1
|
Kishore R, Magadum A. Cell-Specific mRNA Therapeutics for Cardiovascular Diseases and Regeneration. J Cardiovasc Dev Dis 2024; 11:38. [PMID: 38392252 PMCID: PMC10889436 DOI: 10.3390/jcdd11020038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 01/23/2024] [Accepted: 01/24/2024] [Indexed: 02/24/2024] Open
Abstract
Cardiovascular diseases (CVDs) represent a significant global health burden, demanding innovative therapeutic approaches. In recent years, mRNA therapeutics have emerged as a promising strategy to combat CVDs effectively. Unlike conventional small-molecule drugs, mRNA therapeutics enable the direct modulation of cellular functions by delivering specific mRNA molecules to target cells. This approach offers unprecedented advantages, including the ability to harness endogenous cellular machinery for protein synthesis, thus allowing precise control over gene expression without insertion into the genome. This review summarizes the current status of the potential of cell-specific mRNA therapeutics in the context of cardiovascular diseases. First, it outlines the challenges associated with traditional CVD treatments and emphasizes the need for targeted therapies. Subsequently, it elucidates the underlying principles of mRNA therapeutics and the development of advanced delivery systems to ensure cell-specificity and enhanced efficacy. Notably, innovative delivery methods such as lipid nanoparticles and exosomes have shown promise in improving the targeted delivery of mRNA to cardiac cells, activated fibroblasts, and other relevant cell types. Furthermore, the review highlights the diverse applications of cell-specific mRNA therapeutics in addressing various aspects of cardiovascular diseases, including atherosclerosis, myocardial infarction, heart failure, and arrhythmias. By modulating key regulatory genes involved in cardiomyocyte proliferation, inflammation, angiogenesis, tissue repair, and cell survival, mRNA therapeutics hold the potential to intervene at multiple stages of CVD pathogenesis. Despite its immense potential, this abstract acknowledges the challenges in translating cell-specific mRNA therapeutics from preclinical studies to clinical applications like off-target effects and delivery. In conclusion, cell-specific mRNA therapeutics have emerged as a revolutionary gene therapy approach for CVD, offering targeted interventions with the potential to significantly improve patient outcomes.
Collapse
Affiliation(s)
- Raj Kishore
- Department of Cardiovascular Sciences, Temple University, Philadelphia, PA 19140, USA
| | - Ajit Magadum
- Department of Cardiovascular Sciences, Temple University, Philadelphia, PA 19140, USA
| |
Collapse
|
2
|
Magadum A. Modified mRNA Therapeutics for Heart Diseases. Int J Mol Sci 2022; 23:ijms232415514. [PMID: 36555159 PMCID: PMC9779737 DOI: 10.3390/ijms232415514] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 11/26/2022] [Accepted: 11/27/2022] [Indexed: 12/13/2022] Open
Abstract
Cardiovascular diseases (CVD) remain a substantial global health problem and the leading cause of death worldwide. Although many conventional small-molecule treatments are available to support the cardiac function of the patient with CVD, they are not effective as a cure. Among potential targets for gene therapy are severe cardiac and peripheral ischemia, heart failure, vein graft failure, and some forms of dyslipidemias. In the last three decades, multiple gene therapy tools have been used for heart diseases caused by proteins, plasmids, adenovirus, and adeno-associated viruses (AAV), but these remain as unmet clinical needs. These gene therapy methods are ineffective due to poor and uncontrolled gene expression, low stability, immunogenicity, and transfection efficiency. The synthetic modified mRNA (modRNA) presents a novel gene therapy approach which provides a transient, stable, safe, non-immunogenic, controlled mRNA delivery to the heart tissue without any risk of genomic integration, and achieves a therapeutic effect in different organs, including the heart. The mRNA translation starts in minutes, and remains stable for 8-10 days (pulse-like kinetics). The pulse-like expression of modRNA in the heart induces cardiac repair, cardiomyocyte proliferation and survival, and inhibits cardiomyocyte apoptosis post-myocardial infarction (MI). Cell-specific (cardiomyocyte) modRNA translation developments established cell-specific modRNA therapeutics for heart diseases. With these laudable characteristics, combined with its expression kinetics in the heart, modRNA has become an attractive therapeutic for the treatment of CVD. This review discusses new developments in modRNA therapy for heart diseases.
