1
|
Cristallini C, Rossin D, Vanni R, Barbani N, Bulgheresi C, Labardi M, Perveen S, Burchielli S, Terlizzi D, Kusmic C, Del Ry S, Cabiati M, Trouki C, Rossino D, Sergi F, Villano A, Aquaro GD, Scarpellino G, Ruffinatti FA, Amorim S, Pires RA, Reis RL, Rastaldo R, Giachino C. A biodegradable, microstructured, electroconductive and nano-integrated drug eluting patch (MENDEP) for myocardial tissue engineering. Bioact Mater 2025; 50:246-272. [PMID: 40270551 PMCID: PMC12017858 DOI: 10.1016/j.bioactmat.2025.04.008] [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: 12/04/2024] [Revised: 03/24/2025] [Accepted: 04/05/2025] [Indexed: 04/25/2025] Open
Abstract
We produced a microstructured, electroconductive and nano-functionalized drug eluting cardiac patch (MENDEP) designed to attract endogenous precursor cells, favor their differentiation and counteract adverse ventricular remodeling in situ. MENDEP showed mechanical anisotropy and biaxial strength comparable to porcine myocardium, reduced impedance, controlled biodegradability, molecular recognition ability and controlled drug release activity. In vitro, cytocompatibility and cardioinductivity were demonstrated. Migration tests showed the chemoattractive capacity of the patches and conductivity assays showed unaltered cell-cell interactions and cell beating synchronicity. MENDEP was then epicardially implanted in a rat model of ischemia/reperfusion (I/R). Histological, immunofluorescence and biomarker analysis indicated that implantation did not cause damage to the healthy myocardium. After I/R, MENDEP recruited precursor cells into the damaged myocardium and triggered their differentiation towards the vascular lineage. Under the patch, the myocardial tissue appeared well preserved and cardiac gap junctions were correctly distributed at the level of the intercalated discs. The fibrotic area measured in the I/R group was partially reduced in the patch group. Overall, these results demonstrate that MENDEP was fully retained on the epicardial surface of the left ventricle over 4-week implantation period, underwent progressive vascularization, did not perturb the healthy myocardium and showed great potential in repairing the infarcted area.
Collapse
Affiliation(s)
- Caterina Cristallini
- Institute for Chemical and Physical Processes, CNR-IPCF, Via Giuseppe Moruzzi 1, 56124, Pisa, Italy
- Department of Civil and Industrial Engineering, DICI, University of Pisa, Largo Lucio Lazzarino, 56126, Pisa, Italy
| | - Daniela Rossin
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, 10043, Orbassano, Italy
| | - Roberto Vanni
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, 10043, Orbassano, Italy
| | - Niccoletta Barbani
- Institute for Chemical and Physical Processes, CNR-IPCF, Via Giuseppe Moruzzi 1, 56124, Pisa, Italy
- Department of Civil and Industrial Engineering, DICI, University of Pisa, Largo Lucio Lazzarino, 56126, Pisa, Italy
| | - Chiara Bulgheresi
- Department of Civil and Industrial Engineering, DICI, University of Pisa, Largo Lucio Lazzarino, 56126, Pisa, Italy
| | - Massimiliano Labardi
- Institute for Chemical and Physical Processes, CNR-IPCF, Via Giuseppe Moruzzi 1, 56124, Pisa, Italy
| | - Sadia Perveen
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, 10043, Orbassano, Italy
| | | | | | - Claudia Kusmic
- Clinical Physiology Institute, CNR-IFC, Via Giuseppe Moruzzi 1, 56124, Pisa, Italy
| | - Silvia Del Ry
- Clinical Physiology Institute, CNR-IFC, Via Giuseppe Moruzzi 1, 56124, Pisa, Italy
| | - Manuela Cabiati
- Clinical Physiology Institute, CNR-IFC, Via Giuseppe Moruzzi 1, 56124, Pisa, Italy
| | - Cheherazade Trouki
- Institute for Chemical and Physical Processes, CNR-IPCF, Via Giuseppe Moruzzi 1, 56124, Pisa, Italy
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126, Pisa, Italy
| | - Dawid Rossino
- Institute for Chemical and Physical Processes, CNR-IPCF, Via Giuseppe Moruzzi 1, 56124, Pisa, Italy
- Department of Civil and Industrial Engineering, DICI, University of Pisa, Largo Lucio Lazzarino, 56126, Pisa, Italy
| | - Francesca Sergi
- Department of Civil and Industrial Engineering, DICI, University of Pisa, Largo Lucio Lazzarino, 56126, Pisa, Italy
| | - Anthea Villano
- Institute for Chemical and Physical Processes, CNR-IPCF, Via Giuseppe Moruzzi 1, 56124, Pisa, Italy
| | - Giovanni D. Aquaro
- Academic Radiology Unit, Department of Surgical, Medical and Molecular Pathology and of the Critical Area, University of Pisa, Via Paradisa 2, 56124, Pisa, Italy
| | - Giorgia Scarpellino
- Department of Life Sciences and Systems Biology, University of Turin, Via Accademia Albertina 13, 10123, Torino, Italy
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, Via Adolfo Ferrata 9, 27100, Pavia, Italy
| | - Federico A. Ruffinatti
- Department of Life Sciences and Systems Biology, University of Turin, Via Accademia Albertina 13, 10123, Torino, Italy
| | - Sara Amorim
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017, Barco, Guimarães, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Ricardo A. Pires
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017, Barco, Guimarães, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Rui L. Reis
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017, Barco, Guimarães, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Raffaella Rastaldo
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, 10043, Orbassano, Italy
| | - Claudia Giachino
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, 10043, Orbassano, Italy
| |
Collapse
|
2
|
Qin Y, Li M, Liu H. Regulatory T cells: a promising new therapeutic target in ventricular remodeling after myocardial infarction. Front Immunol 2025; 16:1514335. [PMID: 40260235 PMCID: PMC12009920 DOI: 10.3389/fimmu.2025.1514335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Accepted: 03/24/2025] [Indexed: 04/23/2025] Open
Abstract
Myocardial infarction (MI) is one of the leading causes of death worldwide. It is triggered by thrombosis or vascular occlusion. After MI, damaged cardiomyocytes are replaced by scar tissue, leading to systolic and diastolic dysfunction, followed by adverse remodeling. Regulatory T cells (Tregs), as major immune cells, play a crucial role in post-MI inflammation and immunomodulation. Tregs improve cardiac remodeling after MI through various mechanisms, including inhibiting inflammatory cell infiltration, inducing anti-inflammatory macrophages, suppressing cell apoptosis, regulating fibroblast function, and promoting angiogenesis. The modulation of Tregs number or function may provide novel methods for improving post-MI remodeling. This review describes the immunoregulatory roles of Tregs, their regulatory mechanisms in post-MI ventricular remodeling, and the prospects and challenges for clinical application. However, the exact molecular mechanisms of Tregs in ventricular remodeling remain to be investigated. Although most of the current studies are at the preclinical stage, they hold great potential for further application in the future.
Collapse
Affiliation(s)
- Yiran Qin
- Department of Cardiology, Qingpu Hospital Affiliated to Fudan University, Shanghai, China
| | - Mingxuan Li
- Department of Cardiology, Huadong Hospital, Fudan University, Shanghai, China
| | - Haibo Liu
- Department of Cardiology, Qingpu Hospital Affiliated to Fudan University, Shanghai, China
| |
Collapse
|
3
|
Rossin D, Vanni R, Lo Iacono M, Cristallini C, Giachino C, Rastaldo R. APJ as Promising Therapeutic Target of Peptide Analogues in Myocardial Infarction- and Hypertension-Induced Heart Failure. Pharmaceutics 2023; 15:pharmaceutics15051408. [PMID: 37242650 DOI: 10.3390/pharmaceutics15051408] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 04/22/2023] [Accepted: 05/02/2023] [Indexed: 05/28/2023] Open
Abstract
The widely expressed G protein-coupled apelin receptor (APJ) is activated by two bioactive endogenous peptides, apelin and ELABELA (ELA). The apelin/ELA-APJ-related pathway has been found involved in the regulation of many physiological and pathological cardiovascular processes. Increasing studies are deepening the role of the APJ pathway in limiting hypertension and myocardial ischaemia, thus reducing cardiac fibrosis and adverse tissue remodelling, outlining APJ regulation as a potential therapeutic target for heart failure prevention. However, the low plasma half-life of native apelin and ELABELA isoforms lowered their potential for pharmacological applications. In recent years, many research groups focused their attention on studying how APJ ligand modifications could affect receptor structure and dynamics as well as its downstream signalling. This review summarises the novel insights regarding the role of APJ-related pathways in myocardial infarction and hypertension. Furthermore, recent progress in designing synthetic compounds or analogues of APJ ligands able to fully activate the apelinergic pathway is reported. Determining how to exogenously regulate the APJ activation could help to outline a promising therapy for cardiac diseases.
Collapse
Affiliation(s)
- Daniela Rossin
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy
| | - Roberto Vanni
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy
| | - Marco Lo Iacono
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy
| | - Caterina Cristallini
- Institute for Chemical and Physical Processes, IPCF ss Pisa, CNR, 56126 Pisa, Italy
| | - Claudia Giachino
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy
| | - Raffaella Rastaldo
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy
| |
Collapse
|
4
|
Wang J, She Q, Du J. Dapagliflozin attenuates myocardial remodeling in hypertension by activating the circadian rhythm signaling pathway. Arch Pharm Res 2023; 46:117-130. [PMID: 36729273 DOI: 10.1007/s12272-023-01430-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 01/27/2023] [Indexed: 02/03/2023]
Abstract
Sodium-glucose cotransporter 2 inhibitor (SGLT2i) is a new kind of antidiabetic drug which has shown beneficial effects in reducing heart failure-related hospitalization and cardiovascular-related mortality. The mechanisms are complicated. Our study aimed to investigate the effects of dapagliflozin on the myocardium of spontaneously hypertensive rats (SHRs) without heart failure. Wistar-Kyoto rats were used as normal controls. SHRs were randomly divided into the SHR group and the -treated group. After 8 weeks of dapagliflozin treatment, the morphology of heart tissues was examined. The mRNA expression profiles were identified via RNA sequencing (RNA-Seq). Various analysis methods were used to find the differentially expressed genes (DEGs) to predict gene function and coexpression. After dapagliflozin treatment, systolic blood pressure was significantly reduced compared with that in the SHR group. Myocardial remodeling was ameliorated compared with that in the SHR group. After dapagliflozin intervention, 75 DEGs (|log2-fold change | > 0 and Q value < 0.05) were identified in the heart tissues compared to the SHR group. Quantitative real-time PCR analysis confirmed that the expression of the circadian rhythm genes Per3, Bhlhe41, and Nr1d1 was significantly upregulated, while the results were coincident with the RNA-Seq results. Dapagliflozin may effectively inhibit myocardial remodeling and regulate blood pressure. The mechanisms may be related to the activation of the circadian rhythm signaling pathway.
