1
|
Whelan AJ, Mim S, Hunt JP, McKnite AM, Green DJ, Imburgia CE, Momper JD, Stitt G, Watt KM. Interaction of milrinone with extracorporeal life support. THE JOURNAL OF EXTRA-CORPOREAL TECHNOLOGY 2024; 56:167-173. [PMID: 39705580 DOI: 10.1051/ject/2024014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 06/12/2024] [Indexed: 12/22/2024]
Abstract
BACKGROUND Milrinone is commonly prescribed to critically ill patients who need extracorporeal life support such as extracorporeal membrane oxygenation (ECMO) and continuous renal replacement therapy (CRRT). Currently, the effect of ECMO and CRRT on the disposition of milrinone is unknown. METHODS Ex vivo ECMO and CRRT circuits were primed with human blood and then dosed with milrinone to study drug extraction by the circuits. Milrinone percent recovery over time was calculated to determine circuit component interaction with milrinone. RESULTS Milrinone did not exhibit measurable interactions with the ECMO circuit, however, CRRT cleared 99% of milrinone from the experimental circuit within the first 2 hours. CONCLUSION Milrinone dosing adjustments are likely required in patients who are supported with CRRT while dosing adjustments for ECMO based on these ex-vivo results are likely unnecessary. These results will help improve the safety and efficacy of milrinone in patients requiring ECMO and CRRT. Due to the limitations of ex-vivo experiments, future studies of milrinone exposure with ECLS should include patient circuit interactions as well as the physiology of critical illness.
Collapse
Affiliation(s)
- Aviva J Whelan
- Division of Clinical Pharmacology, Department of Pediatrics, University of Utah, 295 Chipeta Way, Salt Lake City, UT 84108, USA - Division of Pediatric Critical Care, Department of Pediatrics, University of Utah, 295 Chipeta Way, Salt Lake City, UT 84108, USA
| | - Sabiha Mim
- Pharmacometric Research Group, Department of Pharmacy, Uppsala University, Husargatan, Uppsala 752 37, Sweden
| | - J Porter Hunt
- Division of Clinical Pharmacology, Department of Pediatrics, University of Utah, 295 Chipeta Way, Salt Lake City, UT 84108, USA
| | - Autumn M McKnite
- Department of Pharmacology and Toxicology, University of Utah, 295 Chipeta Way, Salt Lake City, UT 84108, USA
| | - Danielle J Green
- Division of Clinical Pharmacology, Department of Pediatrics, University of Utah, 295 Chipeta Way, Salt Lake City, UT 84108, USA - Division of Pediatric Critical Care, Department of Pediatrics, University of Utah, 295 Chipeta Way, Salt Lake City, UT 84108, USA
| | - Carina E Imburgia
- Division of Clinical Pharmacology, Department of Pediatrics, University of Utah, 295 Chipeta Way, Salt Lake City, UT 84108, USA
| | - Jeremiah D Momper
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, 9255 Pharmacy Lane, MC 0657, La Jolla, CA 92093, USA
| | - Gideon Stitt
- Division of Clinical Pharmacology, Department of Pediatrics, University of Utah, 295 Chipeta Way, Salt Lake City, UT 84108, USA
| | - Kevin M Watt
- Division of Clinical Pharmacology, Department of Pediatrics, University of Utah, 295 Chipeta Way, Salt Lake City, UT 84108, USA - Division of Pediatric Critical Care, Department of Pediatrics, University of Utah, 295 Chipeta Way, Salt Lake City, UT 84108, USA
| |
Collapse
|
2
|
Chen Q, Xia Y, Liu HN, Chi Y, Li X, Shan LS, Dai B, Zhu Y, Wang YT, Miao X, Sun Q. Synthetic approaches and clinical application of representative small-molecule inhibitors of phosphodiesterase. Eur J Med Chem 2024; 277:116769. [PMID: 39163778 DOI: 10.1016/j.ejmech.2024.116769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 07/25/2024] [Accepted: 08/11/2024] [Indexed: 08/22/2024]
Abstract
Phosphodiesterases (PDEs) constitute a family of enzymes that play a pivotal role in the regulation of intracellular levels of cyclic nucleotides, including cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP). Dysregulation of PDE activity has been implicated in diverse pathological conditions encompassing cardiovascular disorders, pulmonary diseases, and neurological disorders. Small-molecule inhibitors targeting PDEs have emerged as promising therapeutic agents for the treatment of these ailments, some of which have been approved for their clinical use. Despite their success, challenges such as resistance mechanisms and off-target effects persist, urging continuous research for the development of next-generation PDE inhibitors. The objective of this review is to provide an overview of the synthesis and clinical application of representative approved small-molecule PDE inhibitors, with the aim of offering guidance for further advancements in the development of novel PDE inhibitors.
Collapse
Affiliation(s)
- Qingqing Chen
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yu Xia
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - He-Nan Liu
- Shengjing Hospital of China Medical University, Shenyang, China
| | - Yuan Chi
- Shengjing Hospital of China Medical University, Shenyang, China
| | - Xun Li
- Shengjing Hospital of China Medical University, Shenyang, China
| | - Li-Shen Shan
- Shengjing Hospital of China Medical University, Shenyang, China
| | - Bing Dai
- Shengjing Hospital of China Medical University, Shenyang, China
| | - Ying Zhu
- Department of Neurology, The First Hospital of China Medical University, Shenyang, China
| | - Ya-Tao Wang
- Rega Institute for Medical Research, Medicinal Chemistry, KU Leuven, Herestraat 49-Box 1041, 3000, Leuven, Belgium.
| | - Xinxin Miao
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China.
| | - Qian Sun
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
3
|
Noguchi K, Umeda S, Goma M, Ueda C, Tabira S, Furuyama K, Taniguchi M, Nagai A, Matsushita M, Kanae H. Chronotropic and Inotropic Effects of Sudachitin, a Polymethoxyflavone from the Peel of Citrus sudachi on Isolated Rat Atria and Its Underlying Mechanisms. Biol Pharm Bull 2024; 47:2011-2020. [PMID: 39647904 DOI: 10.1248/bpb.b24-00575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/10/2024]
Abstract
Sudachitin, a polymethoxyflavone found in sudachi peel, has been reported to improve hyperlipidemia in humans, and is thus attracting research attention. However, its effect on cardiac function remains unclear. We investigated the mechanisms underlying the chronotropic and inotropic effects of sudachitin on rat atria. Sudachitin (0.3-30 µM) produced concentration-dependent positive chronotropic and inotropic effects. Other polymethoxyflavones, including demethoxysudachitin (0.3-30 µM) and nobiletin (0.3-30 µM), also produced positive chronotropic and inotropic effects; however, the maximum efficacy of all polymethoxyflavones, including sudachitin, was lower than that of isoproterenol. Propranolol (0.1 µM) did not affect the positive chronotropic and inotropic effects of sudachitin. The concentration-response curves for the chronotropic and inotropic effects of dibutyryl-cAMP (1-100 µM) were shifted to the left upon pretreatment with sudachitin (3, 10 µM). Phosphodiesterase inhibitors (3-isobutyl-1-methylxanthine 1 µM or milrinone 10 µM) alone, sudachitin alone (10, 30 µM), and a combination of phosphodiesterase inhibitors and sudachitin exhibited positive chronotropic and inotropic effects, whereas the lack of any interaction between each phosphodiesterase inhibitor and sudachitin indicated an additive effect of the two substances. These results suggest that sudachitin-induced positive chronotropic and inotropic effects similar to those of other polymethoxyflavones, but its maximum efficacy was lower than that of isoproterenol. Both demethoxysudachitin and nobiletin exhibited similar positive chronotropic and inotropic effects, indicating that these effects are not specific to sudachitin, but are common to polymethoxyflavones. The mechanism of action of sudachitin was associated with the enhancement of cAMP-dependent pathways, without the involvement of β-adrenoceptors.
Collapse
Affiliation(s)
- Kazuo Noguchi
- Laboratory of Functional Food Sciences, Department of Health and Bio-Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Mukogawa Women's University
| | - Saki Umeda
- Laboratory of Functional Food Sciences, Department of Health and Bio-Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Mukogawa Women's University
| | - Misaki Goma
- Laboratory of Functional Food Sciences, Department of Health and Bio-Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Mukogawa Women's University
| | - Chinami Ueda
- Laboratory of Functional Food Sciences, Department of Health and Bio-Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Mukogawa Women's University
| | - Sawako Tabira
- Laboratory of Functional Food Sciences, Department of Health and Bio-Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Mukogawa Women's University
| | - Koto Furuyama
- Laboratory of Functional Food Sciences, Department of Health and Bio-Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Mukogawa Women's University
| | - Mirai Taniguchi
- Laboratory of Functional Food Sciences, Department of Health and Bio-Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Mukogawa Women's University
| | - Aino Nagai
- Laboratory of Functional Food Sciences, Department of Health and Bio-Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Mukogawa Women's University
| | - Midori Matsushita
- Laboratory of Functional Food Sciences, Department of Health and Bio-Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Mukogawa Women's University
| | - Haruna Kanae
- Laboratory of Functional Food Sciences, Department of Health and Bio-Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Mukogawa Women's University
| |
Collapse
|
4
|
Britto-Júnior J, Medeiros-Teixeira LR, Lima AT, Dassow LC, Lopes-Martins RÁB, Campos R, Moraes MO, Moraes MEA, Antunes E, De Nucci G. 6-Nitrodopamine Is the Most Potent Endogenous Positive Inotropic Agent in the Isolated Rat Heart. Life (Basel) 2023; 13:2012. [PMID: 37895394 PMCID: PMC10607994 DOI: 10.3390/life13102012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 10/01/2023] [Accepted: 10/02/2023] [Indexed: 10/29/2023] Open
Abstract
BACKGROUND 6-nitrodopamine released from rat isolated atria exerts positive chronotropic action, being more potent than noradrenaline, adrenaline, and dopamine. Here, we determined whether 6-nitrodopamine is released from rat isolated ventricles (RIV) and modulates heart inotropism. METHODS Catecholamines released from RIV were quantified by LC-MS/MS and their effects on heart inotropism were evaluated by measuring left ventricular developed pressure (LVDP) in Langendorff's preparation. RESULTS 6-nitrodopamine was the major released catecholamine from RIV. Incubation with L-NAME (100 µM), but not with tetrodotoxin (1 µM), caused a significant reduction in 6-nitrodopamine basal release. 6-nitrodopamine release was significantly reduced in ventricles obtained from L-NAME chronically treated animals. 6-nitrodopamine (0.01 pmol) caused significant increases in LVDP and dP/dtmax, whereas dopamine and noradrenaline required 10 pmol, and adrenaline required 100 pmol, to induce similar increases in LVDP and dP/dtmax. The infusion of atenolol (10 nM) reduced basal LVDP and blocked the increases in LVDP induced by 6-ND (0.01 pmol), without affecting the increases in LVDP induced by 10 nmol of dopamine and noradrenaline and that induced by adrenaline (100 nmol). CONCLUSIONS 6-nitrodopamine is the major catecholamine released from rat isolated ventricles. It is 1000 times more potent than dopamine and noradrenaline and is selectively blocked by atenolol, indicating that 6-ND is a main regulator of heart inotropism.
Collapse
Affiliation(s)
- José Britto-Júnior
- Department of Pharmacology, Faculty of Medical Sciences, University of Campinas (UNICAMP), Campinas 13083-970, Brazil; (J.B.-J.); (L.R.M.-T.); (A.T.L.); (R.C.); (E.A.)
| | - Lincoln Rangel Medeiros-Teixeira
- Department of Pharmacology, Faculty of Medical Sciences, University of Campinas (UNICAMP), Campinas 13083-970, Brazil; (J.B.-J.); (L.R.M.-T.); (A.T.L.); (R.C.); (E.A.)
| | - Antonio Tiago Lima
- Department of Pharmacology, Faculty of Medical Sciences, University of Campinas (UNICAMP), Campinas 13083-970, Brazil; (J.B.-J.); (L.R.M.-T.); (A.T.L.); (R.C.); (E.A.)
| | - Letícia Costa Dassow
- Laboratory of Biophotonics and Experimental Therapeutics, University Evangélica of Goiás (UniEVANGÉLICA), Anápolis 75083-515, Brazil; (L.C.D.); (R.Á.B.L.-M.)
| | - Rodrigo Álvaro Brandão Lopes-Martins
- Laboratory of Biophotonics and Experimental Therapeutics, University Evangélica of Goiás (UniEVANGÉLICA), Anápolis 75083-515, Brazil; (L.C.D.); (R.Á.B.L.-M.)
| | - Rafael Campos
- Department of Pharmacology, Faculty of Medical Sciences, University of Campinas (UNICAMP), Campinas 13083-970, Brazil; (J.B.-J.); (L.R.M.-T.); (A.T.L.); (R.C.); (E.A.)
- Clinical Pharmacology Unit, Drug Research and Development Center, Federal University of Ceará (UFC), Fortaleza 60020-181, Brazil; (M.O.M.); (M.E.A.M.)
| | - Manoel Odorico Moraes
- Clinical Pharmacology Unit, Drug Research and Development Center, Federal University of Ceará (UFC), Fortaleza 60020-181, Brazil; (M.O.M.); (M.E.A.M.)
| | - Maria Elisabete A. Moraes
- Clinical Pharmacology Unit, Drug Research and Development Center, Federal University of Ceará (UFC), Fortaleza 60020-181, Brazil; (M.O.M.); (M.E.A.M.)
| | - Edson Antunes
- Department of Pharmacology, Faculty of Medical Sciences, University of Campinas (UNICAMP), Campinas 13083-970, Brazil; (J.B.-J.); (L.R.M.-T.); (A.T.L.); (R.C.); (E.A.)
| | - Gilberto De Nucci
- Department of Pharmacology, Faculty of Medical Sciences, University of Campinas (UNICAMP), Campinas 13083-970, Brazil; (J.B.-J.); (L.R.M.-T.); (A.T.L.); (R.C.); (E.A.)
- Laboratory of Biophotonics and Experimental Therapeutics, University Evangélica of Goiás (UniEVANGÉLICA), Anápolis 75083-515, Brazil; (L.C.D.); (R.Á.B.L.-M.)
