1
|
Cerecedo CD, Silva A, Alia VS, Vargas A, Garza A, El Shazly O, Candil AJ. Cilostazol after endovascular therapy for femoropopliteal peripheral arterial disease: A systematic review and meta-analysis. J Vasc Surg 2024:S0741-5214(24)01786-5. [PMID: 39208919 DOI: 10.1016/j.jvs.2024.08.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 08/03/2024] [Accepted: 08/18/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Endovascular therapy (ET) outcomes for femoropopliteal peripheral arterial disease (FP-PAD) remain suboptimal. Cilostazol therapy may improve patency rates and decrease major adverse limb events after ET for FP-PAD. Our goal was to analyze published studies evaluating the use of cilostazol after ET for FP-PAD. METHODS We searched MEDLINE, EMBASE, and CENTRAL for randomized and observational studies (OSs) evaluating cilostazol therapy after ET for FP-PAD. We only included OSs adjusting for confounding variables. We analyzed observational and randomized studies separately and explored heterogeneity by estimating an I2 statistic. A fixed-effects model was chosen if the I2 statistic was low. If the two-sided probability of observing the difference between groups under a true null hypothesis was <5%, we considered this difference statistically significant. RESULTS We screened 2171 studies and included 26 papers in our analysis (5 randomized controlled trials and 21 OSs). All randomized studies were open label. In randomized studies, the odds of restenosis were lower in patients treated with cilostazol (pooled odds ratio, 0.28; 95% confidence interval [CI], 0.18-0.43; P < .01; I2 = 0%). The odds of target lesion revascularization (TLR) were also lower in patients treated with cilostazol (pooled odds ratio, 0.35; 95% CI, 0.22-0.65; P < .01; I2 = 0%). In OSs, we also identified associations between peri-interventional treatment with cilostazol and lower rates of restenosis (pooled hazard ratio [pHR], 0.57; 95% CI, 0.51-0.65; P < .01; I2 = 34%), TLR (pHR, 0.53; 95% CI, 0.36-0.79; P < .01; I2 = 0%), and amputation (pHR, 0.54; 95% CI, 0.32-0.90; P = .02; I2 = 30%). CONCLUSIONS In randomized open-label studies, peri-interventional treatment with cilostazol after ET for FP-PAD decreased the odds of restenosis and TLR (Level 1A). Similarly, in OSs that adjusted for confounding, peri-interventional cilostazol therapy was associated with lower rates of restenosis, TLR, and amputation (Level 2A).
Collapse
Affiliation(s)
- Christian D Cerecedo
- Department of Surgery, University of Texas Rio Grande Valley School of Medicine, Harlingen, TX.
| | - Aaron Silva
- Department of Surgery, University of Texas Rio Grande Valley School of Medicine, Harlingen, TX
| | - Valentine S Alia
- Department of Surgery, University of Texas Rio Grande Valley School of Medicine, Harlingen, TX
| | - Ana Vargas
- Department of Surgery, University of Texas Rio Grande Valley School of Medicine, Harlingen, TX
| | - Alejandro Garza
- Department of Surgery, Doctor's Hospital at Renaissance, Edinburg, TX
| | - Omar El Shazly
- Department of Surgery, Medical College of Georgia at Augusta University, Augusta, GA
| | - Alejandro J Candil
- Department of Surgery, University of Texas Rio Grande Valley School of Medicine, Harlingen, TX
| |
Collapse
|
2
|
Hoelm M, Chowdhury N, Biswas S, Bagchi A, Małecka M. Theoretical Investigations on Free Energy of Binding Cilostazol with Different Cyclodextrins as Complex for Selective PDE3 Inhibition. Molecules 2024; 29:3824. [PMID: 39202903 PMCID: PMC11357564 DOI: 10.3390/molecules29163824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 08/08/2024] [Accepted: 08/09/2024] [Indexed: 09/03/2024] Open
Abstract
Cilostazol is a phosphodiesterase III inhibitor characterized by poor solubility. This limitation can be overcome by using a drug carrier capable of delivering the drug to the target site. Cyclodextrins are essential as drug carriers because of their outstanding complexation abilities and their capacity to improve drug bioavailability. This study comprises two stages: The first involves verifying different cyclodextrins and their complexation abilities towards cilostazol. This was accomplished using molecular docking simulations (MDS) and density functional theory (DFT). Both techniques indicate that the largest Sulfobutyl Ether-β-Cyclodextrin forms the most stable complex with cilostazol. Additionally, other important parameters of the complex are described, including binding sites, dominant interactions, and thermodynamic parameters such as complexation enthalpy, Gibbs free energy, and Gibbs free energy of solvation. The second stage involves a binding study between cilostazol and Phosphodiesterse3 (PDE3). This study was conducted using molecular docking simulations, and the most important energetic parameters are detailed. This is the first such report, and we believe that the results of our predictions will pave the way for future drug development efforts using cyclodextrin-cilostazol complexes as potential therapeutics.
Collapse
Affiliation(s)
- Marta Hoelm
- Department of Physical Chemistry, Faculty of Chemistry, University of Lodz, Pomorska 163/165, 90-236 Lodz, Poland;
| | - Nilkanta Chowdhury
- Department of Biotechnology, Ranchi—Purulia Road Campus, Sidho-Kanho-Birsha University, Purulia 723104, West Bengal, India;
| | - Sima Biswas
- Department of Biochemistry and Biophysics, University of Kalyani, Kalyani 741235, West Bengal, India;
| | - Angshuman Bagchi
- Department of Biochemistry and Biophysics, University of Kalyani, Kalyani 741235, West Bengal, India;
| | - Magdalena Małecka
- Department of Physical Chemistry, Faculty of Chemistry, University of Lodz, Pomorska 163/165, 90-236 Lodz, Poland;
| |
Collapse
|
3
|
Sohn M, Lim S. The Role of Cilostazol, a Phosphodiesterase-3 Inhibitor, in the Development of Atherosclerosis and Vascular Biology: A Review with Meta-Analysis. Int J Mol Sci 2024; 25:2593. [PMID: 38473840 DOI: 10.3390/ijms25052593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 02/07/2024] [Accepted: 02/14/2024] [Indexed: 03/14/2024] Open
Abstract
Atherosclerotic cardiovascular disease (ASCVD) stands as the leading global cause of mortality. Addressing this vital and pervasive condition requires a multifaceted approach, in which antiplatelet intervention plays a pivotal role, together with antihypertensive, antidiabetic, and lipid-lowering therapies. Among the antiplatelet agents available currently, cilostazol, a phosphodiesterase-3 inhibitor, offers a spectrum of pharmacological effects. These encompass vasodilation, the impediment of platelet activation and aggregation, thrombosis inhibition, limb blood flow augmentation, lipid profile enhancement through triglyceride reduction and high-density lipoprotein cholesterol elevation, and the suppression of vascular smooth muscle cell proliferation. However, the role of cilostazol has not been clearly documented in many guidelines for ASCVD. We comprehensively reviewed the cardiovascular effects of cilostazol within randomized clinical trials that compared it to control or active agents and involved individuals with previous coronary artery disease or stroke, as well as those with no previous history of such conditions. Our approach demonstrated that the administration of cilostazol effectively reduced adverse cardiovascular events, although there was less evidence regarding its impact on myocardial infarction. Most studies have consistently reported its favorable effects in reducing intermittent claudication and enhancing ambulatory capacity in patients with peripheral arterial disease. Furthermore, cilostazol has shown promise in mitigating restenosis following coronary stent implantation in patients with acute coronary syndrome. While research from more diverse regions is still needed, our findings shed light on the broader implications of cilostazol in the context of atherosclerosis and vascular biology, particularly for individuals at high risk of ASCVD.
Collapse
Affiliation(s)
- Minji Sohn
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam 13620, Republic of Korea
| | - Soo Lim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam 13620, Republic of Korea
| |
Collapse
|
4
|
Brazão SC, Lima GF, Autran LJ, Mendes ABA, Dos Santos BA, Magliano DC, de Brito FCF, Motta NAV. Subacute administration of cilostazol modulates PLC-γ/PKC-α/p38/NF-kB pathway and plays vascular protective effects through eNOS activation in early stages of atherosclerosis development. Life Sci 2023; 332:122082. [PMID: 37722587 DOI: 10.1016/j.lfs.2023.122082] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 09/08/2023] [Accepted: 09/09/2023] [Indexed: 09/20/2023]
Abstract
AIMS Hypercholesterolemia is an important risk factor for development of cardiovascular disturbances, such as atherosclerosis, and its treatment remains challenging in modern medicine. Cilostazol is a selective inhibitor of phosphodiesterase 3 clinically prescribed for intermittent claudication treatment. Due to its pleiotropic properties, such as lipid lowering, anti-inflammatory, and antioxidant effects, the therapeutic repurposing of cilostazol has become a strategic approach for atherosclerosis treatment. This study aimed to investigate the effects of subacute administration of cilostazol on the aortas of hypercholesterolemic rats, focusing on the signaling pathways involved in these actions. MAIN METHODS A murine model of hypercholesterolemia was employed to mimic the early stages of atherosclerosis development. Vascular reactivity assays were performed on thoracic aorta rings to assess the vascular response, as well as the non-invasive blood pressure was evaluated by plethysmography method. Pro-inflammatory markers and malondialdehyde (MDA) levels were measured to investigate the anti-inflammatory and antioxidant effects of cilostazol. Western Blot analysis was performed in aortas homogenates to evaluate the role of cilostazol on PLC-γ/PKC-α/p38-MAPK/IκB-α/NF-кB and PKA/eNOS/PKG pathways. KEY FINDINGS The hypercholesterolemic diet induced the production of pro-inflammatory mediators such as TNF-α, TXB2, VCAM, and worsened vascular function, marked by increased contractile response, decreased maximum relaxation, and elevated systolic and diastolic blood pressure. Cilostazol seems to counteract the deleterious effects promoted by hypercholesterolemic diet, showing important anti-inflammatory and vasculoprotective properties possibly through the inhibition of the PLC-γ/PKC-α/p38-MAPK/IκB-α/NF-кB pathway and activation of the PKA/eNOS/PKG pathway. SIGNIFICANCE Cilostazol suppressed hypercholesterolemia-induced vascular dysfunction and inflammation. Our data suggest the potential repurposing of cilostazol as a pharmacological treatment for atherosclerosis.
