1
|
Phạm TTT, Murza A, Marsault É, Frampton JP, Rainey JK. Localized apelin-17 analogue-bicelle interactions as a facilitator of membrane-catalyzed receptor recognition and binding. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2024; 1866:184289. [PMID: 38278504 DOI: 10.1016/j.bbamem.2024.184289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 01/17/2024] [Accepted: 01/18/2024] [Indexed: 01/28/2024]
Abstract
The apelinergic system encompasses two peptide ligand families, apelin and apela, along with the apelin receptor (AR or APJ), a class A G-protein-coupled receptor. This system has diverse physiological effects, including modulating heart contraction, vasodilation/constriction, glucose regulation, and vascular development, with involvement in a variety of pathological conditions. Apelin peptides have been previously shown to interact with and become structured upon binding to anionic micelles, consistent with a membrane-catalyzed mechanism of ligand-receptor binding. To overcome the challenges of observing nuclear magnetic resonance (NMR) spectroscopy signals of a dilute peptide in biological environments, 19F NMR spectroscopy, including diffusion ordered spectroscopy (DOSY) and saturation transfer difference (STD) experiments, was used herein to explore the membrane-interactive behaviour of apelin. NMR-optimized apelin-17 analogues with 4-trifluoromethyl-phenylalanine at various positions were designed and tested for bioactivity through ERK activation in stably-AR transfected HEK 293 T cells. Far-UV circular dichroism (CD) spectropolarimetry and 19F NMR spectroscopy were used to compare the membrane interactions of these analogues with unlabelled apelin-17 in both zwitterionic/neutral and net-negative bicelle conditions. Each analogue binds to bicelles with relatively weak affinity (i.e., in fast exchange on the NMR timescale), with preferential interactions observed at the cationic residue-rich N-terminal and mid-length regions of the peptide leaving the C-terminal end unencumbered for receptor recognition, enabling a membrane-anchored fly-casting mechanism of peptide search for the receptor. In all, this study provides further insight into the membrane-interactive behaviour of an important bioactive peptide, demonstrating interactions and biophysical behaviour that cannot be neglected in therapeutic design.
Collapse
Affiliation(s)
- Trần Thanh Tâm Phạm
- Department of Biochemistry & Molecular Biology, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Alexandre Murza
- Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Éric Marsault
- Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - John P Frampton
- Department of Biochemistry & Molecular Biology, Dalhousie University, Halifax, NS B3H 4R2, Canada; School of Biomedical Engineering, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Jan K Rainey
- Department of Biochemistry & Molecular Biology, Dalhousie University, Halifax, NS B3H 4R2, Canada; School of Biomedical Engineering, Dalhousie University, Halifax, NS B3H 4R2, Canada; Department of Chemistry, Dalhousie University, Halifax, NS B3H 4R2, Canada.
| |
Collapse
|
2
|
Song Q, Wang X, Cao Z, Xin C, Zhang J, Li S. The Apelin/APJ System: A Potential Therapeutic Target for Sepsis. J Inflamm Res 2024; 17:313-330. [PMID: 38250143 PMCID: PMC10800090 DOI: 10.2147/jir.s436169] [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: 08/20/2023] [Accepted: 01/01/2024] [Indexed: 01/23/2024] Open
Abstract
Apelin is the native ligand for the G protein-coupled receptor APJ. Numerous studies have demonstrated that the Apelin/APJ system has positive inotropic, anti-inflammatory, and anti-apoptotic effects and regulates fluid homeostasis. The Apelin/APJ system has been demonstrated to play a protective role in sepsis and may serve as a promising therapeutic target for the treatment of sepsis. Better understanding of the mechanisms of the effects of the Apelin/APJ system will aid in the development of novel drugs for the treatment of sepsis. In this review, we provide a brief overview of the physiological role of the Apelin/APJ system and its role in sepsis.
Collapse
Affiliation(s)
- Qing Song
- Intensive Care Unit, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116000, People’s Republic of China
| | - Xi Wang
- Intensive Care Unit, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116000, People’s Republic of China
| | - Zhenhuan Cao
- Intensive Care Unit, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116000, People’s Republic of China
| | - Chun Xin
- Intensive Care Unit, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116000, People’s Republic of China
| | - Jingyuan Zhang
- Intensive Care Unit, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116000, People’s Republic of China
| | - Suwei Li
- Intensive Care Unit, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116000, People’s Republic of China
| |
Collapse
|
3
|
Habibian M, Biniaz S, Moosavi SJ. Protective Role of Short-term Aerobic Exercise Against Zinc Oxide Nanoparticles-Induced Cardiac Oxidative Stress Via Possible Changes of Apelin, Angiotensin II/Angiotensin II Type I Signalling Pathway. Cardiovasc Toxicol 2023:10.1007/s12012-023-09792-8. [PMID: 37184829 DOI: 10.1007/s12012-023-09792-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 04/14/2023] [Indexed: 05/16/2023]
Abstract
This study examined the protective role of short-term aerobic exercise on ZnO NPs-induced cardiac oxidative stress and possible changes of apelin, angiotensin II (AngII) and angiotensin II type I receptor (AT1R) signalling pathway. Thirty-five male Wistar rats were randomized into five groups of seven rats, including control, saline, ZnO NPs, exercise and exercise + ZnO NPs groups. The animal in ZnO NPs and exercise + ZnO NPs groups received 1 mg/kg of ZnO NPs. Rats underwent the treadmill exercise program. Treatments lasted four weeks, 5 days/week. After 4 weeks of treatment, superoxide dismutase (SOD) activity, malondialdehyde (MDA), apelin, Ang II and AT1R concentration were measured in heart tissue.Cardiac MDA, Ang II and AT1R levels significantly increased while SOD activity and apelin levels significantly decreased following ZnO NPs administration. The aerobic exercise induced a significant increase in the SOD activity and apelin levels and a significant decrease in the enhanced MDA, Ang II and AT1R levels in the heart of ZnO NPs-exposed rats. These results suggest that the exercise-induced attenuation of the Ang II-AT1R signalling pathway is mediated by reduced lipid peroxidation, augmented antioxidant defence and enhanced apelin synthesis that may be a protective mechanism to prevent and/or treatment ZnO NPs-induced cardiac oxidative stress.
Collapse
Affiliation(s)
- Masoumeh Habibian
- Department of Physical Education and Sports Sciences, Qaemshahar Branch, Islamic Azad University, Qaemshahar, Iran.
| | - Sara Biniaz
- Department of Physical Education and Sports Sciences, Sari Branch, Islamic Azad University, Sari, Iran
| | - Seyyed Jafar Moosavi
- Department of Physical Education and Sports Sciences, Qaemshahar Branch, Islamic Azad University, Qaemshahar, Iran
| |
Collapse
|
4
|
Pang B, Jiang YR, Xu JY, Shao DX, Hao LY. Apelin/ELABELA-APJ system in cardiac hypertrophy: Regulatory mechanisms and therapeutic potential. Eur J Pharmacol 2023; 949:175727. [PMID: 37062502 DOI: 10.1016/j.ejphar.2023.175727] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 04/03/2023] [Accepted: 04/14/2023] [Indexed: 04/18/2023]
Abstract
Heart failure is one of the most significant public health problems faced by millions of medical researchers worldwide. And pathological cardiac hypertrophy is considered one of the possible factors of increasing the risk of heart failure. Here, we introduce apelin/ELABELA-APJ system as a novel therapeutic target for cardiac hypertrophy, bringing about new directions in clinical treatment. Apelin has been proven to regulate cardiac hypertrophy through various pathways. And an increasing number of studies on ELABELA, the newly discovered endogenous ligand, suggest it can alleviate cardiac hypertrophy through mechanisms similar or different to apelin. In this review, we elaborate on the role that apelin/ELABELA-APJ system plays in cardiac hypertrophy and the intricate mechanisms that apelin/ELABELA-APJ affect cardiac hypertrophy. We also illuminate and make comparisons of the newly designed peptides and small molecules as agonists and antagonists for APJ, updating the breakthroughs in this field.
Collapse
Affiliation(s)
- Bo Pang
- China Medical University-The Queen's University of Belfast Joint College, Queen's University Belfast, Belfast Northern Ireland, BT9 7BL, United Kingdom.
| | - Yin-Ru Jiang
- China Medical University-The Queen's University of Belfast Joint College, Queen's University Belfast, Belfast Northern Ireland, BT9 7BL, United Kingdom.
| | - Jia-Yao Xu
- China Medical University-The Queen's University of Belfast Joint College, Queen's University Belfast, Belfast Northern Ireland, BT9 7BL, United Kingdom.
| | - Dong-Xue Shao
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, 110122, China.
| | - Li-Ying Hao
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, 110122, China.
| |
Collapse
|
5
|
Popov SV, Maslov LN, Mukhomedzyanov AV, Kurbatov BK, Gorbunov AS, Kilin M, Azev VN, Khlestkina MS, Sufianova GZ. Apelin Is a Prototype of Novel Drugs for the Treatment of Acute Myocardial Infarction and Adverse Myocardial Remodeling. Pharmaceutics 2023; 15:pharmaceutics15031029. [PMID: 36986889 PMCID: PMC10056827 DOI: 10.3390/pharmaceutics15031029] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 03/03/2023] [Accepted: 03/16/2023] [Indexed: 03/30/2023] Open
Abstract
In-hospital mortality in patients with ST-segment elevation myocardial infarction (STEMI) is 5-6%. Consequently, it is necessary to develop fundamentally novel drugs capable of reducing mortality in patients with acute myocardial infarction. Apelins could be the prototype for such drugs. Chronic administration of apelins mitigates adverse myocardial remodeling in animals with myocardial infarction or pressure overload. The cardioprotective effect of apelins is accompanied by blockage of the MPT pore, GSK-3β, and the activation of PI3-kinase, Akt, ERK1/2, NO-synthase, superoxide dismutase, glutathione peroxidase, matrix metalloproteinase, the epidermal growth factor receptor, Src kinase, the mitoKATP channel, guanylyl cyclase, phospholipase C, protein kinase C, the Na+/H+ exchanger, and the Na+/Ca2+ exchanger. The cardioprotective effect of apelins is associated with the inhibition of apoptosis and ferroptosis. Apelins stimulate the autophagy of cardiomyocytes. Synthetic apelin analogues are prospective compounds for the development of novel cardioprotective drugs.