- Clinical Pharmacology Unit, Drug Research and Development Center, Federal University of Ceará (UFC), Fortaleza 60020-181, Brazil; (M.O.M.); (M.E.A.M.)
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo (USP), Sāo Paulo 05508-220, Brazil
| |
Collapse
|
5
|
Boonpala P, Saengklub N, Srikam S, Ji-Au W, Panyasing Y, Kumphune S, Kijtawornrat A. Pimobendan prevents cardiac dysfunction, mitigates cardiac mitochondrial dysfunction, and preserves myocyte ultrastructure in a rat model of mitral regurgitation. BMC Vet Res 2023; 19:130. [PMID: 37612694 PMCID: PMC10463781 DOI: 10.1186/s12917-023-03693-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 08/14/2023] [Indexed: 08/25/2023] Open
Abstract
BACKGROUND Pimobendan has been proven to delay the onset of congestive heart failure (CHF) in dogs with mitral regurgitation (MR); however, molecular underlying mechanisms have not been fully elucidated. This study aimed to investigate (1) the effects of pimobendan on cardiac function, cardiac mitochondrial quality and morphology, and cardiac ultrastructure in a rat model of chronic MR and (2) the direct effect of pimobendan on intracellular reactive oxygen species (ROS) production in cardiac cells. MR was surgically induced in 20 Sprague-Dawley rats, and sham procedures were performed on 10 rats. Eight weeks post-surgery, the MR rats were randomly divided into two groups: the MR group and the MR + pimobendan group. Pimobendan (0.15 mg/kg) was administered twice a day via oral gavage for 4 weeks, whereas the sham and MR groups received equivalent volumes of drinking water. Echocardiography was performed at baseline (8 weeks post-surgery) and at the end of the study (4 weeks after treatment). At the end of the study protocol, all rats were euthanized, and their hearts were immediately collected, weighed, and used for transmission electron microscopy and mitochondrial quality assessments. To evaluate the role of pimobendan on intracellular ROS production, preventive or scavenging properties were tested with H2O2-induced ROS generation in rat cardiac myoblasts (H9c2). RESULTS Pimobendan preserved cardiac functions and structure in MR rats. In addition, pimobendan significantly improved mitochondrial quality by attenuating ROS production and depolarization (P < 0.05). The cardiac ultrastructure and mitochondrial morphology were significantly preserved in the MR + pimobendan group. In addition, pimobendan appeared to play as a ROS scavenger, but not as a ROS preventer, in H2O2-induced ROS production in H9c2 cells. CONCLUSIONS Pimobendan demonstrated cardioprotective effects on cardiac function and ultrastructure by preserving mitochondrial quality and acted as an ROS scavenger in a rat model of MR.
Collapse
Affiliation(s)
- Pakit Boonpala
- Department of Physiology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
- Chulalongkorn University Laboratory Animal Center, Chulalongkorn University, Bangkok, Thailand
| | - Nakkawee Saengklub
- Department of Physiology, Faculty of Pharmacy, Mahidol University, Bangkok, Thailand
| | - Sirinapa Srikam
- Department of Pathology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Wilawan Ji-Au
- Department of Pathology, King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| | - Yaowalak Panyasing
- Department of Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| | - Sarawut Kumphune
- Biomedical Engineering Institute (BMEI), Chiang Mai University, Chiang Mai, Thailand
- Biomedical Engineering and Innovation Research Center, Chiang Mai University, Chiang Mai, Thailand
| | - Anusak Kijtawornrat
- Department of Physiology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand.
- Chulalongkorn University Laboratory Animal Center, Chulalongkorn University, Bangkok, Thailand.
| |
Collapse
|
6
|
Xue Y, Li X, Wang Z, Lv Q. Cilostazol regulates the expressions of endothelin‑1 and endothelial nitric oxide synthase via activation of the p38 MAPK signaling pathway in HUVECs. Biomed Rep 2022; 17:77. [DOI: 10.3892/br.2022.1560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 05/12/2022] [Indexed: 11/06/2022] Open
Affiliation(s)
- Ying Xue
- Department of Clinical Pharmacy, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Xiaoye Li
- Department of Clinical Pharmacy, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Zi Wang
- Department of Clinical Pharmacy, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Qianzhou Lv
- Department of Clinical Pharmacy, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| |
Collapse
|
7
|
Chai E, Chen J, Li C, Zhang X, Fan Z, Yang S, Zhao K, Li W, Xiao Z, Zhang Y, Tang F. The Efficacy and Safety of Cilostazol vs. Aspirin for Secondary Stroke Prevention: A Systematic Review and Meta-Analysis. Front Neurol 2022; 13:814654. [PMID: 35242099 PMCID: PMC8885726 DOI: 10.3389/fneur.2022.814654] [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: 11/14/2021] [Accepted: 01/05/2022] [Indexed: 11/16/2022] Open
Abstract
Background Cilostazol is often used in Asia-Pacific countries for stroke prevention. The current systematic review and meta-analysis aimed to evaluate the effectiveness, safety, and adverse outcomes of cilostazol monotherapy compared to aspirin monotherapy for secondary stroke prevention. Methods The researchers conducted a comprehensive research in multiple databases (PubMed, Embase, and Cochrane library) of randomized controlled trials from conception to December 2020. The primary efficacy outcome was the occurrence of any stroke, the primary safety outcome was the bleeding risk, and the primary adverse outcome was the rate of headache and dizziness. The Mantel-Haenszel method was used to calculate a random-effects prediction. Cilostazol and aspirin were compared using a pooled risk assessment with 95% CIs. Results Six studies involving 5,617 patients were included in this review. Compared with aspirin monotherapy, cilostazol was associated with significantly lower rates of any strokes (RR: 0.67; 95% CI: 0.55–0.82) and significantly lower bleeding rates [risk ratio (RR): 0.53; 95% CI: 0.37–0.74]. However, compared with aspirin monotherapy, cilostazol was associated with significantly higher rates of headache (RR: 1.77; 95% CI: 1.41–2.20) and dizziness (RR: 1.28; 95% CI: 1.08–1.52). Conclusions Consistent with previous studies, cilostazol monotherapy is superior to aspirin monotherapy in reducing the rate of any strokes and the bleeding risk after having a stroke. However, the use of cilostazol monotherapy is associated with several adverse life outcomes such as headaches and dizziness.
Collapse
Affiliation(s)
- Erqing Chai
- Cerebrovascular Disease Center, Gansu Provincial Hospital, Lanzhou, China.,Emergency General Hospital, Beijing, China
| | - Jinhua Chen
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Changqing Li
- Cerebrovascular Disease Center, Gansu Provincial Hospital, Lanzhou, China
| | - Xue Zhang
- Intensive Care Unit 1, Gansu Provincial Hospital, Lanzhou, China
| | - Zhiqiang Fan
- First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, China
| | - Shijie Yang
- First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, China
| | - Kaixuan Zhao
- School of Clinical Medicine, Ningxia Medical University, Ningxia, China
| | - Wei Li
- First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, China
| | - Zaixing Xiao
- First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, China
| | - Yichuan Zhang
- School of Clinical Medicine, Ningxia Medical University, Ningxia, China
| | - Futian Tang
- Key Laboratory of Gansu Digestive System Tumor, Lanzhou University Second Hospital, Lanzhou, China
| |
Collapse
|
8
|
Circulating cyclic adenosine monophosphate concentrations in milrinone treated paediatric patients after congenital heart surgery. Cardiol Young 2021; 31:1393-1400. [PMID: 33533327 PMCID: PMC9257900 DOI: 10.1017/s1047951121000251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
BACKGROUND Milrinone is a phosphodiesterase type 3 inhibitor that results in a positive inotropic effect in the heart through an increase in cyclic adenosine monophosphate. The purpose of this study was to evaluate circulating cyclic adenosine monophosphate and milrinone concentrations in milrinone treated paediatric patients undergoing congenital heart surgery. METHODS Single-centre prospective observational pilot study from January 2015 to December 2017 including children aged birth to 18 years. Milrinone and circulating cyclic adenosine monophosphate concentrations were measured at four time points through the first post-operative day and compared between patients with and without low cardiac output syndrome, defined using clinical and laboratory criteria. RESULTS Fifty patients were included. Nine (18%) developed low cardiac output syndrome. For all patients, 22% had single ventricle heart disease. The density and distribution of cyclic adenosine monophosphate concentrations varied between those with and without low cardiac output syndrome but were not significantly different. Milrinone concentrations increased in all patients. Paired t-tests demonstrated an increase in circulating cyclic adenosine monophosphate concentrations during the post-operative period among patients without low cardiac output syndrome. CONCLUSIONS In this prospective observational study, circulating cyclic adenosine monophosphate concentrations increased in those without low cardiac output syndrome during the first 24 post-operative hours and milrinone concentrations increased in all patients. Further study of the utility of cyclic adenosine monophosphate concentrations in milrinone treated patients is necessary.
Collapse
|
9
|
El-Hachem N, Fardoun MM, Slika H, Baydoun E, Eid AH. Repurposing Cilostazol for Raynaud's Phenomenon. Curr Med Chem 2021; 28:2409-2417. [PMID: 32881655 DOI: 10.2174/0929867327666200903114154] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 07/16/2020] [Accepted: 07/21/2020] [Indexed: 11/22/2022]
Abstract
Raynaud 's Phenomenon (RP) results from exaggerated cold-induced vasoconstriction. RP patients suffer from vasospastic attacks and compromised digital blood perfusion leading to a triple color change at the level the fingers. Severe RP may cause ulcers and threaten tissue viability. Many drugs have been used to alleviate the symptoms of RP. These include calcium-channel blockers, cGMP-specific phosphodiesterase type 5 inhibitors, prostacyclin analogs, and angiotensin receptor blockers. Despite their variety, these drugs do not treat RP but rather alleviate its symptoms. To date, no drug for RP has been yet approved by the U.S Food and Drugs Administration. Cilostazol is a selective inhibitor of phosphodiesterase-III, originally prescribed to treat intermittent claudication. Owing to its antiplatelet and vasodilating properties, cilostazol is being repurposed as a potential drug for RP. This review focuses on the different lines of action of cilostazol serving to enhance blood perfusion in RP patients.
Collapse
Affiliation(s)
- Nehme El-Hachem
- Laboratory of Medical Genetics, Institute of Experimental Cardiology, National Medical Research Center of Cardiology, Beirut, Lebanon
| | - Manal M Fardoun
- Department of Biology, American University of Beirut, Beirut, Lebanon
| | - Hasan Slika
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Elias Baydoun
- Department of Biology, American University of Beirut, Beirut, Lebanon
| | - Ali H Eid
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
10
|
Nan Y, Zeng X, Jin Z, Li N, Chen Z, Chen J, Wang D, Wang Y, Lin Z, Ying L. PDE1 or PDE5 inhibition augments NO-dependent hypoxic constriction of porcine coronary artery via elevating inosine 3',5'-cyclic monophosphate level. J Cell Mol Med 2020; 24:14514-14524. [PMID: 33169529 PMCID: PMC7754025 DOI: 10.1111/jcmm.16078] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 10/17/2020] [Accepted: 10/25/2020] [Indexed: 12/30/2022] Open
Abstract
Hypoxic coronary vasospasm may lead to myocardial ischaemia and cardiac dysfunction. Inosine 3',5'-cyclic monophosphate (cIMP) is a putative second messenger to mediate this pathological process. Nevertheless, it remains unclear as to whether levels of cIMP can be regulated in living tissue such as coronary artery and if so, what is the consequence of this regulation on hypoxia-induced vasoconstriction. In the present study, we found that cIMP was a key determinant of hypoxia-induced constriction but not that of the subsequent relaxation response in porcine coronary arteries. Subsequently, coronary arteries were treated with various phosphodiesterase (PDE) inhibitors to identify PDE types that are capable of regulating cIMP levels. We found that inhibition of PDE1 and PDE5 substantially elevated cIMP content in endothelium-denuded coronary artery supplemented with exogenous purified cIMP. However, cGMP levels were far lower than their levels in intact coronary arteries and lower than cIMP levels measured in endothelium-denuded coronary arteries supplemented with exogenous cIMP. The increased cIMP levels induced by PDE1 or PDE5 inhibition further led to augmented hypoxic constriction without apparently affecting the relaxation response. In intact coronary artery, PDE1 or PDE5 inhibition up-regulated cIMP levels under hypoxic condition. Concomitantly, cGMP level increased to a comparable level. Nevertheless, the hypoxia-mediated constriction was enhanced in this situation that was largely compromised by an even stronger inhibition of PDEs. Taken together, these data suggest that cIMP levels in coronary arteries are regulated by PDE1 and PDE5, whose inhibition at a certain level leads to increased cIMP content and enhanced hypoxic constriction.
