tao RR, Han F, Lu YM. Endothelial ErbB4 deficit induces alterations in exploratory behavior and brain energy metabolism in mice. CNS Neurosci Ther 2017; 23:510-517. [PMID: 28421673 DOI: 10.1111/cns.12695] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 03/12/2017] [Accepted: 03/15/2017] [Indexed: 12/17/2022] Open
Abstract
AIMS The receptor tyrosine kinase ErbB4 is present throughout the primate brain and has a distinct functional profile. In this study, we investigate the potential role of endothelial ErbB4 receptor signaling in the brain. RESULTS Here, we show that the endothelial cell-specific deletion of ErbB4 induces decreased exploratory behavior in adult mice. However, the water maze task for spatial memory and the memory reconsolidation test reveal no changes; additionally, we observe no impairment in CaMKII phosphorylation in Cdh5Cre;ErbB4f/f mice, which indicates that the endothelial ErbB4 deficit leads to decreased exploratory activity rather than direct memory deficits. Furthermore, decreased brain metabolism, which was measured using micro-positron emission tomography, is observed in the Cdh5Cre;ErbB4f/f mice. Consistently, the immunoblot data demonstrate the downregulation of brain Glut1, phospho-ULK1 (Ser555), and TIGAR in the endothelial ErbB4 conditional knockout mice. Collectively, our findings suggest that endothelial ErbB4 plays a critical role in regulating brain function, at least in part, through maintaining normal brain energy homeostasis. CONCLUSIONS Targeting ErbB4 or the modulation of endothelial ErbB4 signaling may represent a rational pharmacological approach to treat neurological disorders.
Collapse
Affiliation(s)
- Gang Wu
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.,School of Medicine, Zhejiang University City College, Hangzhou, Zhejiang, China
| | - Xiu-Xiu Liu
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.,School of Medicine, Zhejiang University City College, Hangzhou, Zhejiang, China
| | - Nan-Nan Lu
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Qi-Bing Liu
- School of Pharmacy, Hainan Medical College, Haikou, China
| | - Yun Tian
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Wei-Feng Ye
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Guo-Jun Jiang
- Department of Pharmacy, Zhejiang Xiaoshan Hospital, Hangzhou, Zhejiang, China
| | - Rong-Rong Tao
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Feng Han
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Ying-Mei Lu
- School of Medicine, Zhejiang University City College, Hangzhou, Zhejiang, China
| |
Collapse
|
26
|
Bishopric NH. A Growth Tonic for Heart Failure? JACC Basic Transl Sci 2016; 1:587-589. [PMID: 30167543 PMCID: PMC6113560 DOI: 10.1016/j.jacbts.2016.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
27
|
Gerarduzzi C, de Polo A, Liu XS, El Kharbili M, Little JB, Yuan ZM. Human epidermal growth factor receptor 4 (Her4) Suppresses p53 Protein via Targeting the MDMX-MDM2 Protein Complex: IMPLICATION OF A NOVEL MDMX SER-314 PHOSPHOSITE. J Biol Chem 2016; 291:25937-25949. [PMID: 27777309 DOI: 10.1074/jbc.m116.752303] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Revised: 10/03/2016] [Indexed: 12/11/2022] Open
Abstract
Deregulated receptor tyrosine kinase (RTK) signaling is frequently associated with tumorigenesis and therapy resistance, but its underlying mechanisms still need to be elucidated. In this study, we have shown that the RTK human epidermal growth factor receptor 4 (Her4, also known as Erbb4) can inhibit the tumor suppressor p53 by regulating MDMX-mouse double minute 2 homolog (MDM2) complex stability. Upon activation by either overexpression of a constitutively active vector or ligand binding (Neuregulin-1), Her4 was able to stabilize the MDMX-MDM2 complex, resulting in suppression of p53 transcriptional activity, as shown by p53-responsive element-driven luciferase assay and mRNA levels of p53 target genes. Using a phospho-proteomics approach, we functionally identified a novel Her4-induced posttranslational modification on MDMX at Ser-314, a putative phosphorylation site for the CDK4/6 kinase. Remarkably, inhibition of Ser-314 phosphorylation either with Ser-to-Ala substitution or with a specific inhibitor of CDK4/6 kinase blocked Her4-induced stabilization of MDMX-MDM2 and rescued p53 activity. Our study offers insights into the mechanisms of deregulated RTK-induced carcinogenesis and provides the basis for the use of inhibitors targeting RTK-mediated signals for p53 restoration.
Collapse
Affiliation(s)
- Casimiro Gerarduzzi
- From the John B. Little Center for Radiation Sciences, Harvard T. H. Chan School of Public Health, Boston, Massachusetts 02115
| | - Anna de Polo
- From the John B. Little Center for Radiation Sciences, Harvard T. H. Chan School of Public Health, Boston, Massachusetts 02115
| | - Xue-Song Liu
- From the John B. Little Center for Radiation Sciences, Harvard T. H. Chan School of Public Health, Boston, Massachusetts 02115
| | - Manale El Kharbili
- From the John B. Little Center for Radiation Sciences, Harvard T. H. Chan School of Public Health, Boston, Massachusetts 02115
| | - John B Little
- From the John B. Little Center for Radiation Sciences, Harvard T. H. Chan School of Public Health, Boston, Massachusetts 02115
| | - Zhi-Min Yuan
- From the John B. Little Center for Radiation Sciences, Harvard T. H. Chan School of Public Health, Boston, Massachusetts 02115
| |
Collapse
|
28
|
Repair Injured Heart by Regulating Cardiac Regenerative Signals. Stem Cells Int 2016; 2016:6193419. [PMID: 27799944 PMCID: PMC5075315 DOI: 10.1155/2016/6193419] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 06/27/2016] [Accepted: 06/29/2016] [Indexed: 01/10/2023] Open
Abstract
Cardiac regeneration is a homeostatic cardiogenic process by which the sections of malfunctioning adult cardiovascular tissues are repaired and renewed employing a combination of both cardiomyogenesis and angiogenesis. Unfortunately, while high-quality regeneration can be performed in amphibians and zebrafish hearts, mammalian hearts do not respond in kind. Indeed, a long-term loss of proliferative capacity in mammalian adult cardiomyocytes in combination with dysregulated induction of tissue fibrosis impairs mammalian endogenous heart regenerative capacity, leading to deleterious cardiac remodeling at the end stage of heart failure. Interestingly, several studies have demonstrated that cardiomyocyte proliferation capacity is retained in mammals very soon after birth, and cardiac regeneration potential is correspondingly preserved in some preadolescent vertebrates after myocardial infarction. There is therefore great interest in uncovering the molecular mechanisms that may allow heart regeneration during adult stages. This review will summarize recent findings on cardiac regenerative regulatory mechanisms, especially with respect to extracellular signals and intracellular pathways that may provide novel therapeutics for heart diseases. Particularly, both in vitro and in vivo experimental evidences will be presented to highlight the functional role of these signaling cascades in regulating cardiomyocyte proliferation, cardiomyocyte growth, and maturation, with special emphasis on their responses to heart tissue injury.
Collapse
|