Collapse
Affiliation(s)
- Sergey V Popov
- Tomsk National Research Medical Center, Cardiology Research Institute, The Russian Academy of Sciences, Kyevskaya 111A, Tomsk 634012, Russia
| | - Leonid N Maslov
- Tomsk National Research Medical Center, Cardiology Research Institute, The Russian Academy of Sciences, Kyevskaya 111A, Tomsk 634012, Russia
| | - Alexandr V Mukhomedzyanov
- Tomsk National Research Medical Center, Cardiology Research Institute, The Russian Academy of Sciences, Kyevskaya 111A, Tomsk 634012, Russia
| | - Boris K Kurbatov
- Tomsk National Research Medical Center, Cardiology Research Institute, The Russian Academy of Sciences, Kyevskaya 111A, Tomsk 634012, Russia
| | - Alexandr S Gorbunov
- Tomsk National Research Medical Center, Cardiology Research Institute, The Russian Academy of Sciences, Kyevskaya 111A, Tomsk 634012, Russia
| | - Michail Kilin
- Tomsk National Research Medical Center, Cardiology Research Institute, The Russian Academy of Sciences, Kyevskaya 111A, Tomsk 634012, Russia
| | - Viacheslav N Azev
- Branch of Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, The Russian Academy of Sciences, Pushchino 142290, Russia
| | - Maria S Khlestkina
- Department of Pharmacology, Tyumen State Medical University, Tyumen 625023, Russia
| | - Galina Z Sufianova
- Department of Pharmacology, Tyumen State Medical University, Tyumen 625023, Russia
| |
Collapse
|
6
|
Bernardo VS, Torres FF, da Silva DGH. FoxO3 and oxidative stress: a multifaceted role in cellular adaptation. J Mol Med (Berl) 2023; 101:83-99. [PMID: 36598531 DOI: 10.1007/s00109-022-02281-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 12/14/2022] [Accepted: 12/16/2022] [Indexed: 01/05/2023]
Abstract
Oxidative stress is a major cause of morbidity and mortality in human health and disease. In this review, we focus on the Forkhead Box (Fox) subclass O3 (FoxO3), an extensively studied transcription factor that plays a pleiotropic role in a wide range of physiological and pathological processes by regulating multiple gene regulatory networks involved in the modulation of numerous aspects of cellular metabolism, including fuel metabolism, cell death, and stress resistance. This review will also focus on regulatory mechanisms of FoxO3 expression and activity, such as crucial post-translational modifications and non-coding RNAs. Moreover, this work discusses and evidences some pathways to how this transcription factor and reactive oxygen species regulate each other, which may lead to the pathogenesis of various types of diseases. Therefore, in addition to being a promising therapeutic target, the FoxO3-regulated signaling pathways can also be used as reliable diagnostic and prognostic biomarkers and indicators for drug responsiveness.
Collapse
Affiliation(s)
| | | | - Danilo Grünig Humberto da Silva
- Department of Biology, Universidade Estadual Paulista (UNESP), São Paulo, Brazil.
- Campus de Três Lagoas, Universidade Federal de Mato Grosso Do Sul (CPTL/UFMS), Avenida Ranulpho Marques Leal, 3484, Três Lagoas, Mato Grosso Do Sul, Distrito Industrial-Post code 79613-000, Brazil.
| |
Collapse
|
7
|
Orea-Soufi A, Paik J, Bragança J, Donlon TA, Willcox BJ, Link W. FOXO transcription factors as therapeutic targets in human diseases. Trends Pharmacol Sci 2022; 43:1070-1084. [PMID: 36280450 DOI: 10.1016/j.tips.2022.09.010] [Citation(s) in RCA: 79] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 09/20/2022] [Accepted: 09/28/2022] [Indexed: 11/11/2022]
Abstract
Forkhead box (FOX)O proteins are transcription factors (TFs) with four members in mammals designated FOXO1, FOXO3, FOXO4, and FOXO6. FOXO TFs play a pivotal role in the cellular adaptation to diverse stress conditions. FOXO proteins act as context-dependent tumor suppressors and their dysregulation has been implicated in several age-related diseases. FOXO3 has been established as a major gene for human longevity. Accordingly, FOXO proteins have emerged as potential targets for the therapeutic development of drugs and geroprotectors. In this review, we provide an overview of the most recent advances in our understanding of FOXO regulation and function in various pathological conditions. We discuss strategies targeting FOXOs directly or by the modulation of upstream regulators, shedding light on the most promising intervention points. We also reveal the most relevant clinical indications and discuss the potential, trends, and challenges of modulating FOXO activity for therapeutic purposes.
Collapse
Affiliation(s)
- Alba Orea-Soufi
- Algarve Biomedical Center Research Institute (ABC-RI), University of Algarve, Campus de Gambelas, 8005-139 Faro, Portugal; Algarve Biomedical Center (ABC), University of Algarve, Campus de Gambelas, 8005-139 Faro, Portugal; Faculty of Medicine and Biomedical Sciences, Campus de Gambelas, 8005-139 Faro, Portugal
| | - Jihye Paik
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - José Bragança
- Algarve Biomedical Center Research Institute (ABC-RI), University of Algarve, Campus de Gambelas, 8005-139 Faro, Portugal; Algarve Biomedical Center (ABC), University of Algarve, Campus de Gambelas, 8005-139 Faro, Portugal; Faculty of Medicine and Biomedical Sciences, Campus de Gambelas, 8005-139 Faro, Portugal; Champalimaud Research Program, Champalimaud Center for the Unknown, Lisbon, Portugal
| | - Timothy A Donlon
- Department of Research, Kuakini Medical Center, Honolulu, HI 96817, USA; Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 96813, USA
| | - Bradley J Willcox
- Department of Research, Kuakini Medical Center, Honolulu, HI 96817, USA; Department of Geriatric Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 96813, USA
| | - Wolfgang Link
- Instituto de Investigaciones Biomédicas 'Alberto Sols' (CSIC-UAM), Arturo Duperier 4, 28029-Madrid, Spain.
| |
Collapse
|
8
|
Cha HS, Lee HK, Park SH, Nam MJ. Acetylshikonin induces apoptosis of human osteosarcoma U2OS cells by triggering ROS-dependent multiple signal pathways. Toxicol In Vitro 2022; 86:105521. [DOI: 10.1016/j.tiv.2022.105521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/18/2022] [Accepted: 11/15/2022] [Indexed: 11/19/2022]
|
9
|
Shi Y, Zhao L, Zhang Y, Qin Q, Cong H, Guo Z. Homocysteine promotes cardiac fibrosis by regulating the Akt/FoxO3 pathway. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1732. [PMID: 35071426 PMCID: PMC8743705 DOI: 10.21037/atm-21-5602] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 11/10/2021] [Indexed: 12/19/2022]
Abstract
Background Evaluated plasma homocysteine (Hcy) is an independent risk factor for cardiac fibrosis which is a common feature of cardiovascular disease, although the mechanisms are still unclear. This study aims to explore the mechanism of Hcy-induced cardiac fibrosis. Methods The mRNA and protein levels of Forkhead box O3 (FoxO3) and differentiation markers were detected in primary cardiac fibroblasts (CFs) after 300 µM Hcy treatment. Scratch and transwell migration assay were used to determine the effect of Hcy on proliferation and migration in CFs. The protein levels involved in the fibrotic processes in mice fed with high methionine diet (HMD) for 4 or 8 weeks were investigated by western blot. CFs were infected with FoxO3 recombinant adenovirus to explore the potential role of FoxO3 in Hcy-induced cardiac dysfunction. Results Hcy treatment significantly promoted the differentiation, proliferation and migration of CFs, while FoxO3 activity were decreased in CFs. In HMD hearts, the protein levels of TIMP1, Fibronectin and α-SMA were increased after 4 or 8 weeks, but the FoxO3 activity was decreased. Moreover, the HMD hearts had a higher level of Bcl2 but lower of Bax and LC3II protein. In addition, FoxO3 overexpression attenuates Hcy-induced dysfunction in CFs. Conclusions Hcy promotes myofibroblast activation and resistance to autophagy and apoptosis in CFs, and eventually results in cardiac fibrosis by regulating the Akt/FoxO3 pathway. Thus, FoxO3 is a promising therapeutic target to prevent cardiac remodeling.
Collapse
Affiliation(s)
- Ying Shi
- Tianjin Institute of Cardiovascular Disease, Tianjin Chest Hospital, Tianjin, China.,Key Laboratory of Immune Microenvironment and Disease (Tianjin Medical University), Ministry of Education, Tianjin, China
| | - Lili Zhao
- Tianjin Institute of Cardiovascular Disease, Tianjin Chest Hospital, Tianjin, China
| | - Yifei Zhang
- Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin, China
| | - Qin Qin
- Tianjin Institute of Cardiovascular Disease, Tianjin Chest Hospital, Tianjin, China
| | - Hongliang Cong
- Tianjin Institute of Cardiovascular Disease, Tianjin Chest Hospital, Tianjin, China
| | - Zhigang Guo
- Department of Cardiovascular Surgery, Tianjin Chest Hospital, Tianjin, China
| |
Collapse
|
10
|
Wang P, Luo C, Zhu D, Song Y, Cao L, Luan H, Gao L, Zheng S, Li H, Tian G. Pericardial Adipose Tissue-Derived Leptin Promotes Myocardial Apoptosis in High-Fat Diet-Induced Obese Rats Through Janus Kinase 2/Reactive Oxygen Species/Na+/K+-ATPase Signaling Pathway. J Am Heart Assoc 2021; 10:e021369. [PMID: 34482701 PMCID: PMC8649551 DOI: 10.1161/jaha.121.021369] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Background Pathophysiologic mechanisms underlying cardiac structural and functional changes in obesity are complex and linked to adipocytokines released from pericardial adipose tissue (PAT) and cardiomyocyte apoptosis. Although leptin is involved in various pathological conditions, its role in paracrine action of pericardial adipose tissue on myocardial apoptosis remains unknown. This study was designed to investigate the role of PAT‐derived leptin on myocardial apoptosis in high‐fat diet–induced obese rats. Methods and Results Hearts were isolated from lean or high‐fat diet–induced obese Wistar rats for myocardial remodeling studies. Obese rats had abnormal myocardial structure, diastolic dysfunction, greatly elevated cardiac apoptosis, enhanced cardiac fibrosis, and increased oxidative stress level. ELISA detected significantly higher than circulating leptin level in PAT of obese, but not lean, rats. Western blot and immunohistochemical analyses demonstrated increased leptin receptor density in obese hearts. H9c2 cardiomyoblasts, after being exposed to PAT‐conditioned medium of obese rats, exhibited pronounced reactive oxygen species–mediated apoptosis, which was partially reversed by leptin antagonist. Moreover, leptin derived from PAT of obese rats inhibited Na+/K+‐ATPase activity of H9c2 cells through stimulating reactive oxygen species, thereby activating calcium‐dependent apoptosis. Pretreatment with specific inhibitors revealed that Janus kinase 2/signal transducer and activator of transcription 3 and phosphoinositide 3‐kinase/protein kinase B signaling pathways were involved in leptin‐induced myocardial apoptosis. Conclusions PAT‐derived leptin induces myocardial apoptosis in high‐fat diet–induced obese rats via activating Janus kinase 2/signal transducer and activator of transcription 3/reactive oxygen species signaling pathway and inhibiting its downstream Na+/K+‐ATPase activity.