Collapse
Affiliation(s)
- Stephani Correia Brazão
- Laboratory of Experimental Pharmacology (LAFE), Department of Physiology and Pharmacology, Biomedical Institute, Fluminense Federal University (UFF), Room 310, Valonguinho, 24020-150 Niterói, RJ, Brazil
| | - Gabriel Ferreira Lima
- Laboratory of Experimental Pharmacology (LAFE), Department of Physiology and Pharmacology, Biomedical Institute, Fluminense Federal University (UFF), Room 310, Valonguinho, 24020-150 Niterói, RJ, Brazil
| | - Lis Jappour Autran
- Laboratory of Experimental Pharmacology (LAFE), Department of Physiology and Pharmacology, Biomedical Institute, Fluminense Federal University (UFF), Room 310, Valonguinho, 24020-150 Niterói, RJ, Brazil
| | - Ana Beatriz Araújo Mendes
- Laboratory of Experimental Pharmacology (LAFE), Department of Physiology and Pharmacology, Biomedical Institute, Fluminense Federal University (UFF), Room 310, Valonguinho, 24020-150 Niterói, RJ, Brazil
| | - Beatriz Alexandre Dos Santos
- Laboratory of Morphological and Metabolic Analyses, Department of Morphology Biomedical Institute, Fluminense Federal University (UFF), Brazil
| | - Dangelo Carlo Magliano
- Laboratory of Morphological and Metabolic Analyses, Department of Morphology Biomedical Institute, Fluminense Federal University (UFF), Brazil
| | - Fernanda Carla Ferreira de Brito
- Laboratory of Experimental Pharmacology (LAFE), Department of Physiology and Pharmacology, Biomedical Institute, Fluminense Federal University (UFF), Room 310, Valonguinho, 24020-150 Niterói, RJ, Brazil.
| | - Nadia Alice Vieira Motta
- Laboratory of Experimental Pharmacology (LAFE), Department of Physiology and Pharmacology, Biomedical Institute, Fluminense Federal University (UFF), Room 310, Valonguinho, 24020-150 Niterói, RJ, Brazil
| |
Collapse
|
5
|
Yu B, Li B, Chen T, Yang J, Wang X, Peng B, Hu Q. A NF-κB-Based High-Throughput Screening for Immune Adjuvants and Inhibitors. Inflammation 2023; 46:598-611. [PMID: 36306023 DOI: 10.1007/s10753-022-01758-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 09/19/2022] [Accepted: 10/12/2022] [Indexed: 11/05/2022]
Abstract
The nuclear factor-κB (NF-κB) family is crucial for regulating immune and inflammatory responses. The activation of the immune cell signaling pathway usually activates NF-κB, causing a protective immune response. NF-κB can also cause excessive inflammatory responses by activating a cascade reaction of pro-inflammatory mediators such as cytokines. In this study, we used an NF-κB luciferase reporter gene system. Out of more than 800 compounds screened, four NF-κB agonists were identified with strong activity at nontoxic concentrations. Subsequently, the adjuvant effect was verified on mouse bone marrow-derived dendritic cells (BMDCs) and macrophages RAW264.7. It was found that fostamatinib (R788) disodium increased the production of IL-6, IL-12p40, and TNF-α, indicating that R788 disodium could induce the maturation of antigen-presenting cells (APCs). In addition, three compounds were screened to significantly inhibit NF-κB at nontoxic doses, including dehydrocostus lactone (DHL)-a known NF-κB inhibitor. The results showed that DHL significantly reduced the release of LPS-induced inflammatory cytokines (including TNF-α, IL-6, and IL-12). Our findings indicate that the NF-κB-based high-throughput screening can be used to discover potential immune adjuvants and anti-inflammatory molecules.
Collapse
Affiliation(s)
- Boyang Yu
- The Faculty of Environment and Life, Beijing University of Technology, Beijing, 100124, China
- Beijing International Science and Technology Cooperation Base of Antivirus Drug, Beijing University of Technology, Beijing, 100124, China
| | - Boye Li
- Civil Aviation Medicine Center, Civil Aviation Administration of China, Beijing, 100123, China
| | - Tian Chen
- The Faculty of Environment and Life, Beijing University of Technology, Beijing, 100124, China
- Beijing International Science and Technology Cooperation Base of Antivirus Drug, Beijing University of Technology, Beijing, 100124, China
| | - Jinning Yang
- The Faculty of Environment and Life, Beijing University of Technology, Beijing, 100124, China
- Beijing International Science and Technology Cooperation Base of Antivirus Drug, Beijing University of Technology, Beijing, 100124, China
| | - Xiaoli Wang
- The Faculty of Environment and Life, Beijing University of Technology, Beijing, 100124, China
- Beijing International Science and Technology Cooperation Base of Antivirus Drug, Beijing University of Technology, Beijing, 100124, China
| | - Bo Peng
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Qin Hu
- The Faculty of Environment and Life, Beijing University of Technology, Beijing, 100124, China.
- Beijing International Science and Technology Cooperation Base of Antivirus Drug, Beijing University of Technology, Beijing, 100124, China.
| |
Collapse
|
6
|
Hermes TDA, Mâncio RD, Mizobutti DS, Macedo AB, Kido LA, Cagnon Quitete VHA, Minatel E. Cilostazol attenuates oxidative stress and apoptosis in the quadriceps muscle of the dystrophic mouse experimental model. Int J Exp Pathol 2023; 104:13-22. [PMID: 36565167 PMCID: PMC9845609 DOI: 10.1111/iep.12461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 10/04/2022] [Accepted: 10/18/2022] [Indexed: 12/25/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is the most severe and frequent form of muscular dystrophy. The mdx mouse is one of the most widely used experimental models to understand aspects of the biology of dystrophic skeletal muscles and the mechanisms of DMD. Oxidative stress and apoptosis are present in early stages of the disease in mdx mice. The high production of reactive oxygen species (ROS) causes activation of apoptotic death regulatory proteins due to DNA damage and breakdown of nuclear and mitochondrial membranes. The quadriceps (QUA) muscle of the mdx mouse is a good tool to study oxidative events. Previous studies have demonstrated that cilostazol exerts an anti-oxidant effect by decreasing the production of reactive oxygen species (ROS). The present study aimed to evaluate the ability of cilostazol to modulate oxidative stress and apoptosis in the QUA muscle of mdx mice. Fourteen-day-old mdx mice received cilostazol or saline for 14 days. C57BL/10 mice were used as a control. In the QUA muscle of mdx mice, cilostazol treatment decreased ROS production (-74%), the number of lipofuscin granules (-47%), lipid peroxidation (-11%), and the number of apoptotic cells (-66%). Thus cilostazol showed anti-oxidant and anti-apoptotic action in the QUA muscle of mdx mice.
Collapse
Affiliation(s)
- Túlio de Almeida Hermes
- Department of Structural and Functional Biology, Institute of BiologyState University of Campinas (UNICAMP)São PauloBrazil
- Departament of Anatomy, Institute of Biomedical SciencesFederal University of Alfenas (UNIFAL‐MG)AlfenasBrazil
| | - Rafael Dias Mâncio
- Department of Structural and Functional Biology, Institute of BiologyState University of Campinas (UNICAMP)São PauloBrazil
| | - Daniela Sayuri Mizobutti
- Department of Structural and Functional Biology, Institute of BiologyState University of Campinas (UNICAMP)São PauloBrazil
| | - Aline Barbosa Macedo
- Department of Structural and Functional Biology, Institute of BiologyState University of Campinas (UNICAMP)São PauloBrazil
| | - Larissa Akemi Kido
- Department of Structural and Functional Biology, Institute of BiologyState University of Campinas (UNICAMP)São PauloBrazil
| | | | - Elaine Minatel
- Department of Structural and Functional Biology, Institute of BiologyState University of Campinas (UNICAMP)São PauloBrazil
| |
Collapse
|
7
|
Yanai S, Tago T, Toyohara J, Arasaki T, Endo S. Reversal of spatial memory impairment by phosphodiesterase 3 inhibitor cilostazol is associated with reduced neuroinflammation and increased cerebral glucose uptake in aged male mice. Front Pharmacol 2022; 13:1031637. [PMID: 36618932 PMCID: PMC9810637 DOI: 10.3389/fphar.2022.1031637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 11/30/2022] [Indexed: 12/24/2022] Open
Abstract
The nucleotide second messenger 3', 5'-cyclic adenosine monophosphate (cAMP) and 3', 5'-cyclic guanosine monophosphate (cGMP) mediate fundamental functions of the brain, including learning and memory. Phosphodiesterase 3 (PDE3) can hydrolyze both cAMP and cGMP and appears to be involved in the regulation of their contents in cells. We previously demonstrated that long-term administration of cilostazol, a PDE3 inhibitor, maintained good memory performance in aging mice. Here, we report on studies aimed at determining whether cilostazol also reverses already-impaired memory in aged male mice. One month of oral 1.5% cilostazol administration in 22-month-old mice reversed age-related declines in hippocampus-dependent memory tasks, including the object recognition and the Morris water maze. Furthermore, cilostazol reduced neuroinflammation, as evidenced by immunohistochemical staining, and increased glucose uptake in the brain, as evidence by positron emission tomography (PET) with 2-deoxy-2-[18F]fluoro-d-glucose ([18F]FDG). These results suggest that already-expressed memory impairment in aged male mice that depend on cyclic nucleotide signaling can be reversed by inhibition of PDE3. The reversal of age-related memory impairments may occur in the central nervous system, either through cilostazol-enhanced recall or strengthening of weak memories that otherwise may be resistant to recall.
Collapse
Affiliation(s)
- Shuichi Yanai
- Aging Neuroscience Research Team, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Tetsuro Tago
- Research Team for Neuroimaging, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Jun Toyohara
- Research Team for Neuroimaging, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Tomoko Arasaki
- Aging Neuroscience Research Team, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Shogo Endo
- Aging Neuroscience Research Team, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan,*Correspondence: Shogo Endo,
| |
Collapse
|
8
|
Cilostazol Alleviates NLRP3 Inflammasome-Induced Allodynia/Hyperalgesia in Murine Cerebral Cortex Following Transient Ischemia: Focus on TRPA1/Glutamate and Akt/Dopamine/BDNF/Nrf2 Trajectories. Mol Neurobiol 2022; 59:7194-7211. [PMID: 36127628 PMCID: PMC9616778 DOI: 10.1007/s12035-022-03024-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Accepted: 08/29/2022] [Indexed: 12/02/2022]
Abstract
Global cerebral ischemia/reperfusion (I/R) provokes inflammation that augments neuropathic pain. Cilostazol (CLZ) has pleiotropic effects including neuroprotection in several ravaging central disorders; nonetheless, its potential role in transient central ischemic-induced allodynia and hyperalgesia has not been asserted before. Rats were allocated into 4 groups; sham, sham + CLZ, and 45 min-bilateral carotid occlusion followed by a 48 h-reperfusion period either with or without CLZ (50 mg/kg; p.o) post-treatment. CLZ prolonged latency of hindlimb withdrawal following von Frey filaments, 4 °C cold, and noxious mechanical stimulations. Histopathological alterations and the immunoexpression of glial fibrillary acidic protein induced by I/R were reduced by CLZ in the anterior cingulate cortex (ACC) area, while, CLZ enhanced intact neuronal count. Meanwhile, CLZ modulated cerebral cortical glutamate, dopamine neurotransmission, and transient receptor potential ankyrin 1 (TRPA1). CLZ anti-inflammatory potential was mediated by the downregulated p65 NF-κB and sirtuin-1 enhancement to reduce nucleotide-binding domain-like receptor protein 3 (NLRP3), apoptosis-associated speck-like protein (ASC), active caspase-1, and interleukin-1β, indicative of inflammasome deactivation. It also revealed an antioxidant capacity via boosting nuclear factor E2-related factor (Nrf2) enhancing glutathione through forkhead box protein O3a (FOXO3a) reduction. Additionally, CLZ triggered neuronal survival by promoting the p-content of Akt, TrkB, and CREB as well as BDNF content. A novel approach of CLZ in hindering global cerebral I/R-mediated neuropathy is firstly documented herein to forward its adjunct action via deactivating the NLRP3 inflammasome, besides enhancing Nrf2 axis, neuronal survival, and dopamine neurotransmission as well as inhibiting TRPA1 and excitotoxicity.