Collapse
Affiliation(s)
- Jing Wang
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, No. 74, Linjiang Road, Yuzhong District, Chongqing, 400010, China
| | - Qiang She
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, No. 74, Linjiang Road, Yuzhong District, Chongqing, 400010, China
| | - Jianlin Du
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, No. 74, Linjiang Road, Yuzhong District, Chongqing, 400010, China.
| |
Collapse
|
5
|
Fooladi S, Faramarz S, Dabiri S, Kajbafzadeh A, Nematollahi MH, Mehrabani M. An efficient strategy to recellularization of a rat aorta scaffold: an optimized decellularization, detergent removal, and Apelin-13 immobilization. Biomater Res 2022; 26:46. [PMID: 36138491 PMCID: PMC9502639 DOI: 10.1186/s40824-022-00295-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 09/07/2022] [Indexed: 11/21/2022] Open
Abstract
Background Tissue engineering of native vessels is an alternative approach for patients with vascular disease who lack sufficient saphenous vein or other suitable conduits for autologous vascular graft. Moreover, the harvest of vessels prolongs the surgical procedure and it may lead to the morbidity of donor site in elder patients: therefore, it seems that the use of tissue-engineered vessels would be an attractive and less invasive substitute for autologous vascular grafts. Apelin-13 plays a pivotal role in cell proliferation, survival, and attachment; therefore, covalent attachment of apelin-13 to the acellular scaffolds might be a favorable approach for improving recellularization efficacy. Methods In the present study, the decellularization process was performed using various detergents. Afterward, the efficacy of decellularization procedure was evaluated using multiple approaches including assessment of DNA, hydroxyproline, and GAG content as well as Masson’s trichrome and orcein staining used for collagen and elastin determination. Subsequently, the scaffold was bioconjugated with apelin-13 using the EDC-NHS linker and acellular scaffolds were recellularized using fibroblasts, endothelial cells, and smooth muscle cells. SEM images and characterization methods were also used to evaluate the effect of apelin-13 attachment to the acellular scaffold on tissue recellularization. We also developed a novel strategy to eliminate the remnant detergents from the scaffold and increase cell viability by incubating acellular scaffolds with Bio-Beads SM-2 resin. Testometric tensile testing machine was also used for the assessment of mechanical properties and uniaxial tensile strength of decellularized and recellularized vessels compared to that of native tissues. Results Our results proposed 16-h perfusion of 0.25% sodium dodecyl sulfate (SDS) + 0.5% Triton X-100 combination to the vessel as an optimal decellularization protocol in terms of cell elimination as well as extracellular matrix preservation. Furthermore, the results demonstrated considerable elevation of cell adhesion and proliferation in scaffolds bioconjugated with apelin-13. The immunohistochemical (IHC) staining of CD31, α-SMA, and vimentin markers suggested placement of seeded cells in the suitable sites and considerable elevation of cell attachment within the scaffolds bioconjugated with apelin-13 compared to the non-bioconjugated, and decellularized groups. Moreover, the quantitative analysis of IHC staining of CD31, α-SMA, and vimentin markers suggested considerable elevation in the number of endothelial, smooth muscle, and fibroblast cells in the recellularized scaffolds bioconjugated with apelin-13 group (1.4% ± 0.02, 6.66% ± 0.23, and 9.87% ± 0.13%, respectively) compared to the non-bioconjugated scaffolds (0.03% ± 0.01, 0.28% ± 0.01, and 1.2% ± 0.09%, respectively) and decellularized groups (0.03% ± 0.007, 0.05% ± 0.01, and 0.13% ±0.005%, respectively). Although the maximum strain to the rupture was reduced in tissues decellularized using 0.5% SDS and CHAPS compared to that of native ones (116% ± 6.79, 139.1% ± 3.24, and 164% ± 8.54%, respectively), ultimate stress was decreased in all decellularized and recellularized groups. Besides, our results indicated that cell viability on the 1st, 3rd, and 7th day was 100.79% ± 0.7, 100.34% ± 0.08, and 111.24% ± 1.7% for the decellularized rat aorta conjugated with apelin-13, which was incubated for 48-h with Bio-Beads SM-2, and 73.37% ± 7.99, 47.6% ± 11.69, and 27.3% ± 7.89% for decellularized rat aorta scaffolds conjugated with apelin-13 and washed 48-h by PBS, respectively. These findings reveal that the incubation of the scaffold with Bio-Beads SM-2 is a novel and promising approach for increasing cell viability and growth within the scaffold. Conclusions In conclusion, our results provide a platform in which xenograft vessels are decellularized properly in a short time, and the recellularization process is significantly improved after the bioconjugation of the acellular scaffold with apelin-13 in terms of cell adhesion and viability within the scaffold. Graphical Abstract ![]()
Collapse
Affiliation(s)
- Saba Fooladi
- Student Research Committee, Kerman University of Medical Sciences, Kerman, Iran
| | - Sanaz Faramarz
- Student Research Committee, Kerman University of Medical Sciences, Kerman, Iran
| | - Shahriar Dabiri
- Department of Pathology, Pathology and Stem Cells Research Center, Afzalipour Medical School, Kerman University of Medical Sciences, Kerman, Iran
| | - Abdolmohammad Kajbafzadeh
- Pediatric Urology and Regenerative Medicine Research Center, Gene, Cell and Tissue Research Institute, Children Hospital Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Hadi Nematollahi
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran. .,Department of Clinical Biochemistry, Kerman University of Medical Sciences, Kerman, Iran.
| | - Mehrnaz Mehrabani
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|
6
|
Abstract
Heart regenerative medicine has been gradually evolving from a view of the heart as a nonregenerative organ with terminally differentiated cardiac muscle cells. Understanding the biology of the heart during homeostasis and in response to injuries has led to the realization that cellular communication between all cardiac cell types holds great promise for treatments. Indeed, recent studies highlight new disease-reversion concepts in addition to cardiomyocyte renewal, such as matrix- and vascular-targeted therapies, and immunotherapy with a focus on inflammation and fibrosis. In this review, we will discuss the cross-talk within the cardiac microenvironment and how specific therapies aim to target the hostile cardiac milieu under pathological conditions.
Collapse
Affiliation(s)
- Eldad Tzahor
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Stefanie Dimmeler
- Institute of Cardiovascular Regeneration, Center of Molecular Medicine, Goethe University Frankfurt, 60594 Frankfurt, Germany.,Cardiopulmonary Institute, Goethe University Frankfurt, Frankfurt, Germany.,German Center for Cardiovascular Research, RheinMain, Frankfurt, Germany
| |
Collapse
|
7
|
Liu Y, Xia H, Li M, Chen Y, Wu Y. Prognostic Value of Combining Apelin-12 and Estimated Glomerular Filtration Rate in Patients with ST-Segment Elevation Myocardial Infarction. J Interv Cardiol 2022; 2022:2272928. [PMID: 35847238 PMCID: PMC9249535 DOI: 10.1155/2022/2272928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 04/25/2022] [Accepted: 04/27/2022] [Indexed: 11/17/2022] Open
Abstract
Background Apelin-12 and estimated glomerular filtration rate (eGFR) are considered prognostic factors for ST-segment elevation myocardial infarction (STEMI). However, little is known about whether the combined use of these two biomarkers could enhance the prognostic value. This study aimed to investigate the utility of combining apelin-12 and eGFR for STEMI. Methods Patients were divided into four groups based on median apelin-12 level and eGFR level: A: low apelin-12, low eGFR; B: low apelin-12, high eGFR; C: high apelin-12, low eGFR; and D: high apelin-12, high eGFR. The Cox regression was used to identify prognostic factors. The Kaplan-Meier and the receiver operating characteristic (ROC) curves were generated to evaluate the prognostic value of apelin-12 combined with eGFR in patients with STEMI. Results Among 460 patients, 118 (25.7%) experienced major adverse cardiac events (MACEs) during the entire follow-up of 30 months. The Kaplan-Meier curve analysis revealed that group D had the best prognosis compared with the other three groups. The combination of apelin-12 and eGFR (area under the ROC curve (AUC), 0.699) enhanced the predictive value for MACE compared with either apelin-12 (AUC, 0.617) or eGFR (AUC, 0.596) alone. There was a negative association between apelin-12 and eGFR (r = -0.32, p < 0.001), while no association was observed between the Gensini score and apelin-12 or eGFR. Conclusions This study suggests that both low apelin-12 (<0.76 ng/ml) and low eGFR (<94.06 mL/min/1.73 m2) are associated with poor prognosis in STEMI, indicating that the combination of apelin-12 and eGFR could enhance the prognostic value of patients with STEMI.
Collapse
Affiliation(s)
- Yue Liu
- Department of Cardiology, Second Affiliated Hospital of Nanchang University, No. 1 Mingde Road, Nanchang 330006, Jiangxi, China
| | - Huasong Xia
- Department of Cardiology, Second Affiliated Hospital of Nanchang University, No. 1 Mingde Road, Nanchang 330006, Jiangxi, China
| | - Meng Li
- Department of Cardiology, Second Affiliated Hospital of Nanchang University, No. 1 Mingde Road, Nanchang 330006, Jiangxi, China
| | - Yi Chen
- Department of Cardiology, Second Affiliated Hospital of Nanchang University, No. 1 Mingde Road, Nanchang 330006, Jiangxi, China
| | - Yanqing Wu
- Department of Cardiology, Second Affiliated Hospital of Nanchang University, No. 1 Mingde Road, Nanchang 330006, Jiangxi, China
| |
Collapse
|
8
|
Madonna R. Angiocrine endothelium: From physiology to atherosclerosis and cardiac repair. Vascul Pharmacol 2022; 144:106993. [DOI: 10.1016/j.vph.2022.106993] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 03/25/2022] [Accepted: 03/30/2022] [Indexed: 02/08/2023]
|
9
|
de Oliveira AA, Vergara A, Wang X, Vederas JC, Oudit GY. Apelin pathway in cardiovascular, kidney, and metabolic diseases: Therapeutic role of apelin analogs and apelin receptor agonists. Peptides 2022; 147:170697. [PMID: 34801627 DOI: 10.1016/j.peptides.2021.170697] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 11/12/2021] [Accepted: 11/15/2021] [Indexed: 02/07/2023]
Abstract
The apelin/apelin receptor (ApelinR) signal transduction pathway exerts essential biological roles, particularly in the cardiovascular system. Disturbances in the apelin/ApelinR axis are linked to vascular, heart, kidney, and metabolic disorders. Therefore, the apelinergic system has surfaced as a critical therapeutic strategy for cardiovascular diseases (including pulmonary arterial hypertension), kidney disease, insulin resistance, hyponatremia, preeclampsia, and erectile dysfunction. However, apelin peptides are susceptible to rapid degradation through endogenous peptidases, limiting their use as therapeutic tools and translational potential. These proteases include angiotensin converting enzyme 2, neutral endopeptidase, and kallikrein thereby linking the apelin pathway with other peptide systems. In this context, apelin analogs with enhanced proteolytic stability and synthetic ApelinR agonists emerged as promising pharmacological alternatives. In this review, we focus on discussing the putative roles of the apelin pathway in various physiological systems from function to dysfunction, and emphasizing the therapeutic potential of newly generated metabolically stable apelin analogs and non-peptide ApelinR agonists.
Collapse
Affiliation(s)
- Amanda A de Oliveira
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada; Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Ander Vergara
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada; Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Xiaopu Wang
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada; Department of Physiology, University of Alberta, Edmonton, Alberta, Canada
| | - John C Vederas
- Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada
| | - Gavin Y Oudit
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada; Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada; Department of Physiology, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
10
|
Prasad K. Current Status of Primary, Secondary, and Tertiary Prevention of Coronary Artery Disease. Int J Angiol 2021; 30:177-186. [PMID: 34776817 PMCID: PMC8580611 DOI: 10.1055/s-0041-1731273] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
Fifty percent of all death from cardiovascular diseases is due to coronary artery disease (CAD). This is avoidable if early identification is made. Preventive health care has a major role in the fight against CAD. Atherosclerosis and atherosclerotic plaque rupture are involved in the development of CAD. Modifiable risk factors for CAD are dyslipidemia, diabetes, hypertension, cigarette smoking, obesity, chronic renal disease, chronic infection, high C-reactive protein, and hyperhomocysteinemia. CAD can be prevented by modification of risk factors. This paper defines the primary, secondary, and tertiary prevention of CAD. It discusses the mechanism of risk factor-induced atherosclerosis. This paper describes the CAD risk score and its use in the selection of individuals for primary prevention of CAD. Guidelines for primary, secondary, and tertiary prevention of CAD have been described. Modification of risk factors and use of guidelines for prevention of CAD would prevent, regress, and slow down the progression of CAD, improve the quality of life of patient, and reduce the health care cost.
