1
|
Kowalczyk P, Krych S, Kramkowski K, Jęczmyk A, Hrapkowicz T. Effect of Oxidative Stress on Mitochondrial Damage and Repair in Heart Disease and Ischemic Events. Int J Mol Sci 2024; 25:12467. [PMID: 39596532 PMCID: PMC11594588 DOI: 10.3390/ijms252212467] [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: 10/07/2024] [Revised: 11/10/2024] [Accepted: 11/15/2024] [Indexed: 11/28/2024] Open
Abstract
The literature analysis conducted in this review discusses the latest achievements in the identification of cardiovascular damage induced by oxidative stress with secondary platelet mitochondrial dysfunction. Damage to the platelets of mitochondria as a result of their interactions with reactive oxygen species (ROS) and reactive nitrogen species (RNS) can lead to their numerous ischemic events associated with hypoxia or hyperoxia processes in the cell. Disturbances in redox reactions in the platelet mitochondrial membrane lead to the direct oxidation of cellular macromolecules, including nucleic acids (DNA base oxidation), membrane lipids (lipid peroxidation process) and cellular proteins (formation of reducing groups in repair proteins and amino acid peroxides). Oxidative changes in biomolecules inducing tissue damage leads to inflammation, initiating pathogenic processes associated with faster cell aging or their apoptosis. The consequence of damage to platelet mitochondria and their excessive activation is the induction of cardiovascular and neurodegenerative diseases (Parkinson's and Alzheimer's), as well as carbohydrate metabolism disorders (diabetes). The oxidation of mitochondrial DNA can lead to modifications in its bases, inducing the formation of exocyclic adducts of the ethano and propano type. As a consequence, it disrupts DNA repair processes and conduces to premature neoplastic transformation in critical genes such as the p53 suppressor gene, which leads to the development of various types of tumors. The topic of new innovative methods and techniques for the analysis of oxidative stress in platelet mitochondria based on methods such as a nicking assay, oxygen consumption assay, Total Thrombus formation Analysis System (T-Tas), and continuous-flow left ventricular assist devices (CF-LVADs) was also discussed. They were put together into one scientific and research platform. This will enable the facilitation of faster diagnostics and the identification of platelet mitochondrial damage by clinicians and scientists in order to implement adequate therapeutic procedures and minimize the risk of the induction of cardiovascular diseases, including ischemic events correlated with them. A quantitative analysis of the processes of thrombus formation in cardiovascular diseases will provide an opportunity to select specific anticoagulant and thrombolytic drugs under conditions of preserved hemostasis.
Collapse
Affiliation(s)
- Paweł Kowalczyk
- Department of Animal Nutrition, The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, Instytucka 3, 05-110 Jabłonna, Poland
| | - Sebastian Krych
- Student’s Scientific Association, Department of Cardiac, Vascular and Endovascular Surgery and Transplantology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-055 Katowice, Poland
- Silesian Centre for Heart Diseases in Zabrze, Department of Cardiac, Vascular and Endovascular Surgery and Transplantology, Medical University of Silesia, 40-055 Katowice, Poland;
| | - Karol Kramkowski
- Department of Physical Chemistry, Medical University of Bialystok, Kilińskiego 1, 15-089 Białystok, Poland;
| | - Agata Jęczmyk
- Students’ Scientific Association, III Department of Cardiology, School of Medical Sciences in Zabrze, Medical University of Silesia, 40-055 Katowice, Poland;
| | - Tomasz Hrapkowicz
- Silesian Centre for Heart Diseases in Zabrze, Department of Cardiac, Vascular and Endovascular Surgery and Transplantology, Medical University of Silesia, 40-055 Katowice, Poland;
| |
Collapse
|
2
|
Zheng SS, Perdomo JS. Desialylation and Apoptosis in Immune Thrombocytopenia: Implications for Pathogenesis and Treatment. Curr Issues Mol Biol 2024; 46:11942-11956. [PMID: 39590303 PMCID: PMC11592706 DOI: 10.3390/cimb46110709] [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: 09/16/2024] [Revised: 10/18/2024] [Accepted: 10/22/2024] [Indexed: 11/28/2024] Open
Abstract
Immune thrombocytopenia (ITP) is an autoimmune disease in which platelet autoantibodies play a significant role in its pathogenesis. Regulatory T cell dysfunction and T cell-mediated cytotoxicity also contribute to thrombocytopenia. Current therapies are directed towards immune suppression and modulation as well as stimulation of platelet production with thrombopoietin receptor agonists. Additional mechanisms of the pathogenesis of ITP have been suggested by recent experimental data. One of these processes, known as desialylation, involves antibody-induced removal of terminal sialic acid residues on platelet surface glycoproteins, leading to hepatic platelet uptake and thrombocytopenia. Apoptosis, or programmed platelet death, may also contribute to the pathogenesis of ITP. The extent of the impact of desialylation and apoptosis on ITP, the relative proportion of patients affected, and the role of antibody specificity are still the subject of investigation. This review will discuss both historical and new evidence of the influence of desialylation and apoptosis in the pathogenesis of ITP, with an emphasis on the clinical implications of these developments. Further understanding of both platelet desialylation and apoptosis might change current clinical practice and improve patient outcomes.