Collapse
|
9
|
Albrahim T. Lycopene Modulates Oxidative Stress and Inflammation in Hypercholesterolemic Rats. Pharmaceuticals (Basel) 2022; 15:1420. [PMID: 36422550 PMCID: PMC9693203 DOI: 10.3390/ph15111420] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 11/14/2022] [Accepted: 11/15/2022] [Indexed: 07/30/2023] Open
Abstract
The complicated disorder of hypercholesterolemia has several underlying factors, including genetic and lifestyle factors. Low LDL cholesterol and elevated serum total cholesterol are its defining features. A carotenoid with antioxidant quality is lycopene. Examining lycopene activity in an animal model of hypercholesterolemia induced using food was the aim of this investigation. Triglycerides, LDL cholesterol, HDL cholesterol, and plasma total cholesterol were all measured. Biomarkers of renal and cardiac function were also examined. Apoptotic indicators, pro-inflammatory markers, and oxidative stress were also assessed. Additionally, the mRNA expression of paraoxonase 1 (PON-1), peroxisome proliferator-activated receptor gamma (PPAR-γ), and PPAR-γ coactivator 1 alpha (PGC-1α) in cardiac and renal tissues was examined. Rats showed elevated serum lipid levels, renal and cardiac dysfunction, significant oxidative stress, and pro-inflammatory and apoptotic markers at the end of the study. Treatment with lycopene significantly corrected and restored these changes. Additionally, lycopene markedly increased the mRNA expression of PGC-1α and PON-1, and decreased PPAR-γ expression. It was determined that lycopene has the capacity to modulate the PPAR-γ and PON-1 signaling pathway in order to preserve the cellular energy metabolism of the heart and kidney, which in turn reduces tissue inflammatory response and apoptosis. According to these findings, lycopene may be utilized as a medication to treat hypercholesterolemia. However, further studies should be conducted first to determine the appropriate dose and any adverse effects that may appear after lycopene usage in humans.
Collapse
Affiliation(s)
- Tarfa Albrahim
- Department of Health Sciences, Clinical Nutrition, College of Health and Rehabilitation Sciences, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| |
Collapse
|
10
|
Hada Y, Uchida HA, Umebayashi R, Yoshida M, Wada J. Cilostazol Attenuates AngII-Induced Cardiac Fibrosis in apoE Deficient Mice. Int J Mol Sci 2022; 23:ijms23169065. [PMID: 36012328 PMCID: PMC9408896 DOI: 10.3390/ijms23169065] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/08/2022] [Accepted: 08/11/2022] [Indexed: 11/16/2022] Open
Abstract
Cardiac fibrosis is characterized by the net accumulation of extracellular matrix in the myocardium and is an integral component of most pathological cardiac conditions. Cilostazol, a selective inhibitor of phosphodiesterase type III with anti-platelet, anti-mitogenic, and vasodilating properties, is widely used to treat the ischemic symptoms of peripheral vascular disease. Here, we investigated whether cilostazol has a protective effect against Angiotensin II (AngII)-induced cardiac fibrosis. Male apolipoprotein E-deficient mice were fed either a normal diet or a diet containing cilostazol (0.1% wt/wt). After 1 week of diet consumption, the mice were infused with saline or AngII (1000 ng kg−1 min−1) for 28 days. AngII infusion increased heart/body weight ratio (p < 0.05), perivascular fibrosis (p < 0.05), and interstitial cardiac fibrosis (p < 0.0001), but were significantly attenuated by cilostazol treatment (p < 0.05, respectively). Cilostazol also reduced AngII-induced increases in fibrotic and inflammatory gene expression (p < 0.05, respectively). Furthermore, cilostazol attenuated both protein and mRNA abundance of osteopontin induced by AngII in vivo. In cultured human cardiac myocytes, cilostazol reduced mRNA expression of AngII-induced osteopontin in dose-dependent manner. This reduction was mimicked by forskolin treatment but was cancelled by co-treatment of H-89. Cilostazol attenuates AngII-induced cardiac fibrosis in mice through activation of the cAMP−PKA pathway.
Collapse
Affiliation(s)
- Yoshiko Hada
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Haruhito A. Uchida
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
- Department of Chronic Kidney Disease and Cardiovascular Disease, Okayama University Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
- Correspondence: ; Tel.: +81-86-235-7235
| | - Ryoko Umebayashi
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Masashi Yoshida
- Department of Chronic Kidney Disease and Cardiovascular Disease, Okayama University Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
- Department of Cardiovascular Medicine, Okayama University Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Jun Wada
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| |
Collapse
|
11
|
de Oliveira Lopes R, Lima GF, Mendes ABA, Autran LJ, de Assis Pereira NC, Brazão SC, Alexandre-Santos B, Frantz EDC, Scaramello CBV, Brito FCF, Motta NAV. Cilostazol attenuates cardiac oxidative stress and inflammation in hypercholesterolemic rats. Naunyn Schmiedebergs Arch Pharmacol 2022; 395:789-801. [PMID: 35384464 DOI: 10.1007/s00210-022-02233-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Accepted: 03/16/2022] [Indexed: 10/18/2022]
Abstract
Atherosclerosis is a multifactorial chronic disease associated with pro-inflammatory and pro-oxidative cardiovascular states. Cilostazol, a selective phosphodiesterase 3 inhibitor (PDE3), is clinically used in the treatment of intermittent claudication and secondary prevention of cerebral infarction. The aim of this study was to evaluate the cardioprotective effects of cilostazol and the molecular mechanisms involved in hypercholesterolemic rats. Male Wistar rats were divided into four groups: control group (C) and control + cilostazol group (C+CILO), that were fed a standard chow diet, and hypercholesterolemic diet group (HCD) and HCD + cilostazol (HCD+CILO) that were fed a hypercholesterolemic diet. Cilostazol treatment started after 30 days for C+CILO and HCD+CILO groups. Animals were administered cilostazol once a day for 15 days. Subsequently, serum and left ventricles were extracted for evaluation of lipid profile, inflammatory, and oxidative biomarkers. The HCD group displayed increased serum lipid levels, inflammatory cytokines production, and cardiac NF-kB protein expression and decreased cardiac Nrf2-mediated antioxidant activity. Conversely, the cilostazol treatment improved all these cardiac deleterious effects, inhibiting NF-kB activation and subsequently decreasing inflammatory mediators, reestablishing the antioxidant properties through Nrf2-mediated pathway, including increased SOD, GPx, and catalase expression. Taken together, our results indicated that cilostazol protects hypercholesterolemia-induced cardiac damage by molecular mechanisms targeting the crosstalk between Nrf2 induction and NF-kB inhibition in the heart.
Collapse
Affiliation(s)
- Rosane de Oliveira Lopes
- Laboratory of Experimental Pharmacology (LAFE), Department of Physiology and Pharmacology, Biomedical Institute, Fluminense Federal University (UFF), Room 204-A, Niteroi, RJ, 24210-130, Brazil
| | - Gabriel Ferreira Lima
- Laboratory of Experimental Pharmacology (LAFE), Department of Physiology and Pharmacology, Biomedical Institute, Fluminense Federal University (UFF), Room 204-A, Niteroi, RJ, 24210-130, Brazil
| | - Ana Beatriz Araújo Mendes
- Laboratory of Experimental Pharmacology (LAFE), Department of Physiology and Pharmacology, Biomedical Institute, Fluminense Federal University (UFF), Room 204-A, Niteroi, RJ, 24210-130, Brazil.,Laboratory of Endocrine Physiology Doris Rosenthal, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brazil
| | - Lis Jappour Autran
- Laboratory of Experimental Pharmacology (LAFE), Department of Physiology and Pharmacology, Biomedical Institute, Fluminense Federal University (UFF), Room 204-A, Niteroi, RJ, 24210-130, Brazil
| | - Nikolas Cunha de Assis Pereira
- Laboratory of Experimental Pharmacology (LAFE), Department of Physiology and Pharmacology, Biomedical Institute, Fluminense Federal University (UFF), Room 204-A, Niteroi, RJ, 24210-130, Brazil
| | - Stephani Correia Brazão
- Laboratory of Experimental Pharmacology (LAFE), Department of Physiology and Pharmacology, Biomedical Institute, Fluminense Federal University (UFF), Room 204-A, Niteroi, RJ, 24210-130, Brazil
| | - Beatriz Alexandre-Santos
- Laboratory of Exercise Sciences (LACE), Department of Morphology, Fluminense Federal University (UFF), Niteroi, RJ, Brazil
| | - Eliete Dalla Corte Frantz
- Laboratory of Exercise Sciences (LACE), Department of Morphology, Fluminense Federal University (UFF), Niteroi, RJ, Brazil
| | - Christianne Brêtas Vieira Scaramello
- Laboratory of Experimental Pharmacology (LAFE), Department of Physiology and Pharmacology, Biomedical Institute, Fluminense Federal University (UFF), Room 204-A, Niteroi, RJ, 24210-130, Brazil
| | - Fernanda Carla Ferreira Brito
- Laboratory of Experimental Pharmacology (LAFE), Department of Physiology and Pharmacology, Biomedical Institute, Fluminense Federal University (UFF), Room 204-A, Niteroi, RJ, 24210-130, Brazil.
| | - Nadia Alice Vieira Motta
- Laboratory of Experimental Pharmacology (LAFE), Department of Physiology and Pharmacology, Biomedical Institute, Fluminense Federal University (UFF), Room 204-A, Niteroi, RJ, 24210-130, Brazil
| |
Collapse
|
12
|
Abdallah MS, Ramadan AN, Omara‐Reda H, Mansour NO, Elsokary MA, Elsawah HK, Zaki SA, Abo Mansour HE, Mosalam EM. Double-blind, randomized, placebo-controlled pilot study of the phosphodiesterase-3 inhibitor cilostazol as an adjunctive to antidepressants in patients with major depressive disorder. CNS Neurosci Ther 2021; 27:1540-1548. [PMID: 34545997 PMCID: PMC8611782 DOI: 10.1111/cns.13731] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 09/02/2021] [Accepted: 09/10/2021] [Indexed: 12/28/2022] Open
Abstract
AIMS Cilostazol (CLS) has shown antidepressant effect in cardiovascular patients, post-stroke depression, and animal models through its neurotrophic and antiinflammatory activities. Consequently, we aimed to investigate its safety and efficacy in patients with MDD by conducting double-blind, randomized, placebo-controlled pilot study. METHODS 80 participants with MDD (DSM-IV criteria) and Hamilton Depression Rating Scale (HDRS) score >20 were treated with CLS 50 mg or placebo twice daily plus escitalopram (ESC) 20 mg once daily for six weeks. Patients were evaluated by HDRS scores (weeks 0, 2, 4, and 6). Serum levels of CREB1, BDNF, 5-HT, TNF-α, NF- κB, and FAM19A5 were assessed pre- and post-treatment. RESULTS Co-administration of CLS had markedly decreased HDRS score at all-time points compared to the placebo group (p < 0.001). Early improvement, response, and remission rates after 6 weeks were significantly higher in the CLS group (90%, 90%, 80%, respectively) than in the placebo group (25%, 65%, 50% respectively) (p < 0.001). Moreover, the CLS group was superior to the placebo group in modulation of the measured neurotrophic and inflammatory biomarkers. CONCLUSION CLS is safe and effective short-term adjunctive therapy in patients with MDD with no other comorbid conditions. Trial registration ID:NCT04069819.