Collapse
Affiliation(s)
- Kailash Prasad
- Department of Physiology (APP), College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| |
Collapse
|
11
|
Solomonidis EG, Baker AH, Brittan M. Tissue-selective endothelial arousal revealed by vascular endothelial growth factor gene transfer. Cardiovasc Res 2021; 117:18-20. [PMID: 32584971 DOI: 10.1093/cvr/cvaa172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Emmanouil G Solomonidis
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Andrew H Baker
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Mairi Brittan
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
12
|
Interaction between the apelinergic system and ACE2 in the cardiovascular system: therapeutic implications. Clin Sci (Lond) 2021; 134:2319-2336. [PMID: 32901821 DOI: 10.1042/cs20200479] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 08/27/2020] [Accepted: 09/01/2020] [Indexed: 12/13/2022]
Abstract
The apelinergic system is widely expressed and acts through autocrine and paracrine signaling to exert protective effects, including vasodilatory, metabolic, and inotropic effects on the cardiovascular (CV) system. The apelin pathway's dominant physiological role has delineated therapeutic implications for coronary artery disease, heart failure (HF), aortic aneurysm, pulmonary arterial hypertension (PAH), and transplant vasculopathy. Apelin peptides interact with the renin-angiotensin system (RAS) by promoting angiotensin converting enzyme 2 (ACE2) transcription leading to increased ACE2 protein and activity while also antagonizing the effects of angiotensin II (Ang II). Apelin modulation of the RAS by increasing ACE2 action is limited due to its rapid degradation by proteases, including ACE2, neprilysin (NEP), and kallikrein. Apelin peptides are hence tightly regulated in a negative feedback manner by ACE2. Plasma apelin levels are suppressed in pathological conditions, but its diagnostic and prognostic utility requires further clinical exploration. Enhancing the beneficial actions of apelin peptides and ACE2 axes while complementing existing pharmacological blockade of detrimental pathways is an exciting pathway for developing new therapies. In this review, we highlight the interaction between the apelin and ACE2 systems, discuss their pathophysiological roles and potential for treating a wide array of CV diseases (CVDs).
Collapse
|
13
|
Grillo M, Palmer C, Holmes N, Sang F, Larner AC, Bhosale R, Shaw PE. Stat3 oxidation-dependent regulation of gene expression impacts on developmental processes and involves cooperation with Hif-1α. PLoS One 2020; 15:e0244255. [PMID: 33332446 PMCID: PMC7746180 DOI: 10.1371/journal.pone.0244255] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 12/06/2020] [Indexed: 01/06/2023] Open
Abstract
Reactive oxygen species are bona fide intracellular second messengers that influence cell metabolism and aging by mechanisms that are incompletely resolved. Mitochondria generate superoxide that is dis-mutated to hydrogen peroxide, which in turn oxidises cysteine-based enzymes such as phosphatases, peroxiredoxins and redox-sensitive transcription factors to modulate their activity. Signal Transducer and Activator of Transcription 3 (Stat3) has been shown to participate in an oxidative relay with peroxiredoxin II but the impact of Stat3 oxidation on target gene expression and its biological consequences remain to be established. Thus, we created murine embryonic fibroblasts (MEFs) that express either WT-Stat3 or a redox-insensitive mutant of Stat3 (Stat3-C3S). The Stat3-C3S cells differed from WT-Stat3 cells in morphology, proliferation and resistance to oxidative stress; in response to cytokine stimulation, they displayed elevated Stat3 tyrosine phosphorylation and Socs3 expression, implying that Stat3-C3S is insensitive to oxidative inhibition. Comparative analysis of global gene expression in WT-Stat3 and Stat3-C3S cells revealed differential expression (DE) of genes both under basal conditions and during oxidative stress. Using differential gene regulation pattern analysis, we identified 199 genes clustered into 10 distinct patterns that were selectively responsive to Stat3 oxidation. GO term analysis identified down-regulated genes to be enriched for tissue/organ development and morphogenesis and up-regulated genes to be enriched for cell-cell adhesion, immune responses and transport related processes. Although most DE gene promoters contain consensus Stat3 inducible elements (SIEs), our chromatin immunoprecipitation (ChIP) and ChIP-seq analyses did not detect Stat3 binding at these sites in control or oxidant-stimulated cells, suggesting that oxidised Stat3 regulates these genes indirectly. Our further computational analysis revealed enrichment of hypoxia response elements (HREs) within DE gene promoters, implying a role for Hif-1. Experimental validation revealed that efficient stabilisation of Hif-1α in response to oxidative stress or hypoxia required an oxidation-competent Stat3 and that depletion of Hif-1α suppressed the inducible expression of Kcnb1, a representative DE gene. Our data suggest that Stat3 and Hif-1α cooperate to regulate genes involved in immune functions and developmental processes in response to oxidative stress.
Collapse
Affiliation(s)
- Michela Grillo
- School of Life Sciences, University of Nottingham, Queen’s Medical Centre, Nottingham, United Kingdom
| | - Carolyn Palmer
- School of Life Sciences, University of Nottingham, Queen’s Medical Centre, Nottingham, United Kingdom
| | - Nadine Holmes
- Deep-Seq Unit, School of Life Sciences, University of Nottingham, Queen’s Medical Centre, Nottingham, United Kingdom
| | - Fei Sang
- Deep-Seq Unit, School of Life Sciences, University of Nottingham, Queen’s Medical Centre, Nottingham, United Kingdom
| | - Andrew C. Larner
- Department of Biochemistry and Molecular Biology, Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Rahul Bhosale
- School of Biosciences, University of Nottingham, Sutton Bonington, United Kingdom
| | - Peter E. Shaw
- School of Life Sciences, University of Nottingham, Queen’s Medical Centre, Nottingham, United Kingdom
| |
Collapse
|
14
|
Pioglitazone protects blood vessels through inhibition of the apelin signaling pathway by promoting KLF4 expression in rat models of T2DM. Biosci Rep 2020; 39:221480. [PMID: 31829402 PMCID: PMC6928522 DOI: 10.1042/bsr20190317] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 11/16/2019] [Accepted: 12/02/2019] [Indexed: 02/06/2023] Open
Abstract
Apelin, identified as the endogenous ligand of APJ, exerts various cardiovascular effects. However, the molecular mechanism underlying the regulation of apelin expression in vascular cells is poorly described. Pioglitazone (PIO) and Krüppel-like factor 4 (KLF4) exhibit specific biological functions on vascular physiology and pathophysiology by regulating differentiation- and proliferation-related genes. The present study aimed to investigate the roles of PIO and KLF4 in the transcriptional regulation of apelin in a high-fat diet/streptozotocin rat model of diabetes and in PIO-stimulated vascular smooth muscle cells (VSMCs). Immunohistochemistry, qRT-PCR, and Western blotting assays revealed that the aorta of the Type 2 diabetes mellitus (T2DM) rat models had a high expression of apelin, PIO could decrease the expression of apelin in the PIO-treated rats. In vitro, Western blotting assays and immunofluorescent staining results showed that the basal expression of apelin was decreased but that of KLF4 was increased when VSMCs were stimulated by PIO treatment. Luciferase and chromatin immunoprecipitation assay results suggested that KLF4 bound to the GKLF-binding site of the apelin promoter and negatively regulated the transcription activity of apelin in VSMCs under PIO stimulation. Furthermore, qRT-PCR and Western blotting assay results showed that the overexpression of KLF4 markedly decreased the basal expression of apelin, but the knockdown of KLF4 restored the PIO-induced expression of apelin. In conclusion, PIO inhibited the expression of apelin in T2DM rat models to prevent diabetic macroangiopathy, and negatively regulated the gene transcription of apelin by promoting transcription of KLF4 in the apelin promoter.
Collapse
|
15
|
Helker CS, Eberlein J, Wilhelm K, Sugino T, Malchow J, Schuermann A, Baumeister S, Kwon HB, Maischein HM, Potente M, Herzog W, Stainier DY. Apelin signaling drives vascular endothelial cells toward a pro-angiogenic state. eLife 2020; 9:55589. [PMID: 32955436 PMCID: PMC7567607 DOI: 10.7554/elife.55589] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 09/19/2020] [Indexed: 12/18/2022] Open
Abstract
To form new blood vessels (angiogenesis), endothelial cells (ECs) must be activated and acquire highly migratory and proliferative phenotypes. However, the molecular mechanisms that govern these processes are incompletely understood. Here, we show that Apelin signaling functions to drive ECs into such an angiogenic state. Zebrafish lacking Apelin signaling exhibit defects in endothelial tip cell morphology and sprouting. Using transplantation experiments, we find that in mosaic vessels, wild-type ECs leave the dorsal aorta (DA) and form new vessels while neighboring ECs defective in Apelin signaling remain in the DA. Mechanistically, Apelin signaling enhances glycolytic activity in ECs at least in part by increasing levels of the growth-promoting transcription factor c-Myc. Moreover, APELIN expression is regulated by Notch signaling in human ECs, and its function is required for the hypersprouting phenotype in Delta-like 4 (Dll4) knockdown zebrafish embryos. These data provide new insights into fundamental principles of blood vessel formation and Apelin signaling, enabling a better understanding of vascular growth in health and disease.
Collapse
Affiliation(s)
- Christian Sm Helker
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,Philipps-University Marburg, Faculty of Biology, Cell Signaling and Dynamics, Marburg, Germany
| | - Jean Eberlein
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,Philipps-University Marburg, Faculty of Biology, Cell Signaling and Dynamics, Marburg, Germany
| | - Kerstin Wilhelm
- Angiogenesis and Metabolism Laboratory, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Toshiya Sugino
- Angiogenesis and Metabolism Laboratory, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Julian Malchow
- Philipps-University Marburg, Faculty of Biology, Cell Signaling and Dynamics, Marburg, Germany
| | | | - Stefan Baumeister
- Philipps-University Marburg, Faculty of Biology, Cell Signaling and Dynamics, Marburg, Germany
| | - Hyouk-Bum Kwon
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Hans-Martin Maischein
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Michael Potente
- Angiogenesis and Metabolism Laboratory, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,DZHK (German Center for Cardiovascular Research), partner site Frankfurt Rhine-Main, Berlin, Germany
| | - Wiebke Herzog
- University of Muenster, Muenster, Germany.,Max Planck Institute for Molecular Biomedicine, Muenster, Germany
| | - Didier Yr Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,DZHK (German Center for Cardiovascular Research), partner site Frankfurt Rhine-Main, Berlin, Germany
| |
Collapse
|
16
|
Association of genetic defects in the apelin-AGTRL1 system with myocardial infarction risk in Han Chinese. Gene 2020; 766:145143. [PMID: 32911028 DOI: 10.1016/j.gene.2020.145143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 08/28/2020] [Accepted: 09/02/2020] [Indexed: 11/21/2022]
Abstract
We aimed to test the hypothesis that apelin (APLN) and its receptor AGTRL1 (APLNR) genes may contribute to the pathogenesis of myocardial infarction in Han Chinese. This is a hospital-based, case-control association study, involving 1067 patients with myocardial infarction and 942 healthy controls. Myocardial infarction is diagnosed by electrocardiogram or anatomopathological examination. Eight polymorphisms in APLN gene and 5 in APLNR gene were genotyped using the TaqMan assay. Risk was summarized as odds ratio (OR) and 95% confidence interval (CI). In males, rs56204867-G allele (adjusted OR, 95% CI, p: 0.21, 0.08-0.55, 0.002) and rs2235309-T allele (0.60, 0.42-0.84, 0.004) was associated with a significantly reduced risk of myocardial infarction, and the mutations of rs2235310 was associated with an increased risk (1.41, 1.06-2.52, 0.021), as well as for rs948847-GG genotype (1.85, 1.23-2.91, 0.007). In females, the presence of rs56204867-AG and -GG genotypes was significantly associated with 44% and 50% reduced risk (0.56 and 0.50, 0.40-8.04 and 0.29-0.86, 0.007 and 0.036), respectively; for rs2235310, CC genotype was associated with 72% increased risk (1.72, 1.09-3.22, 0.016), and the odds of myocardial infarction was 3.47 for rs9943582-TT genotype (95% CI: 1.53-7.57, 0.009). The gender-specific association of APLN and APLNR genes with myocardial infarction was reinforced by further linkage and haplotype analyses. Finally, nomograms based on significant polymorphisms are satisfactory, with the C-indexes over 80% for both genders. Taken together, our findings indicate that APLN and APLNR genes are potential candidates in the pathogenesis of myocardial infarction in Han Chinese, and importantly their contribution is gender-dependent.