Collapse
Affiliation(s)
- Ajit Magadum
- Center for Translational Medicine, Temple University, Philadelphia, PA 19140, USA
| |
Collapse
|
3
|
Pi Z, Johnson JA, Meng W, Phillips M, Schumacher WA, Bostwick JS, Gargalovic PS, Onorato JM, Generaux CN, Wang T, He Y, Gordon DA, Wexler RR, Finlay HJ. Identification of 6-Hydroxypyrimidin-4(1 H)-one-3-carboxamides as Potent and Orally Active APJ Receptor Agonists. ACS Med Chem Lett 2021; 12:1766-1772. [PMID: 34795866 DOI: 10.1021/acsmedchemlett.1c00385] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 10/18/2021] [Indexed: 01/01/2023] Open
Abstract
The apelin receptor (APJ) is a significant regulator of cardiovascular function and is involved in heart failure and other cardiovascular diseases. (Pyr1)apelin-13 is one of the endogenous agonists of the APJ receptor. Administration of (Pyr1)apelin-13 increases cardiac output in preclinical models and humans. Recently we disclosed clinical lead BMS-986224 (1), a C3 oxadiazole pyridinone APJ receptor agonist with robust pharmacodynamic effects similar to (Pyr1)apelin-13 in an acute rat pressure-volume loop model. Herein we describe the structure-activity relationship of the carboxamides as oxadiazole bioisosteres at C3 of the pyridinone core and C5 of the respective pyrimidinone core. This study led to the identification of structurally differentiated 6-hydroxypyrimidin-4(1H)-one-3-carboxamide 14a with pharmacodynamic effects comparable to those of compound 1.
Collapse
Affiliation(s)
- Zulan Pi
- Research and Development, Bristol Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543-5400, United States
| | - James A. Johnson
- Research and Development, Bristol Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543-5400, United States
| | - Wei Meng
- Research and Development, Bristol Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543-5400, United States
| | - Monique Phillips
- Research and Development, Bristol Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543-5400, United States
| | - William A. Schumacher
- Research and Development, Bristol Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543-5400, United States
| | - Jeffrey S. Bostwick
- Research and Development, Bristol Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543-5400, United States
| | - Peter S. Gargalovic
- Research and Development, Bristol Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543-5400, United States
| | - Joelle M. Onorato
- Research and Development, Bristol Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543-5400, United States
| | - Claudia N. Generaux
- Research and Development, Bristol Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543-5400, United States
| | - Tao Wang
- Research and Development, Bristol Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543-5400, United States
| | - Yan He
- Research and Development, Bristol Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543-5400, United States
| | - David A. Gordon
- Research and Development, Bristol Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543-5400, United States
| | - Ruth R. Wexler
- Research and Development, Bristol Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543-5400, United States
| | - Heather J. Finlay
- Research and Development, Bristol Myers Squibb Company, P.O. Box 5400, Princeton, New Jersey 08543-5400, United States
| |
Collapse
|
4
|
Agarwal KA, Kyriazis PP, Lecker SH. RAAS-Blockade in COVID-19: The Ace of Spades? Indian J Nephrol 2021; 31:423-424. [PMID: 34584367 PMCID: PMC8443096 DOI: 10.4103/ijn.ijn_322_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 08/28/2020] [Accepted: 09/04/2020] [Indexed: 12/02/2022] Open
Affiliation(s)
- Krishna A. Agarwal
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | | | - Stewart H. Lecker
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| |
Collapse
|
5
|
Gangnus T, Burckhardt BB. Low-volume LC-MS/MS method for the pharmacokinetic investigation of carvedilol, enalapril and their metabolites in whole blood and plasma: Application to a paediatric clinical trial. Drug Test Anal 2020; 13:694-708. [PMID: 33126289 DOI: 10.1002/dta.2949] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 10/01/2020] [Accepted: 10/13/2020] [Indexed: 11/09/2022]
Abstract