Collapse
Affiliation(s)
- Ping Wang
- Department of Cardiology First Affiliated Hospital of Xi'an Jiaotong University Shaanxi China
| | - Chaodi Luo
- Department of Cardiology First Affiliated Hospital of Xi'an Jiaotong University Shaanxi China
| | - Danjun Zhu
- Department of Cardiology First Affiliated Hospital of Xi'an Jiaotong University Shaanxi China
| | - Yan Song
- Department of Ultrasound First Affiliated Hospital of Xi'an Jiaotong University Shaanxi China
| | - Lifei Cao
- Department of Cardiology First Affiliated Hospital of Xi'an Jiaotong University Shaanxi China
| | - Hui Luan
- Department of Cardiology First Affiliated Hospital of Xi'an Jiaotong University Shaanxi China
| | - Lan Gao
- Department of Cardiology First Affiliated Hospital of Xi'an Jiaotong University Shaanxi China
| | - Shuping Zheng
- Department of Cardiology First Affiliated Hospital of Xi'an Jiaotong University Shaanxi China
| | - Hao Li
- Intensive Care Unit First Affiliated Hospital of Xi'an Jiaotong University Shaanxi China
| | - Gang Tian
- Department of Cardiology First Affiliated Hospital of Xi'an Jiaotong University Shaanxi China
| |
Collapse
|
11
|
Zhou F, Wang YK, Zhang CG, Wu BY. miR-19a/b-3p promotes inflammation during cerebral ischemia/reperfusion injury via SIRT1/FoxO3/SPHK1 pathway. J Neuroinflammation 2021; 18:122. [PMID: 34051800 PMCID: PMC8164774 DOI: 10.1186/s12974-021-02172-5] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 05/11/2021] [Indexed: 02/07/2023] Open
Abstract
Background Stroke affects 3–4% of adults and kills numerous people each year. Recovering blood flow with minimal reperfusion-induced injury is crucial. However, the mechanisms underlying reperfusion-induced injury, particularly inflammation, are not well understood. Here, we investigated the function of miR-19a/b-3p/SIRT1/FoxO3/SPHK1 axis in ischemia/reperfusion (I/R). Methods MCAO (middle cerebral artery occlusion) reperfusion rat model was used as the in vivo model of I/R. Cultured neuronal cells subjected to OGD/R (oxygen glucose deprivation/reperfusion) were used as the in vitro model of I/R. MTT assay was used to assess cell viability and TUNEL staining was used to measure cell apoptosis. H&E staining was employed to examine cell morphology. qRT-PCR and western blot were performed to determine levels of miR-19a/b-3p, SIRT1, FoxO3, SPHK1, NF-κB p65, and cytokines like TNF-α, IL-6, and IL-1β. EMSA and ChIP were performed to validate the interaction of FoxO3 with SPHK1 promoter. Dual luciferase assay and RIP were used to verify the binding of miR-19a/b-3p with SIRT1 mRNA. Results miR-19a/b-3p, FoxO3, SPHK1, NF-κB p65, and cytokines were elevated while SIRT1 was reduced in brain tissues following MCAO/reperfusion or in cells upon OGD/R. Knockdown of SPHK1 or FoxO3 suppressed I/R-induced inflammation and cell death. Furthermore, knockdown of FoxO3 reversed the effects of SIRT1 knockdown. Inhibition of the miR-19a/b-3p suppressed inflammation and this suppression was blocked by SIRT1 knockdown. FoxO3 bound SPHK1 promoter and activated its transcription. miR-19a/b-3p directly targeted SIRT1 mRNA. Conclusion miR-19a/b-3p promotes inflammatory responses during I/R via targeting SIRT1/FoxO3/SPHK1 axis.
Collapse
Affiliation(s)
- Feng Zhou
- Research Center of Clinical Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong Province, People's Republic of China.,Department of Neurology, First People's Hospital of Foshan, Foshan, 528000, Guangdong Province, People's Republic of China
| | - Yu-Kai Wang
- Department of Neurology, First People's Hospital of Foshan, Foshan, 528000, Guangdong Province, People's Republic of China
| | - Cheng-Guo Zhang
- Department of Neurology, First People's Hospital of Foshan, Foshan, 528000, Guangdong Province, People's Republic of China.
| | - Bing-Yi Wu
- Research Center of Clinical Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong Province, People's Republic of China.
| |
Collapse
|
12
|
Lou ZL, Zhang CX, Li JF, Chen RH, Wu WJ, Hu XF, Shi HC, Gao WY, Zhao QF. Apelin/APJ-Manipulated CaMKK/AMPK/GSK3 β Signaling Works as an Endogenous Counterinjury Mechanism in Promoting the Vitality of Random-Pattern Skin Flaps. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:8836058. [PMID: 33574981 PMCID: PMC7857910 DOI: 10.1155/2021/8836058] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 12/28/2020] [Indexed: 02/07/2023]
Abstract
A random-pattern skin flap plays an important role in the field of wound repair; the mechanisms that influence the survival of random-pattern skin flaps have been extensively studied but little attention has been paid to endogenous counterinjury substances and mechanism. Previous reports reveal that the apelin-APJ axis is an endogenous counterinjury mechanism that has considerable function in protecting against infection, inflammation, oxidative stress, necrosis, and apoptosis in various organs. As an in vivo study, our study proved that the apelin/APJ axis protected the skin flap by alleviating vascular oxidative stress and the apelin/APJ axis works as an antioxidant stress factor dependent on CaMKK/AMPK/GSK3β signaling. In addition, the apelin/APJ-manipulated CaMKK/AMPK/GSK3β-dependent mechanism improves HUVECs' resistance to oxygen and glucose deprivation/reperfusion (OGD/R), reduces ROS production and accumulation, maintained the normal mitochondrial membrane potential, and suppresses oxidative stress in vitro. Besides, activation of the apelin/APJ axis promotes vascular migration and angiogenesis under relative hypoxia condition through CaMKK/AMPK/GSK3β signaling. In a word, we provide new evidence that the apelin/APJ axis is an effective antioxidant and can significantly improve the vitality of random flaps, so it has potential be a promising clinical treatment.
Collapse
Affiliation(s)
- Zhi-Ling Lou
- Department of Cardiovascular and Thoracic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China
- Children's Heart Center, Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou 325000, China
| | - Chen-Xi Zhang
- The Second School of Medicine, Wenzhou Medical University, Wenzhou 325000, China
- Department of Orthopaedic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou 325000, China
| | - Jia-Feng Li
- The Second School of Medicine, Wenzhou Medical University, Wenzhou 325000, China
- Department of Orthopaedic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou 325000, China
| | - Rui-Heng Chen
- Department of Cardiovascular and Thoracic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou 325000, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou 325000, China
| | - Wei-Jia Wu
- Department of Cardiovascular and Thoracic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou 325000, China
| | - Xiao-Fen Hu
- Zhejiang Chinese Medical University, Hangzhou 310000, China
| | - Hao-Chun Shi
- Department of Cardiovascular and Thoracic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China
- Children's Heart Center, Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou 325000, China
| | - Wei-Yang Gao
- Department of Orthopaedic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou 325000, China
| | - Qi-Feng Zhao
- Department of Cardiovascular and Thoracic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China
- Children's Heart Center, Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China
| |
Collapse
|
13
|
Joshi H, Vastrad B, Joshi N, Vastrad C, Tengli A, Kotturshetti I. Identification of Key Pathways and Genes in Obesity Using Bioinformatics Analysis and Molecular Docking Studies. Front Endocrinol (Lausanne) 2021; 12:628907. [PMID: 34248836 PMCID: PMC8264660 DOI: 10.3389/fendo.2021.628907] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 05/19/2021] [Indexed: 01/01/2023] Open
Abstract
Obesity is an excess accumulation of body fat. Its progression rate has remained high in recent years. Therefore, the aim of this study was to diagnose important differentially expressed genes (DEGs) associated in its development, which may be used as novel biomarkers or potential therapeutic targets for obesity. The gene expression profile of E-MTAB-6728 was downloaded from the database. After screening DEGs in each ArrayExpress dataset, we further used the robust rank aggregation method to diagnose 876 significant DEGs including 438 up regulated and 438 down regulated genes. Functional enrichment analysis was performed. These DEGs were shown to be significantly enriched in different obesity related pathways and GO functions. Then protein-protein interaction network, target genes - miRNA regulatory network and target genes - TF regulatory network were constructed and analyzed. The module analysis was performed based on the whole PPI network. We finally filtered out STAT3, CORO1C, SERPINH1, MVP, ITGB5, PCM1, SIRT1, EEF1G, PTEN and RPS2 hub genes. Hub genes were validated by ICH analysis, receiver operating curve (ROC) analysis and RT-PCR. Finally a molecular docking study was performed to find small drug molecules. The robust DEGs linked with the development of obesity were screened through the expression profile, and integrated bioinformatics analysis was conducted. Our study provides reliable molecular biomarkers for screening and diagnosis, prognosis as well as novel therapeutic targets for obesity.
Collapse
Affiliation(s)
- Harish Joshi
- Department of Endocrinology, Endocrine and Diabetes Care Center, Hubbali, India
| | - Basavaraj Vastrad
- Department of Biochemistry, Basaveshwar College of Pharmacy, Gadag, India
| | - Nidhi Joshi
- Department of Medicine, Dr. D. Y. Patil Medical College, Kolhapur, India
| | - Chanabasayya Vastrad
- Biostatistics and Bioinformatics, Chanabasava Nilaya, Bharthinagar, Dharwad, India
- *Correspondence: Chanabasayya Vastrad,
| | - Anandkumar Tengli
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, Mysuru and JSS Academy of Higher Education & Research, Mysuru, India
| | - Iranna Kotturshetti
- Department of Ayurveda, Rajiv Gandhi Education Society`s Ayurvedic Medical College, Ron, India
| |
Collapse
|
14
|
Lv W, Jiang J, Li Y, Fu L, Meng F, Li J. MiR-302a-3p aggravates myocardial ischemia-reperfusion injury by suppressing mitophagy via targeting FOXO3. Exp Mol Pathol 2020; 117:104522. [PMID: 32866521 DOI: 10.1016/j.yexmp.2020.104522] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 08/22/2020] [Accepted: 08/26/2020] [Indexed: 10/23/2022]
Abstract
OBJECTIVE This study aimed to investigate whether the protection of miR-302a-3p in myocardial ischemia-reperfusion injury (MIRI) is mediated through the suppression of mitophagy. METHODS We constructed a mouse I/R model in vivo by the ligation of left anterior descending coronary artery for 45 min followed by 2 h reperfusion, and an in vitro model by treating mouse cardiomyocytes with hypoxia-reoxygenation (H/R). Knockdown experiments were then performed in vivo and in vitro to determine the effects of miR-302a-3p knockdown on the mitophagy, mitochondrial dysfunction and oxidative stress and apoptosis. The potential targets of miR-302a-3p were further studied by bioinformatics analysis, luciferase assays, quantitative real-time PCR and western blotting. RESULTS MiR-302a-3p expression was significantly upregulated in mice subjected to MIRI and in H/R-treated mouse cardiomyocytes. Functional analyses demonstrated that inhibition of miR-302a-3p protected cardiac tissues against I/R-induced apoptosis and mitophagy, mitochondrial damage and mitochondrial oxidative stress. Furthermore, FOXO3 was identified as the direct target of miR-302a-3p. Mechanistically, knockdown of FOXO3 partially reversed the cardioprotective effects of miR-302a-3p inhibitor. CONCLUSION Our study suggested that inhibition of miR-302a-3p promoted mitochondrial autophagy and inhibited oxidative stress by targeting FOXO3 to suppress myocardial apoptosis, representing a potential target for MIRI treatment.
Collapse
Affiliation(s)
- Wei Lv
- Department of Cardiology, Shengjing Hospital of China Medical University, China
| | - Jinping Jiang
- Department of Cardiology, Shengjing Hospital of China Medical University, China
| | - Yan Li
- Department of Biotherapy, Cancer Research Institute, The First affiliated hospital, China Medical University, China
| | - Liye Fu
- Department of Biotherapy, Cancer Research Institute, The First affiliated hospital, China Medical University, China
| | - Fandong Meng
- Department of Biotherapy, Cancer Research Institute, The First affiliated hospital, China Medical University, China
| | - Jun Li
- Department of Urology, The First affiliated hospital, China Medical University, China.
| |
Collapse
|
15
|
Abbasloo E, Najafipour H, Vakili A. Chronic treatment with apelin, losartan and their combination reduces myocardial infarct size and improves cardiac mechanical function. Clin Exp Pharmacol Physiol 2019; 47:393-402. [PMID: 31630435 DOI: 10.1111/1440-1681.13195] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 09/24/2019] [Accepted: 10/17/2019] [Indexed: 01/15/2023]
Abstract
The renin-angiotensin system (RAS) has a deleterious and apelin/APJ system has protective effect on the ischaemic heart. The collaboration between these systems in the pathophysiology of myocardial infarction is not clear. We determined the effect of chronic pretreatment with apelin, losartan and their combination on ischaemia-reperfusion (IR) injury in the isolated perfused rat heart and on the expression of apelin-13 receptor (APJ) and angiotensin type 1 receptor (AT1R) in the myocardium. During 5 days before the induction of IR, saline (vehicle), apelin-13 (Apl), F13A (apelin antagonist), losartan (Los, AT1R antagonist) and the combination of Apl and Los were administered intraperitoneally in rats. Ischaemia was induced by left anterior descending (LAD) artery occlusion for 30 minutes followed by reperfusion for 55 minutes in the Langendorff isolated heart perfusion system. Pretreatment with Apl, Los and the combination of Apl + Los significantly reduced infarct size by about 30, 33 and 48 percent respectively; and significantly improved the left ventricular function indices such as left ventricular developed pressure (LVDP), left ventricular end-diastolic pressure (LVEDP) and rate pressure product (RPP). IR increased AT1R protein level but it did not change APJ significantly. AT1R expression was reduced in groups treated with Apl, Los and Apl + Los. Findings showed that chronic pretreatment with apelin along with AT1R antagonist had more protective effects against IR injury. Combination therapy may diminish the risk of IR-induced heart damage, by reducing AT1R expression, in the heart of patients with coronary artery disease that are at the risk of MI and reperfusion injury.