Collapse
|
17
|
Masoumi J, Jafarzadeh A, Khorramdelazad H, Abbasloui M, Abdolalizadeh J, Jamali N. Role of Apelin/APJ axis in cancer development and progression. Adv Med Sci 2020; 65:202-213. [PMID: 32087570 DOI: 10.1016/j.advms.2020.02.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 05/26/2019] [Accepted: 02/11/2020] [Indexed: 02/07/2023]
Abstract
Apelin is an endogenous peptide, which is expressed in a vast board of organs such as the brain, placenta, heart, lungs, kidneys, pancreas, testis, prostate and adipose tissues. The apelin receptor, called angiotensin-like-receptor 1 (APJ), is also expressed in the brain, spleen, placenta, heart, liver, intestine, prostate, thymus, testis, ovary, lungs, kidneys, stomach, and adipose tissue. The apelin/APJ axis is involved in a number of physiological and pathological processes. The apelin expression is increased in various kinds of cancer and the apelin/APJ axis plays a key role in the development of tumors through enhancing angiogenesis, metastasis, cell proliferation and also through the development of cancer stem cells and drug resistance. The apelin also stops the apoptosis of cancer cells. The apelin/APJ axis was considered in this review as an attractive therapeutic target for cancer treatment.
Collapse
|
18
|
Mohammadi C, Sameri S, Najafi R. Insight into adipokines to optimize therapeutic effects of stem cell for tissue regeneration. Cytokine 2020; 128:155003. [PMID: 32000014 DOI: 10.1016/j.cyto.2020.155003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 01/18/2020] [Accepted: 01/20/2020] [Indexed: 11/29/2022]
Abstract
Stem cell therapy is considered as a promising regenerative medicine for repairing and treating damaged tissues and/or preventing various diseases. But there are still some obstacles such as low cell migration, poor stem cell engraftment and decreased cell survival that need to be overcome before transplantation. Therefore, a large body of studies has focused on improving the efficiency of stem cell therapy. For instance, preconditioning of stem cells has emerged as an effective strategy to reinforce therapeutic efficacy. Adipokines are signaling molecules, secreted by adipose tissue, which regulate a variety of biological processes in adipose tissue and other organs including the brain, liver, and muscle. In this review article, we shed light on the biological effects of some adipokines including apelin, oncostatin M, omentin-1 and vaspin on stem cell therapy and the most recent preclinical advances in our understanding of how these functions ameliorate stem cell therapy outcome.
Collapse
Affiliation(s)
- Chiman Mohammadi
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Saba Sameri
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Rezvan Najafi
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
| |
Collapse
|
19
|
Leung OM, Li J, Li X, Chan VW, Yang KY, Ku M, Ji L, Sun H, Waldmann H, Tian XY, Huang Y, Lau J, Zhou B, Lui KO. Regulatory T Cells Promote Apelin-Mediated Sprouting Angiogenesis in Type 2 Diabetes. Cell Rep 2020; 24:1610-1626. [PMID: 30089270 DOI: 10.1016/j.celrep.2018.07.019] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 06/27/2018] [Accepted: 07/05/2018] [Indexed: 12/13/2022] Open
Abstract
The role of CD4+ T cells in the ischemic tissues of T2D patients remains unclear. Here, we report that T2D patients' vascular density was negatively correlated with the number of infiltrating CD4+ T cells after ischemic injury. Th1 was the predominant subset, and Th1-derived IFN-γ and TNF-α directly impaired human angiogenesis. We then blocked CD4+ T cell infiltration into the ischemic tissues of both Leprdb/db and diet-induced obese T2D mice. Genome-wide RNA sequencing shows an increased proliferative and angiogenic capability of diabetic ECs in ischemic tissues. Moreover, wire myography shows enhanced EC function and laser Doppler imaging reveals improved post-ischemic blood reperfusion. Mechanistically, functional revascularization after CD4 coreceptor blockade was mediated by Tregs. Genetic lineage tracing via Cdh5-CreER and Apln-CreER and coculture assays further illustrate that Tregs increased vascular density and induced de novo sprouting angiogenesis in a paracrine manner. Taken together, our results reveal that Th1 impaired while Tregs promoted functional post-ischemic revascularization in obesity and diabetes.
Collapse
Affiliation(s)
- Oscar M Leung
- Department of Chemical Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China; Li Ka Shing Institute of Health Sciences, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Jiatao Li
- Department of Chemical Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China; Li Ka Shing Institute of Health Sciences, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Xisheng Li
- Department of Chemical Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Vicken W Chan
- Department of Chemical Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Kevin Y Yang
- Department of Chemical Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Manching Ku
- Next Generation Sequencing Core, Salk Institute for Biological Studies, La Jolla, CA, USA; Department of Paediatrics and Adolescent Medicine, Division of Paediatric Hematology and Oncology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Lu Ji
- Department of Chemical Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China; Li Ka Shing Institute of Health Sciences, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Hao Sun
- Department of Chemical Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China; Li Ka Shing Institute of Health Sciences, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Herman Waldmann
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Xiao Yu Tian
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China; Institute of Vascular Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yu Huang
- Li Ka Shing Institute of Health Sciences, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China; School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China; Institute of Vascular Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - James Lau
- Department of Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Bin Zhou
- The State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Kathy O Lui
- Department of Chemical Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China; Li Ka Shing Institute of Health Sciences, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
20
|
Arababadi MK, Asadikaram P, Asadikaram G. APLN/APJ pathway: The key regulator of macrophage functions. Life Sci 2019; 232:116645. [PMID: 31299236 DOI: 10.1016/j.lfs.2019.116645] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Revised: 06/30/2019] [Accepted: 07/09/2019] [Indexed: 12/16/2022]
Abstract
Macrophages play key roles during cardiovascular diseases (CVD) and their related complications. Apelin (APLN) is a key molecule, whose roles during CVD have been documented previously. Therefore, it has been hypothesized that APLN may perform its roles via modulation of macrophages. Additionally, due to the widespread distribution of the CVD, more effective therapeutic strategies need to be developed to overcome the related complications. This review article collected recent information regarding the roles of APLN on the macrophages and discusses its potential chance to be a target for molecular/cellular therapy of APLN and the APLN treated macrophages for CVD.
Collapse
Affiliation(s)
- Mohammad Kazemi Arababadi
- Immunology of Infectious Diseases Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran; Department of Laboratory Sciences, Faculty of Paramedicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran; Department of Clinical Biochemistry, Afzalipur Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Parisa Asadikaram
- Faculty of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Gholamreza Asadikaram
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran; Department of Clinical Biochemistry, Afzalipur Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|
21
|
Shafiq M, Zhang Y, Zhu D, Zhao Z, Kim DH, Kim SH, Kong D. In situ cardiac regeneration by using neuropeptide substance P and IGF-1C peptide eluting heart patches. Regen Biomater 2018; 5:303-316. [PMID: 30338128 PMCID: PMC6184517 DOI: 10.1093/rb/rby021] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Revised: 06/26/2018] [Accepted: 07/03/2018] [Indexed: 12/23/2022] Open
Abstract
Cardiovascular diseases cause huge socio-economic burden worldwide. Although a mammalian myocardium has its own limited healing capability, scaffold materials capable of releasing stem cell recruiting/engrafting factors may facilitate the regeneration of the infarcted myocardium. The aim of this research was to develop cardiac patches capable of simultaneously eluting substance P (SP) and insulin-like growth factor-1C (IGF-1C) peptide. Polycaprolactone/collagen type 1-based patches with or without SP and IGF-1C peptide were fabricated by co-electrospinning, which exhibited nanofibrous morphology. SP and IGF-1C/SP patches recruited significantly higher numbers of bone marrow-mesenchymal stem cells than that of the negative control and patch-only groups in vitro. The developed patches were transplanted in an infarcted myocardium for up to 14 days. Mice underwent left anterior descending artery ligation and received one of the following treatments: (i) sham, (ii) saline, (iii) patch-only, (iv) IGF-1C patch, (v) SP patch and (vi) IGF-1C/SP patch. SP and IGF-1C/SP patch-treated groups exhibited better heart function and attenuated adverse cardiac remodeling than that of the saline, patch-only and individual peptide containing cardiac patches. SP patch and IGF-1C/SP patch-treated groups also showed higher numbers of CD31-positive vessels and isolectin B4-positive capillaries than that of other groups. IGF-1C/SP-treated group also showed thicker left ventricular wall in comparison to the saline and patch-only groups. Moreover, IGF-1C/SP patches recruited significantly higher numbers of CD29-positive cells and showed less numbers of Tunel-positive cells compared with the other groups. These data suggest that SP and IGF-1C peptides may act synergistically for in situ tissue repair.
Collapse
Affiliation(s)
- Muhammad Shafiq
- Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul, Republic of Korea
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), College of Life Science, Nankai University, Tianjin, China
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology, Cheongryang, Seoul, Republic of Korea
- Department of Chemistry, Center for Tissue Engineering & Regenerative Medicine, Pakistan Institute of Engineering & Applied Sciences (PIEAS), Nilore, Islamabad, Pakistan
| | - Yue Zhang
- Department of Physiology & Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Dashuai Zhu
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), College of Life Science, Nankai University, Tianjin, China
| | - Zongxian Zhao
- Department of Physiology & Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Dong-Hwee Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea
| | - Soo Hyun Kim
- Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul, Republic of Korea
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology, Cheongryang, Seoul, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea
| | - Deling Kong
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), College of Life Science, Nankai University, Tianjin, China
| |
Collapse
|
22
|
Zhang J, Zhou Y, Wu C, Wan Y, Fang C, Li J, Fang W, Yi R, Zhu G, Li J, Wang Y. Characterization of the Apelin/Elabela Receptors (APLNR) in Chickens, Turtles, and Zebrafish: Identification of a Novel Apelin-Specific Receptor in Teleosts. Front Endocrinol (Lausanne) 2018; 9:756. [PMID: 30631305 PMCID: PMC6315173 DOI: 10.3389/fendo.2018.00756] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 11/29/2018] [Indexed: 12/22/2022] Open
Abstract
Apelin receptor(s) (APLNR) are suggested to mediate the actions of apelin and Elabela (ELA) peptides in many physiological processes, including cardiovascular development and food intake in vertebrates. However, the functionality of APLNR has not been examined in most vertebrate groups. Here, we characterized two APLNRs APLNR1, APLNR2) in chickens and red-eared sliders, and three APLNRs in zebrafish (APLNR2a, APLNR2b, APLNR3a), which are homologous to human APLNR. Using luciferase-reporter assays or Western blot, we demonstrated that in chickens, APLNR1 (not APLNR2) expressed in HEK293 cells was potently activated by chicken apelin-36 and ELA-32 and coupled to Gi-cAMP and MAPK/ERK signaling pathways, indicating a crucial role of APLNR1 in mediating apelin/ELA actions; in red-eared sliders, APLNR2 (not APLNR1) was potently activated by apelin-36/ELA-32, suggesting that APLNR2 may mediate apelin/ELA actions; in zebrafish, both APLNR2a and APLNR2b were potently activated by apelin-36/ELA-32 and coupled to Gi-cAMP signaling pathway, as previously proposed, whereas the novel APLNR3a was specifically and potently activated by apelin. Similarly, an apelin-specific receptor (APLNR3b) sharing 57% sequence identity with zebrafish APLNR3a was identified in Nile tilapia. Collectively, our data facilitates the uncovering of the roles of APLNR signaling in different vertebrate groups and suggests a key functional switch between APLNR1 and APLNR2/3 in mediating the actions of ELA and apelin during vertebrate evolution.