Collapse
Affiliation(s)
- Mahmoud S. Abdallah
- Department of Clinical PharmacyFaculty of PharmacyUniversity of Sadat CitySadat CityEgypt
| | - Ahmed N. Ramadan
- Department of NeuropsychiatryFaculty of MedicineMenoufia UniversityShebin El‐KomEgypt
| | - Hend Omara‐Reda
- Department of NeuropsychiatryFaculty of MedicineMenoufia UniversityShebin El‐KomEgypt
| | - Noha O. Mansour
- Clinical Pharmacy and Pharmacy Practice DepartmentFaculty of PharmacyMansoura UniversityMansouraEgypt
| | - Mohamed A. Elsokary
- Department of BiostatisticsHigh Institute of Public HealthAlexandria UniversityAlexandriaEgypt
| | - Hozaifa K. Elsawah
- Department of BiostatisticsHigh Institute of Public HealthAlexandria UniversityAlexandriaEgypt
| | - Shimaa Abdelsattar Zaki
- Department of Clinical Biochemistry and Molecular DiagnosticsNational Liver InstituteMenoufia UniversityShebin El‐KomEgypt
| | - Hend E. Abo Mansour
- Department of BiochemistryFaculty of PharmacyMenoufia UniversityShebin El‐KomEgypt
| | - Esraa M. Mosalam
- Department of BiochemistryFaculty of PharmacyMenoufia UniversityShebin El‐KomEgypt
| |
Collapse
|
13
|
de Havenon A, Sheth KN, Madsen TE, Johnston KC, Turan T, Toyoda K, Elm JJ, Wardlaw JM, Johnston SC, Williams OA, Shoamanesh A, Lansberg MG. Cilostazol for Secondary Stroke Prevention: History, Evidence, Limitations, and Possibilities. Stroke 2021; 52:e635-e645. [PMID: 34517768 PMCID: PMC8478840 DOI: 10.1161/strokeaha.121.035002] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cilostazol is a PDE3 (phosphodiesterase III) inhibitor with a long track record of safety that is Food and Drug Administration and European Medicines Agency approved for the treatment of claudication in patients with peripheral arterial disease. In addition, cilostazol has been approved for secondary stroke prevention in several Asian countries based on trials that have demonstrated a reduction in stroke recurrence among patients with noncardioembolic stroke. The onset of benefit appears after 60 to 90 days of treatment, which is consistent with cilostazol's pleiotropic effects on platelet aggregation, vascular remodeling, blood flow, and plasma lipids. Cilostazol appears safe and does not increase the risk of major bleeding when given alone or in combination with aspirin or clopidogrel. Adverse effects such as headache, gastrointestinal symptoms, and palpitations, however, contributed to a 6% increase in drug discontinuation among patients randomized to cilostazol in a large secondary stroke prevention trial (CSPS.com [Cilostazol Stroke Prevention Study for Antiplatelet Combination]). Due to limitations of prior trials, such as open-label design, premature trial termination, large loss to follow-up, lack of functional or cognitive outcome data, and exclusive enrollment in Asia, the existing trials have not led to a change in clinical practice or guidelines in Western countries. These limitations could be addressed by a double-blind placebo-controlled randomized trial conducted in a broader population. If positive, it would increase the evidence in support of long-term treatment with cilostazol for secondary prevention in the millions of patients worldwide who have experienced a noncardioembolic ischemic stroke.
Collapse
Affiliation(s)
- Adam de Havenon
- Department of Neurology, University of Utah (A.D.); Department of Neurology, Yale University (K.N.S.); Department of Emergency Medicine, Brown University (T.M.); Department of Neurology, University of Virginia (K.J.); Department of Neurology, Medical University of South Carolina (T.T., J.E.); Department of Cerebrovascular Medicine, National Cerebral and Cardiovascular Center, Japan (K.T.); Center for Clinical Brain Sciences, UK Dementia Research Institute, University of Edinburgh (J.M.W.); Dell Medical School (S.C.J.); Department of Neurology, Columbia University (O.W.); Department of Medicine (Neurology), McMaster University/Population Heath Research Institute (A.S.); Department of Neurology, Stanford University (M.L.)
| | - Kevin N. Sheth
- Department of Neurology, University of Utah (A.D.); Department of Neurology, Yale University (K.N.S.); Department of Emergency Medicine, Brown University (T.M.); Department of Neurology, University of Virginia (K.J.); Department of Neurology, Medical University of South Carolina (T.T., J.E.); Department of Cerebrovascular Medicine, National Cerebral and Cardiovascular Center, Japan (K.T.); Center for Clinical Brain Sciences, UK Dementia Research Institute, University of Edinburgh (J.M.W.); Dell Medical School (S.C.J.); Department of Neurology, Columbia University (O.W.); Department of Medicine (Neurology), McMaster University/Population Heath Research Institute (A.S.); Department of Neurology, Stanford University (M.L.)
| | - Tracy E. Madsen
- Department of Neurology, University of Utah (A.D.); Department of Neurology, Yale University (K.N.S.); Department of Emergency Medicine, Brown University (T.M.); Department of Neurology, University of Virginia (K.J.); Department of Neurology, Medical University of South Carolina (T.T., J.E.); Department of Cerebrovascular Medicine, National Cerebral and Cardiovascular Center, Japan (K.T.); Center for Clinical Brain Sciences, UK Dementia Research Institute, University of Edinburgh (J.M.W.); Dell Medical School (S.C.J.); Department of Neurology, Columbia University (O.W.); Department of Medicine (Neurology), McMaster University/Population Heath Research Institute (A.S.); Department of Neurology, Stanford University (M.L.)
| | - Karen C. Johnston
- Department of Neurology, University of Utah (A.D.); Department of Neurology, Yale University (K.N.S.); Department of Emergency Medicine, Brown University (T.M.); Department of Neurology, University of Virginia (K.J.); Department of Neurology, Medical University of South Carolina (T.T., J.E.); Department of Cerebrovascular Medicine, National Cerebral and Cardiovascular Center, Japan (K.T.); Center for Clinical Brain Sciences, UK Dementia Research Institute, University of Edinburgh (J.M.W.); Dell Medical School (S.C.J.); Department of Neurology, Columbia University (O.W.); Department of Medicine (Neurology), McMaster University/Population Heath Research Institute (A.S.); Department of Neurology, Stanford University (M.L.)
| | - Tanya Turan
- Department of Neurology, University of Utah (A.D.); Department of Neurology, Yale University (K.N.S.); Department of Emergency Medicine, Brown University (T.M.); Department of Neurology, University of Virginia (K.J.); Department of Neurology, Medical University of South Carolina (T.T., J.E.); Department of Cerebrovascular Medicine, National Cerebral and Cardiovascular Center, Japan (K.T.); Center for Clinical Brain Sciences, UK Dementia Research Institute, University of Edinburgh (J.M.W.); Dell Medical School (S.C.J.); Department of Neurology, Columbia University (O.W.); Department of Medicine (Neurology), McMaster University/Population Heath Research Institute (A.S.); Department of Neurology, Stanford University (M.L.)
| | - Kazunori Toyoda
- Department of Neurology, University of Utah (A.D.); Department of Neurology, Yale University (K.N.S.); Department of Emergency Medicine, Brown University (T.M.); Department of Neurology, University of Virginia (K.J.); Department of Neurology, Medical University of South Carolina (T.T., J.E.); Department of Cerebrovascular Medicine, National Cerebral and Cardiovascular Center, Japan (K.T.); Center for Clinical Brain Sciences, UK Dementia Research Institute, University of Edinburgh (J.M.W.); Dell Medical School (S.C.J.); Department of Neurology, Columbia University (O.W.); Department of Medicine (Neurology), McMaster University/Population Heath Research Institute (A.S.); Department of Neurology, Stanford University (M.L.)
| | - Jordan J. Elm
- Department of Neurology, University of Utah (A.D.); Department of Neurology, Yale University (K.N.S.); Department of Emergency Medicine, Brown University (T.M.); Department of Neurology, University of Virginia (K.J.); Department of Neurology, Medical University of South Carolina (T.T., J.E.); Department of Cerebrovascular Medicine, National Cerebral and Cardiovascular Center, Japan (K.T.); Center for Clinical Brain Sciences, UK Dementia Research Institute, University of Edinburgh (J.M.W.); Dell Medical School (S.C.J.); Department of Neurology, Columbia University (O.W.); Department of Medicine (Neurology), McMaster University/Population Heath Research Institute (A.S.); Department of Neurology, Stanford University (M.L.)
| | - Joanna M. Wardlaw
- Department of Neurology, University of Utah (A.D.); Department of Neurology, Yale University (K.N.S.); Department of Emergency Medicine, Brown University (T.M.); Department of Neurology, University of Virginia (K.J.); Department of Neurology, Medical University of South Carolina (T.T., J.E.); Department of Cerebrovascular Medicine, National Cerebral and Cardiovascular Center, Japan (K.T.); Center for Clinical Brain Sciences, UK Dementia Research Institute, University of Edinburgh (J.M.W.); Dell Medical School (S.C.J.); Department of Neurology, Columbia University (O.W.); Department of Medicine (Neurology), McMaster University/Population Heath Research Institute (A.S.); Department of Neurology, Stanford University (M.L.)