Collapse
Affiliation(s)
- Shiying Silvia Zheng
- Haematology Research Unit, St. George and Sutherland Clinical Campuses, School of Medicine & Health, University of New South Wales, Kogarah, NSW 2217, Australia;
- Department of Haematology, St. George Hospital, Kogarah, NSW 2217, Australia
| | - José Sail Perdomo
- Haematology Research Group, Central Clinical School, Faculty Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
3
|
Yadav P, Beura SK, Panigrahi AR, Kulkarni PP, Yadav MK, Munshi A, Singh SK. Lysophosphatidylcholine induces oxidative stress and calcium-mediated cell death in human blood platelets. Cell Biol Int 2024; 48:1266-1284. [PMID: 38837523 DOI: 10.1002/cbin.12192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 05/06/2024] [Accepted: 05/11/2024] [Indexed: 06/07/2024]
Abstract
Platelets are essential component of circulation that plays a major role in hemostasis and thrombosis. During activation and its demise, platelets release platelet-derived microvesicles, with lysophosphatidylcholine (LPC) being a prominent component in their lipid composition. LPC, an oxidized low-density lipoprotein, is involved in cellular metabolism, but its higher level is implicated in pathologies like atherosclerosis, diabetes, and inflammatory disorders. Despite this, its impact on platelet function remains relatively unexplored. To address this, we studied LPC's effects on washed human platelets. A multimode plate reader was employed to measure reactive oxygen species and intracellular calcium using H2DCF-DA and Fluo-4-AM, respectively. Flow cytometry was utilized to measure phosphatidylserine expression, mitochondrial membrane potential (ΔΨm), and mitochondrial permeability transition pore (mPTP) formation using FITC-Annexin V, JC-1, and CoCl2/calcein-AM, respectively. Additionally, platelet morphology and its ultrastructure were observed via phase contrast and electron microscopy. Sonoclot and light transmission aggregometry were employed to examine fibrin formation and platelet aggregation, respectively. The findings demonstrate that LPC induced oxidative stress and increased intracellular calcium in platelets, resulting in increased phosphatidylserine expression and reduced ΔΨm. LPC triggered caspase-independent platelet death and mPTP opening via cytosolic and mitochondrial calcium, along with microvesiculation and reduced platelet counts. LPC increased the platelet's size, adopting a balloon-shaped morphology, causing membrane fragmentation and releasing its cellular contents, while inducing a pro-coagulant phenotype with increased fibrin formation and reduced integrin αIIbβ3 activation. Conclusively, this study reveals LPC-induced oxidative stress and calcium-mediated platelet death, necrotic in nature with pro-coagulant properties, potentially impacting inflammation and repair mechanisms during vascular injury.