Collapse
|
23
|
Zhou Y, Wang Y, Qiao S, Yin L. Effects of Apelin on Cardiovascular Aging. Front Physiol 2017; 8:1035. [PMID: 29302260 PMCID: PMC5732982 DOI: 10.3389/fphys.2017.01035] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 11/29/2017] [Indexed: 12/24/2022] Open
Abstract
Apelin is the endogenous ligand of APJ, the orphan G protein-coupled receptor. The apelin-APJ signal transduction pathway is widely expressed in the cardiovascular system and is an important factor in cardiovascular homeostasis. This signal transduction pathway has long been related to diseases with high morbidity in the elderly, such as atherosclerosis, coronary atherosclerotic heart disease, hypertension, calcific aortic valve disease, heart failure and atrial fibrillation. In this review, we discuss the apelin-APJ signal transduction pathway related to age-associated cardiovascular diseases.
Collapse
Affiliation(s)
- Ying Zhou
- Department of Cardiology, China-Japan Friendship Hospital, Beijing, China
| | - Yong Wang
- Department of Cardiology, China-Japan Friendship Hospital, Beijing, China
| | - Shubin Qiao
- Department of Cardiology, Cardiovascular Institute of Fuwai Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Liang Yin
- School of Science, Beijing University of Chemical Technology, Beijing, China
| |
Collapse
|
24
|
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
|
25
|
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
|
26
|
Frump AL, Bonnet S, de Jesus Perez VA, Lahm T. Emerging role of angiogenesis in adaptive and maladaptive right ventricular remodeling in pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol 2017; 314:L443-L460. [PMID: 29097426 DOI: 10.1152/ajplung.00374.2017] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Right ventricular (RV) function is the primary prognostic factor for both morbidity and mortality in pulmonary hypertension (PH). RV hypertrophy is initially an adaptive physiological response to increased overload; however, with persistent and/or progressive afterload increase, this response frequently transitions to more pathological maladaptive remodeling. The mechanisms and disease processes underlying this transition are mostly unknown. Angiogenesis has recently emerged as a major modifier of RV adaptation in the setting of pressure overload. A novel paradigm has emerged that suggests that angiogenesis and angiogenic signaling are required for RV adaptation to afterload increases and that impaired and/or insufficient angiogenesis is a major driver of RV decompensation. Here, we summarize our current understanding of the concepts of maladaptive and adaptive RV remodeling, discuss the current literature on angiogenesis in the adapted and failing RV, and identify potential therapeutic approaches targeting angiogenesis in RV failure.
Collapse
Affiliation(s)
- Andrea L Frump
- Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Department of Medicine, Indiana University School of Medicine , Indianapolis, Indiana
| | - Sébastien Bonnet
- Pulmonary Hypertension Research Group, Institut Universitaire de Cardiologie et de Pneumologie de Québec Research Center, Laval University , Quebec City, Quebec , Canada
| | - Vinicio A de Jesus Perez
- Division of Pulmonary/Critical Care, Stanford University School of Medicine , Stanford, California.,Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University School of Medicine , Stanford, California
| | - Tim Lahm
- Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Department of Medicine, Indiana University School of Medicine , Indianapolis, Indiana.,Richard L. Roudebush Veterans Affairs Medical Center , Indianapolis, Indiana.,Department of Cellular and Integrative Physiology, Indiana University School of Medicine , Indianapolis, Indiana
| |
Collapse
|
27
|
Tatin F, Renaud-Gabardos E, Godet AC, Hantelys F, Pujol F, Morfoisse F, Calise D, Viars F, Valet P, Masri B, Prats AC, Garmy-Susini B. Apelin modulates pathological remodeling of lymphatic endothelium after myocardial infarction. JCI Insight 2017; 2:93887. [PMID: 28614788 DOI: 10.1172/jci.insight.93887] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 05/10/2017] [Indexed: 11/17/2022] Open
Abstract
Lymphatic endothelium serves as a barrier to control fluid balance and immune cell trafficking to maintain tissue homeostasis. Long-term alteration of lymphatic vasculature promotes edema and fibrosis, which is an aggravating factor in the onset of cardiovascular diseases such as myocardial infarction. Apelin is a bioactive peptide that plays a central role in angiogenesis and cardiac contractility. Despite an established role of apelin in lymphangiogenesis, little is known about its function in the cardiac lymphatic endothelium. Here, we show that apelin and its receptor APJ were exclusively expressed on newly formed lymphatic vasculature in a pathological model of myocardial infarction. Using an apelin-knockout mouse model, we identified morphological and functional defects in lymphatic vasculature associated with a proinflammatory status. Surprisingly, apelin deficiency increased the expression of lymphangiogenic growth factors VEGF-C and VEGF-D and exacerbated lymphangiogenesis after myocardial infarction. Conversely, the overexpression of apelin in ischemic heart was sufficient to restore a functional lymphatic vasculature and to reduce matrix remodeling and inflammation. In vitro, the expression of apelin prevented the alteration of cellular junctions in lymphatic endothelial cells induced by hypoxia. In addition, we demonstrated that apelin controls the secretion of the lipid mediator sphingosine-1-phosphate in lymphatic endothelial cells by regulating the level of expression of sphingosine kinase 2 and the transporter SPNS2. Taken together, our results show that apelin plays a key role in lymphatic vessel maturation and stability in pathological settings. Thus, apelin may represent a novel candidate to prevent pathological lymphatic remodeling in diseases.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Fanny Viars
- MetaToul-Lipidomique Core Facility, I2MC INSERM 1048, Toulouse, France
| | | | | | | | | |
Collapse
|
28
|
Hou J, Zhong T, Guo T, Miao C, Zhou C, Long H, Wu H, Zheng S, Wang L, Wang T. Apelin promotes mesenchymal stem cells survival and vascularization under hypoxic-ischemic condition in vitro involving the upregulation of vascular endothelial growth factor. Exp Mol Pathol 2017; 102:203-209. [PMID: 28161441 DOI: 10.1016/j.yexmp.2017.01.015] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Revised: 12/16/2016] [Accepted: 01/29/2017] [Indexed: 01/14/2023]
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) transplantation has been regarded as an optimal therapeutic approach for cardiovascular disease. However, the inferior survival and low vascularization potential of these cells in the local infarct site reduce the therapeutic efficacy. In this study, we investigated the influence of apelin on MSCs survival and vascularization under hypoxic-ischemic condition in vitro and explored the relevant mechanism. METHODS MSCs were obtained from C57BL/6 mice and cultured in vitro. Cells of the third passage were divided into MSCs and MSCs+apelin groups. In the MSCs+apelin group, MSCs were stimulated with apelin-13 (5μM). The two groups experienced exposure to hypoxia (1% O2) and serum deprivation for 24h, using normoxia (20% O2) as a negative control during the process. Human umbilical vein endothelial cells (HUVECs) were used and incubated with conditioned media from both groups to promote vascularization for another 6h. Vascular densities were assessed and relevant biomarkers were detected thereafter. RESULTS Compared with MSCs group, MSCs+apelin group presented more rapid growth. The proliferation rate was much higher. Cells apoptosis percentage was significantly declined both under normoxic and hypoxic conditions. Media produced from MSCs+apelin group triggered HUVECs to form a larger number of vascular branches on matrigel. The expression and secretion of vascular endothelial growth factor (VEGF) were significantly increased. CONCLUSION Apelin could effectively promote MSCs survival and vascularization under hypoxic-ischemic condition in vitro, and this procedure was associated with the upregulation of VEGF. This study provides a new perspective for exploring novel approaches to enhance MSCs survival and vascularization potential.
Collapse
Affiliation(s)
- Jingying Hou
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, 107 Yanjiang Xi Road, Guangzhou, Guangdong 510120, China; Department of Emergency, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, 107 Yanjiang Xi Road, Guangzhou, Guangdong 510120, China
| | - Tingting Zhong
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, 107 Yanjiang Xi Road, Guangzhou, Guangdong 510120, China; Department of Emergency, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, 107 Yanjiang Xi Road, Guangzhou, Guangdong 510120, China
| | - Tianzhu Guo
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, 107 Yanjiang Xi Road, Guangzhou, Guangdong 510120, China; Department of Emergency, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, 107 Yanjiang Xi Road, Guangzhou, Guangdong 510120, China
| | - Changqing Miao
- The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shanxi 710061, China
| | - Changqing Zhou
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, 107 Yanjiang Xi Road, Guangzhou, Guangdong 510120, China; Department of Emergency, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, 107 Yanjiang Xi Road, Guangzhou, Guangdong 510120, China
| | - Huibao Long
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, 107 Yanjiang Xi Road, Guangzhou, Guangdong 510120, China; Department of Emergency, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, 107 Yanjiang Xi Road, Guangzhou, Guangdong 510120, China
| | - Hao Wu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, 107 Yanjiang Xi Road, Guangzhou, Guangdong 510120, China; Department of Emergency, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, 107 Yanjiang Xi Road, Guangzhou, Guangdong 510120, China
| | - Shaoxin Zheng
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, 107 Yanjiang Xi Road, Guangzhou, Guangdong 510120, China; Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, 107 Yanjiang Xi Road, Guangzhou, Guangdong, China
| | - Lei Wang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, 107 Yanjiang Xi Road, Guangzhou, Guangdong 510120, China; Department of Emergency, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, 107 Yanjiang Xi Road, Guangzhou, Guangdong 510120, China
| | - Tong Wang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, 107 Yanjiang Xi Road, Guangzhou, Guangdong 510120, China; Department of Emergency, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, 107 Yanjiang Xi Road, Guangzhou, Guangdong 510120, China; Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, 107 Yanjiang Xi Road, Guangzhou, Guangdong, China.
| |
Collapse
|
29
|
Sans-Roselló J, Casals G, Rossello X, González de la Presa B, Vila M, Duran-Cambra A, Morales-Ruiz M, Ferrero-Gregori A, Jiménez W, Sionis A. Prognostic value of plasma apelin concentrations at admission in patients with ST-segment elevation acute myocardial infarction. Clin Biochem 2016; 50:279-284. [PMID: 27889567 DOI: 10.1016/j.clinbiochem.2016.11.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Revised: 11/15/2016] [Accepted: 11/16/2016] [Indexed: 12/29/2022]
Abstract
BACKGROUND The use of plasma biomarkers is relevant for the prognosis of ST-segment elevation myocardial infarction (STEMI) patients. Apelin, an adipocytokine, plays a pivotal role in the pathophysiology of both ischemia/reperfusion injury and its potential subsequent heart failure. We evaluated apelin concentrations at admission as a biomarker to assess risk of 6-month mortality. METHODS Consecutive patients with STEMI were recruited from January 2012 to January 2013 (n=250). Plasma apelin, brain natriuretic peptide (BNP) and sensitive troponin I (sTnI) were assessed in EDTA-plasma samples obtained at admission. Clinical, hemodynamic and other laboratory variables were also registered. All-cause mortality was assessed at 6-month follow-up. RESULTS Increased plasma apelin concentrations at admission were predictive of 6- month mortality, after adjustment for age, diabetes, systolic blood pressure, heart rate, glomerular filtration rate, Killip class, left ventricular ejection fraction, BNP and sTnI. The combination of apelin with BNP and sTnI further improved the apelin predictive value. Finally, apelin concentrations were associated with markers of ischemic heart failure severity, but not with markers of ischemic insult severity. CONCLUSIONS Increased plasma concentrations of apelin at admission in patients with STEMI were associated with a higher risk of mortality at 6months, adding prognostic value to the provided by BNP. Moreover, apelin levels were also related to markers of ischemic heart failure severity, but not markers of ischemia severity.