Collapse
Affiliation(s)
- Elham Abbasloo
- Physiology, Endocrinology and Metabolism Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Hamid Najafipour
- Physiology, Cardiovascular Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Abedin Vakili
- Physiology Research Center, Semnan University of Medical Sciences, Semnan, Iran
| |
Collapse
|
16
|
Leme Goto P, Cinato M, Merachli F, Vons B, Jimenez T, Marsal D, Todua N, Loi H, Santin Y, Cassel S, Blanzat M, Tronchere H, Dejugnat C, Kunduzova O, Boal F. In vitro and in vivo cardioprotective and metabolic efficacy of vitamin E TPGS/Apelin. J Mol Cell Cardiol 2019; 138:165-174. [PMID: 31836542 DOI: 10.1016/j.yjmcc.2019.12.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 11/29/2019] [Accepted: 12/02/2019] [Indexed: 12/16/2022]
Abstract
AIMS Apelin and vitamin E have been proposed as signaling molecules, but their synergistic role is unknown. The aim of this work was to develop vitamin E TPGS/Apelin system to test their cardioprotective and metabolic efficacy in vitro and in vivo. METHODS FDA-approved surfactant D-α-tocopheryl polyethylene glycol 1000 succinate (TPGS-1000) and Apelin complex were characterized by physico-chemical methods (CMC determination, dynamic light scattering and circular dichroism). In vitro studies were carried out on H9C2 cardiomyoblasts and isolated murine cardiomyocytes. In vivo studies were performed in isoproterenol- and high-fat diet-induced cardiac remodeling models in mice. RESULTS We found that vitamin E TPGS/Apelin provide cardioprotective and metabolic efficacy in vitro and in vivo. In vitro studies revealed that vitamin E TPGS/Apelin reduces hypoxia-induced mitochondrial ROS production in cultured cardiomyocytes and H9C2 cardiomyoblasts. In addition, vitamin E TPGS/Apelin confers apoptotic response to hypoxic stress in cells. In a mouse model of isoproterenol-induced cardiac injury, TPGS is not able to affect cardiac remodeling, however combination of vitamin E TPGS and Apelin counteracts myocardial apoptosis, oxidative stress, hypertrophy and fibrosis. Furthermore, combination treatment attenuated obesity-induced cardiometabolic and fibrotic remodeling in mice. CONCLUSION Together, our data demonstrated the therapeutic benefits of vitamin E TPGS/Apelin complex to combat cardiovascular and metabolic disorders.
Collapse
Affiliation(s)
- Patricia Leme Goto
- Laboratoire des IMRCP, Université de Toulouse, CNRS UMR 5623, Université Toulouse III - Paul Sabatier, France
| | - Mathieu Cinato
- INSERM U1048 I2MC, Toulouse, France; Université Paul Sabatier, Toulouse, France
| | - Fadi Merachli
- Laboratoire des IMRCP, Université de Toulouse, CNRS UMR 5623, Université Toulouse III - Paul Sabatier, France
| | - Bohdana Vons
- Laboratoire des IMRCP, Université de Toulouse, CNRS UMR 5623, Université Toulouse III - Paul Sabatier, France
| | - Tony Jimenez
- Laboratoire des IMRCP, Université de Toulouse, CNRS UMR 5623, Université Toulouse III - Paul Sabatier, France
| | - Dimitri Marsal
- INSERM U1048 I2MC, Toulouse, France; Université Paul Sabatier, Toulouse, France
| | - Nika Todua
- INSERM U1048 I2MC, Toulouse, France; Université Paul Sabatier, Toulouse, France
| | - Halyna Loi
- INSERM U1048 I2MC, Toulouse, France; Université Paul Sabatier, Toulouse, France
| | - Yohan Santin
- INSERM U1048 I2MC, Toulouse, France; Université Paul Sabatier, Toulouse, France
| | - Stéphanie Cassel
- Laboratoire des IMRCP, Université de Toulouse, CNRS UMR 5623, Université Toulouse III - Paul Sabatier, France
| | - Muriel Blanzat
- Laboratoire des IMRCP, Université de Toulouse, CNRS UMR 5623, Université Toulouse III - Paul Sabatier, France
| | - Helene Tronchere
- INSERM U1048 I2MC, Toulouse, France; Université Paul Sabatier, Toulouse, France
| | - Christophe Dejugnat
- Laboratoire des IMRCP, Université de Toulouse, CNRS UMR 5623, Université Toulouse III - Paul Sabatier, France
| | - Oksana Kunduzova
- INSERM U1048 I2MC, Toulouse, France; Université Paul Sabatier, Toulouse, France
| | - Frederic Boal
- INSERM U1048 I2MC, Toulouse, France; Université Paul Sabatier, Toulouse, France.
| |
Collapse
|
17
|
Marsault E, Llorens-Cortes C, Iturrioz X, Chun HJ, Lesur O, Oudit GY, Auger-Messier M. The apelinergic system: a perspective on challenges and opportunities in cardiovascular and metabolic disorders. Ann N Y Acad Sci 2019; 1455:12-33. [PMID: 31236974 PMCID: PMC6834863 DOI: 10.1111/nyas.14123] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 04/11/2019] [Accepted: 05/02/2019] [Indexed: 12/11/2022]
Abstract
The apelinergic pathway has been generating increasing interest in the past few years for its potential as a therapeutic target in several conditions associated with the cardiovascular and metabolic systems. Indeed, preclinical and, more recently, clinical evidence both point to this G protein-coupled receptor as a target of interest in the treatment of not only cardiovascular disorders such as heart failure, pulmonary arterial hypertension, atherosclerosis, or septic shock, but also of additional conditions such as water retention/hyponatremic disorders, type 2 diabetes, and preeclampsia. While it is a peculiar system with its two classes of endogenous ligand, the apelins and Elabela, its intricacies are a matter of continuing investigation to finely pinpoint its potential and how it enables crosstalk between the vasculature and organ systems of interest. In this perspective article, we first review the current knowledge on the role of the apelinergic pathway in the above systems, as well as the associated therapeutic indications and existing pharmacological tools. We also offer a perspective on the challenges and potential ahead to advance the apelinergic system as a target for therapeutic intervention in several key areas.
Collapse
Affiliation(s)
- Eric Marsault
- Department of Pharmacology and Physiology, Institut de Pharmacologie de Sherbrooke, Centre de Recherche du CHUS, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Catherine Llorens-Cortes
- Collège de France, Center for Interdisciplinary Research in Biology, INSERM U1050, CNRS UMR7241, Paris, France
| | - Xavier Iturrioz
- Collège de France, Center for Interdisciplinary Research in Biology, INSERM U1050, CNRS UMR7241, Paris, France
| | - Hyung J. Chun
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Departments of Internal Medicine and Pathology, Yale University School of Medicine, New Haven, Connecticut
| | - Olivier Lesur
- Department of Pharmacology and Physiology, Institut de Pharmacologie de Sherbrooke, Centre de Recherche du CHUS, Université de Sherbrooke, Sherbrooke, Québec, Canada
- Department of Medicine – Division of Intensive Care Units, Centre de Recherche du CHUS, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Gavin Y. Oudit
- Department of Medicine, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada
| | - Mannix Auger-Messier
- Department of Pharmacology and Physiology, Institut de Pharmacologie de Sherbrooke, Centre de Recherche du CHUS, Université de Sherbrooke, Sherbrooke, Québec, Canada
- Department of Medicine – Division of Cardiology, Centre de Recherche du CHUS, Université de Sherbrooke, Sherbrooke, Québec, Canada
| |
Collapse
|
18
|
Read C, Nyimanu D, Williams TL, Huggins DJ, Sulentic P, Macrae RGC, Yang P, Glen RC, Maguire JJ, Davenport AP. International Union of Basic and Clinical Pharmacology. CVII. Structure and Pharmacology of the Apelin Receptor with a Recommendation that Elabela/Toddler Is a Second Endogenous Peptide Ligand. Pharmacol Rev 2019; 71:467-502. [PMID: 31492821 PMCID: PMC6731456 DOI: 10.1124/pr.119.017533] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The predicted protein encoded by the APJ gene discovered in 1993 was originally classified as a class A G protein-coupled orphan receptor but was subsequently paired with a novel peptide ligand, apelin-36 in 1998. Substantial research identified a family of shorter peptides activating the apelin receptor, including apelin-17, apelin-13, and [Pyr1]apelin-13, with the latter peptide predominating in human plasma and cardiovascular system. A range of pharmacological tools have been developed, including radiolabeled ligands, analogs with improved plasma stability, peptides, and small molecules including biased agonists and antagonists, leading to the recommendation that the APJ gene be renamed APLNR and encode the apelin receptor protein. Recently, a second endogenous ligand has been identified and called Elabela/Toddler, a 54-amino acid peptide originally identified in the genomes of fish and humans but misclassified as noncoding. This precursor is also able to be cleaved to shorter sequences (32, 21, and 11 amino acids), and all are able to activate the apelin receptor and are blocked by apelin receptor antagonists. This review summarizes the pharmacology of these ligands and the apelin receptor, highlights the emerging physiologic and pathophysiological roles in a number of diseases, and recommends that Elabela/Toddler is a second endogenous peptide ligand of the apelin receptor protein.