Evidence-based pharmacotherapy with carvedilol and enalapril in children suffering from heart failure is insufficient owing to limited pharmacokinetic data. Although a few data sets regarding enalapril, its metabolite enalaprilat and carvedilol in children have been published, pharmacokinetic data on carvedilol metabolites are missing. However, for both drug substances, their active metabolites contribute substantially to drug efficacy. As data can hardly be derived from adults owing to the unknown impacts of enzymatic maturation and ontogeny during childhood, customised assays are important to facilitate paediatric evidence-based pharmacotherapy. Considering ethical paediatric constraints, a low-volume liquid chromatography coupled to mass spectrometry (LC-MS/MS) assay was developed using whole blood or plasma for the quantification of enalapril, enalaprilat, carvedilol, O-desmethyl carvedilol, 4- and 5-hydroxyphenyl carvedilol as well as 3- and 8-hydroxy carvedilol. To facilitate broader applications in adults, the elderly and children, a wide calibration range-between 0.024/0.049 and 50.000 ng/ml-was achieved with good linearity (r ≥ 0.995 for all analytes). In compliance with international bioanalytical guidelines, accuracy, precision, sensitivity and internal standard normalised matrix effects were further successfully validated with the exception of those for 3-hydroxy carvedilol, which was therefore assessed semi-quantitatively. Distinct haematocrits did not impact matrix effects or recoveries when analysing whole blood. Blood-to-plasma ratios were determined for all analytes to form the basis for pharmacokinetic modelling. Finally, incurred sample reanalysis of paediatric samples confirmed the reproducibility of the developed low-volume LC-MS/MS method during study sample analysis. The assay facilitates the reliable generation of important data and contributes towards a safe drug therapy in children.
Collapse
Affiliation(s)
- Tanja Gangnus
- Institute of Clinical Pharmacy and Pharmacotherapy, Heinrich Heine University, Düesseldorf, Germany
| | - Bjoern B Burckhardt
- Institute of Clinical Pharmacy and Pharmacotherapy, Heinrich Heine University, Düesseldorf, Germany
| | -
- Institute of Clinical Pharmacy and Pharmacotherapy, Heinrich Heine University, Düesseldorf, Germany
| |
Collapse
|
6
|
Njegic A, Wilson C, Cartwright EJ. Targeting Ca 2 + Handling Proteins for the Treatment of Heart Failure and Arrhythmias. Front Physiol 2020; 11:1068. [PMID: 33013458 PMCID: PMC7498719 DOI: 10.3389/fphys.2020.01068] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 08/04/2020] [Indexed: 12/18/2022] Open
Abstract
Diseases of the heart, such as heart failure and cardiac arrhythmias, are a growing socio-economic burden. Calcium (Ca2+) dysregulation is key hallmark of the failing myocardium and has long been touted as a potential therapeutic target in the treatment of a variety of cardiovascular diseases (CVD). In the heart, Ca2+ is essential for maintaining normal cardiac function through the generation of the cardiac action potential and its involvement in excitation contraction coupling. As such, the proteins which regulate Ca2+ cycling and signaling play a vital role in maintaining Ca2+ homeostasis. Changes to the expression levels and function of Ca2+-channels, pumps and associated intracellular handling proteins contribute to altered Ca2+ homeostasis in CVD. The remodeling of Ca2+-handling proteins therefore results in impaired Ca2+ cycling, Ca2+ leak from the sarcoplasmic reticulum and reduced Ca2+ clearance, all of which contributes to increased intracellular Ca2+. Currently, approved treatments for targeting Ca2+ handling dysfunction in CVD are focused on Ca2+ channel blockers. However, whilst Ca2+ channel blockers have been successful in the treatment of some arrhythmic disorders, they are not universally prescribed to heart failure patients owing to their ability to depress cardiac function. Despite the progress in CVD treatments, there remains a clear need for novel therapeutic approaches which are able to reverse pathophysiology associated with heart failure and arrhythmias. Given that heart failure and cardiac arrhythmias are closely associated with altered Ca2+ homeostasis, this review will address the molecular changes to proteins associated with both Ca2+-handling and -signaling; their potential as novel therapeutic targets will be discussed in the context of pre-clinical and, where available, clinical data.