Collapse
Affiliation(s)
- Jordi Sans-Roselló
- Acute and Intensive Cardiovascular Care Unit, Department of Cardiology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute IIB-Sant Pau, Barcelona, Spain
| | - Gregori Casals
- Service of Biochemistry and Molecular Genetics, Hospital Clínic, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Xavier Rossello
- Acute and Intensive Cardiovascular Care Unit, Department of Cardiology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute IIB-Sant Pau, Barcelona, Spain
| | - Bernardino González de la Presa
- Service of Biochemistry and Molecular Genetics, Hospital Clínic, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Montserrat Vila
- Acute and Intensive Cardiovascular Care Unit, Department of Cardiology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute IIB-Sant Pau, Barcelona, Spain
| | - Albert Duran-Cambra
- Acute and Intensive Cardiovascular Care Unit, Department of Cardiology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute IIB-Sant Pau, Barcelona, Spain
| | - Manuel Morales-Ruiz
- Service of Biochemistry and Molecular Genetics, Hospital Clínic, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Andreu Ferrero-Gregori
- Acute and Intensive Cardiovascular Care Unit, Department of Cardiology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute IIB-Sant Pau, Barcelona, Spain
| | - Wladimiro Jiménez
- Service of Biochemistry and Molecular Genetics, Hospital Clínic, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain,; Department of Biomedicine, University of Barcelona, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain
| | - Alessandro Sionis
- Acute and Intensive Cardiovascular Care Unit, Department of Cardiology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute IIB-Sant Pau, Barcelona, Spain.
| |
Collapse
|
30
|
Chung WJ, Cho A, Byun K, Moon J, Ge X, Seo HS, Moon E, Dash R, Yang PC. Apelin-13 infusion salvages the peri-infarct region to preserve cardiac function after severe myocardial injury. Int J Cardiol 2016; 222:361-367. [PMID: 27500765 DOI: 10.1016/j.ijcard.2016.07.263] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2016] [Revised: 07/22/2016] [Accepted: 07/30/2016] [Indexed: 11/30/2022]
Abstract
BACKGROUND Apelin-13 (A13) regulates cardiac homeostasis. However, the effects and mechanism of A13 infusion after an acute myocardial injury (AMI) have not been elucidated. This study assesses the restorative effects and mechanism of A13 on the peri-infarct region in murine AMI model. METHODS 51 FVB/N mice (12weeks, 30g) underwent AMI. A week following injury, continuous micro-pump infusion of A13 (0.5μg/g/day) and saline was initiated for 4-week duration. Dual contrast MRI was conducted on weeks 1, 2, 3, and 5, consisting of delayed-enhanced and manganese-enhanced MRI. Four mice in each group were followed for an extended period of 4weeks without further infusion and underwent MRI scans on weeks 7 and 9. RESULTS A13 infusion demonstrated preserved LVEF compared to saline from weeks 1 to 4 (21.9±3.2% to 23.1±1.7%* vs. 23.5±1.7% to 16.9±2.8%, *p=0.02), which persisted up to 9weeks post-MI (+1.4%* vs. -9.4%, *p=0.03). Mechanistically, dual contrast MRI demonstrated significant decrease in the peri-infarct and scar % volume in A13 group from weeks 1 to 4 (15.1 to 7.4% and 34.3 to 25.1%, p=0.02, respectively). This was corroborated by significant increase in 5-ethynyl-2'-deoxyuridine (EdU(+)) cells by A13 vs. saline groups in the peri-infarct region (16.5±3.1% vs. 8.1±1.6%; p=0.04), suggesting active cell mitosis. Finally, significantly enhanced mobilization of CD34(+) cells in the peripheral blood and up-regulation of APJ, fibrotic, and apoptotic genes in the peri-infarct region were found. CONCLUSIONS A13 preserves cardiac performance by salvaging the peri-infarct region and may contribute to permanent restoration of the severely injured myocardium.
Collapse
Affiliation(s)
- Wook-Jin Chung
- Department of Cardiovascular Medicine, Stanford University, Stanford, CA, USA; Department of Cardiovascular Medicine, Gachon University, Incheon, Republic of Korea; Gachon Cardiovascular Research Institute, Gachon University, Incheon, Republic of Korea
| | - Ahryon Cho
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Kyunghee Byun
- Gachon Cardiovascular Research Institute, Gachon University, Incheon, Republic of Korea; Department of Anatomy and Cell Biology, Gachon University, Incheon, Republic of Korea; Center for Genomics and Proteomics & Stem Cell Core Facility, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Republic of Korea
| | - Jeongsik Moon
- Gachon Cardiovascular Research Institute, Gachon University, Incheon, Republic of Korea; Center for Genomics and Proteomics & Stem Cell Core Facility, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Republic of Korea
| | - Xiaohu Ge
- Department of Cardiovascular Medicine, Stanford University, Stanford, CA, USA
| | - Hye-Sun Seo
- Division of Cardiology, Soon Chun Hyang University College of Medicine, Bucheon, Republic of Korea
| | - Ejung Moon
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | - Rajesh Dash
- Department of Cardiovascular Medicine, Stanford University, Stanford, CA, USA
| | - Phillip C Yang
- Department of Cardiovascular Medicine, Stanford University, Stanford, CA, USA.
| |
Collapse
|
31
|
Liang D, Han D, Fan W, Zhang R, Qiao H, Fan M, Su T, Ma S, Li X, Chen J, Wang Y, Ren J, Cao F. Therapeutic efficacy of apelin on transplanted mesenchymal stem cells in hindlimb ischemic mice via regulation of autophagy. Sci Rep 2016; 6:21914. [PMID: 26902855 PMCID: PMC4763210 DOI: 10.1038/srep21914] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 02/02/2016] [Indexed: 01/15/2023] Open
Abstract
Mesenchymal stem cells (MSCs)-based therapy provides a promising avenue for the management of peripheral arterial disease (PAD). However, engrafted MSCs are subjected to acute cell death in the ischemic microenvironment. Apelin has been shown to protect bone marrow MSCs against apoptosis although the mechanism of action remains elusive. Here we demonstrated that apelin promoted functional survival of AD-MSCs in ischemic hindlimbs and provoked a synergetic effect with AD-MSCs to restore hindlimb blood perfusion and limb functions. Further in vitro studies revealed that a biphasic response in autophagy was induced by apelin in AD-MSCs during hypoxia and hypoxia/reoxygenation (H/R) stages to exert cytoprotective effects against H/R injury. Mechanistically, apelin increased the viability of AD-MSCs via promoting protective autophagy during hypoxia, which was accompanied with activation of AMPK and inhibition of mammalian target of rapamycin (mTOR). To the contrary, apelin suppressed autophagic cell death during reoxygenation, which was accompanied with activation of Akt and inhibition of Beclin1. Our findings indicated that apelin facilitated AD-MSCs-based therapy in PAD, possibly through promoting survival of AD-MSCs by way of autophagy regulation. Our data support the promises of apelin as a novel strategy to improve MSC-based therapy for PAD, possibly through autophagy modulation in MSCs.
Collapse
Affiliation(s)
- Dong Liang
- Department of Cardiology, Chinese PLA General Hospital, Beijing, 100853, China.,Department of Cardiology, Armed Police Corps Hospital of Shaanxi, Xi'an, Shaanxi 710032, China
| | - Dong Han
- Department of Cardiology, Chinese PLA General Hospital, Beijing, 100853, China.,Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Weiwei Fan
- Department of Cardiology, Chinese PLA General Hospital, Beijing, 100853, China.,Department of Cardiology, the 175th Hospital of Chinese PLA, the Affiliated Southeast Hospital of Xiamen University, Zhangzhou, Fujian 363000, China
| | - Ran Zhang
- Department of Cardiology, Chinese PLA General Hospital, Beijing, 100853, China
| | - Hongyu Qiao
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Miaomiao Fan
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Tao Su
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Sai Ma
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Xiujuan Li
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Jiangwei Chen
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Yabin Wang
- Department of Cardiology, Chinese PLA General Hospital, Beijing, 100853, China
| | - Jun Ren
- Center for Cardiovascular Research and Alternative Medicine, University of Wyoming, Laramie, WY 82071, USA
| | - Feng Cao
- Department of Cardiology, Chinese PLA General Hospital, Beijing, 100853, China.,Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| |
Collapse
|
32
|
He L, Liu Q, Hu T, Huang X, Zhang H, Tian X, Yan Y, Wang L, Huang Y, Miquerol L, Wythe JD, Zhou B. Genetic lineage tracing discloses arteriogenesis as the main mechanism for collateral growth in the mouse heart. Cardiovasc Res 2016; 109:419-30. [PMID: 26768261 PMCID: PMC4752045 DOI: 10.1093/cvr/cvw005] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 12/29/2015] [Indexed: 12/21/2022] Open
Abstract
Aims Capillary and arterial endothelial cells share many common molecular markers in both the neonatal and adult hearts. Herein, we aim to establish a genetic tool that distinguishes these two types of vessels in order to determine the cellular mechanism underlying collateral artery formation. Methods and results Using Apln-GFP and Apln-LacZ reporter mice, we demonstrate that APLN expression is enriched in coronary vascular endothelial cells. However, APLN expression is reduced in coronary arterial endothelial cells. Genetic lineage tracing, using an Apln-CreER mouse line, robustly labelled capillary endothelial cells, but not arterial endothelial cells. We leveraged this differential activity of Apln-CreER to study collateral artery formation following myocardial infarction (MI). In a neonatal heart MI model, we found that Apln-CreER-labelled capillary endothelial cells do not contribute to the large collateral arteries. Instead, these large collateral arteries mainly arise from pre-existing, infrequently labelled coronary arteries, indicative of arteriogenesis. Furthermore, in an adult heart MI model, Apln-CreER activity also distinguishes large and small diameter arteries from capillaries. Lineage tracing in this setting demonstrated that most large and small coronary arteries in the infarcted myocardium and border region are derived not from capillaries, but from pre-existing arteries. Conclusion Apln-CreER-mediated lineage tracing distinguishes capillaries from large arteries, in both the neonatal and adult hearts. Through genetic fate mapping, we demonstrate that pre-existing arteries, but not capillaries, extensively contribute to collateral artery formation following myocardial injury. These results suggest that arteriogenesis is the major mechanism underlying collateral vessel formation.