Collapse
Affiliation(s)
- Cai Read
- Experimental Medicine and Immunotherapeutics, University of Cambridge, Centre for Clinical Investigation, Addenbrooke's Hospital, Cambridge, United Kingdom (C.R., D.N., T.L.W., D.J.H., P.S., R.G.C.M., P.Y., J.J.M., A.P.D.); The Centre for Molecular Informatics, Department of Chemistry, University of Cambridge, Cambridge, United Kingdom (D.J.H., R.C.G.); and Computational and Systems Medicine, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, United Kingdom (R.C.G.)
| | - Duuamene Nyimanu
- Experimental Medicine and Immunotherapeutics, University of Cambridge, Centre for Clinical Investigation, Addenbrooke's Hospital, Cambridge, United Kingdom (C.R., D.N., T.L.W., D.J.H., P.S., R.G.C.M., P.Y., J.J.M., A.P.D.); The Centre for Molecular Informatics, Department of Chemistry, University of Cambridge, Cambridge, United Kingdom (D.J.H., R.C.G.); and Computational and Systems Medicine, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, United Kingdom (R.C.G.)
| | - Thomas L Williams
- Experimental Medicine and Immunotherapeutics, University of Cambridge, Centre for Clinical Investigation, Addenbrooke's Hospital, Cambridge, United Kingdom (C.R., D.N., T.L.W., D.J.H., P.S., R.G.C.M., P.Y., J.J.M., A.P.D.); The Centre for Molecular Informatics, Department of Chemistry, University of Cambridge, Cambridge, United Kingdom (D.J.H., R.C.G.); and Computational and Systems Medicine, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, United Kingdom (R.C.G.)
| | - David J Huggins
- Experimental Medicine and Immunotherapeutics, University of Cambridge, Centre for Clinical Investigation, Addenbrooke's Hospital, Cambridge, United Kingdom (C.R., D.N., T.L.W., D.J.H., P.S., R.G.C.M., P.Y., J.J.M., A.P.D.); The Centre for Molecular Informatics, Department of Chemistry, University of Cambridge, Cambridge, United Kingdom (D.J.H., R.C.G.); and Computational and Systems Medicine, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, United Kingdom (R.C.G.)
| | - Petra Sulentic
- Experimental Medicine and Immunotherapeutics, University of Cambridge, Centre for Clinical Investigation, Addenbrooke's Hospital, Cambridge, United Kingdom (C.R., D.N., T.L.W., D.J.H., P.S., R.G.C.M., P.Y., J.J.M., A.P.D.); The Centre for Molecular Informatics, Department of Chemistry, University of Cambridge, Cambridge, United Kingdom (D.J.H., R.C.G.); and Computational and Systems Medicine, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, United Kingdom (R.C.G.)
| | - Robyn G C Macrae
- Experimental Medicine and Immunotherapeutics, University of Cambridge, Centre for Clinical Investigation, Addenbrooke's Hospital, Cambridge, United Kingdom (C.R., D.N., T.L.W., D.J.H., P.S., R.G.C.M., P.Y., J.J.M., A.P.D.); The Centre for Molecular Informatics, Department of Chemistry, University of Cambridge, Cambridge, United Kingdom (D.J.H., R.C.G.); and Computational and Systems Medicine, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, United Kingdom (R.C.G.)
| | - Peiran Yang
- Experimental Medicine and Immunotherapeutics, University of Cambridge, Centre for Clinical Investigation, Addenbrooke's Hospital, Cambridge, United Kingdom (C.R., D.N., T.L.W., D.J.H., P.S., R.G.C.M., P.Y., J.J.M., A.P.D.); The Centre for Molecular Informatics, Department of Chemistry, University of Cambridge, Cambridge, United Kingdom (D.J.H., R.C.G.); and Computational and Systems Medicine, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, United Kingdom (R.C.G.)
| | - Robert C Glen
- Experimental Medicine and Immunotherapeutics, University of Cambridge, Centre for Clinical Investigation, Addenbrooke's Hospital, Cambridge, United Kingdom (C.R., D.N., T.L.W., D.J.H., P.S., R.G.C.M., P.Y., J.J.M., A.P.D.); The Centre for Molecular Informatics, Department of Chemistry, University of Cambridge, Cambridge, United Kingdom (D.J.H., R.C.G.); and Computational and Systems Medicine, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, United Kingdom (R.C.G.)
| | - Janet J Maguire
- Experimental Medicine and Immunotherapeutics, University of Cambridge, Centre for Clinical Investigation, Addenbrooke's Hospital, Cambridge, United Kingdom (C.R., D.N., T.L.W., D.J.H., P.S., R.G.C.M., P.Y., J.J.M., A.P.D.); The Centre for Molecular Informatics, Department of Chemistry, University of Cambridge, Cambridge, United Kingdom (D.J.H., R.C.G.); and Computational and Systems Medicine, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, United Kingdom (R.C.G.)
| | - Anthony P Davenport
- Experimental Medicine and Immunotherapeutics, University of Cambridge, Centre for Clinical Investigation, Addenbrooke's Hospital, Cambridge, United Kingdom (C.R., D.N., T.L.W., D.J.H., P.S., R.G.C.M., P.Y., J.J.M., A.P.D.); The Centre for Molecular Informatics, Department of Chemistry, University of Cambridge, Cambridge, United Kingdom (D.J.H., R.C.G.); and Computational and Systems Medicine, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, United Kingdom (R.C.G.)
| |
Collapse
|
19
|
Chang Z, Xia J, Wu H, Peng W, Jiang F, Li J, Liang C, Zhao H, Park K, Song G, Kim S, Huang R, Zheng L, Cai D, Qi X. Forkhead box O3 protects the heart against paraquat-induced aging-associated phenotypes by upregulating the expression of antioxidant enzymes. Aging Cell 2019; 18:e12990. [PMID: 31264342 PMCID: PMC6718552 DOI: 10.1111/acel.12990] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 05/10/2019] [Accepted: 05/23/2019] [Indexed: 11/30/2022] Open
Abstract
Paraquat (PQ) promotes cell senescence in brain tissue, which contributes to Parkinson's disease. Furthermore, PQ induces heart failure and oxidative damage, but it remains unknown whether and how PQ induces cardiac aging. Here, we demonstrate that PQ induces phenotypes associated with senescence of cardiomyocyte cell lines and results in cardiac aging‐associated phenotypes including cardiac remodeling and dysfunction in vivo. Moreover, PQ inhibits the activation of Forkhead box O3 (FoxO3), an important longevity factor, both in vitro and in vivo. We found that PQ‐induced senescence phenotypes, including proliferation inhibition, apoptosis, senescence‐associated β‐galactosidase activity, and p16INK4a expression, were significantly enhanced by FoxO3 deficiency in cardiomyocytes. Notably, PQ‐induced cardiac remolding, apoptosis, oxidative damage, and p16INK4a expression in hearts were exacerbated by FoxO3 deficiency. In addition, both in vitro deficiency and in vivo deficiency of FoxO3 greatly suppressed the activation of antioxidant enzymes including catalase (CAT) and superoxide dismutase 2 (SOD2) in the presence of PQ, which was accompanied by attenuation in cardiac function. The direct in vivo binding of FoxO3 to the promoters of the Cat and Sod2 genes in the heart was verified by chromatin immunoprecipitation (ChIP). Functionally, overexpression of Cat or Sod2 alleviated the PQ‐induced senescence phenotypes in FoxO3‐deficient cardiomyocyte cell lines. Overexpression of FoxO3 and CAT in hearts greatly suppressed the PQ‐induced heart injury and phenotypes associated with aging. Collectively, these results suggest that FoxO3 protects the heart against an aging‐associated decline in cardiac function in mice exposed to PQ, at least in part by upregulating the expression of antioxidant enzymes and suppressing oxidative stress.
Collapse
Affiliation(s)
- Zao‐Shang Chang
- Key Laboratory of Regenerative Medicine of Ministry of Education, Department of Developmental & Regenerative Biology Jinan University Guangzhou China
| | - Jing‐Bo Xia
- Key Laboratory of Regenerative Medicine of Ministry of Education, Department of Developmental & Regenerative Biology Jinan University Guangzhou China
| | - Hai‐Yan Wu
- Key Laboratory of Regenerative Medicine of Ministry of Education, Department of Developmental & Regenerative Biology Jinan University Guangzhou China
| | - Wen‐Tao Peng
- Key Laboratory of Regenerative Medicine of Ministry of Education, Department of Developmental & Regenerative Biology Jinan University Guangzhou China
| | - Fu‐Qing Jiang
- Key Laboratory of Regenerative Medicine of Ministry of Education, Department of Developmental & Regenerative Biology Jinan University Guangzhou China
| | - Jing Li
- Department of Surgery, Union Hospital, Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Chi‐Qian Liang
- Key Laboratory of Regenerative Medicine of Ministry of Education, Department of Developmental & Regenerative Biology Jinan University Guangzhou China
| | - Hui Zhao
- Key Laboratory of Regenerative Medicine of Ministry of Education, School of Biomedical Sciences, Faculty of Medicine The Chinese University of Hong Kong Hong Kong SAR China
| | - Kyu‐Sang Park
- Department of Physiology, Wonju College of Medicine Yonsei University Wonju Korea
| | - Guo‐Hua Song
- Institute of Atherosclerosis TaiShan Medical University Tai'an China
| | - Soo‐Ki Kim
- Department of Microbiology Wonju College of Medicine, Yonsei University Wonju Korea
| | - Ruijin Huang
- Institute of Anatomy, Department of Neuroanatomy, Medical Faculty Bonn Rheinische Friedrich-Wilhelms-University of Bonn Bonn Germany
| | - Li Zheng
- School of Environmental Science and Engineering Guangdong University of Technology Guangzhou China
| | - Dong‐Qing Cai
- Key Laboratory of Regenerative Medicine of Ministry of Education, Department of Developmental & Regenerative Biology Jinan University Guangzhou China
| | - Xu‐Feng Qi
- Key Laboratory of Regenerative Medicine of Ministry of Education, Department of Developmental & Regenerative Biology Jinan University Guangzhou China
| |
Collapse
|
20
|
Sawada N, Daimon M, Kawata T, Nakao T, Kimura K, Nakanishi K, Kurano M, Hirokawa M, Xu B, Yamanaka Y, Kato TS, Watanabe M, Yatomi Y, Komuro I. The Significance of the Effect of Visceral Adiposity on Left Ventricular Diastolic Function in the General Population. Sci Rep 2019; 9:4435. [PMID: 30872595 PMCID: PMC6418254 DOI: 10.1038/s41598-018-37137-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 11/27/2018] [Indexed: 02/08/2023] Open
Abstract
We evaluated the association between visceral adiposity and left ventricular (LV) diastolic function in association with plasma adiponectin levels in 213 subjects without overt cardiac diseases. Abdominal visceral fat area was quantified by computed tomography. Excessive visceral fat was significantly associated with impaired diastolic parameters including E/A, E′ and E/E′. Although serum adiponectin levels decreased with increased visceral adiposity, there was no independent association between serum adiponectin levels and diastolic parameters, which suggest that the role of adiponectin in this association might be indirect.
Collapse
Affiliation(s)
- Naoko Sawada
- Department of Cardiovascular Medicine, The University of Tokyo Hospital, Tokyo, Japan
| | - Masao Daimon
- Department of Cardiovascular Medicine, The University of Tokyo Hospital, Tokyo, Japan. .,Department of Clinical Laboratory, The University of Tokyo Hospital, Tokyo, Japan.