Collapse
Affiliation(s)
- Alexandra Njegic
- Division of Cardiovascular Sciences, The University of Manchester, Manchester, United Kingdom.,Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Claire Wilson
- Division of Cardiovascular Sciences, The University of Manchester, Manchester, United Kingdom.,Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Elizabeth J Cartwright
- Division of Cardiovascular Sciences, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
7
|
Awwad ZM, El-Ganainy SO, ElMallah AI, Khattab MM, El-Khatib AS. Telmisartan and captopril ameliorate pregabalin-induced heart failure in rats. Toxicology 2019; 428:152310. [PMID: 31629013 DOI: 10.1016/j.tox.2019.152310] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Revised: 09/19/2019] [Accepted: 10/15/2019] [Indexed: 12/17/2022]
Abstract
Pregabalin (PRG) is highly effective in the treatment of epilepsy, neuropathic pain and anxiety disorders. Despite its potential benefits, PRG administration has been reported to induce or exacerbate heart failure (HF). It has been previously documented that overactivation of the renin angiotensin system (RAS) is involved in HF pathophysiological mechanism. The target of the current study was to examine the possible cardioprotective effect of telmisartan (Tel), an angiotensin II type 1 receptor (AT1R) blocker, compared with that of captopril (Cap), an angiotensin converting enzyme (ACE) inhibitor, in ameliorating PRG-induced HF in rats by assessing morphometric, echocardiographic and histopathological parameters. Furthermore, to investigate the role of RAS blockade by the two drugs in guarding against PRG-induced changes in cardiac angiotensin 1-7 (Ang 1-7) and angiotensin II (Ang II) levels, in addition to myocardial expression of ACE2, ACE, Mas receptor (MasR) and AT1R. Results showed that PRG administration induced morphometric, echocardiographic and histopathological deleterious alterations and significantly elevated cardiac Ang II, ACE and AT1R levels, while reduced Ang 1-7, ACE2 and MasR cardiac levels. Concurrent treatment with either Tel or Cap reversed PRG-induced morphometric, echocardiographic and histopathological abnormalities and revealed prominent protection against PRG-induced HF via downregulation of ACE/Ang II/AT1R and upregulation of ACE2/Ang 1-7/MasR axes. These are the first findings to demonstrate that the potential benefits of Tel and Cap are mediated by counteracting the altered balance between the RAS axes induced by PRG. Hence; Tel and Cap may attenuate PRG-induced HF partially through stimulation of ACE2/Ang 1-7/MasR pathway.
Collapse
Affiliation(s)
- Zeinab M Awwad
- Department of Pharmacology and Therapeutics, Faculty of Pharmacy and Drug Manufacturing, Pharos University in Alexandria, Alexandria, Egypt.
| | - Samar O El-Ganainy
- Department of Pharmacology and Therapeutics, Faculty of Pharmacy and Drug Manufacturing, Pharos University in Alexandria, Alexandria, Egypt
| | - Ahmed I ElMallah
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Mahmoud M Khattab
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Aiman S El-Khatib
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| |
Collapse
|
8
|
Kim H, Baik SY, Yang SJ, Kim TM, Lee SH, Cho JH, Choi IY, Kim JH, Yoon KH, Kim HS. Clinical experiences and case review of angiotensin II receptor blocker-related angioedema in Korea. Basic Clin Pharmacol Toxicol 2018; 124:115-122. [PMID: 30003686 DOI: 10.1111/bcpt.13097] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 07/10/2018] [Indexed: 12/01/2022]
Abstract
Although angiotensin-converting enzyme inhibitor-related angioedema is well known, angiotensin II receptor blocker (ARB)-related angioedema has not been extensively studied because of its lower incidence. Therefore, ARB-related angioedema is likely to be overlooked in the clinical setting. We analysed the medical records of adults who had been prescribed ARB and diagnosed with angioedema between 2009 and 2015. All adults over the age of 18 years who were initially administered ARB between 1 January 2009 and 31 December 2015 were selected as participants in this study. To confirm whether the angioedema was actually due to the administration of ARB, we conducted a chart review. A total of 35 584 patients were prescribed ARB for the first time when visiting the Seoul St. Mary's Hospital during the study period. Twenty-four patients diagnosed with angioedema for other reasons prior to their first prescription of ARB were excluded from this study. ARB-related angioedema was suspected in six of 35 560 patients (0.02%) who were initially prescribed ARB during the study period. The manifestation of ARB-related angioedema ranged from several days (1/6 case) to several years (3/6 cases). Some patients continued taking ARB with intermittent antihistamine or steroid therapy. In such cases, angioedema symptoms improved but did not completely resolve. Its diagnosis can be delayed and the symptoms may be recurrent as symptoms improve with antihistamine use. In some cases, the same person had different reactions depending on the type of ARB. Definitively diagnosing ARB-related angioedema is difficult, and physicians often overlook angioedema without suspecting that it is an adverse effect of ARB. Close attention of physicians and improved patient education can reduce the incidence of ARB-related angioedema.