Collapse
Affiliation(s)
- Lingjuan He
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Qiaozhen Liu
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Tianyuan Hu
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xiuzhen Huang
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Hui Zhang
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xueying Tian
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yan Yan
- Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Li Wang
- Institute of Vascular Medicine, Shenzhen Research Institute, and Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong, China
| | - Yu Huang
- Institute of Vascular Medicine, Shenzhen Research Institute, and Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong, China
| | - Lucile Miquerol
- Aix Marseille Universite, CNRS, IBDM UMR 7288, Marseille 13288, France
| | - Joshua D Wythe
- Department of Molecular Physiology and Biophysics, Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA
| | - Bin Zhou
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China ShanghaiTech University, Shanghai 201210, China
| |
Collapse
|
33
|
Fibroblast Growth Factor-9 Activates c-Kit Progenitor Cells and Enhances Angiogenesis in the Infarcted Diabetic Heart. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2016:5810908. [PMID: 26682010 PMCID: PMC4670684 DOI: 10.1155/2016/5810908] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 08/09/2015] [Indexed: 12/20/2022]
Abstract
We hypothesized that fibroblast growth factor-9 (FGF-9) would enhance angiogenesis via activating c-kit positive stem cells in the infarcted nondiabetic and diabetic heart. In brief, animals were divided into three groups: Sham, MI, and MI+FGF-9. Two weeks following MI or sham surgery, our data suggest that treatment with FGF-9 significantly diminished vascular apoptosis compared to the MI group in both C57BL/6 and db/db mice (p < 0.05). Additionally, the number of c-kit+ve/SM α-actin+ve cells and c-kit+ve/CD31+ve cells were greatly enhanced in the MI+FGF-9 groups relative to the MI suggesting FGF-9 enhances c-Kit cell activation and their differentiation into vascular smooth muscle cells and endothelial cells, respectively (p < 0.05). Histology shows that the total number of vessels were quantified for all groups and our data suggest that the FGF-9 treated groups had significantly more vessels than their MI counterparts (p < 0.05). Finally, echocardiographic data suggests a significant improvement in left ventricular output, as indicated by fractional shortening and ejection fraction in both nondiabetic and diabetic animals treated with FGF-9 (p < 0.05). Overall, our data suggests FGF-9 has the potential to attenuate vascular cell apoptosis, activate c-Kit progenitor cells, and enhance angiogenesis and neovascularization in C57BL/6 and db/db mice leading to improved cardiac function.
Collapse
|
34
|
Stöhr R, Kappel BA, Carnevale D, Cavalera M, Mavilio M, Arisi I, Fardella V, Cifelli G, Casagrande V, Rizza S, Cattaneo A, Mauriello A, Menghini R, Lembo G, Federici M. TIMP3 interplays with apelin to regulate cardiovascular metabolism in hypercholesterolemic mice. Mol Metab 2015; 4:741-52. [PMID: 26500845 PMCID: PMC4588459 DOI: 10.1016/j.molmet.2015.07.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Revised: 07/23/2015] [Accepted: 07/27/2015] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVE Tissue inhibitor of metalloproteinase 3 (TIMP3) is an extracellular matrix (ECM) bound protein, which has been shown to be downregulated in human subjects and experimental models with cardiometabolic disorders, including type 2 diabetes mellitus, hypertension and atherosclerosis. The aim of this study was to investigate the effects of TIMP3 on cardiac energy homeostasis during increased metabolic stress conditions. METHODS ApoE(-/-)TIMP3(-/-) and ApoE(-/-) mice on a C57BL/6 background were subjected to telemetric ECG analysis and experimental myocardial infarction as models of cardiac stress induction. We used Western blot, qRT-PCR, histology, metabolomics, RNA-sequencing and in vivo phenotypical analysis to investigate the molecular mechanisms of altered cardiac energy metabolism. RESULTS ApoE(-/-)TIMP3(-/-) revealed decreased lifespan. Telemetric ECG analysis showed increased arrhythmic episodes, and experimental myocardial infarction by left anterior descending artery (LAD) ligation resulted in increased peri-operative mortality together with increased scar formation, ventricular dilatation and a reduction of cardiac function after 4 weeks in the few survivors. Hearts of ApoE(-/-)TIMP3(-/-) exhibited accumulation of neutral lipids when fed a chow diet, which was exacerbated by a high fat, high cholesterol diet. Metabolomics analysis revealed an increase in circulating markers of oxidative stress with a reduction in long chain fatty acids. Using whole heart mRNA sequencing, we identified apelin as a putative modulator of these metabolic defects. Apelin is a regulator of fatty acid oxidation, and we found a reduction in the levels of enzymes involved in fatty acid oxidation in the left ventricle of ApoE(-/-)TIMP3(-/-) mice. Injection of apelin restored the hitherto identified metabolic defects of lipid oxidation. CONCLUSION TIMP3 regulates lipid metabolism as well as oxidative stress response via apelin. These findings therefore suggest that TIMP3 maintains metabolic flexibility in the heart, particularly during episodes of increased cardiac stress.
Collapse
Affiliation(s)
- Robert Stöhr
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
- Department of Internal Medicine I, University Hospital Aachen, Pauwelsstraße 30, 52074 Aachen, Germany
| | - Ben Arpad Kappel
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
- Department of Internal Medicine I, University Hospital Aachen, Pauwelsstraße 30, 52074 Aachen, Germany
| | - Daniela Carnevale
- Department of Angiocardioneurology and Translational Medicine, IRCCS Neuromed, 86077 Pozzilli, IS, Italy
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Michele Cavalera
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Maria Mavilio
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Ivan Arisi
- Genomics Facility, European Brain Research Institute, Rome, Italy
| | - Valentina Fardella
- Department of Angiocardioneurology and Translational Medicine, IRCCS Neuromed, 86077 Pozzilli, IS, Italy
| | - Giuseppe Cifelli
- Department of Angiocardioneurology and Translational Medicine, IRCCS Neuromed, 86077 Pozzilli, IS, Italy
| | - Viviana Casagrande
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Stefano Rizza
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Antonino Cattaneo
- European Brain Research Institute, Rome, Italy
- Scuola Normale Superiore, Pisa, Italy
| | - Alessandro Mauriello
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Rossella Menghini
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Giuseppe Lembo
- Department of Angiocardioneurology and Translational Medicine, IRCCS Neuromed, 86077 Pozzilli, IS, Italy
| | - Massimo Federici
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
- Center for Atherosclerosis, Department of Medicine, Policlinico Tor Vergata, 00133 Rome, Italy
- Corresponding author. Department of Systems Medicine, University of Rome “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy. Tel.: +39 06 72596889; fax: +39 06 72596890.
| |
Collapse
|
35
|
Pisarenko O, Shulzhenko V, Studneva I, Pelogeykina Y, Timoshin A, Anesia R, Valet P, Parini A, Kunduzova O. Structural apelin analogues: mitochondrial ROS inhibition and cardiometabolic protection in myocardial ischaemia reperfusion injury. Br J Pharmacol 2015; 172:2933-45. [PMID: 25521429 PMCID: PMC4459014 DOI: 10.1111/bph.13038] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Revised: 10/17/2014] [Accepted: 11/26/2014] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND AND PURPOSE Mitochondria-derived oxidative stress is believed to be crucially involved in cardiac ischaemia reperfusion (I/R) injury, although currently no therapies exist that specifically target mitochondrial reactive oxygen species (ROS) production. The present study was designed to evaluate the potential effects of the structural analogues of apelin-12, an adipocyte-derived peptide, on mitochondrial ROS generation, cardiomyocyte apoptosis, and metabolic and functional recovery to myocardial I/R injury. EXPERIMENTAL APPROACH In cultured H9C2 cardiomyoblasts and adult cardiomyocytes, oxidative stress was induced by hypoxia reoxygenation. Isolated rat hearts were subjected to 35 min of global ischaemia and 30 min of reperfusion. Apelin-12, apelin-13 and structural apelin-12 analogues, AI and AII, were infused during 5 min prior to ischaemia. KEY RESULTS In cardiac cells, mitochondrial ROS production was inhibited by the structural analogues of apelin, AI and AII, in comparison with the natural peptides, apelin-12 and apelin-13. Treatment of cardiomyocytes with AI and AII decreased cell apoptosis concentration-dependently. In a rat model of I/R injury, pre-ischaemic infusion of AI and AII markedly reduced ROS formation in the myocardial effluent and attenuated cell membrane damage. Prevention of oxidative damage by AI and AII was associated with the improvement of functional and metabolic recovery after I/R in the heart. CONCLUSIONS AND IMPLICATIONS These data provide the evidence for the potential of the structural apelin analogues in selective reduction of mitochondrial ROS generation and myocardial apoptosis and form the basis for a promising therapeutic strategy in the treatment of oxidative stress-related heart disease.
Collapse
Affiliation(s)
- Oleg Pisarenko
- Russian Cardiology Research-and-Production ComplexMoscow, Russia
| | | | - Irina Studneva
- Russian Cardiology Research-and-Production ComplexMoscow, Russia
| | | | | | - Rodica Anesia
- National Institute of Health and Medical Research (INSERM) U1048Toulouse, France
- University of Toulouse, UPS, Institute of Metabolic and Cardiovascular DiseasesToulouse, France
| | - Philippe Valet
- National Institute of Health and Medical Research (INSERM) U1048Toulouse, France
- University of Toulouse, UPS, Institute of Metabolic and Cardiovascular DiseasesToulouse, France
| | - Angelo Parini
- National Institute of Health and Medical Research (INSERM) U1048Toulouse, France
- University of Toulouse, UPS, Institute of Metabolic and Cardiovascular DiseasesToulouse, France
| | - Oksana Kunduzova
- National Institute of Health and Medical Research (INSERM) U1048Toulouse, France
- University of Toulouse, UPS, Institute of Metabolic and Cardiovascular DiseasesToulouse, France
| |
Collapse
|
36
|
Sevá Pessôa B, Becher PM, Van Veghel R, De Vries R, Tempel D, Sneep S, Van Beusekom H, Van Der Velden VHJ, Westermann D, Danser AHJ, Roks AJM. Effect of a stable Angiotensin-(1-7) analogue on progenitor cell recruitment and cardiovascular function post myocardial infarction. J Am Heart Assoc 2015; 4:jah3823. [PMID: 25655571 PMCID: PMC4345874 DOI: 10.1161/jaha.114.001510] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Background Angiotensin‐(1–7) improves cardiac function and remodeling after myocardial infarction (MI). This may involve recruitment of hematopoietic progenitor cells that support angiogenesis. However, angiotensin‐(1–7) is rapidly metabolized in plasma and tissue. The authors investigated in mice the effect of a metabolically stable angiotensin‐(1–7) analogue, cyclic angiotensin‐(1–7), on progenitor cell recruitment and on the heart post MI, when given in the angiogenesis phase of remodeling. Methods and Results Angiogenic progenitor cell recruitment was measured by using flow cytometry 24 and 72 hours after a daily bolus injection of cyclic angiotensin‐(1–7) in healthy C57BL/6 mice. Further, mice underwent MI or sham surgery and subsequently received saline or 2 different doses of cyclic angiotensin‐(1–7) for 3 or 9 weeks. Cyclic angiotensin‐(1–7) increased circulating hematopoietic progenitor cells at 24 hours but not 72 hours. Post MI, cyclic angiotensin‐(1–7) diminished cardiomyocyte hypertrophy and reduced myogenic tone, without altering cardiovascular function or cardiac histology at 9 weeks. Importantly, cyclic angiotensin‐(1–7)–treated mice had reduced cardiac capillary density at 3 weeks after MI but not after 9 weeks. Finally, cyclic angiotensin‐(1–7) decreased tube formation by cultured human umbilical vein endothelial cells. Conclusions Our results suggest that cyclic angiotensin‐(1–7), when given early after MI, recruits progenitor cells but does not lead to improved angiogenesis, most likely because it simultaneously exerts antiangiogenic effect in adult endothelial cells. Apparently, optimal treatment with cyclic angiotensin‐(1–7) depends on the time point of onset of application after MI.