Collapse
Affiliation(s)
- Pooja Yadav
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Ghudda, Bathinda, India
| | - Samir K Beura
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Ghudda, Bathinda, India
| | - Abhishek R Panigrahi
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Ghudda, Bathinda, India
| | - Paresh P Kulkarni
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Mithlesh K Yadav
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Ghudda, Bathinda, India
| | - Anjana Munshi
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Ghudda, Bathinda, India
| | - Sunil K Singh
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Ghudda, Bathinda, India
- Department of Biochemistry, School of Basic Sciences, Central University of Punjab, Ghudda, Bathinda, India
| |
Collapse
|
4
|
Sołtysik BK, Karolczak K, Kostka T, Stephenson SS, Watala C, Kostka J. Contribution of Physical Activity to the Oxidative and Antioxidant Potential in 60-65-Year-Old Seniors. Antioxidants (Basel) 2023; 12:1200. [PMID: 37371930 DOI: 10.3390/antiox12061200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 05/19/2023] [Accepted: 05/30/2023] [Indexed: 06/29/2023] Open
Abstract
Both acute exercise and regular physical activity (PA) are directly related to the redox system. However, at present, there are data suggesting both positive and negative relationships between the PA and oxidation. In addition, there is a limited number of publications differentiating the relationships between PA and numerous markers of plasma and platelets targets for the oxidative stress. In this study, in a population of 300 participants from central Poland (covering the age range between 60 and 65 years), PA was assessed as regards energy expenditure (PA-EE) and health-related behaviors (PA-HRB). Total antioxidant potential (TAS), total oxidative stress (TOS) and several other markers of an oxidative stress, monitored in platelet and plasma lipids and proteins, were then determined. The association of PA with oxidative stress was determined taking into the account basic confounders, such as age, sex and the set of the relevant cardiometabolic factors. In simple correlations, platelet lipid peroxides, free thiol and amino groups of platelet proteins, as well as the generation of superoxide anion radical, were inversely related with PA-EE. In multivariate analyses, apart from other cardiometabolic factors, a significant positive impact of PA-HRB was revealed for TOS (inverse relationship), while in the case of PA-EE, the effect was found to be positive (inverse association) for lipid peroxides and superoxide anion but negative (lower concentration) for free thiol and free amino groups in platelets proteins. Therefore, the impact of PA may be different on oxidative stress markers in platelets as compared to plasma proteins and also dissimilar on platelet lipids and proteins. These associations are more visible for platelets than plasma markers. For lipid oxidation, PA seems to have protective effect. In the case of platelets proteins, PA tends to act as pro-oxidative factor.
Collapse
Affiliation(s)
- Bartłomiej K Sołtysik
- Department of Geriatrics, Medical University of Lodz, Haller Square No. 1, 90-419 Łódź, Poland
| | - Kamil Karolczak
- Department of Hemostatic Disorders, Medical University of Lodz, Mazowiecka Street 6/8, 92-215 Łódź, Poland
| | - Tomasz Kostka
- Department of Geriatrics, Medical University of Lodz, Haller Square No. 1, 90-419 Łódź, Poland
| | - Serena S Stephenson
- Department of Geriatrics, Medical University of Lodz, Haller Square No. 1, 90-419 Łódź, Poland
| | - Cezary Watala
- Department of Hemostatic Disorders, Medical University of Lodz, Mazowiecka Street 6/8, 92-215 Łódź, Poland
| | - Joanna Kostka
- Department of Gerontology, Medical University of Lodz, Milionowa Street No. 14, 93-113 Łódź, Poland
| |
Collapse
|
5
|
Hosseini E, Nodeh FK, Ghasemzadeh M. Gamma irradiation induces a pro-apoptotic state in longer stored platelets, without progressing to an overt apoptosis by day 7 of storage. Apoptosis 2023:10.1007/s10495-023-01841-5. [PMID: 37127837 DOI: 10.1007/s10495-023-01841-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/01/2023] [Indexed: 05/03/2023]
Abstract
BACKGROUND Although gamma-irradiation to platelet products is a standard method to prevent the risk of TA-GVHD in vulnerable recipients, it induces some proteomic and redox changes, of which irradiation-induced ROS increments may potentiate platelet mitochondrial dysfunction. However, whether these changes cause platelet apoptosis, or affect their viability during storage, is the main subject of this study. METHODS PLT-rich plasma PC was split into two bags, one kept as control while other was subjected to gamma-irradiation. Within 7-days storage, cytosolic and mitochondrial levels of cytochrome c and pro-apoptotic molecules of Bak and Bax were evaluated by western-blotting. Intraplatelet active caspase (using FAM-DEVD-FMK) and PS-exposure were detected by flowcytometry. Caspase activity in platelet lysate was also confirmed by immunofluorescence detection of Caspase-3/7 Substrate N-Ac-DEVD-N'-MC-R110 while platelet viability was evaluated with MTT assays. RESULTS Cytosolic cytochrome c gradually increased while its mitochondrial content steadily declined during 7 days of storage. In a contrary trend, reverse patterns were observed for Bak and Bax expressions. Gamma-irradiated platelets showed higher release of mitochondrial cytochrome c that reflected by higher cytosolic cytochrome c levels on day 7 of storage. Concurrently mitochondrial pro-apoptotic Bak and Bax proteins increased on day 7 in irradiated products. However, gamma-irradiation didn't significantly increase caspase activity or PS-exposure, nor did it decrease platelet viability. CONCLUSION Here, consistent with studies on "gamma-irradiation-induced oxidative stress", we showed that gamma-ray also increases platelet pro-apoptotic signals during storage, although not strongly enough to affect platelet viability by overt apoptosis induction. Conclusively, whether supplementing ROS scavengers or antioxidants to irradiated platelets can improve their quality during storage may be of interest for future research.