Collapse
Affiliation(s)
- Hyunah Kim
- College of Pharmacy, Sookmyung Women's University, Seoul, Korea
| | - Seo Yeon Baik
- Department of Medical Informatics, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - So Jung Yang
- Department of Medical Informatics, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Tong Min Kim
- Department of Medical Informatics, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Seung-Hwan Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jae Hyoung Cho
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - In Young Choi
- Department of Medical Informatics, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Ju Han Kim
- Division of Biomedical Informatics, Systems Biomedical Informatics Research Centre, Seoul National University College of Medicine, Seoul, Korea
| | - Kun-Ho Yoon
- Department of Medical Informatics, College of Medicine, The Catholic University of Korea, Seoul, Korea.,Division of Endocrinology and Metabolism, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Hun-Sung Kim
- Department of Medical Informatics, College of Medicine, The Catholic University of Korea, Seoul, Korea.,Division of Endocrinology and Metabolism, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
9
|
Yan MJ, Tian ZS, Zhao ZH, Yang P. MiR-31a-5p protects myocardial cells against apoptosis by targeting Tp53. Mol Med Rep 2017; 17:3898-3904. [PMID: 29286111 DOI: 10.3892/mmr.2017.8357] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 09/11/2017] [Indexed: 11/06/2022] Open
Abstract
The pathogenesis and progression of heart failure (HF) involves multiple mechanisms, including the increased activity of the renin-angiotensin-aldosterone system, apoptosis and differential expression of microRNAs (miRNAs/miRs). Our previous study revealed an increase in miR‑31a‑5p levels in the failing hearts of a rat HF model. In the present study, whether and how miR‑31a‑5p mediates angiotensin II (AngII)‑induced apoptosis in the cardiac H9C2 cell line, was investigated using molecular biological approaches, including reverse transcription followed by quantitative polymerase chain reaction, western blotting, RNA arrays, and mutagenesis. It was demonstrated that AngII stimulation increased apoptosis and decreased miR‑31a‑5p expression, which coincided with increased tumor protein p53 (Tp53) levels. Overexpression of miR‑31a‑5p significantly suppressed the AngII‑induced apoptotic rate and caspase‑3 activity, while suppression of miR‑31a‑5p did the opposite. A total of 16 proapoptotic genes that were downregulated and 4 antiapoptotic genes that were upregulated in the miR‑31a‑5p‑overexpressed cells were identified. It was also revealed that Tp53 mRNA contained the seed sequence in its 3'‑untranslated region for miR‑31a‑5p binding. The luciferase reporter analysis showed that miR‑31a‑5p repressed the luciferase activity of the wild‑type seed sequence, but not the mutated seed sequence fused to a reporter construct. Thus, it was demonstrated that miR‑31a‑5p mediated AngII‑triggered apoptosis in myocardial cells at least partially through targeting Tp53. These findings advance the understanding of the functional interaction between miRNAs and Tp53 in the setting of cardiac diseases. Further work is required to explore whether miR‑31a‑5p can serve as a therapeutic target for HF treatment in vivo.