| | - S. Claiborne Johnston
- Department of Neurology, University of Utah (A.D.); Department of Neurology, Yale University (K.N.S.); Department of Emergency Medicine, Brown University (T.M.); Department of Neurology, University of Virginia (K.J.); Department of Neurology, Medical University of South Carolina (T.T., J.E.); Department of Cerebrovascular Medicine, National Cerebral and Cardiovascular Center, Japan (K.T.); Center for Clinical Brain Sciences, UK Dementia Research Institute, University of Edinburgh (J.M.W.); Dell Medical School (S.C.J.); Department of Neurology, Columbia University (O.W.); Department of Medicine (Neurology), McMaster University/Population Heath Research Institute (A.S.); Department of Neurology, Stanford University (M.L.)
| | - Olajide A. Williams
- Department of Neurology, University of Utah (A.D.); Department of Neurology, Yale University (K.N.S.); Department of Emergency Medicine, Brown University (T.M.); Department of Neurology, University of Virginia (K.J.); Department of Neurology, Medical University of South Carolina (T.T., J.E.); Department of Cerebrovascular Medicine, National Cerebral and Cardiovascular Center, Japan (K.T.); Center for Clinical Brain Sciences, UK Dementia Research Institute, University of Edinburgh (J.M.W.); Dell Medical School (S.C.J.); Department of Neurology, Columbia University (O.W.); Department of Medicine (Neurology), McMaster University/Population Heath Research Institute (A.S.); Department of Neurology, Stanford University (M.L.)
| | - Ashkan Shoamanesh
- Department of Neurology, University of Utah (A.D.); Department of Neurology, Yale University (K.N.S.); Department of Emergency Medicine, Brown University (T.M.); Department of Neurology, University of Virginia (K.J.); Department of Neurology, Medical University of South Carolina (T.T., J.E.); Department of Cerebrovascular Medicine, National Cerebral and Cardiovascular Center, Japan (K.T.); Center for Clinical Brain Sciences, UK Dementia Research Institute, University of Edinburgh (J.M.W.); Dell Medical School (S.C.J.); Department of Neurology, Columbia University (O.W.); Department of Medicine (Neurology), McMaster University/Population Heath Research Institute (A.S.); Department of Neurology, Stanford University (M.L.)
| | - Maarten G. Lansberg
- Department of Neurology, University of Utah (A.D.); Department of Neurology, Yale University (K.N.S.); Department of Emergency Medicine, Brown University (T.M.); Department of Neurology, University of Virginia (K.J.); Department of Neurology, Medical University of South Carolina (T.T., J.E.); Department of Cerebrovascular Medicine, National Cerebral and Cardiovascular Center, Japan (K.T.); Center for Clinical Brain Sciences, UK Dementia Research Institute, University of Edinburgh (J.M.W.); Dell Medical School (S.C.J.); Department of Neurology, Columbia University (O.W.); Department of Medicine (Neurology), McMaster University/Population Heath Research Institute (A.S.); Department of Neurology, Stanford University (M.L.)
| |
Collapse
|
14
|
Kherallah RY, Khawaja M, Olson M, Angiolillo D, Birnbaum Y. Cilostazol: a Review of Basic Mechanisms and Clinical Uses. Cardiovasc Drugs Ther 2021; 36:777-792. [PMID: 33860901 DOI: 10.1007/s10557-021-07187-x] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/05/2021] [Indexed: 12/24/2022]
Abstract
Primarily used in the treatment of intermittent claudication, cilostazol is a 2-oxyquinolone derivative that works through the inhibition of phosphodiesterase III and related increases in cyclic adenosine monophosphate (cAMP) levels. However, cilostazol has been implicated in a number of other basic pathways including the inhibition of adenosine reuptake, the inhibition of multidrug resistance protein 4, among others. It has been observed to exhibit antiplatelet, antiproliferative, vasodilatory, and ischemic-reperfusion protective properties. As such, cilostazol has been investigated for clinical use in a variety of settings including intermittent claudication, as an adjunctive for reduction of restenosis after coronary and peripheral endovascular interventions, and in the prevention of secondary stroke, although its widespread implementation for indications other than intermittent claudication has been limited by relatively modest effect sizes and lack of studies in western populations. In this review, we highlight the pleiotropic effects of cilostazol and the evidence for its clinical use.
Collapse
Affiliation(s)
- Riyad Y Kherallah
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Muzamil Khawaja
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Michael Olson
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Dominick Angiolillo
- Division of Cardiology, University of Florida College of Medicine, Jacksonville, FL, USA
| | - Yochai Birnbaum
- Section of Cardiology, Department of Medicine, Baylor College of Medicine, 7200 Cambridge Street, Houston, TX, USA.
| |
Collapse
|
15
|
Motta NAV, Autran LJ, Brazão SC, Lopes RDO, Scaramello CBV, Lima GF, Brito FCFD. Could cilostazol be beneficial in COVID-19 treatment? Thinking about phosphodiesterase-3 as a therapeutic target. Int Immunopharmacol 2021; 92:107336. [PMID: 33418248 PMCID: PMC7768212 DOI: 10.1016/j.intimp.2020.107336] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 11/25/2020] [Accepted: 12/22/2020] [Indexed: 01/25/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes coronavirus disease 2019 (COVID-19) that has emerged and rapidly spread across the world. The COVID-19 severity is associated to viral pneumonia with additional extrapulmonary complications. Hyperinflammation, dysfunctional immune response and hypercoagulability state are associated to poor prognosis. Therefore, the repositioning of multi-target drugs to control the hyperinflammation represents an important challenge for the scientific community. Cilostazol, a selective phosphodiesterase type-3 inhibitor (PDE-3), is an antiplatelet and vasodilator drug, that presents a range of pleiotropic effects, such as antiapoptotic, anti-inflammatory, antioxidant, and cardioprotective activities. Cilostazol also can inhibit the adenosine uptake, which enhances intracellular cAMP levels. In the lungs, elevated cAMP promotes anti-fibrotic, vasodilator, antiproliferative effects, as well as mitigating inflammatory events. Interestingly, a recent study evaluated antiplatelet FDA-approved drugs through molecular docking-based virtual screening on viral target proteins. This study revealed that cilostazol is a promising drug against COVID-19 by inhibiting both main protease (Mpro) and Spike glycoprotein, reinforcing its use as a promising therapeutic approach for COVID-19. Considering the complexity associated to COVID-19 pathophysiology and observing its main mechanisms, this article raises the hypothesis that cilostazol may act on important targets in development of the disease. This review highlights the importance of drug repurposing to address such an urgent clinical demand safely, effectively and at low cost, reinforcing the main pharmacological actions, to support the hypothesis that a multi-target drug such as cilostazol could play an important role in the treatment of COVID-19.
Collapse
Affiliation(s)
- Nadia Alice Vieira Motta
- Laboratory of Experimental Pharmacology (LAFE), Department of Physiology and Pharmacology, Biomedical Institute, Fluminense Federal University (UFF), Room 204-A, 24420-210 Niterói, RJ, Brazil
| | - Lis Jappour Autran
- Laboratory of Experimental Pharmacology (LAFE), Department of Physiology and Pharmacology, Biomedical Institute, Fluminense Federal University (UFF), Room 204-A, 24420-210 Niterói, RJ, Brazil
| | - Stephani Correia Brazão
- Laboratory of Experimental Pharmacology (LAFE), Department of Physiology and Pharmacology, Biomedical Institute, Fluminense Federal University (UFF), Room 204-A, 24420-210 Niterói, RJ, Brazil
| | - Rosane de Oliveira Lopes
- Laboratory of Experimental Pharmacology (LAFE), Department of Physiology and Pharmacology, Biomedical Institute, Fluminense Federal University (UFF), Room 204-A, 24420-210 Niterói, RJ, Brazil
| | - Christianne Brêtas Vieira Scaramello
- Laboratory of Experimental Pharmacology (LAFE), Department of Physiology and Pharmacology, Biomedical Institute, Fluminense Federal University (UFF), Room 204-A, 24420-210 Niterói, RJ, Brazil
| | - Gabriel Ferreira Lima
- Laboratory of Experimental Pharmacology (LAFE), Department of Physiology and Pharmacology, Biomedical Institute, Fluminense Federal University (UFF), Room 204-A, 24420-210 Niterói, RJ, Brazil
| | - Fernanda Carla Ferreira de Brito
- Laboratory of Experimental Pharmacology (LAFE), Department of Physiology and Pharmacology, Biomedical Institute, Fluminense Federal University (UFF), Room 204-A, 24420-210 Niterói, RJ, Brazil.
| |
Collapse
|
16
|
Barrett TJ, Schlegel M, Zhou F, Gorenchtein M, Bolstorff J, Moore KJ, Fisher EA, Berger JS. Platelet regulation of myeloid suppressor of cytokine signaling 3 accelerates atherosclerosis. Sci Transl Med 2020; 11:11/517/eaax0481. [PMID: 31694925 DOI: 10.1126/scitranslmed.aax0481] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 07/19/2019] [Accepted: 10/04/2019] [Indexed: 12/19/2022]
Abstract
Platelets are best known as mediators of hemostasis and thrombosis; however, their inflammatory effector properties are increasingly recognized. Atherosclerosis, a chronic vascular inflammatory disease, represents the interplay between lipid deposition in the artery wall and unresolved inflammation. Here, we reveal that platelets induce monocyte migration and recruitment into atherosclerotic plaques, resulting in plaque platelet-macrophage aggregates. In Ldlr -/- mice fed a Western diet, platelet depletion decreased plaque size and necrotic area and attenuated macrophage accumulation. Platelets drive atherogenesis by skewing plaque macrophages to an inflammatory phenotype, increasing myeloid suppressor of cytokine signaling 3 (SOCS3) expression and reducing the Socs1:Socs3 ratio. Platelet-induced Socs3 expression regulates plaque macrophage reprogramming by promoting inflammatory cytokine production (Il6, Il1b, and Tnfa) and impairing phagocytic capacity, dysfunctions that contribute to unresolved inflammation and sustained plaque growth. Translating our data to humans with cardiovascular disease, we found that women with, versus without, myocardial infarction have up-regulation of SOCS3, lower SOCS1:SOCS3, and increased monocyte-platelet aggregate. A second cohort of patients with lower extremity atherosclerosis demonstrated that SOCS3 and the SOCS1:SOCS3 ratio correlated with platelet activity and inflammation. Collectively, these data provide a causative link between platelet-mediated myeloid inflammation and dysfunction, SOCS3, and cardiovascular disease. Our findings define an atherogenic role of platelets and highlight how, in the absence of thrombosis, platelets contribute to inflammation.