Collapse
Affiliation(s)
- Ehteramolsadat Hosseini
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Iranian Blood Transfusion Organization Building, Hemmat Exp. Way, Next to the Milad Tower, Tehran, 14665-1157, Iran
| | - Fatemeh Kiani Nodeh
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Iranian Blood Transfusion Organization Building, Hemmat Exp. Way, Next to the Milad Tower, Tehran, 14665-1157, Iran
| | - Mehran Ghasemzadeh
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Iranian Blood Transfusion Organization Building, Hemmat Exp. Way, Next to the Milad Tower, Tehran, 14665-1157, Iran.
| |
Collapse
|
6
|
Jain K, Gu SX, Hwa J. Cross talk on "endogenous SOD2 (superoxide dismutase) regulates platelet-dependent thrombin generation and thrombosis during aging" SOD2 in platelets: with age comes responsibility. JOURNAL OF THROMBOSIS AND HAEMOSTASIS : JTH 2023; 21:1077-1081. [PMID: 36716965 DOI: 10.1016/j.jtha.2023.01.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 01/13/2023] [Accepted: 01/18/2023] [Indexed: 01/29/2023]
Affiliation(s)
- Kanika Jain
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine Yale University School of Medicine, New Haven, Connecticut, USA
| | - Sean X Gu
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine Yale University School of Medicine, New Haven, Connecticut, USA; Department of Laboratory Medicine, Yale University School of Medicine, Yale New Haven Hospital, New Haven, Connecticut, USA
| | - John Hwa
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine Yale University School of Medicine, New Haven, Connecticut, USA.
| |
Collapse
|
7
|
Abstract
There is a crucial need for platelet transfusion during an emergency-surgery and treatment of platelet disorders. The unavailability of donors has furthermore increased the demand for platelet storage. Platelets have limited shelf life due to bacterial contamination and storage lesions. Temperature, materials, oxygen availability, media, platelet processing and manufacturing methods influence the platelet quality and viability during storage. The conception of various platelet additive solutions along with the advent of plastic storage during the 1980s led to enormous developments in platelet storage strategies. Cold storage of platelets gained attention despite its inability to contribute to platelet survival post-transfusion as it offers faster haemostasis. Several developments in platelet storage strategies over the years have improved the quality and shelf-life of stored platelets. Despite the progress, the efficacy of platelets during storage beyond a week has not been achieved. Antioxidants as additives have been explored in platelet storage and have proven to enhance the efficacy of platelets during prolonged storage. However, the molecular interactions of antioxidants in platelets can provide a better understanding of their mechanism of action. Optimization of dosage concentrations of antioxidants is also a critical parameter to be considered as they tend to exhibit toxicity at certain levels. This review provides comprehensive insights into the critical factors affecting platelet storage and the evolution of platelet storage. It also emphasizes the role of antioxidants as additives in platelet storage solutions and their future prospects towards better platelet banking.
Collapse
Affiliation(s)
- Vani Rajashekaraiah
- Department of Biotechnology, School of Sciences, JAIN (Deemed-to-be University), #34, 1st Cross, JC Road, Bengaluru, 560027, India.
| | - Magdaline Christina Rajanand
- Department of Biotechnology, School of Sciences, JAIN (Deemed-to-be University), #34, 1st Cross, JC Road, Bengaluru, 560027, India
| |
Collapse
|
8
|
Coenzyme Q10 Attenuates Human Platelet Aggregation Induced by SARS-CoV-2 Spike Protein via Reducing Oxidative Stress In Vitro. Int J Mol Sci 2022; 23:ijms232012345. [PMID: 36293203 PMCID: PMC9604356 DOI: 10.3390/ijms232012345] [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: 09/09/2022] [Revised: 10/04/2022] [Accepted: 10/05/2022] [Indexed: 12/23/2022] Open
Abstract
Platelet hyperreactivity and oxidative stress are the important causes of thrombotic disorders in patients with COVID-19. Oxidative stress, induced by the excessive generation of reactive oxygen species (ROS), could increase platelet function and the risk of thrombus formation. Coenzyme Q10 (CoQ10), exhibits strong antioxidative activity and anti-platelet effect. However, the effects and mechanisms of CoQ10 on attenuating platelet aggregation induced by spike protein have never been studied. This study aims to investigate whether the SARS-CoV-2 spike protein potentiates human platelet function via ROS signaling and the protective effect of CoQ10 in vitro. Using a series of platelet function assays, we found that spike protein potentiated platelet aggregation and oxidative stress, such as ROS level, mitochondrial membrane potential depolarization, and lipid damage level (MDA and 8-iso-PGF2α) in vitro. Furthermore, CoQ10 attenuated platelet aggregation induced by spike protein. As an anti-platelet mechanism, we showed that CoQ10 significantly decreased the excess production of ROS induced by spike protein. Our findings show that the protective effect of CoQ10 on spike protein-potentiated platelet aggregation is probably associated with its strong antioxidative ability.