Collapse
Affiliation(s)
- Meng-Jie Yan
- Department of Internal Medicine and Cardiology, China‑Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Zhi-Sen Tian
- Department of Orthopedics, China‑Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Zhi-Hui Zhao
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun, Jilin 130033, P.R. China
| | - Ping Yang
- Department of Internal Medicine and Cardiology, China‑Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| |
Collapse
|
10
|
Atherogenic Cytokines Regulate VEGF-A-Induced Differentiation of Bone Marrow-Derived Mesenchymal Stem Cells into Endothelial Cells. Stem Cells Int 2015; 2015:498328. [PMID: 26106428 PMCID: PMC4464597 DOI: 10.1155/2015/498328] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 05/11/2015] [Indexed: 11/17/2022] Open
Abstract
Coronary artery stenting or angioplasty procedures frequently result in long-term endothelial dysfunction or loss and complications including arterial thrombosis and myocardial infarction. Stem cell-based therapies have been proposed to support endothelial regeneration. Mesenchymal stem cells (MSCs) differentiate into endothelial cells (ECs) in the presence of VEGF-A in vitro. Application of VEGF-A and MSC-derived ECs at the interventional site is a complex clinical challenge. In this study, we examined the effect of atherogenic cytokines (IL-6, TNFα, and Ang II) on EC differentiation and function. MSCs (CD44(+), CD73(+), CD90(+), CD14(-), and CD45(-)) were isolated from the bone marrow of Yucatan microswine. Naïve MSCs cultured in differentiation media containing VEGF-A (50 ng/mL) demonstrated increased expression of EC-specific markers (vWF, PECAM-1, and VE-cadherin), VEGFR-2 and Sox18, and enhanced endothelial tube formation. IL-6 or TNFα caused a dose-dependent attenuation of EC marker expression in VEGF-A-stimulated MSCs. In contrast, Ang II enhanced EC marker expression in VEGF-A-stimulated MSCs. Addition of Ang II to VEGF-A and IL-6 or TNFα was sufficient to rescue the EC phenotype. Thus, Ang II promotes but IL-6 and TNFα inhibit VEGF-A-induced differentiation of MSCs into ECs. These findings have important clinical implications for therapies intended to increase cardiac vascularity and reendothelialize coronary arteries following intervention.
Collapse
|
11
|
Ikhapoh IA, Pelham CJ, Agrawal DK. Synergistic effect of angiotensin II on vascular endothelial growth factor-A-mediated differentiation of bone marrow-derived mesenchymal stem cells into endothelial cells. Stem Cell Res Ther 2015; 6:4. [PMID: 25563650 PMCID: PMC4417220 DOI: 10.1186/scrt538] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Revised: 12/18/2014] [Accepted: 12/18/2014] [Indexed: 12/31/2022] Open
Abstract
INTRODUCTION Increased levels of angiotensin II (Ang II) and activity of Ang II receptor type 1 (AT1R) elicit detrimental effects in cardiovascular disease. However, the role of Ang II receptor type 2 (AT2R) remains poorly defined. Mesenchymal stem cells (MSCs) replenish and repair endothelial cells in the cardiovascular system. Herein, we investigated a novel role of angiotensin signaling in enhancing vascular endothelial growth factor (VEGF)-A-mediated differentiation of MSCs into endothelial cells (ECs). METHODS Bone marrow was aspirated from the femurs of Yucatan microswine. MSCs were extracted via ficoll density centrifugation technique and were strongly immunopositive for MSC markers, CD44, CD90, and CD105, but negative for hematopoietic markers, CD14 and CD45. Subsequently, naïve MSCs were differentiated for 10 days in varying concentrations and combinations of VEGF-A, Ang II, and AT1R or AT2R antagonists. Markers specific to ECs were determined by FACS analysis. RESULTS AT1R and AT2R expression and cellular localization was demonstrated in MSCs stimulated with VEGF-A and Ang II via quantitative RT-PCR and immunofluorescence, respectively. Differentiation of naïve MSCs in media containing Ang II (2 ng/ml) plus low-dose VEGF-A (2 ng/ml) produced a significantly higher percentage of cells that were positive for expression of EC markers (for example, platelet endothelial cell adhesion molecule, vascular endothelial Cadherin and von Willebrand factor) compared to VEGF-A alone. Ang II alone failed to induce EC marker expression. MSCs differentiated with the combination of Ang II and VEGF-A were capable of forming capillary tubes using an in vitro angiogenesis assay. Induction of EC marker expression was greatly attenuated by co-treatment of Ang II/VEGF-A with the AT2R antagonist PD123319, but not the AT1R antagonist telmisartan. CONCLUSIONS We report the presence of functional AT2R receptor on porcine bone marrow-derived MSCs, where it positively regulates EC differentiation. These findings have significant implications toward therapeutic approaches based on activation of AT2R, which could be a means to stimulate regeneration of damaged endothelium and prevent vascular thrombosis.