Collapse
Affiliation(s)
- Yan Nan
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Xueqin Zeng
- Department of Pathophysiology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Zhiyi Jin
- Department of Pathophysiology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Na Li
- Department of Pathophysiology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China.,Department of Pathology, Wenzhou Central Hospital, Wenzhou, China
| | - Zhengju Chen
- Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, China
| | - Jiantong Chen
- Department of Pathophysiology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Dezhong Wang
- Institute of Life Sciences, Wenzhou University, Wenzhou, China
| | - Yang Wang
- Department of Pathophysiology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Zhenlang Lin
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Lei Ying
- Department of Pathophysiology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
11
|
Xue Y, Wang Z, Wu H, Li X, Chen J, Lv Q. Cilostazol increases adenosine plasma concentration in patients with acute coronary syndrome. J Clin Pharm Ther 2020; 46:328-332. [PMID: 33052628 DOI: 10.1111/jcpt.13284] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 09/09/2020] [Accepted: 09/15/2020] [Indexed: 12/17/2022]
Abstract
WHAT IS KNOWN AND OBJECTIVE Cilostazol is a specific and strong inhibitor of phosphodiesterase (PDE) type III which can suppress the platelet aggregation by increasing cyclic adenosine monophosphate (cAMP) levels. The clinical benefit of cilostazol in ACS patients suggested that the drug may have non-platelet-directed properties. Some in vitro and animal studies also indicated that the 'pleiotropic' properties of cilostazol might be related to the interaction with adenosine metabolism. Adenosine is an important regulatory metabolite and an inhibitor of platelet activation. However, no human study has been conducted to determine whether cilostazol could increase the adenosine plasma concentration in vivo. As a result, this study aimed to investigate the impact of cilostazol on adenosine plasma concentration (APC) in acute coronary syndrome (ACS) patients. METHODS We prospectively analysed 149 ACS patients undergoing percutaneous coronary intervention (PCI) with drug-eluting stents. The included patients were divided into two groups according to the presence (cilostazol group, n = 64) or absence (aspirin group, n = 85) of aspirin intolerance. The inhibition of platelet aggregation (IPA), APC and cAMP concentration was measured. Patient characteristics, medications and 30-day clinical outcomes were examined. RESULTS Patients receiving cilostazol had a significantly higher adenosine and cAMP plasma concentration than patients receiving aspirin (3.00 ± 0.67 vs 2.56 ± 0.74 mol/L, P < .001; 28.10 ± 14.74 vs 20.48 ± 11.35 pmol/mL, P = .0014). Cilostazol was associated with a higher inhibition rate of ADP induced platelet aggregation than aspirin (63.35 ± 26.71 vs 52.2 ± 28.35, P = .036). The plasma levels of adenosine and cAMP showed a positive correlation with ADP induced platelet aggregation. WHAT IS NEW AND CONCLUSION Cilostazol increases adenosine concentration compared with aspirin. Its potent antiplatelet effect in ACS patients may be partly mediated by adenosine.
Collapse
Affiliation(s)
- Ying Xue
- Department of Clinical Pharmacy, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zi Wang
- Department of Clinical Pharmacy, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Hongyi Wu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiaoye Li
- Department of Clinical Pharmacy, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jiahui Chen
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qianzhou Lv
- Department of Clinical Pharmacy, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
12
|
Kim HN, Seo BR, Kim H, Koh JY. Cilostazol restores autophagy flux in bafilomycin A1-treated, cultured cortical astrocytes through lysosomal reacidification: roles of PKA, zinc and metallothionein 3. Sci Rep 2020; 10:9175. [PMID: 32514052 PMCID: PMC7280249 DOI: 10.1038/s41598-020-66292-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 05/18/2020] [Indexed: 12/21/2022] Open
Abstract
Cilostazol, a phosphodiesterase 3 inhibitor, reduces the amyloid-beta (Aβ) burden in mouse models of Alzheimer disease by as yet unidentified mechanisms. In the present study, we examined the possibility that cilostazol ameliorates lysosomal dysfunction. Astrocytes treated with bafilomycin A1 (BafA1) exhibited markedly reduced DND-189 and acridine orange (AO) fluorescence, indicating reduced lysosomal acidity. In both cases, BafA1-induced alkalization was reversed by addition of cilostazol, dibutyryl cAMP or forskolin. All three agents significantly increased free zinc levels in lysosomes, and addition of the zinc chelator TPEN abrogated lysosomal reacidification. These treatments did not raise free zinc levels or reverse BafA1-mediated lysosomal alkalization in metallothionein 3 (Mt3)-null astrocytes, indicating that the increases in zinc in astrocytes were derived mainly from Mt3. Lastly, in FITC-Aβ-treated astrocytes, cilostazol reversed lysosomal alkalization, increased cathepsin D activity, and reduced Aβ accumulation in astrocytes. Cilostazol also reduced mHtt aggregate formation in GFP-mHttQ74–expressing astrocytes. Collectively, our results present the novel finding that cAMP/PKA can overcome the v-ATPase blocking effect of BafA1 in a zinc- and Mt3-dependent manner.
Collapse
Affiliation(s)
- Ha Na Kim
- Neural Injury Lab, Biomedical Research Center, Asan Institute for Life Sciences, Asan Medical Center, Seoul, Korea
| | - Bo-Ra Seo
- Neural Injury Lab, Biomedical Research Center, Asan Institute for Life Sciences, Asan Medical Center, Seoul, Korea
| | - Hyunjin Kim
- Department of Neurology, University of Ulsan College of Medicine, Seoul, Korea; Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jae-Young Koh
- Department of Neurology, University of Ulsan College of Medicine, Seoul, Korea; Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea. .,Neural Injury Lab, Biomedical Research Center, Asan Institute for Life Sciences, Asan Medical Center, Seoul, Korea.
| |
Collapse
|
13
|
Zhao S, Zhong Z, Qi G, Shi L, Tian W. Effects of Cilostazol-Based Triple Antiplatelet Therapy Versus Dual Antiplatelet Therapy After Coronary Drug-Eluting Stent Implantation: An Updated Meta-Analysis of the Randomized Controlled Trials. Clin Drug Investig 2019; 39:1-13. [PMID: 30251232 DOI: 10.1007/s40261-018-0711-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND AND OBJECTIVE The results of studies on cilostazol-based triple antiplatelet therapy (TAT) after drug-eluting stent (DES) implantation were inconsistent. To assess the effects of TAT compared with dual antiplatelet therapy (DAT) after DES/second-generation DES implantation, we performed a meta-analysis of randomized controlled trials (RCTs). METHODS All relevant studies evaluated were identified by searching the PubMed, EMBASE, Cochrane Library, and ISI Web of Science databases without time and language limitation. Subgroup analyses were performed to evaluate the efficacy and safety of TAT after second-generation DES implantation. RESULTS Eleven RCTs involving a total of 4684 patients were included. The meta-analysis showed TAT was associated with significant beneficial effects on angiographic findings of in-stent restenosis [risk ratio (RR) 0.645, 95% confidence interval (CI) 0.470-0.885; P = 0.007], in-segment restenosis (RR 0.606, 95% CI 0.450-0.817; P = 0.001), in-stent late loss (RR - 0.095, 95% CI - 0.136 to - 0.054; P < 0.0001), in-segment late loss (RR - 0.100, 95% CI - 0.139 to - 0.061; P < 0.0001), target lesion revascularization (TLR) (RR 0.570, 95% CI 0.430-0.755; P < 0.0001), and target vessel revascularization (TVR) (RR 0.523, 95% CI 0.380-0.719; P < 0.0001). No significant difference was found in outcomes of all-cause death, cardiac death, definite/probable stent thrombosis (ST), non-fatal myocardial infarction (MI), overall bleeding, and major bleeding between the two groups, as well as some minor adverse effects including palpitations, thrombocytopenia, neutropenia, and hepatic dysfunction. However, the incidence rate of rash, gastrointestinal disorders, and headache was significantly higher in TAT. The second-generation DES subgroup showed similar results, except for the indicators of all-cause death (RR 2.161, 95% CI 1.007-4.635; P = 0.048) and hepatic dysfunction (RR 0.176, 95% CI 0.031-0.995; P = 0.049). CONCLUSIONS Compared with DAT, cilostazol-based TAT can significantly improve the angiographic findings of in-stent and in-segment late loss, in-stent and in-segment restenosis, TLR, and TVR after DES/second-generation DES implantation. However, no benefits were observed in outcomes of all-cause death, cardiac death, ST, and MI.
Collapse
Affiliation(s)
- Shijie Zhao
- Department of Geriatric Cardiology, First Affiliated Hospital, China Medical University, No. 155 Nanjingbei Street, 110001 Heping District, Shenyang, China
| | - Zhaoshuang Zhong
- Department of Respiratory, Central Hospital, Shenyang Medical College, Shenyang, China
| | - Guoxian Qi
- Department of Geriatric Cardiology, First Affiliated Hospital, China Medical University, No. 155 Nanjingbei Street, 110001 Heping District, Shenyang, China
| | - Liye Shi
- Department of Geriatric Cardiology, First Affiliated Hospital, China Medical University, No. 155 Nanjingbei Street, 110001 Heping District, Shenyang, China
| | - Wen Tian
- Department of Geriatric Cardiology, First Affiliated Hospital, China Medical University, No. 155 Nanjingbei Street, 110001 Heping District, Shenyang, China.
| |
Collapse
|
14
|
Fan ZG, Zhang WL, Xu B, Ji J, Tian NL, He SH. Comparisons between ticagrelor and clopidogrel following percutaneous coronary intervention in patients with acute coronary syndrome: a comprehensive meta-analysis. DRUG DESIGN DEVELOPMENT AND THERAPY 2019; 13:719-730. [PMID: 30863011 PMCID: PMC6388955 DOI: 10.2147/dddt.s196535] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Background The efficacy and safety of ticagrelor following percutaneous coronary intervention for patients with acute coronary syndrome remains unclear. This study sought to evaluate clinical outcomes of ticagrelor as part of dual-antiplatelet treatment for these patients. Methods PubMed, MEDLINE, Embase, and other Internet sources were searched for eligible citations. The primary end point was major adverse cardiovascular and cerebrovascular events, consisting of cardiovascular death, myocardial infarction, and stroke. The secondary end point was the occurrence of definite/probable stent thrombosis (ST). The risk of bleeding was chosen to be the safety end point. Results Eleven clinical trials – six randomized trials and five observational trials – were finally analyzed. A tendency toward reduction in the risk of major adverse cardiovascular and cerebrovascular events was observed only with respect to ticagrelor (OR 0.83, 95% CI 0.66–1.03; P=0.091), which might have resulted from the lower risk of cardiovascular death (OR 0.78, 95% CI 0.68–0.89; P<0.001). The overall incidence of ST differed significantly between the ticagrelor group and the clopidogrel group (OR 0.74, 95% CI 0.59–0.93; P=0.009), but the risk of bleeding, regardless of major or minor bleeding, increased significantly. Conclusion As part of dual-antiplatelet treatment following percutaneous coronary intervention, ticagrelor significantly reduced the risk of cardiovascular death and ST in acute coronary syndrome patients, but at the cost of bleeding. More powerful relevant randomized trials are still warranted to guide clinical decision-making.
Collapse
Affiliation(s)
- Zhong-Guo Fan
- Department of Cardiology, Subei People's Hospital of Jiangsu Province, Yangzhou University, Yangzhou, Jiangsu, China,
| | - Wen-Ling Zhang
- Department of Geriatric Gastroenterology, First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Bing Xu
- Department of Cardiology, Subei People's Hospital of Jiangsu Province, Yangzhou University, Yangzhou, Jiangsu, China,
| | - Jun Ji
- Department of Cardiology, Subei People's Hospital of Jiangsu Province, Yangzhou University, Yangzhou, Jiangsu, China,
| | - Nai-Liang Tian
- Department of Cardiology, Nanjing First Hospital, Nanjing Heart Center, Nanjing Medical University, Nanjing, Jiangsu, China,
| | - Sheng-Hu He
- Department of Cardiology, Subei People's Hospital of Jiangsu Province, Yangzhou University, Yangzhou, Jiangsu, China,
| |
Collapse
|
15
|
Wu CK, Lin JW, Wu LC, Chang CH. Risk of Heart Failure Hospitalization Associated With Cilostazol in Diabetes: A Nationwide Case-Crossover Study. Front Pharmacol 2019; 9:1467. [PMID: 30666197 PMCID: PMC6330376 DOI: 10.3389/fphar.2018.01467] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 11/30/2018] [Indexed: 01/08/2023] Open
Abstract
Background and Objective: It has been suggested to avoid cilostazol, the first-line therapy for peripheral arterial disease, in patients with congestive heart failure (HF). The objective of this study was to evaluate the risk of hospitalization for heart failure (HHF) associated with cilostazol use in the patients of diabetes mellitus. Methods: This case-crossover study retrieved records on diabetic patients > 20 years of age who were hospitalized for heart failure during the period of 2009-2011 from the Taiwan National Health Insurance Database. The "current" period was defined as 1-30 days prior to HHF whereas the 91-120 days prior to HHF served as the "reference" period. The exposure status just preceding the event is compared with exposure of the same person in one or more referent remote to the event. Adjusted odds ratios (OR) were used to estimate time-varying discordant exposure by the ratio of the number exposed to cilostazol only during the case period to the number exposed to cilostazol only during the control period. Results: A total of 47,506 diabetic patients were included in the analysis (average age: 72.7 ± 12.4, percentage of males: 48%). A total of 399 patients (0.84%) received cilostazol only in the current period, and 252 (0.53%) received cilostazol only in the reference period. After adjustment for other medications, a significant association was found between cilostazol and HHF (OR: 1.35, 95% CI: 1.14-1.59). After further adjustment for time-varying co-morbidities the ORs remained essentially the same. Sensitivity analyses using different definitions of control period (ranging from 31-60, 61-90, to 121-150 days before index date) yielded adjusted ORs of 1.43 (95% CI: 1.14-1.79), 1.31 (95% CI: 1.09-1.57) and 1.23 (95% CI: 1.06-1.44), respectively suggesting the robustness of our study findings. Conclusion: Use of cilostazol may be positively related to the risk of HHF. Further studies are warranted to explore the underlying mechanisms and to confirm the association.
Collapse
Affiliation(s)
- Cho-Kai Wu
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.,Department of Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Jou-Wei Lin
- Department of Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan.,Cardiovascular Center, National Taiwan University Hospital Yunlin Branch, Douliu, Taiwan
| | - Li-Chiu Wu
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - Chia-Hsuin Chang
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.,Department of Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan.,Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
16
|
Fukushima R, Kawaguchi T, Yamada S, Yoshimura A, Hirao D, Oomori T. Effects of cilostazol on the heart rate in healthy dogs. J Vet Med Sci 2018; 80:1707-1715. [PMID: 30249936 PMCID: PMC6261822 DOI: 10.1292/jvms.18-0240] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Appropriate dosages of cilostazol have not been studied in veterinary patients, and the degrees of heart rate (HR) increase have not been studied in dogs administered cilostazol. Therefore,
this study aimed to investigate the degrees of HR increase in healthy dogs administered cilostazol. Thirty healthy beagle dogs (15 males and 15 females; age, 5–8 years) were divided into 3
groups of 10 dogs each and orally administered 2.5, 5, or 10 mg/kg cilostazol (twice a day at 8:00 AM and 8:00 PM for 10 days). Higher HR increases were seen in the 5 mg/kg group than in the
2.5 mg/kg group at all time points except 7:00 AM, 9:00 AM, 1:00 PM, and 4:00 PM (P<0.01). Higher HR increases were also observed in the 10 mg/kg group than in the 2.5
mg/kg group at all time points except 4:00 PM (P<0.01). The 10 mg/kg group showed higher HR increases than the 5 mg/kg group at all time points except 6:00 AM, 7:00 AM,
6:00 PM, and 7:00 PM (P<0.05 for 4:00 PM and 5:00 PM; P<0.01 for the other time points). These results together show that the HR of healthy dogs
increased in a dose-dependent manner after cilostazol administration twice a day at doses of 5 to 10 mg/kg. These results provide a useful basis for choosing cilostazol in the treatment of
bradyarrhythmia in dogs.