| | - Takayuki Kawata
- Department of Cardiovascular Medicine, The University of Tokyo Hospital, Tokyo, Japan
| | - Tomoko Nakao
- Department of Cardiovascular Medicine, The University of Tokyo Hospital, Tokyo, Japan.,Department of Clinical Laboratory, The University of Tokyo Hospital, Tokyo, Japan
| | - Koichi Kimura
- Department of Cardiovascular Medicine, The University of Tokyo Hospital, Tokyo, Japan
| | - Koki Nakanishi
- Department of Cardiovascular Medicine, The University of Tokyo Hospital, Tokyo, Japan
| | - Makoto Kurano
- Department of Clinical Laboratory, The University of Tokyo Hospital, Tokyo, Japan
| | - Megumi Hirokawa
- Department of Cardiovascular Medicine, The University of Tokyo Hospital, Tokyo, Japan
| | - Boqing Xu
- Department of Clinical Laboratory, The University of Tokyo Hospital, Tokyo, Japan
| | - Yuko Yamanaka
- Department of Cardiovascular Medicine, Jichi Medical University, Tochigi, Japan
| | - Tomoko S Kato
- Department of Cardiology, The Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Tokyo, Japan
| | - Masafumi Watanabe
- Department of Cardiovascular Medicine, The University of Tokyo Hospital, Tokyo, Japan
| | - Yutaka Yatomi
- Department of Clinical Laboratory, The University of Tokyo Hospital, Tokyo, Japan
| | - Issei Komuro
- Department of Cardiovascular Medicine, The University of Tokyo Hospital, Tokyo, Japan
| |
Collapse
|
21
|
Ding L, Gong C, Zhao J, Liu X, Li T, Rao S, Wang S, Liu Y, Peng S, Xiao W, Xiong C, Wang R, Liang S, Xu H. Noncoding transcribed ultraconserved region (T‐UCR) UC.48+ is a novel regulator of high‐fat diet induced myocardial ischemia/reperfusion injury. J Cell Physiol 2018; 234:9849-9861. [DOI: 10.1002/jcp.27674] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 10/02/2018] [Indexed: 12/11/2022]
Affiliation(s)
- Lu Ding
- Department of Physiology JiangXi Medical College of Nanchang University Nanchang Jiangxi China
- Clinical Department, JiangXi Medical College of Nanchang University Nanchang Jiangxi China
| | - Chengxin Gong
- Department of Science and Education Chest Hospital of Jiangxi Province Nanchang Jiangxi China
| | - Jiani Zhao
- Clinical Department, JiangXi Medical College of Nanchang University Nanchang Jiangxi China
| | - Xingzi Liu
- Clinical Department, JiangXi Medical College of Nanchang University Nanchang Jiangxi China
| | - Tao Li
- Clinical Department, JiangXi Medical College of Nanchang University Nanchang Jiangxi China
| | - Shenqiang Rao
- Department of Physiology JiangXi Medical College of Nanchang University Nanchang Jiangxi China
| | - Shuo Wang
- Clinical Department, JiangXi Medical College of Nanchang University Nanchang Jiangxi China
| | - Yuanyuan Liu
- Department of Physiology JiangXi Medical College of Nanchang University Nanchang Jiangxi China
| | - Shanping Peng
- Basic Medical Sciences, JiangXi Medical College of Nanchang University Nanchang Jiangxi China
| | - Wen Xiao
- Department of Pathology Jiangxi Medical College of Nanchang University Nanchang Jiangxi China
| | - Chaopeng Xiong
- Department of Nursing The Second Affliated Hospital of Nanchang University Nanchang Jiangxi China
| | - Rumeng Wang
- Department of Physiology JiangXi Medical College of Nanchang University Nanchang Jiangxi China
| | - Shangdong Liang
- Department of Physiology JiangXi Medical College of Nanchang University Nanchang Jiangxi China
| | - Hong Xu
- Department of Physiology JiangXi Medical College of Nanchang University Nanchang Jiangxi China
| |
Collapse
|
22
|
Wang W, Zhang D, Yang R, Xia W, Qian K, Shi Z, Brown R, Zhou H, Xi Y, Shi L, Chen L, Xu F, Sun X, Zhu D, Gong DW. Hepatic and cardiac beneficial effects of a long-acting Fc-apelin fusion protein in diet-induced obese mice. Diabetes Metab Res Rev 2018; 34:e2997. [PMID: 29577579 DOI: 10.1002/dmrr.2997] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 02/10/2018] [Accepted: 02/11/2018] [Indexed: 12/22/2022]
Abstract
BACKGROUND Apelin is a peptide ligand of the G-protein-coupled receptor APJ and exhibits anti-diabetes and anti-heart failure activities. However, short serum half-life of the apelin peptide limits its potential clinical applications. This study aimed to develop a long-acting apelin analog. METHODS To extend apelin's in vivo half-life, we made a recombinant protein by fusing the IgG Fc fragment to apelin-13 (Fc-apelin-13), conducted pharmacokinetics studies in mice, and determined in vitro biological activities in suppressing cyclic adenosine monophosphate and activating extracellular signal-regulated kinase signalling by reporter assays. We investigated the effects of Fc-apelin-13 on food intake, body weight, fasting blood glucose and insulin levels, glucose tolerance test, hepatic steatosis, and cardiac function and fibrosis by subcutaneous administration of Fc-apelin-13 in diet-induced obese mice for 4 weeks. RESULTS The estimated half-life of Fc-apelin-13 in blood was approximately 33 hours. Reporter assays showed that Fc-apelin-13 was active in suppressing cyclic adenosine monophosphate response element and activating serum response element activities. Four weeks of Fc-apelin-13 treatment in obese mice did not affect food intake and body weight, but resulted in a significant improvement of glucose tolerance, and a decrease in hepatic steatosis and fibrosis, as well as in serum alanine transaminase levels. Moreover, cardiac stroke volume and output were increased and cardiac fibrosis was decreased in the treated mice. CONCLUSIONS Fc-apelin-13 fusion protein has an extended in vivo half-life and exerts multiple benefits on obese mice with respect to the improvement of glucose disposal, amelioration of liver steatosis and heart fibrosis, and increase of cardiac output. Hence, Fc-apelin-13 is potentially a therapeutic for obesity-associated disease conditions.
Collapse
Affiliation(s)
- Weimin Wang
- Department of Endocrinology, Drum Tower Hospital of Nanjing Medical University, Nanjing, China
- Division of Endocrinology, Department of Medicine and Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Dongming Zhang
- Division of Endocrinology, Department of Medicine and Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Rongze Yang
- Division of Endocrinology, Department of Medicine and Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Wei Xia
- Division of Endocrinology, Department of Medicine and Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Kun Qian
- Division of Endocrinology, Department of Medicine and Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Zhengrong Shi
- Division of Endocrinology, Department of Medicine and Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Robert Brown
- Division of Endocrinology, Department of Medicine and Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Huifen Zhou
- Division of Endocrinology, Department of Medicine and Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Yue Xi
- Division of Endocrinology, Department of Medicine and Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Lin Shi
- Division of Endocrinology, Department of Medicine and Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Ling Chen
- Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Feng Xu
- Division of Endocrinology, Department of Medicine and Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Nephrology, The Second Hospital of Jilin University, Changchun, China
| | - Xiaojian Sun
- Division of Endocrinology, Department of Medicine and Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Dalong Zhu
- Department of Endocrinology, Drum Tower Hospital of Nanjing Medical University, Nanjing, China
| | - Da-Wei Gong
- Division of Endocrinology, Department of Medicine and Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
23
|
Timotin A, Cinato M, Boal F, Dejean S, Anesia R, Arnaut O, Lagente C, Roncalli J, Desmoulin F, Tronchere H, Kunduzova O. Differential protein profiling as a potential multi-marker approach for obese patients with heart failure: A retrospective study. Sci Rep 2018; 8:7894. [PMID: 29784904 PMCID: PMC5962559 DOI: 10.1038/s41598-018-26118-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 05/03/2018] [Indexed: 01/04/2023] Open
Abstract
Identification of novel circulating biomarkers predicting death and major cardio-metabolic events in obese patients with heart failure (HF) remains a research priority. In this study, we compared multi-marker profile of non-obese (NOB) and obese (OB) HF patients in relation to mortality outcome. The new multiplex proximity extension assay technology was used to analyze the levels of 92 proteins in plasma samples from HF patients according to body mass index (BMI) categories. At 2-year follow-up, all-cause mortality rates were significantly greater in NOB patients (BMI < 30 kg/m2) compared to the OB patients (BMI > 30 kg/m2) with HF (odds ratio 26; 95% CI: 1.14–624, p < 0,04). Quantitative proteomic analysis revealed thirteen distinct proteins expression profiles of OB and NOB HF patients. Among these proteins, RAGE, CXCL6, CXCL1, CD40, NEMO, VEGF-A, KLK6, PECAM1, PAR1, MMP1, BNP and NTproBNP were down-regulated, whereas leptin was up-regulated in OB HF patients. In addition, an inverse correlation between plasma BNP levels and leptin in OB HF patients was observed (r = −0.58 p = 0.02). This study identifies specific plasma protein signature in OB and NOB patients with HF in relation to mortality outcome.
Collapse
Affiliation(s)
- Andrei Timotin
- INSERM U1048, University of Toulouse, Toulouse, Cedex 4, 31432, France
| | - Mathieu Cinato
- INSERM U1048, University of Toulouse, Toulouse, Cedex 4, 31432, France
| | - Frederic Boal
- INSERM U1048, University of Toulouse, Toulouse, Cedex 4, 31432, France
| | - Sebastien Dejean
- Toulouse Mathematics Institute, University of Toulouse, Toulouse, Cedex 4, France
| | - Rodica Anesia
- INSERM U1048, University of Toulouse, Toulouse, Cedex 4, 31432, France
| | - Oleg Arnaut
- INSERM U1048, University of Toulouse, Toulouse, Cedex 4, 31432, France
| | - Christine Lagente
- Department of Cardiology, Toulouse University Hospital, Toulouse, Cedex 9, France
| | - Jerome Roncalli
- INSERM U1048, University of Toulouse, Toulouse, Cedex 4, 31432, France.,Department of Cardiology, Toulouse University Hospital, Toulouse, Cedex 9, France
| | - Franck Desmoulin
- Toulouse NeuroImaging Center, ToNIC, University of Toulouse, Inserm U1214, Toulouse, Cedex 3, France
| | - Helene Tronchere
- INSERM U1048, University of Toulouse, Toulouse, Cedex 4, 31432, France
| | - Oksana Kunduzova
- INSERM U1048, University of Toulouse, Toulouse, Cedex 4, 31432, France.
| |
Collapse
|
24
|
Over-expression of growth differentiation factor 15 (GDF15) preventing cold ischemia reperfusion (I/R) injury in heart transplantation through Foxo3a signaling. Oncotarget 2018; 8:36531-36544. [PMID: 28388574 PMCID: PMC5482674 DOI: 10.18632/oncotarget.16607] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 03/02/2017] [Indexed: 12/21/2022] Open
Abstract
Ischemia reperfusion (I/R) injury which inevitably occurs during heart transplantation is the major factor leading to organ failure and graft rejection. In order to develop new therapies to prevent I/R injury, we used both a murine heart transplantation model with 24 hour cold I/R and an in vitro cell culture system to determine whether growth differentiation factor 15 (GDF15) is a protective factor in preventing I/R injury in heart transplantation and to further investigate underlying mechanisms of I/R injury. We found that cold I/R caused severe damage to the endocardium, epicardium and myocardium of heart grafts from wild type C57BL/6 mice, whereas grafts from GDF15 transgenic (TG) mice showed less damage as demonstrated by decreased cell apoptosis/death, decreased neutrophils infiltration and the preservation of the normal structure of the heart. Over-expression of GDF15 reduced expression of phosphorylated RelA p65, pre-inflammatory and pro-apoptotic genes while it enhanced Foxo3a phosphorylation in vitro and in vivo. Over-expression of GDF15 inhibited cell apoptosis/death and reduced neutrophil infiltration. In conclusion, this study, for the first time, demonstrates that GDF15 is a promising target for preventing cold I/R injury in heart transplantation. This study also shows that the resultant protective effects are mediated by the Foxo3 and NFκB signaling pathways.
Collapse
|
25
|
Tronchere H, Cinato M, Timotin A, Guitou L, Villedieu C, Thibault H, Baetz D, Payrastre B, Valet P, Parini A, Kunduzova O, Boal F. Inhibition of PIKfyve prevents myocardial apoptosis and hypertrophy through activation of SIRT3 in obese mice. EMBO Mol Med 2018; 9:770-785. [PMID: 28396567 PMCID: PMC5452048 DOI: 10.15252/emmm.201607096] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
PIKfyve is an evolutionarily conserved lipid kinase that regulates pleiotropic cellular functions. Here, we identify PIKfyve as a key regulator of cardiometabolic status and mitochondrial integrity in chronic diet‐induced obesity. In vitro, we show that PIKfyve is critical for the control of mitochondrial fragmentation and hypertrophic and apoptotic responses to stress. We also provide evidence that inactivation of PIKfyve by the selective inhibitor STA suppresses excessive mitochondrial ROS production and apoptosis through a SIRT3‐dependent pathway in cardiomyoblasts. In addition, we report that chronic STA treatment improves cardiometabolic profile in a mouse model of cardiomyopathy linked to obesity. We provide evidence that PIKfyve inhibition reverses obesity‐induced cardiac mitochondrial damage and apoptosis by activating SIRT3. Furthermore, treatment of obese mice with STA improves left ventricular function and attenuates cardiac hypertrophy. In contrast, STA is not able to reduce isoproterenol‐induced cardiac hypertrophy in SIRT3.KO mice. Altogether, these results unravel a novel role for PIKfyve in obesity‐associated cardiomyopathy and provide a promising therapeutic strategy to combat cardiometabolic complications in obesity.