Collapse
|
9
|
Platelet Redox Imbalance in Hypercholesterolemia: A Big Problem for a Small Cell. Int J Mol Sci 2022; 23:ijms231911446. [PMID: 36232746 PMCID: PMC9570056 DOI: 10.3390/ijms231911446] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 09/23/2022] [Accepted: 09/24/2022] [Indexed: 11/17/2022] Open
Abstract
The imbalance between reactive oxygen species (ROS) synthesis and their scavenging by anti-oxidant defences is the common soil of many disorders, including hypercholesterolemia. Platelets, the smallest blood cells, are deeply involved in the pathophysiology of occlusive arterial thrombi associated with myocardial infarction and stroke. A great deal of evidence shows that both increased intraplatelet ROS synthesis and impaired ROS neutralization are implicated in the thrombotic process. Hypercholesterolemia is recognized as cause of atherosclerosis, cerebro- and cardiovascular disease, and, closely related to this, is the widespread acceptance that it strongly contributes to platelet hyperreactivity via direct oxidized LDL (oxLDL)-platelet membrane interaction via scavenger receptors such as CD36 and signaling pathways including Src family kinases (SFK), mitogen-activated protein kinases (MAPK), and nicotinamide adenine dinucleotide phosphate (NADPH) oxidase. In turn, activated platelets contribute to oxLDL generation, which ends up propagating platelet activation and thrombus formation through a mechanism mediated by oxidative stress. When evaluating the effect of lipid-lowering therapies on thrombogenesis, a large body of evidence shows that the effects of statins and proprotein convertase subtilisin/kexin type 9 inhibitors are not limited to the reduction of LDL-C but also to the down-regulation of platelet reactivity mainly by mechanisms sensitive to intracellular redox balance. In this review, we will focus on the role of oxidative stress-related mechanisms as a cause of platelet hyperreactivity and the pathophysiological link of the pleiotropism of lipid-lowering agents to the beneficial effects on platelet function.
Collapse
|
10
|
Water-Soluble Tomato Concentrate, a Potential Antioxidant Supplement, Can Attenuate Platelet Apoptosis and Oxidative Stress in Healthy Middle-Aged and Elderly Adults: A Randomized, Double-Blinded, Crossover Clinical Trial. Nutrients 2022; 14:nu14163374. [PMID: 36014880 PMCID: PMC9412583 DOI: 10.3390/nu14163374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 08/14/2022] [Accepted: 08/15/2022] [Indexed: 11/17/2022] Open
Abstract
Increased oxidative stress and platelet apoptotic in middle-aged and elderly adults are important risk factors for atherosclerotic cardiovascular disease (ASCVD). Therefore, it is of great significance to control the oxidative stress and platelet apoptosis in middle-aged and elderly adults. Previous acute clinical trials have shown that water-soluble tomato concentrate (WSTC) from fresh tomatoes could exert antiplatelet benefits after 3 h or 7 h, but its effects on platelet apoptosis and oxidative stress are still unknown, especially in healthy middle-aged and elderly adults. This current study aimed to examine the efficacies of WSTC on platelet apoptosis and oxidative stress in healthy middle-aged and elderly adults via a randomized double-blinded placebo-controlled crossover clinical trial (10 weeks in total). A total of 52 healthy middle-aged and elderly adults completed this trial. The results showed that WSTC could increase the serum total antioxidant capacity levels (p < 0.05) and decrease the serum malondialdehyde levels (p < 0.05) after a 4-week WSTC supplementation in healthy middle-aged and elderly adults. Platelet endogenous reactive oxygen species generation (p < 0.05), mitochondrial membrane potential dissipation (p < 0.05) and phosphatidylserine exposure (p < 0.05) were attenuated. In addition, our present study also found that WSTC could inhibit platelet aggregation and activation induced by collagen or ADP after intervention (p < 0.05), while having no effects on adverse events (p > 0.05). The results suggest that WSTC can inhibit oxidative stress and its related platelet apoptosis, which may provide a basis for the primary prevention of WSTC in ASCVD.