Collapse
Affiliation(s)
- Ryuji Fukushima
- Animal Medical Center, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo 183-8509, Japan
| | - Takae Kawaguchi
- Animal Medical Center, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo 183-8509, Japan
| | - Shusaku Yamada
- Animal Medical Center, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo 183-8509, Japan
| | - Aritada Yoshimura
- Animal Medical Center, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo 183-8509, Japan
| | - Daiki Hirao
- Animal Medical Center, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo 183-8509, Japan
| | - Takahiro Oomori
- Animal Medical Center, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo 183-8509, Japan
| |
Collapse
|
17
|
Choi HI, Kim DY, Choi SJ, Shin CY, Hwang ST, Kim KH, Kwon O. The effect of cilostazol, a phosphodiesterase 3 (PDE3) inhibitor, on human hair growth with the dual promoting mechanisms. J Dermatol Sci 2018; 91:60-68. [DOI: 10.1016/j.jdermsci.2018.04.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 03/23/2018] [Accepted: 04/03/2018] [Indexed: 02/07/2023]
|
18
|
Ercu M, Klussmann E. Roles of A-Kinase Anchoring Proteins and Phosphodiesterases in the Cardiovascular System. J Cardiovasc Dev Dis 2018; 5:jcdd5010014. [PMID: 29461511 PMCID: PMC5872362 DOI: 10.3390/jcdd5010014] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 02/16/2018] [Accepted: 02/18/2018] [Indexed: 12/13/2022] Open
Abstract
A-kinase anchoring proteins (AKAPs) and cyclic nucleotide phosphodiesterases (PDEs) are essential enzymes in the cyclic adenosine 3′-5′ monophosphate (cAMP) signaling cascade. They establish local cAMP pools by controlling the intensity, duration and compartmentalization of cyclic nucleotide-dependent signaling. Various members of the AKAP and PDE families are expressed in the cardiovascular system and direct important processes maintaining homeostatic functioning of the heart and vasculature, e.g., the endothelial barrier function and excitation-contraction coupling. Dysregulation of AKAP and PDE function is associated with pathophysiological conditions in the cardiovascular system including heart failure, hypertension and atherosclerosis. A number of diseases, including autosomal dominant hypertension with brachydactyly (HTNB) and type I long-QT syndrome (LQT1), result from mutations in genes encoding for distinct members of the two classes of enzymes. This review provides an overview over the AKAPs and PDEs relevant for cAMP compartmentalization in the heart and vasculature and discusses their pathophysiological role as well as highlights the potential benefits of targeting these proteins and their protein-protein interactions for the treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Maria Ercu
- Max Delbrück Center for Molecular Medicine Berlin (MDC), Berlin 13125, Germany.
| | - Enno Klussmann
- Max Delbrück Center for Molecular Medicine Berlin (MDC), Berlin 13125, Germany.
- DZHK (German Centre for Cardiovascular Research), partner site Berlin 13347, Germany.
| |
Collapse
|
19
|
The efficacy and safety of cilostazol as an alternative to aspirin in Chinese patients with aspirin intolerance after coronary stent implantation: a combined clinical study and computational system pharmacology analysis. Acta Pharmacol Sin 2018; 39:205-212. [PMID: 28933424 DOI: 10.1038/aps.2017.85] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 05/19/2017] [Indexed: 12/18/2022]
Abstract
Dual antiplatelet therapy (DAT) with aspirin and clopidogrel is the standard regimen to achieve rapid platelet inhibition and prevent thrombotic events. Currently, little information is available regarding alternative antiplatelet therapy in patients with an allergy or intolerance to aspirin. Although cilostazol is already a common alternative to aspirin in clinical practice in China, its efficacy and safety remain to be determined. We retrospectively analyzed 613 Chinese patients who had undergone primary percutaneous coronary intervention (PCI). Among them, 405 patients received standard DAT (aspirin plus clopidogrel) and 205 patients were identified with intolerance to aspirin and received alternative DAT (cilostazol plus clopidogrel). There were no significant differences between the two groups in their baseline clinical characteristics. The main outcomes of the study included major adverse cardiac events (MACEs) and bleeding events during 12 months of follow-up. The MACEs endpoint was reached in 10 of 205 patients treated with cilostazol (4.9%) and in 34 of 408 patients treated with aspirin (8.3%). No statistically significant difference was observed in MACEs between the two groups. However, patients in the cilostazol group had less restenosis than did patients in the aspirin group (1.5% vs 4.9%, P=0.035). The occurrence of bleeding events tended to be lower in the cilostazol group (0.49% vs 2.7%, P=0.063). These clinical observations were further analyzed using network system pharmacology analysis, and the outcomes were consistent with clinical observations and preclinical data reports. We conclude that in Chinese patients with aspirin intolerance undergoing coronary stent implantation, the combination of clopidogrel with cilostazol may be an efficacious and safe alternative to the standard DAT regimen.
Collapse
|
20
|
Yanai S, Toyohara J, Ishiwata K, Ito H, Endo S. Long-term cilostazol administration ameliorates memory decline in senescence-accelerated mouse prone 8 (SAMP8) through a dual effect on cAMP and blood-brain barrier. Neuropharmacology 2016; 116:247-259. [PMID: 27979612 DOI: 10.1016/j.neuropharm.2016.12.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 11/18/2016] [Accepted: 12/09/2016] [Indexed: 12/12/2022]
Abstract
Phosphodiesterases (PDEs), which hydrolyze and inactivate 3', 5'-cyclic adenosine monophosphate (cAMP) and 3', 5'-cyclic guanosine monophosphate (cGMP), play an important role in synaptic plasticity that underlies memory. Recently, several PDE inhibitors were assessed for their possible therapeutic efficacy in treating cognitive disorders. Here, we examined how cilostazol, a selective PDE3 inhibitor, affects brain functions in senescence-accelerated mouse prone 8 (SAMP8), an animal model of age-related cognitive impairment. Long-term administration of cilostazol restored the impaired context-dependent conditioned fear memory of SAMP8 to match that in normal aging control substrain SAMR1. Cilostazol also increased the number of cells containing phosphorylated cAMP-responsive element binding protein (CREB), a downstream component of the cAMP pathway. Finally, cilostazol improves blood-brain barrier (BBB) integrity, demonstrated by reduced extravasation of 2-deoxy-2-18F-fluoro-d-glucose and Evans Blue dye in the brains of SAMP8. This improvement in BBB integrity was associated with an increased amount of zona occludens protein 1 (ZO-1) and occludin proteins, components of tight junctions integral to the BBB. The results suggest that long-term administration of cilostazol exerts its beneficial effects on age-related cognitive impairment through a dual mechanism: by enhancing the cAMP system in the brain and by maintaining or improving BBB integrity.
Collapse
Affiliation(s)
- Shuichi Yanai
- Aging Neuroscience Research Team, Tokyo Metropolitan Institute of Gerontology, Itabashi, Tokyo 173-0015, Japan
| | - Jun Toyohara
- Research Team for Neuroimaging, Tokyo Metropolitan Institute of Gerontology, Itabashi, Tokyo 173-0015, Japan
| | - Kiichi Ishiwata
- Research Team for Neuroimaging, Tokyo Metropolitan Institute of Gerontology, Itabashi, Tokyo 173-0015, Japan; Institute of Cyclotron and Drug Discovery Research, Southern TOHOKU Research Institute for Neuroscience, Koriyama, Fukushima 963-8052, Japan; Department of Biofunctional Imaging, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Hideki Ito
- Department of CNS Research, Otsuka Pharmaceutical Co., Ltd., Tokushima 771-0192, Japan
| | - Shogo Endo
- Aging Neuroscience Research Team, Tokyo Metropolitan Institute of Gerontology, Itabashi, Tokyo 173-0015, Japan.
| |
Collapse
|
21
|
Fan ZG, Ding GB, Li XB, Gao XF, Gao YL, Tian NL. The clinical outcomes of triple antiplatelet therapy versus dual antiplatelet therapy for high-risk patients after coronary stent implantation: a meta-analysis of 11 clinical trials and 9,553 patients. DRUG DESIGN DEVELOPMENT AND THERAPY 2016; 10:3435-3448. [PMID: 27799743 PMCID: PMC5076804 DOI: 10.2147/dddt.s119616] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Background The optimal antiplatelet regimen after in-coronary intervention among patients presenting with complex coronary artery lesions or acute coronary syndrome (ACS) has remained unclear. This study sought to evaluate the clinical outcomes of triple antiplatelet treatment (TAPT) (cilostazol added to aspirin plus clopidogrel) in these patients. Methods The PubMed, EMBASE, MEDLINE, and other Internet sources were searched for relevant articles. The primary end point was major adverse cardiac events (MACE), including all-cause mortality, myocardial infarction, and target vessel revascularization. The incidence of definite/probable stent thrombosis and bleeding were analyzed as the safety end points. Results Eleven clinical trials involving 9,553 patients were analyzed. The risk of MACE was significantly decreased following TAPT after stent implantation in the ACS subgroup (odds ratio [OR]: 0.72; 95% confidence interval [CI]: 0.61–0.85; P<0.001), which might mainly result from the lower risk of all-cause mortality in this subset (OR: 0.62; 95% CI: 0.48–0.80; P<0.001). The risk of bleeding was not increased with respect to TAPT. Conclusion TAPT after stent implantation was associated with feasible benefits on reducing the risk of MACE, especially on reducing the incidence of all-cause mortality among patients suffering from ACS, without higher incidence of bleeding. Larger and more powerful randomized trials are still warranted to prove the superiority of TAPT for such patients.
Collapse
Affiliation(s)
- Zhong-Guo Fan
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing
| | - Guo-Bin Ding
- Department of Cardiology, Taixing People's Hospital, Yangzhou University, Taizhou
| | - Xiao-Bo Li
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing; Department of Cardiology, Nanjing Heart Center, Nanjing, People's Republic of China
| | - Xiao-Fei Gao
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing; Department of Cardiology, Nanjing Heart Center, Nanjing, People's Republic of China
| | - Ya-Li Gao
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing
| | - Nai-Liang Tian
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing; Department of Cardiology, Nanjing Heart Center, Nanjing, People's Republic of China
| |
Collapse
|
22
|
Patocskai B, Barajas-Martinez H, Hu D, Gurabi Z, Koncz I, Antzelevitch C. Cellular and ionic mechanisms underlying the effects of cilostazol, milrinone, and isoproterenol to suppress arrhythmogenesis in an experimental model of early repolarization syndrome. Heart Rhythm 2016; 13:1326-34. [PMID: 26820510 DOI: 10.1016/j.hrthm.2016.01.024] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Indexed: 02/07/2023]
Abstract
BACKGROUND Early repolarization syndrome (ERS) is associated with polymorphic ventricular tachycardia (PVT) and ventricular fibrillation, leading to sudden cardiac death. OBJECTIVE The present study tests the hypothesis that the transient outward potassium current (Ito)-blocking effect of phosphodiesterase-3 (PDE-3) inhibitors plays a role in reversing repolarization heterogeneities responsible for arrhythmogenesis in experimental models of ERS. METHODS Transmembrane action potentials (APs) were simultaneously recorded from epicardial and endocardial regions of coronary-perfused canine left ventricular (LV) wedge preparations, together with a transmural pseudo-electrocardiogram. The Ito agonist NS5806 (7-15 μM) and L-type calcium current (ICa) blocker verapamil (2-3 μM) were used to induce an early repolarization pattern and PVT. RESULTS After stable induction of arrhythmogenesis, the PDE-3 inhibitors cilostazol and milrinone or isoproterenol were added to the coronary perfusate. All were effective in restoring the AP dome in the LV epicardium, thus abolishing the repolarization defects responsible for phase 2 reentry and PVT. Arrhythmic activity was suppressed in 7 of 8 preparations by cilostazol (10 μM), 6 of 7 by milrinone (2.5 μM), and 7 of 8 by isoproterenol (0.1-1 μM). Using voltage clamp techniques applied to LV epicardial myocytes, both cilostazol (10 μM) and milrinone (2.5 μM) were found to reduce Ito by 44.4% and 40.4%, respectively, in addition to their known effects to augment ICa. CONCLUSION Our findings suggest that PDE-3 inhibitors exert an ameliorative effect in the setting of ERS by producing an inward shift in the balance of current during the early phases of the epicardial AP via inhibition of Ito as well as augmentation of ICa, thus reversing the repolarization defects underlying the development of phase 2 reentry and ventricular tachycardia/ventricular fibrillation.