Collapse
MESH Headings
- Angiotensin II/pharmacology
- Angiotensin II Type 1 Receptor Blockers/pharmacology
- Angiotensin II Type 2 Receptor Blockers/pharmacology
- Animals
- Antigens, CD/metabolism
- Benzimidazoles/pharmacology
- Benzoates/pharmacology
- Bone Marrow Cells/cytology
- Cadherins/metabolism
- Cell Differentiation/drug effects
- Drug Synergism
- Endothelial Cells/cytology
- Endothelial Cells/metabolism
- Femur/cytology
- Imidazoles/pharmacology
- Mesenchymal Stem Cells/cytology
- Mesenchymal Stem Cells/drug effects
- Mesenchymal Stem Cells/metabolism
- Microscopy, Fluorescence
- Pyridines/pharmacology
- RNA Interference
- RNA, Small Interfering/metabolism
- Real-Time Polymerase Chain Reaction
- Receptor, Angiotensin, Type 1/genetics
- Receptor, Angiotensin, Type 1/metabolism
- Receptor, Angiotensin, Type 2/genetics
- Receptor, Angiotensin, Type 2/metabolism
- Swine
- Telmisartan
- Vascular Endothelial Growth Factor A/antagonists & inhibitors
- Vascular Endothelial Growth Factor A/genetics
- Vascular Endothelial Growth Factor A/pharmacology
- von Willebrand Factor/metabolism
Collapse
Affiliation(s)
- Izuagie Attairu Ikhapoh
- Department of Medical Microbiology and Immunology, Creighton School of Medicine, 2500 California Plaza, Omaha, NE, 68178-0405, USA.
| | - Christopher J Pelham
- Department of Biomedical Sciences, Creighton School of Medicine, 2500 California Plaza, Omaha, NE, 68178, USA.
| | - Devendra K Agrawal
- Department of Medical Microbiology and Immunology, Creighton School of Medicine, 2500 California Plaza, Omaha, NE, 68178-0405, USA.
- Department of Biomedical Sciences, Creighton School of Medicine, 2500 California Plaza, Omaha, NE, 68178, USA.
- Center for Clinical and Translational Science, CRISS II Room 510, Creighton School of Medicine, 2500 California Plaza, Omaha, NE, 68178-0405, USA.
| |
Collapse
|
12
|
The brain-heart connection: frontal cortex and left ventricle angiotensinase activities in control and captopril-treated hypertensive rats-a bilateral study. Int J Hypertens 2013; 2013:156179. [PMID: 23476743 PMCID: PMC3583112 DOI: 10.1155/2013/156179] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2012] [Accepted: 01/04/2013] [Indexed: 11/18/2022] Open
Abstract
The model of neurovisceral integration suggests that the frontal cortex (FC) and the cardiovascular function are reciprocally and asymmetrically connected. We analyzed several angiotensinase activities in the heart left ventricle (VT) of control and captopril-treated SHR, and we search for a relationship between these activities and those determined in the left and right FC. Captopril was administered in drinking water for 4 weeks. Samples from the left VT and from the left and right FC were obtained. Soluble and membrane-bound enzymatic activities were measured fluorometrically using arylamides as substrates. The weight of heart significantly decreased after treatment with captopril, mainly, due to the reduction of the left VT weight. In the VT, no differences for soluble activities were observed between control and treated SHR. In contrast, a generalized significant reduction was observed for membrane-bound activities. The most significant correlations between FC and VT were observed in the right FC of the captopril-treated group. The other correlations, right FC versus VT and left FC versus VT in controls and left FC versus VT in the captopril group, were few and low. These results confirm that the connection between FC and cardiovascular system is asymmetrically organized.