Collapse
Affiliation(s)
- Tessa J Barrett
- Marc and Ruti Bell Program in Vascular Biology, Division of Cardiology, Department of Medicine, New York University School of Medicine, New York, NY 10016, USA
| | - Martin Schlegel
- Marc and Ruti Bell Program in Vascular Biology, Division of Cardiology, Department of Medicine, New York University School of Medicine, New York, NY 10016, USA
| | - Felix Zhou
- Marc and Ruti Bell Program in Vascular Biology, Division of Cardiology, Department of Medicine, New York University School of Medicine, New York, NY 10016, USA
| | - Mike Gorenchtein
- Marc and Ruti Bell Program in Vascular Biology, Division of Cardiology, Department of Medicine, New York University School of Medicine, New York, NY 10016, USA
| | - Jennifer Bolstorff
- Marc and Ruti Bell Program in Vascular Biology, Division of Cardiology, Department of Medicine, New York University School of Medicine, New York, NY 10016, USA
| | - Kathryn J Moore
- Marc and Ruti Bell Program in Vascular Biology, Division of Cardiology, Department of Medicine, New York University School of Medicine, New York, NY 10016, USA
| | - Edward A Fisher
- Marc and Ruti Bell Program in Vascular Biology, Division of Cardiology, Department of Medicine, New York University School of Medicine, New York, NY 10016, USA
| | - Jeffrey S Berger
- Marc and Ruti Bell Program in Vascular Biology, Division of Cardiology, Department of Medicine, New York University School of Medicine, New York, NY 10016, USA.
| |
Collapse
|
17
|
Inosine, an endogenous purine nucleoside, avoids early stages of atherosclerosis development associated to eNOS activation and p38 MAPK/NF-kB inhibition in rats. Eur J Pharmacol 2020; 882:173289. [PMID: 32565337 DOI: 10.1016/j.ejphar.2020.173289] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 06/03/2020] [Accepted: 06/16/2020] [Indexed: 01/22/2023]
Abstract
Atherosclerosis is a multifactorial chronic disease, initiated by an endothelial dysfunction. Adenosine and its analogs can change a variety of inflammatory diseases and has shown important effects at different disease models. Inosine is a stable analogous of adenosine, but its effects in inflammatory diseases, like atherosclerosis, have not yet been studied. The aim of this study was to evaluate the pharmacological properties of inosine, administered sub chronically in a hypercholesterolemic model. Male Wistar rats were divided into four groups: control group (C) and control + inosine (C + INO) received standard chow, hypercholesterolemic diet group (HCD) and HCD + inosine (HCD + INO) were fed a hypercholesterolemic diet. At 31st experimentation day, the treatment with inosine was performed for C + INO and HCD + INO groups once daily in the last 15 days. We observed that the hypercholesterolemic diet promoted an increase in lipid levels and inflammatory cytokines production, while inosine treatment strongly decreased these effects. Additionally, HCD group presented a decrease in maximum relaxation acetylcholine induced and an increase in contractile response phenylephrine induced when compared to the control group, as well as it has presented an enhancement in collagen and ADP-induced platelet aggregation. On the other hand, inosine treatment promoted a decrease in contractile response to phenylephrine, evoked an improvement in endothelium-dependent vasorelaxant response and presented antiplatelet properties. Moreover, inosine activated eNOS and reduced p38 MAPK/NF-κB pathway in aortic tissues. Taken together, the present results indicate inosine as a potential drug for the treatment of cardiovascular disorders such as atherosclerosis.
Collapse
|
18
|
Sanders O, Rajagopal L. Phosphodiesterase Inhibitors for Alzheimer's Disease: A Systematic Review of Clinical Trials and Epidemiology with a Mechanistic Rationale. J Alzheimers Dis Rep 2020; 4:185-215. [PMID: 32715279 PMCID: PMC7369141 DOI: 10.3233/adr-200191] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/17/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Preclinical studies, clinical trials, and reviews suggest increasing 3',5'-cyclic adenosine monophosphate (cAMP) and 3',5'-cyclic guanosine monophosphate (cGMP) with phosphodiesterase inhibitors is disease-modifying in Alzheimer's disease (AD). cAMP/protein kinase A (PKA) and cGMP/protein kinase G (PKG) signaling are disrupted in AD. cAMP/PKA and cGMP/PKG activate cAMP response element binding protein (CREB). CREB binds mitochondrial and nuclear DNA, inducing synaptogenesis, memory, and neuronal survival gene (e.g., brain-derived neurotrophic factor) and peroxisome proliferator-activated receptor-γ coactivator-1α (PGC1α). cAMP/PKA and cGMP/PKG activate Sirtuin-1, which activates PGC1α. PGC1α induces mitochondrial biogenesis and antioxidant genes (e.g.,Nrf2) and represses BACE1. cAMP and cGMP inhibit BACE1-inducing NFκB and tau-phosphorylating GSK3β. OBJECTIVE AND METHODS We review efficacy-testing clinical trials, epidemiology, and meta-analyses to critically investigate whether phosphodiesteraseinhibitors prevent or treat AD. RESULTS Caffeine and cilostazol may lower AD risk. Denbufylline and sildenafil clinical trials are promising but preliminary and inconclusive. PF-04447943 and BI 409,306 are ineffective. Vinpocetine, cilostazol, and nicergoline trials are mixed. Deprenyl/selegiline trials show only short-term benefits. Broad-spectrum phosphodiesterase inhibitor propentofylline has been shown in five phase III trials to improve cognition, dementia severity, activities of daily living, and global assessment in mild-to-moderate AD patients on multiple scales, including the ADAS-Cogand the CIBIC-Plus in an 18-month phase III clinical trial. However, two books claimed based on a MedScape article an 18-month phase III trial failed, so propentofylline was discontinued. Now, propentofylline is used to treat canine cognitive dysfunction, which, like AD, involves age-associated wild-type Aβ deposition. CONCLUSION Phosphodiesterase inhibitors may prevent and treat AD.
Collapse
|
19
|
Satti HH, Khaleel EF, Badi RM, Elrefaie AO, Mostafa DG. Antiplatelet activity of astaxanthin in control- and high cholesterol-fed rats mediated by down-regulation of P2Y 12, inhibition of NF-κB, and increasing intracellular levels of cAMP. Platelets 2020; 32:469-478. [PMID: 32379559 DOI: 10.1080/09537104.2020.1756237] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
This study evaluated the antiplatelet effect of the plant carotenoid, astaxanthin (ASTX) in rats fed either control or high cholesterol plus cholic acid diet (HCCD) and possible underlying mechanisms. Adult male Wistar rats were divided into four groups (n = 8/each), namely, control (fed normal diet), control + ASTX (10 mg/kg/day), HCCD-fed rats, and HCCD + ASTX-treated rats. Diets and treatments were orally administered daily for 30 days. In both control and HCCD-fed rats, ASTX significantly increased fecal levels of triglycerides and cholesterol, reduced platelet count, prolonged bleeding time, and inhibited platelet aggregation. It also reduced platelet levels of reactive oxygen species (ROS) and Bcl-2; thromboxane B2 (TXB2) release; and the expression of P2Y12, P-selectin, and CD36 receptors. Moreover, the activity NF-κB p65 and Akt was inhibited. Concomitantly, it increased the protein levels of cleaved caspase-3 and vasodilator-stimulated phosphoprotein (p-VASP) as well as intracellular levels of cAMP. However, in HCCD-fed rats, the effects of ASTX were associated with reduced serum levels of ox-LDL-c and fasting plasma glucose levels. In conclusion, antiplatelet effects of ASTX involve ROS scavenging, inhibiting NF-κB activity, down-regulating P2Y12 expression, and increasing intracellular levels of cAMP that are attributed to its antioxidant, hypolipidemic, and anti-inflammatory effects.
Collapse
Affiliation(s)
- Huda H Satti
- Department of Pathology, College of Medicine, King Khalid University, Abha, Kingdom of Saudi Arabia.,Department of Pathology, University of Khartoum, Khartoum, Sudan
| | - Eman F Khaleel
- Department of Medical Physiology, College of Medicine, King Khalid University, Abha, Kingdom of Saudi Arabia.,Faculty of Medicine, Department of Medical Physiology, Cairo University, Cairo, Egypt
| | - Rehab M Badi
- Department of Medical Physiology, College of Medicine, King Khalid University, Abha, Kingdom of Saudi Arabia.,Faculty of Medicine, Department of Physiology, University of Khartoum, Khartoum, Sudan
| | - Amany O Elrefaie
- Department of Pathology, College of Medicine, King Khalid University, Abha, Kingdom of Saudi Arabia.,National Liver Institute, Department of Pathology, Menoufyia University, Menoufyia, Egypt
| | - Dalia G Mostafa
- Department of Medical Physiology, College of Medicine, King Khalid University, Abha, Kingdom of Saudi Arabia.,Faculty of Medicine, Department of Medical Physiology, Assiut University, Assiut, Egypt
| |
Collapse
|
20
|
Silva NLC, Motta NAV, Soares MA, Araujo OMO, Espíndola LCP, Colombo APV, Lopes RT, Brito FCF, Miranda ALP, Tributino JLM. Periodontal status, vascular reactivity, and platelet aggregation changes in rats submitted to hypercholesterolemic diet and periodontitis. J Periodontal Res 2020; 55:453-463. [PMID: 31994219 DOI: 10.1111/jre.12730] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 12/14/2019] [Accepted: 12/28/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND OBJECTIVES Periodontitis can corroborate with development and progression of atherosclerosis and a possible bidirectional interaction between both pathologies has been hypothesized. The aim of this work was to study the interactions between diet-induced hypercholesterolemia and ligature-induced periodontitis in Wistar rats submitted to both conditions. MATERIAL AND METHODS Animals were divided into four experimental groups: C (control: standard diet without periodontitis), Perio (periodontitis plus standard diet), HC (high cholesterol diet without periodontitis), and HC + Perio (high cholesterol diet plus periodontitis). The diets were offered for 45 days and a silk ligature was applied in the lower first molars of Perio and HC-Perio animals on day 34 and maintained for 11 days until euthanasia. The mandibles were excised, and alveolar bone loss was determined by macroscopic and micro-tomographic (µ-CT) imaging. Blood samples were obtained, and platelet aggregation was induced in plasma rich in platelets by adenosine diphosphate (ADP) and collagen. Endothelium-dependent vascular reactivity and protein expression of endothelial (eNOS), phosphorylated endothelial (peNOS), and inducible (iNOS) nitric oxide synthases were evaluated in aorta samples. RESULTS The HC diet combined with periodontitis (HC + Perio group) was associated with an increased alveolar bone loss, when compared to the other groups. Both in Perio and HC groups, platelet aggregation induced by ADP or collagen was increased, while maximum aortic relaxation induced by acetylcholine was decreased. Periodontitis or HC diet alone decreased the expression of peNOS and HC diet increased the expression of iNOS. In contrast, no additive or synergistic effects were found in vascular reactivity or in platelet aggregation when the two conditions were associated (HC + Perio group). CONCLUSION Hypercholesterolemia accelerated the process of bone loss induced by periodontitis while a high cholesterol diet or periodontitis individually increased platelet aggregation and vascular reactivity in rats without additive or synergistic effects, when associated.