Collapse
Affiliation(s)
- Bence Patocskai
- Masonic Medical Research Laboratory, Utica, New York; Department of Pharmacology and Pharmacotherapy, University of Szeged, Szeged, Hungary
| | | | - Dan Hu
- Masonic Medical Research Laboratory, Utica, New York
| | - Zsolt Gurabi
- Masonic Medical Research Laboratory, Utica, New York; Department of Pharmacology and Pharmacotherapy, University of Szeged, Szeged, Hungary
| | - István Koncz
- Masonic Medical Research Laboratory, Utica, New York; Department of Pharmacology and Pharmacotherapy, University of Szeged, Szeged, Hungary
| | | |
Collapse
|
23
|
Chen J, Meng H, Xu L, Liu J, Kong D, Chen P, Gong X, Bai J, Zou F, Yang Z, Li C, Eikelboom JW. Efficacy and safety of cilostazol based triple antiplatelet treatment versus dual antiplatelet treatment in patients undergoing coronary stent implantation: an updated meta-analysis of the randomized controlled trials. J Thromb Thrombolysis 2015; 39:23-34. [PMID: 24869717 DOI: 10.1007/s11239-014-1090-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The aim of this study was to obtain best estimates of the efficacy and safety of cilostazol-based triple antiplatelet therapy (TAPT: aspirin, clopidogrel and cilostazol) compared with dual antiplatelet therapy (DAPT: aspirin and clopidogrel) in patients undergoing coronary stent implantation. We searched the literature to identify all randomized clinical trials examining efficacy and safety of TAPT versus DAPT in patients undergoing coronary stent implantation. Major efficacy outcomes were death, non-fatal myocardial infarction (MI), ischemic stroke and stent thrombosis (ST) and the safety outcome was bleeding. Data were analyzed using the Review Manager 5.0.0 software. A total of 19 trials involving 7,464 patients were included. TAPT and DAPT were associated with similar rates of death, non-fatal MI, ischemic stroke and ST, but compared with DAPT, TAPT had lower rates of target lesion revascularization (TLR) (RR 0.67, 95 % CI 0.56-0.82, P < 0.0001) and target vessel revascularization (TVR) (RR 0.65, 95 % CI 0.55-0.77, P < 0.00001), as well as less late loss of minimal lumen diameter (mean difference -0.14, 95 % CI -0.17--0.11, P < 0.00001), and less binary angiographic restenosis (RR 0.54, 95 % CI 0.45-0.65, P < 0.00001). TAPT and DAPT had similar rates of bleeding, but TAPT had significantly higher rates of headache, palpitation, rash and gastrointestinal side-effects. Cilostazol-based TAPT compared with DAPT is associated with improved angiographic outcomes and decreased risk of TLR and TVR but does not reduce major cardiovascular events and is associated with an increase in minor adverse events.
Collapse
Affiliation(s)
- Jun Chen
- Department of Cardiology, First Affiliated Hospital of Nanjing Medical University, 300, Guangzhou Road, Nanjing, 210029, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Hashimoto A, Tanaka M, Takeda S, Ito H, Nagano K. Cilostazol Induces PGI2 Production via Activation of the Downstream Epac-1/Rap1 Signaling Cascade to Increase Intracellular Calcium by PLCε and to Activate p44/42 MAPK in Human Aortic Endothelial Cells. PLoS One 2015; 10:e0132835. [PMID: 26181635 PMCID: PMC4504471 DOI: 10.1371/journal.pone.0132835] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Accepted: 06/19/2015] [Indexed: 12/31/2022] Open
Abstract
Background Cilostazol, a selective phosphodiesterase 3 (PDE3) inhibitor, is known as an anti-platelet drug and acts directly on platelets. Cilostazol has been shown to exhibit vascular protection in ischemic diseases. Although vascular endothelium-derived prostaglandin I2 (PGI2) plays an important role in vascular protection, it is unknown whether cilostazol directly stimulates PGI2 synthesis in endothelial cells. Here, we elucidate the mechanism of cilostazol-induced PGI2 stimulation in endothelial cells. Methods and Results Human aortic endothelial cells (HAECs) were stimulated with cilostazol and PGI2 accumulation in the culture media was measured. Cilostazol increased PGI2 synthesis via the arachidonic acid pathway. Cilostazol-induced intracellular calcium also promoted PGI2 synthesis via the inositol 1,4,5-trisphosphate receptor. Using RNAi, silencing of PDE3B abolished the induction effect of cilostazol on PGI2 synthesis and intracellular cAMP accumulation. Inhibition of the exchange protein, which was directly activated by cyclic AMP 1 (Epac-1) and its downstream signal the Ras-like small GTPase (Rap-1), abolished cilostazol-induced PGI2 synthesis, but this did not take place via protein kinase A (PKA). Inhibition of downstream signaling, such as mitogen-activated protein kinase (MAPK), phosphoinositide 3-kinase (PI3K) γ, and phospholipase C (PLC) ε, suppressed cilostazol-induced PGI2 synthesis. Conclusions The PDE3/Epac-1/Rap-1 signaling pathway plays an important role in cilostazol-induced PGI2 synthesis. Namely, stimulation of HAECs with cilostazol induces intracellular calcium elevation via the Rap-1/PLCε/IP3 pathway, along with MAPK activation via direct activation by Epac-1/Rap-1 and indirect activation by Epac-1/Rap-1/PI3Kγ, resulting in synergistically induced PGI2 synthesis.
Collapse
Affiliation(s)
- Ayako Hashimoto
- First Institute of New Drug Discovery, Otsuka Pharmaceutical Co., Ltd., Tokushima-shi, Tokushima, Japan
| | - Michinori Tanaka
- Medical Chemistry Research Institute, Otsuka Pharmaceutical Co., Ltd. Tokushima-shi, Tokushima, Japan
| | - Satoshi Takeda
- Qs'Research Institute, Otsuka Pharmaceutical Co., Ltd. Tokushima-shi, Tokushima, Japan
| | - Hideki Ito
- First Institute of New Drug Discovery, Otsuka Pharmaceutical Co., Ltd., Tokushima-shi, Tokushima, Japan
| | - Keisuke Nagano
- First Institute of New Drug Discovery, Otsuka Pharmaceutical Co., Ltd., Tokushima-shi, Tokushima, Japan
| |
Collapse
|
25
|
Maass PG, Aydin A, Luft FC, Schächterle C, Weise A, Stricker S, Lindschau C, Vaegler M, Qadri F, Toka HR, Schulz H, Krawitz PM, Parkhomchuk D, Hecht J, Hollfinger I, Wefeld-Neuenfeld Y, Bartels-Klein E, Mühl A, Kann M, Schuster H, Chitayat D, Bialer MG, Wienker TF, Ott J, Rittscher K, Liehr T, Jordan J, Plessis G, Tank J, Mai K, Naraghi R, Hodge R, Hopp M, Hattenbach LO, Busjahn A, Rauch A, Vandeput F, Gong M, Rüschendorf F, Hübner N, Haller H, Mundlos S, Bilginturan N, Movsesian MA, Klussmann E, Toka O, Bähring S. PDE3A mutations cause autosomal dominant hypertension with brachydactyly. Nat Genet 2015; 47:647-53. [PMID: 25961942 DOI: 10.1038/ng.3302] [Citation(s) in RCA: 118] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 04/17/2015] [Indexed: 01/02/2023]
Abstract
Cardiovascular disease is the most common cause of death worldwide, and hypertension is the major risk factor. Mendelian hypertension elucidates mechanisms of blood pressure regulation. Here we report six missense mutations in PDE3A (encoding phosphodiesterase 3A) in six unrelated families with mendelian hypertension and brachydactyly type E (HTNB). The syndrome features brachydactyly type E (BDE), severe salt-independent but age-dependent hypertension, an increased fibroblast growth rate, neurovascular contact at the rostral-ventrolateral medulla, altered baroreflex blood pressure regulation and death from stroke before age 50 years when untreated. In vitro analyses of mesenchymal stem cell-derived vascular smooth muscle cells (VSMCs) and chondrocytes provided insights into molecular pathogenesis. The mutations increased protein kinase A-mediated PDE3A phosphorylation and resulted in gain of function, with increased cAMP-hydrolytic activity and enhanced cell proliferation. Levels of phosphorylated VASP were diminished, and PTHrP levels were dysregulated. We suggest that the identified PDE3A mutations cause the syndrome. VSMC-expressed PDE3A deserves scrutiny as a therapeutic target for the treatment of hypertension.
Collapse
Affiliation(s)
- Philipp G Maass
- 1] Experimental and Clinical Research Center (ECRC), a joint cooperation between the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany. [2] Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Atakan Aydin
- 1] Experimental and Clinical Research Center (ECRC), a joint cooperation between the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany. [2] Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Friedrich C Luft
- 1] Experimental and Clinical Research Center (ECRC), a joint cooperation between the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany. [2] Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany. [3] Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Carolin Schächterle
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Anja Weise
- Institute of Human Genetics, Jena University Hospital, Friedrich Schiller University, Jena, Germany
| | - Sigmar Stricker
- 1] Max Planck Institute for Molecular Genetics, Berlin, Germany. [2] Institute for Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Carsten Lindschau
- 1] Department of Nephrology, Hannover University Medical School, Hannover, Germany. [2] Staatliche Technikerschule Berlin, Berlin, Germany
| | - Martin Vaegler
- 1] Experimental and Clinical Research Center (ECRC), a joint cooperation between the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany. [2] Department of Urology, Laboratory of Tissue Engineering, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Fatimunnisa Qadri
- 1] Experimental and Clinical Research Center (ECRC), a joint cooperation between the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany. [2] Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Hakan R Toka
- 1] Division of Nephrology and Hypertension, Eastern Virginia Medical School, Norfolk, Virginia, USA. [2] Division of Nephrology, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Herbert Schulz
- 1] Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany. [2] Cologne Center for Genomics (CCG), University of Cologne, Cologne, Germany
| | - Peter M Krawitz
- 1] Max Planck Institute for Molecular Genetics, Berlin, Germany. [2] Institute for Medical Genetics and Human Genetics, Charité Universitätsmedizin Berlin, Berlin, Germany. [3] Berlin Brandenburg Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Dmitri Parkhomchuk
- 1] Max Planck Institute for Molecular Genetics, Berlin, Germany. [2] Institute for Medical Genetics and Human Genetics, Charité Universitätsmedizin Berlin, Berlin, Germany. [3] Berlin Brandenburg Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Jochen Hecht
- 1] Max Planck Institute for Molecular Genetics, Berlin, Germany. [2] Berlin Brandenburg Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Irene Hollfinger
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Yvette Wefeld-Neuenfeld
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Eireen Bartels-Klein
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Astrid Mühl
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Martin Kann
- 1] Department II of Medicine, University of Cologne, Cologne, Germany. [2] Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | | | - David Chitayat
- 1] Division of Clinical and Metabolic Genetics, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada. [2] Prenatal Diagnosis and Medical Genetics Program, Department of Obstetrics and Gynecology, Mount Sinai Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Martin G Bialer
- 1] Division of Medical Genetics, North Shore/LIJ Health System, Manhasset, New York, USA. [2] Department of Pediatrics, North Shore/LIJ Health System, Manhasset, New York, USA
| | - Thomas F Wienker
- 1] Max Planck Institute for Molecular Genetics, Berlin, Germany. [2] Institute for Medical Biometry, Informatics and Epidemiology, University of Bonn, Bonn, Germany
| | - Jürg Ott
- 1] Institute of Psychology, Chinese Academy of Sciences, Beijing, China. [2] Statistical Genetics, Rockefeller University, New York, New York, USA
| | - Katharina Rittscher
- Institute of Human Genetics, Jena University Hospital, Friedrich Schiller University, Jena, Germany
| | - Thomas Liehr
- Institute of Human Genetics, Jena University Hospital, Friedrich Schiller University, Jena, Germany
| | - Jens Jordan
- Institute of Clinical Pharmacology, Hannover Medical School, Hannover, Germany
| | - Ghislaine Plessis
- Centre Hospitalier Universitaire de Caen, Cytogénétique Postnatale et Génétique Clinique, Caen, France
| | - Jens Tank
- Institute of Clinical Pharmacology, Hannover Medical School, Hannover, Germany
| | - Knut Mai
- Experimental and Clinical Research Center (ECRC), a joint cooperation between the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Ramin Naraghi
- Department of Neurosurgery, Bundeswehrkrankenhaus Ulm, Ulm, Germany
| | - Russell Hodge
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Maxwell Hopp
- Department of Pediatrics, Griffith Base Hospital, Griffith, New South Wales, Australia
| | - Lars O Hattenbach
- Department of Ophthalmology, Hospital Ludwigshafen, Ludwigshafen, Germany
| | | | - Anita Rauch
- Institute for Medical Genetics, University of Zurich, Zurich, Switzerland
| | - Fabrice Vandeput
- 1] Cardiology Section, Veterans Affairs Salt Lake City Health Care System, Salt Lake City, Utah, USA. [2] Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA. [3] Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, Utah, USA
| | - Maolian Gong
- 1] Experimental and Clinical Research Center (ECRC), a joint cooperation between the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany. [2] Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Franz Rüschendorf
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Norbert Hübner
- 1] Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany. [2] DZHK (German Centre for Cardiovascular Research), Berlin, Germany. [3] Charité Universitätsmedizin, Berlin, Germany
| | - Hermann Haller
- Department of Nephrology, Hannover University Medical School, Hannover, Germany
| | - Stefan Mundlos
- 1] Max Planck Institute for Molecular Genetics, Berlin, Germany. [2] Institute for Medical Genetics and Human Genetics, Charité Universitätsmedizin Berlin, Berlin, Germany. [3] Berlin Brandenburg Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Nihat Bilginturan
- Department of Pediatric Oncology, Hacettepe University, Ankara, Turkey
| | - Matthew A Movsesian
- 1] Cardiology Section, Veterans Affairs Salt Lake City Health Care System, Salt Lake City, Utah, USA. [2] Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA. [3] Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, Utah, USA
| | - Enno Klussmann
- 1] Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany. [2] DZHK (German Centre for Cardiovascular Research), Berlin, Germany
| | - Okan Toka
- Department of Pediatric Cardiology, Children's Hospital, Friedrich Alexander University Erlangen, Erlangen, Germany
| | - Sylvia Bähring
- 1] Experimental and Clinical Research Center (ECRC), a joint cooperation between the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany. [2] Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| |
Collapse
|
26
|
Sahin M, Alizade E, Pala S, Alici G, Ozkan B, Akgun T, Emiroglu Y, Demir S, Yazicioglu MV, Turkmen MM. The effect of cilostazol on right heart function and pulmonary pressure. Cardiovasc Ther 2014; 31:e88-93. [PMID: 23911039 DOI: 10.1111/1755-5922.12041] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
INTRODUCTION Cilostazol is a PDE3 inhibitor and used to treat peripheral arterial disease. There are few reports on the influence of cilostazol on heart. AIMS The aim of this study was to assess this effect on right ventricular function and pulmonary artery pressure. METHODS Forty patients with normal left and right ventricular ejection fraction and mild or moderate pulmonary artery hypertension were enrolled in the study. Right ventricular function was assessed by tricuspid annular plane systolic excursion (TAPSE), fractional area change (FAC), tissue Doppler imaging (TDI), and two-dimensional speckle-tracking echocardiography (2D-STE) before and after oral administration of cilostazol. Also pulmonary artery pressure assessed before and after administration of cilostazol. RESULTS After cilostazol administration, there were significant increases in the TAPSE (1.9 ± 0.3 cm vs. 2.2 ± 0.3 cm, P < 0.001). Peak longitudinal strain (-18.7 ± 4.5% vs. -21.3 ± 3.7%, P = 0.001), isovolumetric acceleration (IVA) (176.6 ± 62.7 cm/sec(2) vs. 200.6 ± 61.9 cm/sec(2) , P = 0.025), right ventricular FAC increased significantly (37.6 ± 8.0% vs. 41.5 ± 8.9%, P < 0.001). Pulmonary artery pressure decreased significantly (39.9 ± 7.9 vs. 36.6 ± 5.5 mmHg, P = 0.001) after cilostazol administration. CONCLUSION Our study demonstrated that cilostazol improved right ventricular systolic function and reduced pulmonary artery pressure.