Collapse
|
13
|
Kitzman DW, Hundley WG, Brubaker PH, Morgan TM, Moore JB, Stewart KP, Little WC. A randomized double-blind trial of enalapril in older patients with heart failure and preserved ejection fraction: effects on exercise tolerance and arterial distensibility. Circ Heart Fail 2010; 3:477-85. [PMID: 20516425 DOI: 10.1161/circheartfailure.109.898916] [Citation(s) in RCA: 106] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND Exercise intolerance is the primary symptom in older patients with heart failure and preserved ejection fraction (HFPEF); however, little is known regarding its mechanisms and therapy. METHODS AND RESULTS Seventy-one stable elderly (70+/-1 years) patients (80% women) with compensated HFPEF and controlled blood pressure were randomized into a 12-month follow-up double-blind trial of enalapril 20 mg/d versus placebo. Assessments were peak exercise oxygen consumption; 6-minute walk test; Minnesota Living with HF Questionnaire; MRI; Doppler echocardiography; and vascular ultrasound. Compliance by pill count was excellent (94%). Twenty-five patients in the enalapril group versus 34 in the placebo group completed the 12-month follow-up. During follow-up, there was no difference in the primary outcome of peak exercise oxygen consumption (enalapril, 14.5+/-3.2 mL/kg/min; placebo, 14.3+/-3.4 mL/kg/min; P=0.99), or in 6-minute walk distance, aortic distensibility (the primary mechanistic outcome), left ventricle mass, or neurohormonal profile. The effect size of enalapril on peak exercise oxygen consumption was small (0.7%; 95% CI, 4.2% to 5.6%). There was a trend toward improved Minnesota Living with HF Questionnaire total score (P=0.07), a modest reduction in systolic blood pressure at peak exercise (P=0.02), and a marginal improvement in carotid arterial distensibility (P=0.04). CONCLUSIONS In stable, older patients with compensated HFPEF and controlled blood pressure, 12 months of enalapril did not improve exercise capacity or aortic distensibility. These data, combined with those from large clinical event trials, suggest that angiotensin inhibition does not substantially improve key long-term clinical outcomes in this group of patients. This finding contrasts sharply with observations in HF with reduced EF and highlights our incomplete understanding of this important and common disorder.
Collapse
Affiliation(s)
- Dalane W Kitzman
- Cardiology Section, Department of Internal Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA.
| | | | | | | | | | | | | |
Collapse
|
14
|
Abstract
Anemia is a complex issue in patients with heart failure (HF). In past years, clinicians accepted anemia as a given or an "accessory" diagnosis in HF patients. This attitude has changed since understanding of the causes and morbidity of anemia in HF has improved and with the introduction of targeted treatments. Increasing health care costs have stimulated vigorous debate about the cost-effectiveness of such treatments. It behooves clinicians to understand the effectiveness of specific treatments, risks and benefits, and costs. This review addresses the impact of anemia's prevalence, etiology, associated outcomes, and treatments on the economic burden of HF patients.
Collapse
|
15
|
Mendis B, Page SR. Candesartan: widening indications for this angiotensin II receptor blocker? Expert Opin Pharmacother 2009; 10:1995-2007. [PMID: 19563275 DOI: 10.1517/14656560903092197] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Candesartan cilexetil is one of a number of drugs of the angiotensin II receptor blocker (ARB) class. Their principal mode of action involves competitive blockade of the angiotensin II type 1 receptor, thereby modulating the activity of the rennin-angiotensin-aldosterone system. Angiotensin II receptor blocker therapy has been proven to be well tolerated and effective in the management of hypertension, chronic heart failure with left ventricular dysfunction and the prevention and progression of diabetic renal disease. Candesartan is a highly potent, long-acting and selective angiotensin II type 1 receptor blocker. It was launched in 1998 for the treatment of hypertension. Its use has increased dramatically, with recently published data suggesting benefit in the treatment of stroke, heart failure, diabetic renal disease and most recently in preventing the development of or delaying the progression of diabetic retinopathy. In this article we review the literature on the use of ARB drugs in general before focusing on candesartan.
Collapse
Affiliation(s)
- B Mendis
- Nottingham University Hospitals NHS Trust, QMC Campus, Department of Diabetes and Endocrinology, Derby Road, Nottingham NG72UH, UK.
| | | |
Collapse
|
16
|
Tschöpe C, Schultheiss HP. [Cardioprotection by means of Candesartan in cardiac insufficiency. CHARM overall partial evaluation (Candesartan in heart failure assessment of reduction in mortality and morbidity)]. Internist (Berl) 2006; 47:1084-6. [PMID: 16988804 DOI: 10.1007/s00108-006-1710-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Affiliation(s)
- C Tschöpe
- Abteilung für Kardiologie und Pulmologie, Charité Universitätsmedizin Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, 12200, Berlin, Germany.
| | | |
Collapse
|