Collapse
Affiliation(s)
- Helene Tronchere
- INSERM U1048 I2MC, Toulouse, Cedex 4, France.,Université Paul Sabatier, Toulouse, France
| | - Mathieu Cinato
- INSERM U1048 I2MC, Toulouse, Cedex 4, France.,Université Paul Sabatier, Toulouse, France
| | - Andrei Timotin
- INSERM U1048 I2MC, Toulouse, Cedex 4, France.,Université Paul Sabatier, Toulouse, France
| | - Laurie Guitou
- INSERM U1048 I2MC, Toulouse, Cedex 4, France.,Université Paul Sabatier, Toulouse, France
| | - Camille Villedieu
- CarMeN Laboratory, Inserm U1060, Univ-Lyon, Université Claude Bernard Lyon 1, Bron, France
| | - Helene Thibault
- CarMeN Laboratory, Inserm U1060, Univ-Lyon, Université Claude Bernard Lyon 1, Bron, France
| | - Delphine Baetz
- CarMeN Laboratory, Inserm U1060, Univ-Lyon, Université Claude Bernard Lyon 1, Bron, France
| | - Bernard Payrastre
- INSERM U1048 I2MC, Toulouse, Cedex 4, France.,Université Paul Sabatier, Toulouse, France
| | - Philippe Valet
- INSERM U1048 I2MC, Toulouse, Cedex 4, France.,Université Paul Sabatier, Toulouse, France
| | - Angelo Parini
- INSERM U1048 I2MC, Toulouse, Cedex 4, France.,Université Paul Sabatier, Toulouse, France
| | - Oksana Kunduzova
- INSERM U1048 I2MC, Toulouse, Cedex 4, France.,Université Paul Sabatier, Toulouse, France
| | - Frederic Boal
- INSERM U1048 I2MC, Toulouse, Cedex 4, France .,Université Paul Sabatier, Toulouse, France
| |
Collapse
|
26
|
Abstract
Apelin and apela (ELABELA/ELA/Toddler) are two peptide ligands for a class A G-protein-coupled receptor named the apelin receptor (AR/APJ/APLNR). Ligand-AR interactions have been implicated in regulation of the adipoinsular axis, cardiovascular system, and central nervous system alongside pathological processes. Each ligand may be processed into a variety of bioactive isoforms endogenously, with apelin ranging from 13 to 55 amino acids and apela from 11 to 32, typically being cleaved C-terminal to dibasic proprotein convertase cleavage sites. The C-terminal region of the respective precursor protein is retained and is responsible for receptor binding and subsequent activation. Interestingly, both apelin and apela exhibit isoform-dependent variability in potency and efficacy under various physiological and pathological conditions, but most studies focus on a single isoform. Biophysical behavior and structural properties of apelin and apela isoforms show strong correlations with functional studies, with key motifs now well determined for apelin. Unlike its ligands, the AR has been relatively difficult to characterize by biophysical techniques, with most characterization to date being focused on effects of mutagenesis. This situation may improve following a recently reported AR crystal structure, but there are still barriers to overcome in terms of comprehensive biophysical study. In this review, we summarize the three components of the apelinergic system in terms of structure-function correlation, with a particular focus on isoform-dependent properties, underlining the potential for regulation of the system through multiple endogenous ligands and isoforms, isoform-dependent pharmacological properties, and biological membrane-mediated receptor interaction. © 2018 American Physiological Society. Compr Physiol 8:407-450, 2018.
Collapse
Affiliation(s)
- Kyungsoo Shin
- Department of Biochemistry & Molecular Biology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Calem Kenward
- Department of Biochemistry & Molecular Biology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Jan K Rainey
- Department of Biochemistry & Molecular Biology, Dalhousie University, Halifax, Nova Scotia, Canada
- Department of Chemistry, Dalhousie University, Halifax, Nova Scotia, Canada
| |
Collapse
|
27
|
Renaud-Gabardos E, Tatin F, Hantelys F, Lebas B, Calise D, Kunduzova O, Masri B, Pujol F, Sicard P, Valet P, Roncalli J, Chaufour X, Garmy-Susini B, Parini A, Prats AC. Therapeutic Benefit and Gene Network Regulation by Combined Gene Transfer of Apelin, FGF2, and SERCA2a into Ischemic Heart. Mol Ther 2017; 26:902-916. [PMID: 29249393 DOI: 10.1016/j.ymthe.2017.11.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 11/06/2017] [Accepted: 11/10/2017] [Indexed: 01/16/2023] Open
Abstract
Despite considerable advances in cardiovascular disease treatment, heart failure remains a public health challenge. In this context, gene therapy appears as an attractive approach, but clinical trials using single therapeutic molecules result in moderate benefit. With the objective of improving ischemic heart failure therapy, we designed a combined treatment, aimed to simultaneously stimulate angiogenesis, prevent cardiac remodeling, and restore contractile function. We have previously validated IRES-based vectors as powerful tools to co-express genes of interest. Mono- and multicistronic lentivectors expressing fibroblast growth factor 2 (angiogenesis), apelin (cardioprotection), and/or SERCA2a (contractile function) were produced and administrated by intramyocardial injection into a mouse model of myocardial infarction. Data reveal that combined treatment simultaneously improves vessel number, heart function parameters, and fibrosis prevention, due to FGF2, SERCA2a, and apelin, respectively. Furthermore, addition of SERCA2a in the combination decreases cardiomyocyte hypertrophy. Large-scale transcriptome analysis reveals that the triple treatment is the most efficient in restoring angiogenic balance as well as expression of genes involved in cardiac function and remodeling. Our study validates the concept of combined treatment of ischemic heart disease with apelin, FGF2, and SERCA2a and shows that such therapeutic benefit is mediated by a more effective recovery of gene network regulation.
Collapse
Affiliation(s)
| | - Florence Tatin
- UMR 1048-I2MC, Université de Toulouse, INSERM, FHU IMPACT, 31432 Toulouse, France
| | - Fransky Hantelys
- UMR 1048-I2MC, Université de Toulouse, INSERM, FHU IMPACT, 31432 Toulouse, France
| | - Benoît Lebas
- UMR 1048-I2MC, Université de Toulouse, INSERM, FHU IMPACT, 31432 Toulouse, France; Centre Hospitalier Universitaire de Toulouse, 31059 Toulouse, France
| | - Denis Calise
- UMS 006, Université de Toulouse, INSERM, 31432 Toulouse, France
| | - Oksana Kunduzova
- UMR 1048-I2MC, Université de Toulouse, INSERM, FHU IMPACT, 31432 Toulouse, France
| | - Bernard Masri
- UMR 1048-I2MC, Université de Toulouse, INSERM, FHU IMPACT, 31432 Toulouse, France
| | - Françoise Pujol
- UMR 1048-I2MC, Université de Toulouse, INSERM, FHU IMPACT, 31432 Toulouse, France
| | - Pierre Sicard
- UMR 1048-I2MC, Université de Toulouse, INSERM, FHU IMPACT, 31432 Toulouse, France
| | - Philippe Valet
- UMR 1048-I2MC, Université de Toulouse, INSERM, FHU IMPACT, 31432 Toulouse, France
| | - Jérôme Roncalli
- UMR 1048-I2MC, Université de Toulouse, INSERM, FHU IMPACT, 31432 Toulouse, France; Centre Hospitalier Universitaire de Toulouse, 31059 Toulouse, France
| | - Xavier Chaufour
- UMR 1048-I2MC, Université de Toulouse, INSERM, FHU IMPACT, 31432 Toulouse, France; Centre Hospitalier Universitaire de Toulouse, 31059 Toulouse, France
| | - Barbara Garmy-Susini
- UMR 1048-I2MC, Université de Toulouse, INSERM, FHU IMPACT, 31432 Toulouse, France
| | - Angelo Parini
- UMR 1048-I2MC, Université de Toulouse, INSERM, FHU IMPACT, 31432 Toulouse, France
| | - Anne-Catherine Prats
- UMR 1048-I2MC, Université de Toulouse, INSERM, FHU IMPACT, 31432 Toulouse, France.
| |
Collapse
|
28
|
Yang L, Zheng T, Wu H, Xin W, Mou X, Lin H, Chen Y, Wu X. Predictive value of apelin-12 in patients with ST-elevation myocardial infarction with different renal function: a prospective observational study. BMJ Open 2017; 7:e018595. [PMID: 29150476 PMCID: PMC5701982 DOI: 10.1136/bmjopen-2017-018595] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
OBJECTIVES To investigate factors predicting the onset of major adverse cardiovascular events (MACEs) after primary percutaneous coronary intervention (pPCI) for patients with ST-segment elevation myocardial infarction (STEMI) . BACKGROUND Apelin-12 plays an essential role in cardiovascular homoeostasis. However, current knowledge of its predictive prognostic value is limited. METHODS 464 patients with STEMI (63.0±11.9 years, 355 men) who underwent successful pPCI were enrolled and followed for 2.5 years. Multivariate cox regression analysis and receiver operating characteristic (ROC) curve analysis were performed to determine the factors predicting MACEs. RESULTS 118 patients (25.4%) experienced MACEs in the follow-up period. Multivariate cox regression analysis found low apelin-12 (HR=0.132, 95% CI 0.060 to 0.292, P<0.001), low left ventricular ejection fraction (HR=0.965, 95% CI 0.941 to 0.991, P=0.007), low estimated glomerular filtration rate (eGFR) (HR=0.985, 95% CI 0.977 to 0.993, P<0.001), Killip's classification>I (HR=0.610, 95% CI 0.408 to 0.912, P=0.016) and pathological Q-wave (HR=1.536, 95% CI 1.058 to 2.230, P=0.024) were independent predictors of MACEs in the 2.5 year follow-up period. Low apelin-12 also predicted poorer in-hospital prognosis and MACEs in the 2.5 years follow-up period compared with Δapelin-12 (P=0.0115) and eGFR (P=0.0071) among patients with eGFR>90 mL/min×1.73 m2. Further analysis showed Δapelin-12 <20% was associated with MACEs in patients whose apelin-12 was below 0.76 ng/mL (P=0.0075) on admission. CONCLUSIONS Patients with STEMI receiving pPCI with lower apelin-12 are more likely to suffer MACEs in hospital and 2.5 years postprocedure, particularly in those with normal eGFR levels.