Collapse
Affiliation(s)
- Natália L C Silva
- Laboratory of Studies in Experimental Pharmacology (LEFEx), Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Nadia A V Motta
- Laboratory of Experimental Pharmacology (LAFE), Department of Physiology and Pharmacology, Biomedical Institute, Fluminense Federal University, Niterói, Brazil
| | - Mariana A Soares
- Laboratory of Studies in Experimental Pharmacology (LEFEx), Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Olga M O Araujo
- Laboratory of Nuclear Instrumentation (LIN), Nuclear Engineering Program (PEN), Institute Alberto Luiz de Coimbra of Post-Graduation and Research in Engineering (COPPE-UFRJ), Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Laís Christina P Espíndola
- Department of Medical Microbiology, Institute of Microbiology Paulo de Góes, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ana Paula V Colombo
- Department of Medical Microbiology, Institute of Microbiology Paulo de Góes, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ricardo Tadeu Lopes
- Laboratory of Nuclear Instrumentation (LIN), Nuclear Engineering Program (PEN), Institute Alberto Luiz de Coimbra of Post-Graduation and Research in Engineering (COPPE-UFRJ), Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fernanda Carla F Brito
- Laboratory of Experimental Pharmacology (LAFE), Department of Physiology and Pharmacology, Biomedical Institute, Fluminense Federal University, Niterói, Brazil
| | - Ana Luísa P Miranda
- Laboratory of Studies in Experimental Pharmacology (LEFEx), Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Jorge L M Tributino
- Laboratory of Molecular Pharmacology (LFM), Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
21
|
Kojok K, El-Kadiry AEH, Merhi Y. Role of NF-κB in Platelet Function. Int J Mol Sci 2019; 20:E4185. [PMID: 31461836 PMCID: PMC6747346 DOI: 10.3390/ijms20174185] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 08/25/2019] [Accepted: 08/26/2019] [Indexed: 01/04/2023] Open
Abstract
Platelets are megakaryocyte-derived fragments lacking nuclei and prepped to maintain primary hemostasis by initiating blood clots on injured vascular endothelia. Pathologically, platelets undergo the same physiological processes of activation, secretion, and aggregation yet with such pronouncedness that they orchestrate and make headway the progression of atherothrombotic diseases not only through clot formation but also via forcing a pro-inflammatory state. Indeed, nuclear factor-κB (NF-κB) is largely implicated in atherosclerosis and its pathological complication in atherothrombotic diseases due to its transcriptional role in maintaining pro-survival and pro-inflammatory states in vascular and blood cells. On the other hand, we know little on the functions of platelet NF-κB, which seems to function in other non-genomic ways to modulate atherothrombosis. Therein, this review will resemble a rich portfolio for NF-κB in platelets, specifically showing its implications at the levels of platelet survival and function. We will also share the knowledge thus far on the effects of active ingredients on NF-κB in general, as an extrapolative method to highlight the potential therapeutic targeting of NF-κB in coronary diseases. Finally, we will unzip a new horizon on a possible extra-platelet role of platelet NF-κB, which will better expand our knowledge on the etiology and pathophysiology of atherothrombosis.
Collapse
Affiliation(s)
- Kevin Kojok
- The Laboratory of Thrombosis and Hemostasis, Montreal Heart Institute, Research Centre, 5000 Belanger Street, Montreal, H1T 1C8, QC, Canada
- Faculty of Medicine, Université de Montréal, Montreal, H3T 1J4, QC, Canada
| | - Abed El-Hakim El-Kadiry
- The Laboratory of Thrombosis and Hemostasis, Montreal Heart Institute, Research Centre, 5000 Belanger Street, Montreal, H1T 1C8, QC, Canada
- Faculty of Medicine, Université de Montréal, Montreal, H3T 1J4, QC, Canada
| | - Yahye Merhi
- The Laboratory of Thrombosis and Hemostasis, Montreal Heart Institute, Research Centre, 5000 Belanger Street, Montreal, H1T 1C8, QC, Canada.
- Faculty of Medicine, Université de Montréal, Montreal, H3T 1J4, QC, Canada.
| |
Collapse
|
22
|
Vardenafil and cilostazol can improve vascular reactivity in rats with diabetes mellitus and rheumatoid arthritis co-morbidity. Life Sci 2019; 229:67-79. [PMID: 31085245 DOI: 10.1016/j.lfs.2019.05.024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 04/27/2019] [Accepted: 05/10/2019] [Indexed: 12/17/2022]
Abstract
Endothelial dysfunction and vascular reactivity defects secondary to metabolic and immunological disorders carry risk of serious cardiovascular complications. Here, the effects of the phosphodiesterase (PDE) inhibitors vardenafil and cilostazol were examined against rheumatoid arthritis (RA)/diabetes mellitus (DM)-co-morbidity-induced endothelial dysfunction and vascular reactivity defects. After setting of RA/DM-co-morbidity model, rats were divided into a normal control group, an RA/DM-co-morbidity group, and two treatment groups receiving oral vardenafil (10 mg/kg/day) and cilostazol (30 mg/kg/day) for 21 days after RA/DM-co-morbidity induction. Aorta was isolated for biochemical estimations of the pro-inflammatory vasoconstrictor molecules angiotensin-II (Ang-II) and endothelin-1 (ET-1), the adhesion molecules P-selectin and vascular cell adhesion molecule-1 (VCAM-1), the energy sensor adenosine-5'-monophosphate-activated protein kinase (AMPK), and the vasodilator anti-inflammatory molecule vasoactive intestinal peptide (VIP) using enzyme-linked immunosorbent assay (ELISA) and western blot analysis. Immunohistochemical estimations of endothelial nitric oxide synthase (eNOS) and matrix metalloproteinase (MMP)-2 were performed coupled with histopathological examination using routine hematoxylin and eosin (H&E) and special Masson trichrome staining. The in vitro study was conducted using aortic strips where cumulative concentration response curves were done for the endothelium-dependent relaxing factor acetylcholine and the endothelium-independent relaxing factor sodium nitroprusside after submaximal contraction with phenylephrine. Vardenafil and cilostazol significantly improved endothelial integrity biomarkers in vivo supported with histopathological findings in addition to improved vasorelaxation in vitro. Apart from their known PDE inhibition, up-regulation of vascular AMPK and eNOS coupled with down-regulation of Ang-II, ET-1, P-selectin, VCAM-1 and MMP-2 may explain vardenafil and cilostazol protective effect against RA/DM-co-morbidity-induced endothelial dysfunction and vascular reactivity defects.
Collapse
|
23
|
Piazza S, Pacchetti B, Fumagalli M, Bonacina F, Dell'Agli M, Sangiovanni E. Comparison of Two Ginkgo biloba L. Extracts on Oxidative Stress and Inflammation Markers in Human Endothelial Cells. Mediators Inflamm 2019; 2019:6173893. [PMID: 31341420 PMCID: PMC6614955 DOI: 10.1155/2019/6173893] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 05/22/2019] [Accepted: 06/10/2019] [Indexed: 11/24/2022] Open
Abstract
Atherosclerosis is characterized by interaction between immune and vascular endothelial cells which is mediated by adhesion molecules occurring on the surface of the vascular endothelium leading to massive release of proinflammatory mediators. Ginkgo biloba L. (Ginkgoaceae) standardized extracts showing beneficial effects are commonly prepared by solvent extraction, and acetone is used according to the European Pharmacopoeia recommendations; the well-known Ginkgo biloba acetone extract EGb761® is the most clinically investigated. However, in some countries, the allowed amount of solvent is limited to ethanol, thus implying that the usage of a standardized Ginkgo biloba ethanol extract may be preferred in all those cases, such as for food supplements. The present paper investigates if ethanol and acetone extracts, with comparable standardization, may be considered comparable in terms of biological activity, focusing on the radical scavenging and anti-inflammatory activities. Both the extracts showed high inhibition of TNFα-induced VCAM-1 release (41.1-43.9 μg/mL), which was partly due to the NF-κB pathway impairment. Besides ROS decrease, cAMP increase following treatment with ginkgo extracts was addressed and proposed as further molecular mechanism responsible for the inhibition of endothelial E-selectin. No statistical difference was observed between the extracts. The present study demonstrates for the first time that ethanol and acetone extracts show comparable biological activities in human endothelial cell, thus providing new insights into the usage of ethanol extracts in those countries where restrictions in amount of acetone are present.
Collapse
Affiliation(s)
- Stefano Piazza
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milano, Italy
| | | | - Marco Fumagalli
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milano, Italy
| | - Fabrizia Bonacina
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milano, Italy
| | - Mario Dell'Agli
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milano, Italy
| | - Enrico Sangiovanni
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milano, Italy
| |
Collapse
|
24
|
El Awdan SA, Abdel Rahman RF, Ibrahim HM, Hegazy RR, El Marasy SA, Badawi M, Arbid MS. Regression of fibrosis by cilostazol in a rat model of thioacetamide-induced liver fibrosis: Up regulation of hepatic cAMP, and modulation of inflammatory, oxidative stress and apoptotic biomarkers. PLoS One 2019; 14:e0216301. [PMID: 31067255 PMCID: PMC6505801 DOI: 10.1371/journal.pone.0216301] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 04/17/2019] [Indexed: 12/20/2022] Open
Abstract
In liver fibrosis, conversion of fibroblasts to profibrogenic myofibroblasts significantly drives the development of the disease. A crucial role of cyclic adenosine monophosphate (cAMP) in regulation of fibroblast function has been reported. Increase in cAMP levels has been found to decrease fibroblast proliferation, inhibit their conversion to myofibroblast, and stimulate their death. cAMP is generated by adenyl cyclase (AC), and degraded by cyclic nucleotide phosphodiesterase (PDE). In this study, the antifibrotic effect of a PDE inhibitor, cilostazol (Cilo), on a rat model of liver fibrosis induced by thioacetamide (TAA) was investigated. Four groups of rats were used; the first group received the vehicles and served as the normal control group, while liver fibrosis was induced in the other groups using (TAA, 200 mg/kg/biweekly for 8 successive weeks, ip). The last two groups were treated with Cilo (50 and 100 mg/kg/day, po, respectively). Induction of liver fibrosis in TAA-treated rats was observed as evidenced by the biochemical and histopathological findings. On the other hand, a potent antifibrotic effect was observed in the groups treated with Cilo, with preference to the higher dose. In these groups, a significant increase in the liver content of cAMP was demonstrated that was accompanied by reduction in the hepatic expression of key fibrogenic cytokines, growth factors, and inflammatory biomarkers, including interleukin-6, tumor necrosis factor-alpha, nuclear factor kappa B, and transforming growth factor-beta as compared to TAA group. Moreover, amelioration of TAA-induced oxidative stress and apoptosis in the liver has been observed. These findings reveal the antifibrotic effect of Cilo against TAA-induced liver fibrosis in rats, and suggest regulation of cAMP pathway, together with the modulation of oxidative stress, inflammation, and apoptosis as mechanistic cassette underlines this effect.