Collapse
Affiliation(s)
- Muslum Sahin
- Department of Cardiology, Kartal Kosuyolu High Speciality Education and Research Hospital, Istanbul, Turkey
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Chattipakorn SC, Thummasorn S, Sanit J, Chattipakorn N. Phosphodiesterase-3 inhibitor (cilostazol) attenuates oxidative stress-induced mitochondrial dysfunction in the heart. JOURNAL OF GERIATRIC CARDIOLOGY : JGC 2014; 11:151-7. [PMID: 25009566 PMCID: PMC4076456 DOI: 10.3969/j.issn.1671-5411.2014.02.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Revised: 05/05/2014] [Accepted: 05/30/2014] [Indexed: 11/28/2022]
Abstract
Background Cilostazol is a type 3 phosphodiesterase inhibitor which has been previously demonstrated to prevent the occurrence of tachyarrhythmia and improve defibrillation efficacy. However, the mechanism for this beneficial effect is still unclear. Since cardiac mitochondria have been shown to play a crucial role in fatal cardiac arrhythmias and that oxidative stress is one of the main contributors to arrhythmia generation, we tested the effects of cilostazol on cardiac mitochondria under severe oxidative stress. Methods Mitochondria were isolated from rat hearts and treated with H2O2 to induce oxidative stress. Cilostazol, at various concentrations, was used to study its protective effects. Pharmacological interventions, including a mitochondrial permeability transition pore (mPTP) blocker, cyclosporine A (CsA), and an inner membrane anion channel (IMAC) blocker, 4′-chlorodiazepam (CDP), were used to investigate the mechanistic role of cilostazol on cardiac mitochondria. Cardiac mitochondrial reactive oxygen species (ROS) production, mitochondrial membrane potential change and mitochondrial swelling were determined as indicators of cardiac mitochondrial function. Results Cilostazol preserved cardiac mitochondrial function when exposed to oxidative stress by preventing mitochondrial depolarization, mitochondrial swelling, and decreasing ROS production. Conclusions Our findings suggest that cardioprotective effects of cilostazol reported previously could be due to its prevention of cardiac mitochondrial dysfunction caused by severe oxidative stress.
Collapse
Affiliation(s)
- Siriporn C Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand ; Faculty of Dentistry, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Savitree Thummasorn
- Cardiac Electrophysiology Research and Training Center, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Jantira Sanit
- Cardiac Electrophysiology Research and Training Center, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Nipon Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand ; Biomedical Engineering Center, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
28
|
Yanai S, Semba Y, Ito H, Endo S. Cilostazol improves hippocampus-dependent long-term memory in mice. Psychopharmacology (Berl) 2014; 231:2681-93. [PMID: 24464529 DOI: 10.1007/s00213-014-3442-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Accepted: 12/31/2013] [Indexed: 12/15/2022]
Abstract
RATIONALE Phosphodiesterases (PDEs) play an important role in the regulation of intracellular signaling mediated by cyclic adenosine monophosphate (cAMP). Recently, several PDE inhibitors were assessed for their possible cognitive enhancing properties. However, little is known about the effect of PDE3 inhibitors on memory function. OBJECTIVES We examined how the PDE3 inhibitor cilostazol affects C57BL/6 J mice as they perform various behavioral tasks. After behavioral assessment, brains of the mice were analyzed immunohistochemically to quantify the phosphorylation of cAMP-responsive element binding protein (CREB), a downstream component of the cAMP pathway. RESULTS Oral administration of cilostazol significantly enhanced recollection of the exact platform location in the Morris water maze probe test. Cilostazol also improved context-dependent long-term fear memory, without affecting short-term memory. No apparent effect was observed in cue-dependent fear memory. The results suggest that cilostazol selectively improves hippocampus-dependent long-term memory in these tasks. Cilostazol also significantly increased the number of phosphorylated-CREB-positive cells in hippocampal dentate gyrus. CONCLUSIONS These results suggest that cilostazol may exert its beneficial effects on learning and memory by enhancing the cAMP system in hippocampus, where it increases intracellular cAMP activity.
Collapse
Affiliation(s)
- Shuichi Yanai
- Aging Neuroscience Research Team, Tokyo Metropolitan Institute of Gerontology, Itabashi, Tokyo, 173-0015, Japan
| | | | | | | |
Collapse
|
29
|
Fuentes E, Palomo I. Regulatory mechanisms of cAMP levels as a multiple target for antiplatelet activity and less bleeding risk. Thromb Res 2014; 134:221-6. [PMID: 24830902 DOI: 10.1016/j.thromres.2014.04.027] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Revised: 04/22/2014] [Accepted: 04/25/2014] [Indexed: 12/19/2022]
Abstract
Platelet activation is a critical component of atherothrombosis. The multiple pathways of platelet activation limit the effect of specific receptor/pathway inhibitors, resulting in limited clinical efficacy. Recent research has confirmed that combination therapy results in enhanced antithrombotic efficacy without increasing bleeding risk. In this way, the best-known inhibitor and turn off signaling in platelet activation is cAMP. In this article we discuss the mechanisms of regulation of intraplatelet cAMP levels, a) platelet-dependent pathway: Gi/Gs protein-coupled receptors, phosphodiesterase inhibition and activation of PPARs and b) platelet-independent pathway: inhibition of adenosine uptake by erythrocytes. With respect to the association between intraplatelet cAMP levels and bleeding risk it is possible to establish that compounds/drugs with pleitropic effect for increased intraplatelet cAMP level could have an antithrombotic activity with less risk of bleeding.
Collapse
Affiliation(s)
- Eduardo Fuentes
- Department of Clinical Biochemistry and Immunohaematology, Faculty of Health Sciences, Interdisciplinary Excellence Research Program on Healthy Aging (PIEI-ES), Universidad de Talca, Talca, Chile; Centro de Estudios en Alimentos Procesados (CEAP), CONICYT-Regional, Gore Maule, R09I2001, Chile.
| | - Iván Palomo
- Department of Clinical Biochemistry and Immunohaematology, Faculty of Health Sciences, Interdisciplinary Excellence Research Program on Healthy Aging (PIEI-ES), Universidad de Talca, Talca, Chile; Centro de Estudios en Alimentos Procesados (CEAP), CONICYT-Regional, Gore Maule, R09I2001, Chile.
| |
Collapse
|
30
|
Collinson DJ, Donnelly R. Cilostazol: improving walking distance in patients with intermittent claudication. Expert Rev Cardiovasc Ther 2014; 2:503-9. [PMID: 15225110 DOI: 10.1586/14779072.2.4.503] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Intermittent claudication is a common, disabling symptom of peripheral arterial disease that limits walking distance and is associated with an increased cardiovascular risk of acute limb- or life-threatening complications. Very few patients with intermittent claudication (<7%) are suitable candidates for surgical revascularization, yet in contrast to the treatment of stable angina, few effective medical therapies (apart from exercise) are available for the symptomatic relief of intermittent claudication. The phosphodiesterase-3 inhibitor, cilostazol (Pletal, Otsuka Pharmaceuticals Ltd), is the first symptom-relieving treatment for intermittent claudication that has been evaluated successfully in large multicenter placebo-controlled, double-blind clinical trials (involving >2000 patients). A meta-analysis of the eight major efficacy studies with cilostazol has shown significant improvements in pain-free and maximum walking distance, and a good overall safety and tolerability profile. Thus, in the UK, USA and Japan, cilostazol administered at 100 mg twice daily is licensed for symptom relief in patients with stable, moderate-to-severe intermittent claudication, as an adjunct to nonpharmacological approaches such as exercise.
Collapse
Affiliation(s)
- Deborah J Collinson
- University of Nottinham Medical School, Derby City General Hospital, Uttoxeter Road, Derby, DE22 3DT, UK
| | | |
Collapse
|
31
|
Baek AE, Kanthi Y, Sutton NR, Liao H, Pinsky DJ. Regulation of ecto-apyrase CD39 (ENTPD1) expression by phosphodiesterase III (PDE3). FASEB J 2013; 27:4419-28. [PMID: 23901069 DOI: 10.1096/fj.13-234625] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The ectoenzyme CD39 suppresses thrombosis and inflammation by suppressing ATP and ADP to AMP. However, mechanisms of CD39 transcriptional and post-translational regulation are not well known. Here we show that CD39 levels are modulated by inhibition of phosphodiesterase 3 (PDE3). RAW macrophages and human umbilical vein endothelial cells (HUVECs) were treated with the PDE3 inhibitors cilostazol and milrinone, then analyzed using qRT-PCR, immunoprecipitation/Western blot, immunofluorescent staining, radio-thin-layer chromatography, a malachite green assay, and ELISA. HUVECs expressed elevated CD39 protein (2-fold [P<0.05] for cilostazol and 2.5-fold [P<0.01] for milrinone), while macrophage CD39 mRNA and protein were both elevated after PDE3 inhibition. HUVEC ATPase activity increased by 25% with cilostazol and milrinone treatment (P<0.05 and P<0.01, respectively), as did ADPase activity (47% and 61%, P<0.001). There was also a dose-dependent elevation of soluble CD39 after treatment with 8-Br-cAMP, with maximal elevation of 60% more CD39 present compared to controls (1 mM, P<0.001). Protein harvested after 8-Br-cAMP treatment showed that ubiquitination of CD39 was decreased by 43% compared to controls. A DMSO or PBS vehicle control was included for each experiment based on solubility of cilostazol, milrinone, and 8-Br-cAMP. These results indicate that PDE3 inhibition regulates endothelial CD39 at a post-translational level.
Collapse
Affiliation(s)
- Amy E Baek
- 17240 Medical Science Research Bldg. III, 1150 W. Medical Center Dr., Ann Arbor, MI 48109, USA.
| | | | | | | | | |
Collapse
|
32
|
TSUTSUMI M, AIKAWA H, NII K, ETOU H, SAKAMOTO K, KURESHIMA M, INOUE R, YOSHIDA H, MATSUMOTO Y, NARITA S, KAZEKAWA K. Cilostazol Reduces Periprocedural Hemodynamic Depression in Carotid Artery Stenting. Neurol Med Chir (Tokyo) 2013; 53:163-70. [DOI: 10.2176/nmc.53.163] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Masanori TSUTSUMI
- Department of Neurosurgery and Neuroradiology, Fukuoka University Chikushi Hospital
| | - Hiroshi AIKAWA
- Department of Neurosurgery and Neuroradiology, Fukuoka University Chikushi Hospital
| | - Kouhei NII
- Department of Neurosurgery and Neuroradiology, Fukuoka University Chikushi Hospital
| | - Housei ETOU
- Department of Neurosurgery and Neuroradiology, Fukuoka University Chikushi Hospital
| | - Kimiya SAKAMOTO
- Department of Neurosurgery and Neuroradiology, Fukuoka University Chikushi Hospital
| | - Makoto KURESHIMA
- Department of Neurosurgery and Neuroradiology, Fukuoka University Chikushi Hospital
| | - Ritsurou INOUE
- Department of Neurosurgery and Neuroradiology, Fukuoka University Chikushi Hospital
| | - Hidenori YOSHIDA
- Department of Neurosurgery and Neuroradiology, Fukuoka University Chikushi Hospital
| | - Yoshihisa MATSUMOTO
- Department of Neurosurgery and Neuroradiology, Fukuoka University Chikushi Hospital
| | - Sumito NARITA
- Department of Neurosurgery and Neuroradiology, Fukuoka University Chikushi Hospital
| | - Kiyoshi KAZEKAWA
- Department of Neurosurgery and Neuroradiology, Fukuoka University Chikushi Hospital
| |
Collapse
|
33
|
Affiliation(s)
- Jeffrey S Berger
- Divisions of Cardiology and Vascular Surgery, New York University School of Medicine, New York, NY, USA
| | | |
Collapse
|
34
|
Synthesis and evaluation of novel 2-pyridone derivatives as inhibitors of phosphodiesterase3 (PDE3): A target for heart failure and platelet aggregation. Bioorg Med Chem Lett 2012; 22:6010-5. [PMID: 22897945 DOI: 10.1016/j.bmcl.2012.05.019] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2012] [Revised: 04/16/2012] [Accepted: 05/02/2012] [Indexed: 01/24/2023]
|
35
|
Trian T, Burgess JK, Niimi K, Moir LM, Ge Q, Berger P, Liggett SB, Black JL, Oliver BG. β2-Agonist induced cAMP is decreased in asthmatic airway smooth muscle due to increased PDE4D. PLoS One 2011; 6:e20000. [PMID: 21611147 PMCID: PMC3096656 DOI: 10.1371/journal.pone.0020000] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2010] [Accepted: 04/22/2011] [Indexed: 11/19/2022] Open
Abstract
Background and Objective Asthma is associated with airway narrowing in response to bronchoconstricting stimuli and increased airway smooth muscle (ASM) mass. In addition, some studies have suggested impaired β-agonist induced ASM relaxation in asthmatics, but the mechanism is not known. Objective To characterize the potential defect in β-agonist induced cAMP in ASM derived from asthmatic in comparison to non-asthmatic subjects and to investigate its mechanism. Methods We examined β2-adrenergic (β2AR) receptor expression and basal β-agonist and forskolin (direct activator of adenylyl cyclase) stimulated cAMP production in asthmatic cultured ASM (n = 15) and non-asthmatic ASM (n = 22). Based on these results, PDE activity, PDE4D expression and cell proliferation were determined. Results In the presence of IBMX, a pan PDE inhibitor, asthmatic ASM had ∼50% lower cAMP production in response to isoproterenol, albuterol, formoterol, and forskolin compared to non-asthmatic ASM. However when PDE4 was specifically inhibited, cAMP production by the agonists and forskolin was normalized in asthmatic ASM. We then measured the amount and activity of PDE4, and found ∼2-fold greater expression and activity in asthmatic ASM compared to non-asthmatic ASM. Furthermore, inhibition of PDE4 reduced asthmatic ASM proliferation but not that of non-asthmatic ASM. Conclusion Decreased β-agonist induced cAMP in ASM from asthmatics results from enhanced degradation due to increased PDE4D expression. Clinical manifestations of this dysregulation would be suboptimal β-agonist-mediated bronchodilation and possibly reduced control over increasing ASM mass. These phenotypes appear to be “hard-wired” into ASM from asthmatics, as they do not require an inflammatory environment in culture to be observed.