Collapse
Affiliation(s)
- Lingchang Yang
- Department of Emergency Medicine, The First People’s Hospital of Taizhou, Taizhou, Zhejiang, China
| | - Ting Zheng
- Department of Emergency Medicine, The First People’s Hospital of Taizhou, Taizhou, Zhejiang, China
| | - Haopeng Wu
- Department of Emergency Medicine, The First People’s Hospital of Taizhou, Taizhou, Zhejiang, China
| | - Wenwei Xin
- Department of Emergency Medicine, The First People’s Hospital of Taizhou, Taizhou, Zhejiang, China
| | - Xiongneng Mou
- Department of Emergency Medicine, The First People’s Hospital of Taizhou, Taizhou, Zhejiang, China
| | - Hui Lin
- Department of Emergency Medicine, The First People’s Hospital of Taizhou, Taizhou, Zhejiang, China
| | - Yide Chen
- Department of Emergency Medicine, The First People’s Hospital of Taizhou, Taizhou, Zhejiang, China
| | - Xiaoyu Wu
- Department of Emergency Medicine, The First People’s Hospital of Taizhou, Taizhou, Zhejiang, China
| |
Collapse
|
29
|
Zhang W, Zhang DG, Liang X, Zhang WL, Ma JX. Effects of apelin on retinal microglial cells in a rat model of oxygen-induced retinopathy of prematurity. J Cell Biochem 2017; 119:2900-2910. [PMID: 29091306 DOI: 10.1002/jcb.26473] [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: 06/11/2017] [Accepted: 10/24/2017] [Indexed: 12/19/2022]
Abstract
This study explores the effects of apelin on retinal microglial cells in rat models of oxygen-induced retinopathy of prematurity (ROP). Totally, 274 rats were selected for establishing oxygen-induced retinopathy (OIR) models, and 92 healthy rats for control group. OIR rats were assigned into OIR, 10-5 g/L apelin, 10-4 g/L apelin, and 10-3 g/L apelin groups. Immunohistochemistry was employed to determine morphology of microglial cells and cell number. CDllb, ionized calcium-binding adapter molecule 1 (IBA-1), TNF-α, and iNOS mRNA and protein expressions were identified using RT-qPCR and Western blotting, respectively. ELISA was employed to determine the levels of VEGF and glial fibrillary acidic protein (GFAP). The amoeboid microglial cells were found in the OIR and 10-3 g/L apelin groups, while bipolar microglial cells were found in the normal control, 10-5 g/L apelin and 10-4 g/L apelin groups. In the 1, 2, 3, and 4th week after apelin treatment, there were significantly decreased bipolar microglial cells, lower mRNA and protein expressions of CDllb, IBA-1, TNF-α and iNOS, and the levels of VEGF and GFAP in the 10-4 g/L apelin group than in the OIR, 10-3 g/L apelin and 10-5 g/L apelin groups. The differences between the normal control and 10-4 g/L apelin groups are not significant. Compared with the OIR group, the 10-5 g/L apelin and 10-3 g/L apelin groups presented decreased microglial cells and mRNA and protein expressions of CDllb, IBA-1, TNF-α, and iNOS. Appropriate concentration of apelin may reduce retinal microglial cells in a rat model of oxygen-induced ROP.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Ophthalmology, the Second Hospital of Hebei Medical University, Shijiazhuang, P.R. China.,Department of Ophthalmology, Shanxi Dayi Hospital, Taiyuan, P.R. China
| | - Ding-Guo Zhang
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, P.R. China
| | - Xing Liang
- Department of Ophthalmology, Shanxi Dayi Hospital, Taiyuan, P.R. China
| | - Wei-Liang Zhang
- Department of Ophthalmology, Shanxi Dayi Hospital, Taiyuan, P.R. China
| | - Jing-Xue Ma
- Department of Ophthalmology, the Second Hospital of Hebei Medical University, Shijiazhuang, P.R. China
| |
Collapse
|
30
|
Zarrinkalam E, Heidarianpour A, Department of exercise physiology, faculty of physical education and sport science, Bu-Ali Sina University, Hamedan, Iran, Department of exercise physiology, faculty of physical education and sport science, Bu-Ali Sina University, Hamedan, Iran. Effect of 8-Week Aerobic, Strength and Concurrent Training on Circulating Apelin in Morphine-Dependent Rats. MEDICAL LABORATORY JOURNAL 2017. [DOI: 10.29252/mlj.11.5.27] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
|
31
|
Leung HW, Foo G, Banumurthy G, Chai X, Ghosh S, Mitra-Ganguli T, VanDongen AMJ. The effect of Bacopa monnieri on gene expression levels in SH-SY5Y human neuroblastoma cells. PLoS One 2017; 12:e0182984. [PMID: 28832626 PMCID: PMC5568221 DOI: 10.1371/journal.pone.0182984] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Accepted: 07/27/2017] [Indexed: 12/22/2022] Open
Abstract
Bacopa monnieri is a plant used as a nootropic in Ayurveda, a 5000-year-old system of traditional Indian medicine. Although both animal and clinical studies supported its role as a memory enhancer, the molecular and cellular mechanism underlying Bacopa's nootropic action are not understood. In this study, we used deep sequencing (RNA-Seq) to identify the transcriptome changes upon Bacopa treatment on SH-SY5Y human neuroblastoma cells. We identified several genes whose expression levels were regulated by Bacopa. Biostatistical analysis of the RNA-Seq data identified biological pathways and molecular functions that were regulated by Bacopa, including regulation of mRNA translation and transmembrane transport, responses to oxidative stress and protein misfolding. Pathway analysis using the Ingenuity platform suggested that Bacopa may protect against brain damage and improve brain development. These newly identified molecular and cellular determinants may contribute to the nootropic action of Bacopa and open up a new direction of investigation into its mechanism of action.
Collapse
Affiliation(s)
- How-Wing Leung
- Program in Neuroscience and Behavioral Disorders, Duke-NUS Medical School, Singapore, Singapore
| | - Gabriel Foo
- Program in Neuroscience and Behavioral Disorders, Duke-NUS Medical School, Singapore, Singapore
| | | | - Xiaoran Chai
- Program in Neuroscience and Behavioral Disorders, Duke-NUS Medical School, Singapore, Singapore
| | - Sujoy Ghosh
- Program in Neuroscience and Behavioral Disorders, Duke-NUS Medical School, Singapore, Singapore
| | | | - Antonius M J VanDongen
- Program in Neuroscience and Behavioral Disorders, Duke-NUS Medical School, Singapore, Singapore
| |
Collapse
|
32
|
Targeting the apelin pathway as a novel therapeutic approach for cardiovascular diseases. Biochim Biophys Acta Mol Basis Dis 2017; 1863:1942-1950. [DOI: 10.1016/j.bbadis.2016.11.007] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 10/14/2016] [Accepted: 11/01/2016] [Indexed: 01/01/2023]
|
33
|
Association of apelin and apelin receptor with the risk of coronary artery disease: a meta-analysis of observational studies. Oncotarget 2017; 8:57345-57355. [PMID: 28915675 PMCID: PMC5593646 DOI: 10.18632/oncotarget.17360] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 04/04/2017] [Indexed: 12/16/2022] Open
Abstract
It is well established that apelin-APLNR (apelin receptor) pathway plays a central role in cardiovascular system. In this meta-analysis, we summarized published results on circulating apelin concentration in association with coronary artery disease (CAD), apelin and APLNR genetic polymorphism(s) in predisposition to CAD risk and circulating apelin changes after surgical treatment for CAD. The results from 15 articles were pooled. Two authors independently took charge of literature search, article selection and information collection. Overall, circulating apelin concentration was significantly lower in CAD patients (N=1021) than in controls (N=654) (weighted mean difference [WMD]: -1.285 ng/mL, 95% confidence interval [CI]: -1.790 to -0.780, P<0001), with significant heterogeneity (I2=99.3%) but without publication bias. For the association of APLNR gene rs9943582 polymorphism with CAD (patients/controls: 5975/4717), the mutant T allele was associated with a 5.2% increased risk relative to the wild C allele (odds ratio: 1.052, 95% CI: 0.990 to 1.117, P=0.100), without heterogeneity (I2=0.0%) or publication bias. Circulating apelin was increased significantly after surgical treatment for CAD (N=202) (WMD: 2.011 ng/mL, 95% CI: 0.541 to 3.481, P=0.007), with significant heterogeneity (I2=98.0%). Stratified analyses showed that circulating apelin was significantly reduced in studies with age- and sex-matched patients and controls (WMD: -1.881 ng/mL, 95% CI: -2.457 to -1.304, P<0.001) and with total sample size ≥125 (WMD: -1.657 ng/mL, 95% CI: -2.378 to -0.936, P<0.001), relative to studies without matching reports and with total sample size <125. In brief, our results suggested that circulating apelin was a prominent athero-protective marker against the development of CAD.
Collapse
|
34
|
Topuz M, Oz F, Akkus O, Sen O, Topuz AN, Bulut A, Ozbicer S, Okar S, Koc M, Gur M. Plasma apelin-12 levels may predict in-hospital major adverse cardiac events in ST-elevation myocardial infarction and the relationship between apelin-12 and the neutrophil/lymphocyte ratio in patients undergoing primary coronary intervention. Perfusion 2016; 32:206-213. [PMID: 27770057 DOI: 10.1177/0267659116676335] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
OBJECTIVE We aimed to investigate the compliance of plasma apelin-12 levels to show angiographic properties and hospital MACE in patients with ST-elevation myocardial infarction (STEMI) undergoing primary percutaneous coronary intervention (PCI). MATERIAL AND METHODS The association of apelin-12 levels with the N/L ratio on admission was assessed in 170 consecutive patients with primary STEMI undergoing primary PCI. All patient SYNTAX scores and thrombolysis in myocardial infarction (TIMI) flow grades were also assessed. Patients were divided into two groups according to their TIMI flow grade. Patients with a TIMI 0-2 flow and TIMI 3 flow with grade 0/1 myocardial blush grade (MBG) score were defined as the no-reflow group and patients with TIMI grade 3 flow with ⩾2 MBG were considered as the normal flow group. RESULTS Baseline apelin-12 levels were significantly lower in the no-reflow group than in the normal flow group (3.3±1.81 vs 6.2±1.74, p<0.001). In-hospital events, including death, myocardial infarction (MI) and re-infarction were significantly higher in patients in the no-reflow group than normal flow group (23% vs 7%, p<0.001). Apelin-12 level was negative correlated with the N/L ratio (r= -0.352, p<0.001), Hs-Crp (r=-0.272, p=0.01) and SYNTAX score (r= -0.246, p=0.029). In the multivariate regression analysis, apelin-12, presence of no-reflow and the SYNTAX score were independent predictors of in-hospital MACE (odds ratio [OR] 1.41, 95% confidence interval (CI) [1.27 to 1.67], p=0.001 for apelin-12, OR 1.085, [0.981 to 1.203], p<0.001 for no-reflow and OR 0.201, 95% CI [0.05 to 0.47], p= 0.004 for SYNTAX score). CONCLUSION We have shown that lower apelin-12 level on admission is associated with higher SYNTAX scores and no-reflow phenomenon and may be used as a prognostic marker for hospital MACE in patients with STEMI.
Collapse
Affiliation(s)
- Mustafa Topuz
- 1 Cardiology, Adana Numune Education and Research Hospital, Adana, Turkey
| | - Fahrettin Oz
- 1 Cardiology, Adana Numune Education and Research Hospital, Adana, Turkey
| | - Oguz Akkus
- 1 Cardiology, Adana Numune Education and Research Hospital, Adana, Turkey
| | - Omer Sen
- 1 Cardiology, Adana Numune Education and Research Hospital, Adana, Turkey
| | - Ayse Nur Topuz
- 2 Family Medicine, Cukurova Universty Medicine Faculty, Adana, Turkey
| | - Atilla Bulut
- 1 Cardiology, Adana Numune Education and Research Hospital, Adana, Turkey
| | - Suleyman Ozbicer
- 1 Cardiology, Adana Numune Education and Research Hospital, Adana, Turkey
| | - Sefa Okar
- 1 Cardiology, Adana Numune Education and Research Hospital, Adana, Turkey
| | - Mevlut Koc
- 1 Cardiology, Adana Numune Education and Research Hospital, Adana, Turkey
| | - Mustafa Gur
- 1 Cardiology, Adana Numune Education and Research Hospital, Adana, Turkey
| |
Collapse
|