Collapse
Affiliation(s)
- Sally A. El Awdan
- Pharmacology Department, Medical Division, National Research Centre, Giza, Egypt
| | | | - Heba M. Ibrahim
- Pharmacology Department, Medical Division, National Research Centre, Giza, Egypt
| | - Rehab R. Hegazy
- Pharmacology Department, Medical Division, National Research Centre, Giza, Egypt
| | - Salma A. El Marasy
- Pharmacology Department, Medical Division, National Research Centre, Giza, Egypt
| | - Manal Badawi
- Pathology Department, Medical Division, National Research Centre, Giza, Egypt
| | - Mahmoud S. Arbid
- Pharmacology Department, Medical Division, National Research Centre, Giza, Egypt
| |
Collapse
|
25
|
Cho O, Jang YJ, Park KY, Heo TH. Beneficial anti-inflammatory effects of combined rosuvastatin and cilostazol in a TNF-driven inflammatory model. Pharmacol Rep 2019; 71:266-271. [DOI: 10.1016/j.pharep.2018.12.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 11/07/2018] [Accepted: 12/14/2018] [Indexed: 02/07/2023]
|
26
|
Park KY, Heo TH. Combination therapy with cilostazol and pravastatin improves antiatherogenic effects in low-density lipoprotein receptor knockout mice. Cardiovasc Ther 2018; 36:e12476. [PMID: 30378752 DOI: 10.1111/1755-5922.12476] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Revised: 09/29/2018] [Accepted: 10/29/2018] [Indexed: 12/22/2022] Open
Abstract
AIMS Despite the therapeutic efficacy of statins and antiplatelet agents for atherosclerosis, monotherapy with each drug alone is often insufficient to achieve the patient's therapeutic goals. We previously showed that combined statin/antiplatelet agent/anti-tumor necrosis factor (TNF) agent therapy (pravastatin/sarpogrelate/etanercept) reduces atherosclerotic lesions by inhibiting TNF, an atherogenic cytokine that contributes to the progression of arteriosclerosis. In addition, our previous study showed that combined treatment with pravastatin and cilostazol is effective for reducing TNF-driven inflammation through anti-TNF activity. Therefore, in the present study, we evaluated the additive effects of combined pravastatin and cilostazol therapy on atherosclerotic progression using low-density lipoprotein receptor (LDLR) knockout (KO) mice. METHODS Ten-week-old LDLR KO mice were fed a high-fat, high-cholesterol diet and orally administered pravastatin and cilostazol alone or in combination. Body weight, plasma lipid levels, and the levels of intracellular adhesion molecules and inflammatory cytokines were analyzed. In addition, aortas and aortic roots were stained with Oil Red O, and atherosclerotic plaques were quantified. RESULTS The atherosclerotic plaques in the combined pravastatin and cilostazol treatment groups were significantly reduced compared to those in each drug monotherapy group. The combination therapy group also showed the downregulation of ICAM-1, MOMA-2, TNF, interleukin (IL)-6, triglyceride, total cholesterol, and low-density lipoprotein levels and the upregulation of high-density lipoprotein levels compared to those of the pravastatin- or cilostazol-treated groups. CONCLUSIONS Our results suggest that combination therapy with pravastatin and cilostazol exerts beneficial effects by decreasing atherosclerotic lesion progression and improving the pro-inflammatory state in the vascular endothelium. These effects are mediated by the reduction in adhesion molecule expression, immune cell infiltration, and cytokine levels and the antiatherosclerotic modulation of serum cholesterol levels. Therefore, we conclude that combined treatment with pravastatin and cilostazol may be a more effective antiatherosclerotic strategy than treatment with either agent alone.
Collapse
Affiliation(s)
- Kyung-Yeon Park
- Department of Pharmacy, Integrated Research Institute of Pharmaceutical Sciences, and BK21 PLUS Team for Creative Leader Program for Pharmacomics-based Future Pharmacy, College of Pharmacy, The Catholic University of Korea, Bucheon-si, Korea
| | - Tae-Hwe Heo
- Department of Pharmacy, Integrated Research Institute of Pharmaceutical Sciences, and BK21 PLUS Team for Creative Leader Program for Pharmacomics-based Future Pharmacy, College of Pharmacy, The Catholic University of Korea, Bucheon-si, Korea
| |
Collapse
|
27
|
Beute J, Lukkes M, Koekoek EP, Nastiti H, Ganesh K, de Bruijn MJ, Hockman S, van Nimwegen M, Braunstahl GJ, Boon L, Lambrecht BN, Manganiello VC, Hendriks RW, KleinJan A. A pathophysiological role of PDE3 in allergic airway inflammation. JCI Insight 2018; 3:94888. [PMID: 29367458 DOI: 10.1172/jci.insight.94888] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 12/12/2017] [Indexed: 01/04/2023] Open
Abstract
Phosphodiesterase 3 (PDE3) and PDE4 regulate levels of cyclic AMP, which are critical in various cell types involved in allergic airway inflammation. Although PDE4 inhibition attenuates allergic airway inflammation, reported side effects preclude its application as an antiasthma drug in humans. Case reports showed that enoximone, which is a smooth muscle relaxant that inhibits PDE3, is beneficial and lifesaving in status asthmaticus and is well tolerated. However, clinical observations also showed antiinflammatory effects of PDE3 inhibition. In this study, we investigated the role of PDE3 in a house dust mite-driven (HDM-driven) allergic airway inflammation (AAI) model that is characterized by T helper 2 cell activation, eosinophilia, and reduced mucosal barrier function. Compared with wild-type (WT) littermates, mice with a targeted deletion of the PDE3A or PDE3B gene showed significantly reduced HDM-driven AAI. Therapeutic intervention in WT mice showed that all hallmarks of HDM-driven AAI were abrogated by the PDE3 inhibitors enoximone and milrinone. Importantly, we found that enoximone also reduced the upregulation of the CD11b integrin on mouse and human eosinophils in vitro, which is crucial for their recruitment during allergic inflammation. This study provides evidence for a hitherto unknown antiinflammatory role of PDE3 inhibition in allergic airway inflammation and offers a potentially novel treatment approach.
Collapse
Affiliation(s)
- Jan Beute
- Department of Pulmonary Medicine, Erasmus MC, 's-Gravendijkwal, Rotterdam, Netherlands
| | - Melanie Lukkes
- Department of Pulmonary Medicine, Erasmus MC, 's-Gravendijkwal, Rotterdam, Netherlands
| | - Ewout P Koekoek
- Department of Pulmonary Medicine, Erasmus MC, 's-Gravendijkwal, Rotterdam, Netherlands
| | - Hedwika Nastiti
- Department of Pulmonary Medicine, Erasmus MC, 's-Gravendijkwal, Rotterdam, Netherlands
| | - Keerthana Ganesh
- Department of Pulmonary Medicine, Erasmus MC, 's-Gravendijkwal, Rotterdam, Netherlands
| | | | - Steve Hockman
- Cardiovascular and Pulmonary Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland USA
| | - Menno van Nimwegen
- Department of Pulmonary Medicine, Erasmus MC, 's-Gravendijkwal, Rotterdam, Netherlands
| | | | - Louis Boon
- Epirus Biopharmaceuticals Netherlands Yalelaan, Utrecht, Netherlands
| | - Bart N Lambrecht
- Department of Pulmonary Medicine, Erasmus MC, 's-Gravendijkwal, Rotterdam, Netherlands.,VIB Center for Inflammation Research, Ghent University, Ghent, Belgium
| | - Vince C Manganiello
- Cardiovascular and Pulmonary Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland USA
| | - Rudi W Hendriks
- Department of Pulmonary Medicine, Erasmus MC, 's-Gravendijkwal, Rotterdam, Netherlands
| | - Alex KleinJan
- Department of Pulmonary Medicine, Erasmus MC, 's-Gravendijkwal, Rotterdam, Netherlands
| |
Collapse
|
28
|
Ohnuki Y, Ohnuki Y, Kohara S, Shimizu M, Takizawa S. Dual Therapy with Aspirin and Cilostazol May Improve Platelet Aggregation in Noncardioembolic Stroke Patients: A Pilot Study. Intern Med 2017; 56:1307-1313. [PMID: 28566591 PMCID: PMC5498192 DOI: 10.2169/internalmedicine.56.7760] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Objective Some previous studies have found clinical benefit of dual antiplatelet therapy with aspirin and cilostazol for prevention of secondary stroke, but the physiological mechanism involved remains unknown. We aimed to clarify the effects of aspirin/cilostazol therapy on the platelet and endothelial functions of patients with acute noncardioembolic ischemic stroke, in comparison to patients who were treated with aspirin alone. Methods The present randomized prospective pilot study enrolled 24 patients within a week after the onset of noncardioembolic ischemic stroke. The patients were randomly allocated to receive aspirin (100 mg/day) (A group; 11 patients) or cilostazol (200 mg/day) plus aspirin (100 mg/day) (CA group; 13 patients). We measured platelet aggregation, platelet activation, and the thrombomodulin (TM), highly sensitive C-reactive protein (hs-CRP), intercellular adhesion molecule-1 (ICAM-1), vascular cell adhesion molecule-1 (VCAM-1) and von Willebrand (vWF) antigen levels and vWF activity over a 4-week period after enrollment. Results There was no significant difference in the platelet functions of the A and CA groups. However, the platelet aggregation induced by adenosine diphosphate (ADP) was decreased at 2 and 4 weeks (p<0.05) after treatment in comparison to the pre-treatment values in the CA group, but not in the A group. Platelet activation, and the hs-CRP, TM, ICAM-1, VCAM-1 and vWF values did not significantly decrease after treatment in either group. Conclusion Although there were no significant differences in platelet aggregation, platelet activation or the endothelial biomarker levels of the A and CA groups, dual therapy with aspirin and cilostazol inhibited platelet aggregation in comparison to the pre-treatment values, similarly to patients who received aspirin alone. This may suggest the clinical usefulness of dual therapy with aspirin and cilostazol in the treatment of patients with noncardioembolic ischemic stroke.
Collapse
Affiliation(s)
- Yoichi Ohnuki
- Division of Neurology, Department of Internal Medicine, Tokai University School of Medicine, Japan
| | - Yuko Ohnuki
- Department of Molecular Life Science, Basic Medical Science and Molecular Medicine, Tokai University School of Medicine, Japan
| | - Saori Kohara
- Division of Neurology, Department of Internal Medicine, Tokai University School of Medicine, Japan
| | - Mie Shimizu
- Division of Neurology, Department of Internal Medicine, Tokai University School of Medicine, Japan
| | - Shunya Takizawa
- Division of Neurology, Department of Internal Medicine, Tokai University School of Medicine, Japan
| |
Collapse
|