Collapse
Affiliation(s)
- Thomas Trian
- Cell Biology, Woolcock Institute of Medical Research, Sydney, New South Wales, Australia
- Discipline of Pharmacology, School of Medical Sciences, The University of Sydney, Sydney, New South Wales, Australia
| | - Janette K. Burgess
- Cell Biology, Woolcock Institute of Medical Research, Sydney, New South Wales, Australia
- Discipline of Pharmacology, School of Medical Sciences, The University of Sydney, Sydney, New South Wales, Australia
| | - Kyoko Niimi
- Cell Biology, Woolcock Institute of Medical Research, Sydney, New South Wales, Australia
- Discipline of Pharmacology, School of Medical Sciences, The University of Sydney, Sydney, New South Wales, Australia
| | - Lyn M. Moir
- Cell Biology, Woolcock Institute of Medical Research, Sydney, New South Wales, Australia
- Discipline of Pharmacology, School of Medical Sciences, The University of Sydney, Sydney, New South Wales, Australia
| | - Qi Ge
- Cell Biology, Woolcock Institute of Medical Research, Sydney, New South Wales, Australia
- Discipline of Pharmacology, School of Medical Sciences, The University of Sydney, Sydney, New South Wales, Australia
| | - Patrick Berger
- Centre de Recherce Cardio-Thoracique de Bordeaux, Université Bordeaux Segalen, INSERM, Bordeaux, France
| | - Stephen B. Liggett
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Judith L. Black
- Cell Biology, Woolcock Institute of Medical Research, Sydney, New South Wales, Australia
- Discipline of Pharmacology, School of Medical Sciences, The University of Sydney, Sydney, New South Wales, Australia
| | - Brian G. Oliver
- Cell Biology, Woolcock Institute of Medical Research, Sydney, New South Wales, Australia
- Discipline of Pharmacology, School of Medical Sciences, The University of Sydney, Sydney, New South Wales, Australia
- * E-mail:
| |
Collapse
|
36
|
Kanlop N, Chattipakorn S, Chattipakorn N. Effects of cilostazol in the heart. J Cardiovasc Med (Hagerstown) 2011; 12:88-95. [PMID: 21200326 DOI: 10.2459/jcm.0b013e3283439746] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Cilostazol is a selective phosphodiesterase 3 (PDE3) inhibitor approved by the Food and Drug Administration for treatment of intermittent claudication. It has also been used in bradyarrhythmic patients to increase heart rates. Recently, cilostazol has been shown to prevent ventricular fibrillation in patients with Brugada syndrome. Cilostazol is hypothesized to suppress transient outward potassium (Ito) current and increase inward calcium current, thus, maintaining the dome (phase 2) of action potential, decreasing transmural dispersion of repolarization and preventing ventricular fibrillation. Although many PDE3 inhibitors have been shown to increase cardiac arrhythmia in heart failure, cilostazol has presented effects that are different from other PDE3 inhibitors, especially adenosine uptake inhibition. Owing to this effect, cilostazol could be an effective cardioprotective drug, with its beneficial effects in preventing arrhythmia. In this review, the cardiac electrophysiological effects of cilostazol are presented and its possible cardioprotective effects, particularly in preventing ventricular fibrillation, are discussed, with emphasis on the need to further verify its clinical benefits.
Collapse
Affiliation(s)
- Natnicha Kanlop
- Cardiac Electrophysiology Unit, Department of Physiology, Thailand
| | | | | |
Collapse
|
37
|
Yoshikawa H, Suzuki M, Hashimoto G, Otsuka T, Sugi K. Impact of Cilostazol on Left Ventricular Geometry and Function: Assessment by Tissue Doppler Imaging and Two-Dimensional Speckle-Tracking Echocardiography. Echocardiography 2011; 28:431-7. [DOI: 10.1111/j.1540-8175.2010.01357.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
|
38
|
Kanlop N, Shinlapawittayatorn K, Sungnoon R, Weerateerangkul P, Chattipakorn S, Chattipakorn N. Cilostazol attenuates ventricular arrhythmia induction and improves defibrillation efficacy in swine. Can J Physiol Pharmacol 2010; 88:422-8. [PMID: 20555410 DOI: 10.1139/y09-127] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Previous reports demonstrated that cilostazol, a phosphodiesterase 3 inhibitor, affected cellular electrophysiology and reduced episodes of ventricular fibrillation (VF) in patients with Brugada syndrome. However, its effects on VF induction and defibrillation efficacy have never been investigated. We tested the hypothesis that cilostazol increases the VF threshold (VFT) and decreases the upper limit of vulnerability (ULV) and the defibrillation threshold (DFT). A total of 48 pigs were randomly assigned to defibrillation and VF induction studies. The diastolic pacing threshold (DPT), VFT, ULV, DFT, and effective refractory period were determined before and after the infusion of cilostazol at 6 mg/kg, 3 mg/kg, or vehicle. The DPT was significantly increased after administration of 3 and 6 mg/kg cilostazol. The ULV and DFT were significantly decreased after administration of 6 mg/kg cilostazol only. The ULV in the 6 mg/kg group had 12% lower peak voltage and 25% lower total energy, and the DFT had 13% lower peak voltage and 25% lower total energy. The VFT was not altered in any experimental group. This study shows that cilostazol administration significantly increased the DPT, which was associated with significantly reduced DFT and ULV.
Collapse
Affiliation(s)
- Natnicha Kanlop
- Cardiac Electrophysiology Unit, Department of Physiology, and Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | | | | | | | | | | |
Collapse
|
39
|
Adderley SP, Sridharan M, Bowles EA, Stephenson AH, Ellsworth ML, Sprague RS. Protein kinases A and C regulate receptor-mediated increases in cAMP in rabbit erythrocytes. Am J Physiol Heart Circ Physiol 2009; 298:H587-93. [PMID: 20008267 DOI: 10.1152/ajpheart.00975.2009] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Activation of the beta-adrenergic receptor (beta-AR) or the prostacyclin receptor (IPR) results in increases in cAMP and ATP release from erythrocytes. cAMP levels depend on a balance between synthesis via adenylyl cyclase and hydrolysis by phosphodiesterases (PDEs). Previously, we reported that cAMP increases associated with activation of the beta-AR and IPR in rabbit and human erythrocytes are tightly regulated by distinct PDEs. Importantly, inhibitors of these PDEs potentiated both increases in cAMP and ATP release. It has been shown that increases in protein kinase (PK) activity can activate PDE3 and PDE4. Both PKA and PKC are present in the erythrocyte and can phosphorylate and activate these PDEs. Here we investigate the hypothesis that PKA regulates PDE activity associated with the beta-AR and both PKA and PKC regulate the PDE activity associated with the IPR in rabbit erythrocytes. Pretreatment of erythrocytes with the PKA inhibitor, H89 (10 microM), in the presence of the PDE4 inhibitor, rolipram (10 microM), augmented isoproterenol (1 microM)-induced cAMP increases. In contrast, in the presence of the PDE3 inhibitor, cilostazol (10 microM), pretreatment of erythrocytes with either H89 (1 microM) or two chemically dissimilar inhibitors of PKC, calphostin C (1 microM) or GFX109203X (1 microM), potentiated iloprost (1 microM)-induced cAMP increases. Furthermore, pretreatment of erythrocytes with both H89 and GFX109203X in the presence of cilostazol augmented the iloprost-induced increases in cAMP to a greater extent than either PK inhibitor individually. These results support the hypothesis that PDEs associated with receptor-mediated increases in cAMP in rabbit erythrocytes are regulated by kinases specific to the receptor's signaling pathway.
Collapse
Affiliation(s)
- Shaquria P Adderley
- Department of Pharmacological and Physiological Science, Saint Louis University School of Medicine, Saint Louis, Missouri, USA.
| | | | | | | | | | | |
Collapse
|
40
|
Jensen JT, Stouffer RL, Stanley JE, Zelinski MB. Evaluation of the phosphodiesterase 3 inhibitor ORG 9935 as a contraceptive in female macaques: initial trials. Contraception 2009; 81:165-71. [PMID: 20103457 DOI: 10.1016/j.contraception.2009.09.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2009] [Revised: 09/28/2009] [Accepted: 09/30/2009] [Indexed: 10/20/2022]
Abstract
BACKGROUND The study was conducted to determine whether a phosphodiesterase (PDE) 3 inhibitor has potential as a novel contraceptive in primates. METHODS Regularly cycling adult female cynomolgus macaques of proven fertility (n=16) were treated for 7 months with placebo (controls) or the PDE3 inhibitor ORG 9935 as a daily food treat (150 mg/kg) or as a weekly depot injection (150 mg/kg, sc). After 1 month, a male of proven fertility was introduced into each group. Females underwent weekly monitoring of progesterone (P) and ultrasound evaluation for pregnancy if P remained elevated (1.0 ng/mL) >3 weeks. ORG 9935 values were evaluated using high-performance liquid chromatography. RESULTS Overall, the pregnancy rate in ORG 9935-treated monkeys (4/8, 50%) did not differ from controls (7/8, 88%; p=.5). However, no animal became pregnant in a cycle when the serum level of ORG 9935 exceeded 300 nmol/L. Moreover, two treated monkeys who mated throughout the treatment phase and did not conceive became pregnant within four cycles after stopping ORG 9935. The other two animals were discontinued prematurely from the protocol. CONCLUSIONS These results demonstrate that ORG 9935 may prevent pregnancy in primates at serum concentrations above 300 nmol/L and that the effect is reversible.
Collapse
Affiliation(s)
- Jeffrey T Jensen
- Department of Obstetrics and Gynecology, Oregon Health and Science University, Portland, OR 97239, USA.
| | | | | | | |
Collapse
|
41
|
Dobesh PP, Stacy ZA, Persson EL. Pharmacologic therapy for intermittent claudication. Pharmacotherapy 2009; 29:526-53. [PMID: 19397462 DOI: 10.1592/phco.29.5.526] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Peripheral artery disease, defined as atherosclerosis in the lower extremities, affects nearly 8.5 million people in the United States. Due to the frequent asymptomatic manifestation of peripheral artery disease, diagnosis may be delayed and its true incidence underestimated. However, some patients may experience aching pain, numbness, weakness, or fatigue, a condition termed intermittent claudication. Peripheral atherosclerosis is associated with cardiovascular risk and physical impairment; therefore, treatment goals are aimed at decreasing cardiovascular risk, as well as improving quality of life. Little debate exists regarding the management of cardiovascular risk reduction, which consists of both antiplatelet therapy and risk factor modification. Despite recently published guidelines, the treatment of intermittent claudication is less well established and the management remains controversial and uncertain. Exercise remains the first-line therapy for intermittent claudication; however, pharmacologic treatment is often necessary. Although only two prescription drugs have been approved by the U.S. Food and Drug Administration for the treatment of intermittent claudication, several supplements and investigational agents have been evaluated. Therapeutic optimization should balance the anticipated improvements in quality of life with the potential safety risks.
Collapse
Affiliation(s)
- Paul P Dobesh
- College of Pharmacy, University of Nebraska Medical Center, 986045 Nebraska Medical Center, Omaha, NE 68198-6045, USA.
| | | | | |
Collapse
|
42
|
Wang F, Gao L, Gong B, Hu J, Li M, Guan Q, Zhao J. Tissue-specific expression of PPAR mRNAs in diabetic rats and divergent effects of cilostazol. Can J Physiol Pharmacol 2008; 86:465-71. [PMID: 18641696 DOI: 10.1139/y08-043] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Cilostazol and ligands of peroxisome proliferator-activated receptors (PPARs) have been effectively used to alleviate diabetic complications, but the common and tissue-specific expression patterns of PPARs in different tissues in diabetic patients and those treated with cilostazol have not been reported. Here, we aimed to assess the effects of diabetes and cilostazol on mRNA expression of PPARalpha and PPARgamma in the aorta, renal cortex, and retina of diabetic rats treated with cilostazol for 8 weeks. PPARalpha mRNA expression showed uniform downregulation in all these tissues in diabetic rats, and this effect was reversed by cilostazol treatment. Surprisingly, PPARgamma mRNA expression was reduced in the renal cortex and retina, yet increased in the aorta of diabetic rats, although cilostazol still reversed these changes. Interestingly, cilostazol, a well-known phosphodiesterase 3 inhibitor and cAMP elevator, augmented cAMP content only in the aorta, but showed no significant effects in the renal cortex of diabetic rats. In conclusion, mRNA expression of PPARs is tissue-specific in diabetes and may be differently affected by cilostazol, possibly because of its tissue-specific effects on cAMP content.