Collapse
Affiliation(s)
- Bruno Sevá Pessôa
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC, Erasmus University Medical Center, Rotterdam, The Netherlands (B.S.P., R.V.V., R.D.V., J.D., A.M.R.)
| | - Peter Moritz Becher
- Department of General and Interventional Cardiology, University Heart Center Hamburg Eppendorf, Germany (P.M.B., D.W.)
| | - Richard Van Veghel
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC, Erasmus University Medical Center, Rotterdam, The Netherlands (B.S.P., R.V.V., R.D.V., J.D., A.M.R.)
| | - René De Vries
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC, Erasmus University Medical Center, Rotterdam, The Netherlands (B.S.P., R.V.V., R.D.V., J.D., A.M.R.)
| | - Dennie Tempel
- Division of Cardiology and Pulmonology, Department of Interventional Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands (D.T.)
| | - Stefan Sneep
- Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus MC, Erasmus University Medical Center, Rotterdam, The Netherlands (S.S., H.V.B.)
| | - Heleen Van Beusekom
- Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus MC, Erasmus University Medical Center, Rotterdam, The Netherlands (S.S., H.V.B.)
| | - Vincent H J Van Der Velden
- Department of Immunology, Erasmus MC, Erasmus University Medical Center, Rotterdam, The Netherlands (V.J.V.D.V.)
| | - Dirk Westermann
- Department of General and Interventional Cardiology, University Heart Center Hamburg Eppendorf, Germany (P.M.B., D.W.)
| | - A H Jan Danser
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC, Erasmus University Medical Center, Rotterdam, The Netherlands (B.S.P., R.V.V., R.D.V., J.D., A.M.R.)
| | - Anton J M Roks
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC, Erasmus University Medical Center, Rotterdam, The Netherlands (B.S.P., R.V.V., R.D.V., J.D., A.M.R.)
| |
Collapse
|
37
|
Genetic targeting of sprouting angiogenesis using Apln-CreER. Nat Commun 2015; 6:6020. [PMID: 25597280 PMCID: PMC4309445 DOI: 10.1038/ncomms7020] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Accepted: 12/02/2014] [Indexed: 02/07/2023] Open
Abstract
Under pathophysiological conditions in adults, endothelial cells (ECs) sprout from pre-existing blood vessels to form new ones by a process termed angiogenesis. During embryonic development, Apelin (APLN) is robustly expressed in vascular ECs. In adult mice, however, APLN expression in the vasculature is significantly reduced. Here we show that APLN expression is reactivated in adult ECs after ischaemia insults. In models of both injury ischaemia and tumor angiogenesis, we find that Apln-CreER genetically labels sprouting but not quiescent vasculature. By leveraging this specific activity, we demonstrate that abolishment of the VEGF-VEGFR2 signalling pathway as well as ablation of sprouting ECs diminished tumour vascularization and growth without compromising vascular homeostasis in other organs. Collectively, we show that Apln-CreER distinguishes sprouting vessels from stabilized vessels in multiple pathological settings. The Apln-CreER line described here will greatly aid future mechanistic studies in both vascular developmental biology and adult vascular diseases.
Collapse
|
38
|
Bai B, Cai X, Jiang Y, Karteris E, Chen J. Heterodimerization of apelin receptor and neurotensin receptor 1 induces phosphorylation of ERK(1/2) and cell proliferation via Gαq-mediated mechanism. J Cell Mol Med 2014; 18:2071-81. [PMID: 25164432 PMCID: PMC4244021 DOI: 10.1111/jcmm.12404] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Accepted: 07/18/2014] [Indexed: 01/25/2023] Open
Abstract
Dimerization of G protein-coupled receptors (GPCRs) is crucial for receptor function including agonist affinity, efficacy, trafficking and specificity of signal transduction, including G protein coupling. Emerging data suggest that the cardiovascular system is the main target of apelin, which exerts an overall neuroprotective role, and is a positive regulator of angiotensin-converting enzyme 2 (ACE2) in heart failure. Moreover, ACE2 cleaves off C-terminal residues of vasoactive peptides including apelin-13, and neurotensin that activate the apelin receptor (APJ) and neurotensin receptor 1 (NTSR1) respectively, that belong to the A class of GPCRs. Therefore, based on the similar mode of modification by ACE2 at peptide level, the homology at amino acid level and the capability of forming dimers with other GPCRs, we have been suggested that APJ and NTSR1 can form a functional heterodimer. Using co-immunoprecipitation, BRET and FRET, we provided conclusive evidence of heterodimerization between APJ and NTSR1 in a constitutive and induced form. Upon agonist stimulation, hetrodimerization enhanced ERK1/2 activation and increased proliferation via activation of Gq α-subunits. These novel data provide evidence for a physiological role of APJ/NTSR1 heterodimers in terms of ERK1/2 activation and increased intracellular calcium and induced cell proliferation and provide potential new pharmaceutical targets for cardiovascular disease.
Collapse
Affiliation(s)
- Bo Bai
- Neurobiology Institute, Jining Medical University, Jining, Shandong, China
| | | | | | | | | |
Collapse
|
39
|
Xie F, Lv D, Chen L. ELABELA: a novel hormone in cardiac development acting as a new endogenous ligand for the APJ receptor. Acta Biochim Biophys Sin (Shanghai) 2014; 46:620-2. [PMID: 24829400 DOI: 10.1093/abbs/gmu032] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Affiliation(s)
- Feng Xie
- Learning Key Laboratory for Pharmaco-proteomics, Institute of Pharmacy and Pharmacology, University of South China, Hengyang 421001, China
| | - Deguan Lv
- Learning Key Laboratory for Pharmaco-proteomics, Institute of Pharmacy and Pharmacology, University of South China, Hengyang 421001, China
| | - Linxi Chen
- Learning Key Laboratory for Pharmaco-proteomics, Institute of Pharmacy and Pharmacology, University of South China, Hengyang 421001, China
| |
Collapse
|
40
|
Pauli A, Norris ML, Valen E, Chew GL, Gagnon JA, Zimmerman S, Mitchell A, Ma J, Dubrulle J, Reyon D, Tsai SQ, Joung JK, Saghatelian A, Schier AF. Toddler: an embryonic signal that promotes cell movement via Apelin receptors. Science 2014; 343:1248636. [PMID: 24407481 DOI: 10.1126/science.1248636] [Citation(s) in RCA: 486] [Impact Index Per Article: 44.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
It has been assumed that most, if not all, signals regulating early development have been identified. Contrary to this expectation, we identified 28 candidate signaling proteins expressed during zebrafish embryogenesis, including Toddler, a short, conserved, and secreted peptide. Both absence and overproduction of Toddler reduce the movement of mesendodermal cells during zebrafish gastrulation. Local and ubiquitous production of Toddler promote cell movement, suggesting that Toddler is neither an attractant nor a repellent but acts globally as a motogen. Toddler drives internalization of G protein-coupled APJ/Apelin receptors, and activation of APJ/Apelin signaling rescues toddler mutants. These results indicate that Toddler is an activator of APJ/Apelin receptor signaling, promotes gastrulation movements, and might be the first in a series of uncharacterized developmental signals.
Collapse
Affiliation(s)
- Andrea Pauli
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Yu XH, Tang ZB, Liu LJ, Qian H, Tang SL, Zhang DW, Tian GP, Tang CK. Apelin and its receptor APJ in cardiovascular diseases. Clin Chim Acta 2014; 428:1-8. [DOI: 10.1016/j.cca.2013.09.001] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2013] [Revised: 08/31/2013] [Accepted: 09/01/2013] [Indexed: 12/29/2022]
|
42
|
Myocardial injection of apelin-overexpressing bone marrow cells improves cardiac repair via upregulation of Sirt3 after myocardial infarction. PLoS One 2013; 8:e71041. [PMID: 24039710 PMCID: PMC3765164 DOI: 10.1371/journal.pone.0071041] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Accepted: 06/26/2013] [Indexed: 12/29/2022] Open
Abstract
Our previous study shows that treatment with apelin increases bone marrow cells (BMCs) recruitment and promotes cardiac repair after myocardial infarction (MI). The objective of this study was to investigate whether overexpression of apelin in BMCs improved cell therapy and accelerated cardiac repair and functional recovery in post-MI mice. Mouse myocardial infarction was achieved by coronary artery ligation and BMCs overexpressing apelin (apelin-BMCs) or GFP (GFP-BMCs) were injected into ischemic area immediately after surgery. In vitro, exposure of cultured BMCs to apelin led to a gradual increase in SDF-1á and CXCR4 expression. Intramyocardial delivery of apelin-BMCs in post-MI mice resulted in a significant increase number of APJ+/c-kit+/Sca1+ cells in the injected area compared to GFP-BMCs treated post-MI mice. Treatment with apelin-BMCs increased expression of VEGF, Ang-1 and Tie-2 in post-MI mice. Apelin-BMCs treatment also significantly increased angiogenesis and attenuated cardiac fibrosis formation in post-MI mice. Most importantly, treatment with apelin-BMCs significantly improved left ventricular (LV) systolic function in post-MI mice. Mechanistically, Apelin-BMCs treatment led to a significant increase in Sirtuin3 (Sirt3) expression and reduction of reactive oxygen species (ROS) formation. Treatment of cultured BMCs with apelin also increased Notch3 expression and Akt phosphorylation. Apelin treatment further attenuated stress-induced apoptosis whereas knockout of Sirt3 abolished anti-apoptotic effect of apelin in cultured BMCs. Moreover, knockout of Sirt3 significantly attenuated apelin-BMCs-induced VEGF expression and angiogenesis in post-MI mice. Knockout of Sirt3 further blunted apelin-BMCs-mediated improvement of cardiac repair and systolic functional recovery in post-MI mice. These data suggest that apelin improves BMCs therapy on cardiac repair and systolic function in post-MI mice. Upregulation of Sirt3 may contribute to the protective effect of apelin-BMCs therapy.
Collapse
|
43
|
Mechanism underlying the inhibitory effect of Apelin-13 on glucose deprivation-induced autophagy in rat cardiomyocytes. Exp Ther Med 2013; 5:797-802. [PMID: 23403698 PMCID: PMC3570085 DOI: 10.3892/etm.2013.902] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2012] [Accepted: 01/08/2013] [Indexed: 12/03/2022] Open
Abstract
The aim of the present study was to investigate the effect of Apelin-13 on cardiomyocyte autophagy and to determine the underlying mechanism of this effect. To establish an autophagic model system, the cardiomyocytes of Sprague Dawley rats (postnatal day 3) were cultured and divided into five groups: normal control (Co), glucose deprivation (GD), GD+Apelin-13, GD+Apelin-13 treated with the Akt-specific inhibitor triciribine (GD+Apelin-13+Triciribine) and triciribine alone (Triciribine). The intracellular autophagosomes were then observed using transmission electron microscopy (TEM) and the expression levels of cellular autophagy-related protein microtubule-associated protein 1 light chain 3 (LC3), phosphatidylinositol-3-kinase (PI3K) and mammalian target of rapamycin (mTOR) protein were measured using western blotting. Compared with the Co group, the ratio of LC3-II/LC3-I increased significantly in all treatment groups, with the exception of the Triciribine group (P<0.05). Compared with the GD group, the ratio of LC3-II/LC3-I was significantly decreased, and the PI3K and mTOR expression was significantly enhanced in the GD+Apelin-13 and GD+Apelin-13+Triciribine groups (P<0.05). Compared with the GD+Apelin-13 group, the ratio of LC3-II/LC3-I increased significantly (P<0.05) and the PI3K expression remained unchanged in the GD+Apelin-13+Triciribine group (P>0.05), but mTOR expression was significantly reduced (P<0.05). GD led to increased numbers of autophagosomes and augmented the LC3-II/LC3-I ratio (P<0.05). Apelin-13 pretreatment attenuated GD-induced cardiomyocte injury, decreased the autophagosome number and the ratio of LC3-II/LC3-I (P<0.05), enhanced PI3K activity (P<0.05) and upregulated the phosphorylation levels of the Akt and mTOR proteins (P<0.05). The Akt-specific inhibitor triciribine weakened the protective role of Apelin-13 (P<0.05). To a certain extent, Apelin-13 inhibited GD-induced cardiomyocyte autophagy, which may be related in part to the activation of the PI3K/Akt/mTOR signaling pathway.
Collapse
|