Collapse
Affiliation(s)
- Furong Wang
- Provincial Hospital affiliated to Shandong University, Jinan, Shandong 250021, China
| | | | | | | | | | | | | |
Collapse
|
43
|
Sanganalmath SK, Barta J, Takeda N, Kumamoto H, Dhalla NS. Antiplatelet therapy mitigates cardiac remodeling and dysfunction in congestive heart failure due to myocardial infarctionThis article is one of a selection of papers published in the special issue Bridging the Gap: Where Progress in Cardiovascular and Neurophysiologic Research Meet. Can J Physiol Pharmacol 2008; 86:180-9. [DOI: 10.1139/y08-005] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Antiplatelet agents such as sarpogrelate (SAR), a 5-hydroxytryptamine antagonist, and cilostazol (CIL), a phosphodiesterase-III inhibitor, are used in the management of peripheral vascular disease. In this study, we tested the hypothesis that both SAR and CIL prevent cardiac remodeling and improve cardiac function in congestive heart failure (CHF) due to myocardial infarction (MI). Post-MI rats (3 weeks after the occlusion of coronary artery) received either vehicle (MI+V, n = 36), SAR (MI+SAR; 5 mg·kg–1·day–1, n = 35) or CIL (MI+CIL; 5 mg·kg–1·day–1, n = 34) from day 21 to day 56. Sham-operated rats (n = 29) served as controls. Electrocardiographic, echocardiographic, and hemodynamic parameters were measured on day 56. Treatment of infarcted animals with SAR or CIL significantly improved the left ventricular (LV) dimensions, LV fractional shortening, cardiac output, stroke volume, mean arterial pressure, LV diastolic function, and LV systolic pressure, as well as rates of LV pressure development and pressure decay. Although cardiac hypertrophy was reduced, both SAR and CIL had no effect on infarct size or MI-associated QTc prolongation. However, SAR decreased whereas CIL increased the incidence of ventricular arrhythmias and the mean number of episodes in infarcted animals. Mortality during the treatment period was decreased by 17% with SAR and increased by 10% with CIL, but these changes were not significant statistically. The data in this study suggest that both SAR and CIL prevent cardiac remodeling and improve cardiac function in MI-induced CHF; however, CIL unlike SAR increased the incidence of arrhythmias and adversely affected patient mortality.
Collapse
Affiliation(s)
- Santosh K. Sanganalmath
- Institute of Cardiovascular Sciences, St. Boniface General Hospital Research Centre, 351 Taché Avenue, and Department of Physiology, Faculty of Medicine, University of Manitoba, Winnipeg, MB R2H 2A6, Canada
- Department of Internal Medicine, Aoto Hospital, Jikei University, Tokyo, Japan
- Business and Marketing Department, Mitsubishi Pharma Europe Ltd., London
| | - Judit Barta
- Institute of Cardiovascular Sciences, St. Boniface General Hospital Research Centre, 351 Taché Avenue, and Department of Physiology, Faculty of Medicine, University of Manitoba, Winnipeg, MB R2H 2A6, Canada
- Department of Internal Medicine, Aoto Hospital, Jikei University, Tokyo, Japan
- Business and Marketing Department, Mitsubishi Pharma Europe Ltd., London
| | - Nobuakira Takeda
- Institute of Cardiovascular Sciences, St. Boniface General Hospital Research Centre, 351 Taché Avenue, and Department of Physiology, Faculty of Medicine, University of Manitoba, Winnipeg, MB R2H 2A6, Canada
- Department of Internal Medicine, Aoto Hospital, Jikei University, Tokyo, Japan
- Business and Marketing Department, Mitsubishi Pharma Europe Ltd., London
| | - Hideo Kumamoto
- Institute of Cardiovascular Sciences, St. Boniface General Hospital Research Centre, 351 Taché Avenue, and Department of Physiology, Faculty of Medicine, University of Manitoba, Winnipeg, MB R2H 2A6, Canada
- Department of Internal Medicine, Aoto Hospital, Jikei University, Tokyo, Japan
- Business and Marketing Department, Mitsubishi Pharma Europe Ltd., London
| | - Naranjan S. Dhalla
- Institute of Cardiovascular Sciences, St. Boniface General Hospital Research Centre, 351 Taché Avenue, and Department of Physiology, Faculty of Medicine, University of Manitoba, Winnipeg, MB R2H 2A6, Canada
- Department of Internal Medicine, Aoto Hospital, Jikei University, Tokyo, Japan
- Business and Marketing Department, Mitsubishi Pharma Europe Ltd., London
| |
Collapse
|
44
|
Soeding PE, Royse CF, Wright CE, Royse AG, Angus JA. Inoprotection: the perioperative role of levosimendan. Anaesth Intensive Care 2008; 35:845-62. [PMID: 18084975 DOI: 10.1177/0310057x0703500602] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Levosimendan is emerging as a novel cardioprotective inotrope. Levosimendan augments myocardial contractility by sensitising contractile myofilaments to calcium without increasing myosin adenosine triphosphatase activity or oxygen consumption. Levosimendan activates cellular adenosine triphosphate-dependent potassium channels, a mechanism which is postulated to protect cells from ischaemia in a manner similar to ischaemic preconditioning. Levosimendan may therefore protect the ischaemic myocardium during ischaemia-reperfusion as well as improve the contractile function of the heart. Adenosine triphosphate-dependent potassium channel activation by levosimendan may also be protective in other tissues, such as coronary vascular endothelium, kidney and brain. Clinical trials in patients with decompensated heart failure and myocardial ischaemia show levosimendan to improve haemodynamic performance and potentially improve survival. This paper reviews the known pharmacology of levosimendan, the clinical experience with the drug to date and the potential use of levosimendan as a cardioprotective agent during surgery.
Collapse
Affiliation(s)
- P E Soeding
- Cardiovascular Therapeutics Unit, Department of Pharmacology, University of Melbourne, Melbourne, Victoria, Australia
| | | | | | | | | |
Collapse
|
45
|
Ozaki K, Maeda C, Takayama T, Hoyano M, Yanagawa T, Tsuchida K, Takahashi K, Miida T, Oda H. Dynamic intraventricular obstruction in acute myocardial infarction with administration of cilostazol. Circ J 2007; 71:608-12. [PMID: 17384467 DOI: 10.1253/circj.71.608] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Dynamic intraventricular obstruction is a less well-known mechanical complication of acute myocardial infarction (AMI). Its hallmark is the development of a new systolic murmur, and echocardiography is necessary for diagnosis. We describe a case of a 74-year-old woman with dynamic intraventricular obstruction complicating AMI. Serial echocardiography suggested that the intraventricular gradient was a consequence of basal hyperkinesis, which was a reciprocal response to akinesis of the apical wall. Cilostazol, which was administered to prevent subacute stent thrombosis after percutaneous coronary intervention, might have contributed to the transient intraventricular obstruction.
Collapse
Affiliation(s)
- Kazuyuki Ozaki
- Department of Cardiology, Niigata City General Hospital.
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Feldman AM, McNamara DM. Reevaluating the role of phosphodiesterase inhibitors in the treatment of cardiovascular disease. Clin Cardiol 2006; 25:256-62. [PMID: 12058787 PMCID: PMC6654250 DOI: 10.1002/clc.4960250603] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
First developed for clinical use in the late 1980s, the phosphodiesterase inhibitors were found to increase the levels of the ubiquitous second messenger cyclic adenosine monophosphate and could effect changes in vascular tone, cardiac function, and other cellular events. After several early studies using high doses of phosphodiesterase inhibitors in patients with severe heart failure suggested adverse consequences, they fell out of favor. However, recent investigations of phosphodiesterase inhibitors in patients with intermittent claudication have demonstrated profound benefits. Furthermore, these agents have proven useful in prevention of cerebral infarction and coronary restenosis, and their use in the treatment of heart failure is being reevaluated. The reemergence of phosphodiesterase inhibitors can be attributed to a better understanding of dosing and drug-specific pharmacology, the use of concomitant medications, and a recognition of unique ancillary properties; however, their use still requires caution.
Collapse
Affiliation(s)
- Arthur M Feldman
- The Cardiovascular Institute, University of Pittsburgh Health System, University of Pittsburgh Medical Center, Pennsylvania 15213, USA,.
| | | |
Collapse
|
47
|
Papp Z, Csapó K, Pollesello P, Haikala H, Edes I. Pharmacological Mechanisms Contributing to the Clinical Efficacy of Levosimendan. ACTA ACUST UNITED AC 2006; 23:71-98. [PMID: 15867949 DOI: 10.1111/j.1527-3466.2005.tb00158.x] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Acute decompensation of chronic heart failure is a direct life-threatening situation with short-term mortality approaching 30%. A number of maladaptive changes are amplified within the cardiovascular system during the progression of chronic heart failure that makes the decompensation phase difficult to handle. Levosimendan is a new Ca2+-sensitizer for the treatment of acutely decompensated heart failure that has proved to be effective during the decompensation of chronic heart failure and acute myocardial infarction. Levosimendan differs from other cardiotonic agents that are used for acute heart failure in that it utilizes a unique dual mechanism of action: Ca2+-sensitization through binding to troponin C in the myocardium, and the opening of ATP-sensitive K+ channels in vascular smooth muscle. In general, these mechanisms evoke positive inotropy and vasodilation. Clinical studies suggested long-term benefits on mortality following short-term administration. It may, therefore, be inferred that levosimendan has additional effects on the cardiovascular system that are responsible for the prolongation of survival. Results of preclinical and clinical investigations suggest that the combination of levosimendan-induced cardiac and vascular changes has favorable effects on the coronary, pulmonary and peripheral circulations. Redistribution of the circulating blood offers an improved hemodynamic context for the development of a positive inotropic effect through Ca2+-sensitization of the contractile filaments, without a proportionate increase in myocardial oxygen consumption or the development of arrhythmias. Activation of ATP-sensitive K+ channels, both on sarcolemma and mitochondria, may protect against myocardial ischemia, and decreased levels of cytokines may prevent the development of further myocardial remodeling. Collectively, these effects of levosimendan shift the disturbed cardiovascular parameters towards normalization, thereby halting the perpetuation of the vicious cycle of heart failure progression. This may contribute to stabilization of the circulation and improved life expectancy of patients with chronic heart failure.
Collapse
Affiliation(s)
- Zoltán Papp
- Division of Clinical Physiology, Institute of Cardiology, University of Debrecen, Medical and Health Science Center, Medical School, P.O. BOX 1, H-4004 Debrecen, Hungary.
| | | | | | | | | |
Collapse
|
48
|
A prospective randomized antiplatelet trial of cilostazol versus clopidogrel in patients with bare metal stent. Chin Med J (Engl) 2006. [DOI: 10.1097/00029330-200603010-00002] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
49
|
Szilágyi S, Pollesello P, Levijoki J, Haikala H, Bak I, Tósaki A, Borbély A, Edes I, Papp Z. Two inotropes with different mechanisms of action: contractile, PDE-inhibitory and direct myofibrillar effects of levosimendan and enoximone. J Cardiovasc Pharmacol 2006; 46:369-76. [PMID: 16116344 DOI: 10.1097/01.fjc.0000175454.69116.9] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
We characterized the Ca2+-sensitizing and phosphodiesterase (PDE)-inhibitory potentials of levosimendan and enoximone to assess their contributions to the positive inotropic effects of these drugs. In guinea pig hearts perfused in the working-heart mode, the maximal increase in cardiac output (55%, P<0.05) was attained at 50 nM levosimendan. The corresponding value for enoximone (36%) was significantly smaller (P<0.05) and was observed at a higher concentration (500 nM). In permeabilized myocyte-sized preparations levosimendan evoked a maximal increase of 55.8+/-8% (mean+/-SEM) in isometric force production via Ca2+ sensitization (pCa 6.2, EC50 8.4 nM). Enoximone up to a concentration of 10 microM failed to influence the isometric force. The PDE-inhibitory effects were probed on the PDE III and PDE IV isoforms. Levosimendan proved to be a 1300-fold more potent and a 90-fold more selective PDE III inhibitor (IC50 for PDE III 1.4 nM, and IC50 for PDE IV 11 microM, selectivity factor approximately 8000) than enoximone (IC50 for PDE III 1.8 microM, and IC50 for PDE IV 160 microM, selectivity factor approximately 90). Hence, our data support the hypothesis that levosimendan exerts positive inotropy via a Ca2+-sensitizing mechanism, whereas enoximone does so via PDE inhibition with a limited PDE III versus PDE IV selectivity.
Collapse
Affiliation(s)
- Szabolcs Szilágyi
- Division of Clinical Physiology, Institute of Cardiology, University of Debrecen, Medical and Health Science Center, Faculty of Medicine, Debrecen, Hungary
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Nakamura K, Ikomi F, Ohhashi T. Cilostazol, an Inhibitor of Type 3 Phosphodiesterase, Produces Endothelium-Independent Vasodilation in Pressurized Rabbit Cerebral Penetrating Arterioles. J Vasc Res 2005; 43:86-94. [PMID: 16286783 DOI: 10.1159/000089723] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2005] [Accepted: 08/23/2005] [Indexed: 11/19/2022] Open
Abstract
We investigated the effects of cilostazol, a potent inhibitor of cGMP-inhibited cAMP phosphodiesterase, on mechanical activity of isolated pressurized rabbit cerebral penetrating arterioles with special reference to the function of the endothelium. Both cilostazol and milrinone, another inhibitor of cAMP phosphodiesterase, produced vasodilation of the cerebral penetrating arterioles in a dose-dependent manner. Pretreatment with selective inhibitors of cyclooxygenase or nitric oxide synthase, or chemical denudation of the endothelial cells caused no significant effect on the cilostazol-mediated vasodilation of the cerebral arterioles. A selective large-conductance calcium-activated potassium channel inhibitor, iberiotoxin, and a selective protein kinase A inhibitor, H-89, caused no significant effect on the cilostazol-mediated vasodilation. In the cerebral arterioles, low concentration (10(-6)M) of cilostazol or milrinone caused a significant shift of the dose-vasodilatory response curve for adenosine to the left. These findings suggest that cilostazol produces vasodilation independent of the presence of the endothelium or activation of endogenous vasodilative prostaglandins, nitric oxide, calcium-activated potassium channel and protein kinase A. In conclusion, the vasodilator action of cilostazol may, in part, contribute to the beneficial effect of preventing lacunar cerebral infarction in patients with functional damage of the endothelium in cerebral penetrating arterioles.
Collapse
Affiliation(s)
- Kazuya Nakamura
- Department of Physiology, Shinshu University School of Medicine, Matsumoto, Japan
| | | | | |
Collapse
|