Collapse
|
11
|
Gu SX, Dayal S. Redox Mechanisms of Platelet Activation in Aging. Antioxidants (Basel) 2022; 11:995. [PMID: 35624860 PMCID: PMC9137594 DOI: 10.3390/antiox11050995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 05/14/2022] [Accepted: 05/17/2022] [Indexed: 02/01/2023] Open
Abstract
Aging is intrinsically linked with physiologic decline and is a major risk factor for a broad range of diseases. The deleterious effects of advancing age on the vascular system are evidenced by the high incidence and prevalence of cardiovascular disease in the elderly. Reactive oxygen species are critical mediators of normal vascular physiology and have been shown to gradually increase in the vasculature with age. There is a growing appreciation for the complexity of oxidant and antioxidant systems at the cellular and molecular levels, and accumulating evidence indicates a causal association between oxidative stress and age-related vascular disease. Herein, we review the current understanding of mechanistic links between oxidative stress and thrombotic vascular disease and the changes that occur with aging. While several vascular cells are key contributors, we focus on oxidative changes that occur in platelets and their mediation in disease progression. Additionally, we discuss the impact of comorbid conditions (i.e., diabetes, atherosclerosis, obesity, cancer, etc.) that have been associated with platelet redox dysregulation and vascular disease pathogenesis. As we continue to unravel the fundamental redox mechanisms of the vascular system, we will be able to develop more targeted therapeutic strategies for the prevention and management of age-associated vascular disease.
Collapse
Affiliation(s)
- Sean X. Gu
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT 06511, USA;
| | - Sanjana Dayal
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
- Iowa City VA Healthcare System, Iowa City, IA 52246, USA
| |
Collapse
|
12
|
Ghosh AK, Soberanes S, Lux E, Shang M, Aillon RP, Eren M, Budinger GRS, Miyata T, Vaughan DE. Pharmacological inhibition of PAI-1 alleviates cardiopulmonary pathologies induced by exposure to air pollutants PM 2.5. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 287:117283. [PMID: 34426376 PMCID: PMC8434953 DOI: 10.1016/j.envpol.2021.117283] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 04/16/2021] [Accepted: 04/28/2021] [Indexed: 05/09/2023]
Abstract
Numerous studies have established that acute or chronic exposure to environmental pollutants like particulate matter (PM) leads to the development of accelerated aging related pathologies including pulmonary and cardiovascular diseases, and thus air pollution is one of the major global threats to human health. Air pollutant particulate matter 2.5 (PM2.5)-induced cellular dysfunction impairs tissue homeostasis and causes vascular and cardiopulmonary damage. To test a hypothesis that elevated plasminogen activator inhibitor-1 (PAI-1) levels play a pivotal role in air pollutant-induced cardiopulmonary pathologies, we examined the efficacy of a drug-like novel inhibitor of PAI-1, TM5614, in treating PM2.5-induced vascular and cardiopulmonary pathologies. Results from biochemical, histological, and immunohistochemical studies revealed that PM2.5 increases the circulating levels of PAI-1 and thrombin and that TM5614 treatment completely abrogates these effects in plasma. PM2.5 significantly augments the levels of pro-inflammatory cytokine interleukin-6 (IL-6) in bronchoalveolar lavage fluid (BALF), and this also can be reversed by TM5614, indicating its efficacy in amelioration of PM2.5-induced increases in inflammatory and pro-thrombotic factors. TM5614 reduces PM2.5-induced increased levels of inflammatory markers cluster of differentiation 107 b (Mac3) and phospho-signal transducer and activator of transcription-3 (pSTAT3), adhesion molecule vascular cell adhesion molecule 1 (VCAM1), and apoptotic marker cleaved caspase 3. Longer exposure to PM2.5 induces pulmonary and cardiac thrombosis, but TM5614 significantly ameliorates PM2.5-induced vascular thrombosis. TM5614 also reduces PM2.5-induced increased blood pressure and heart weight. In vitro cell culture studies revealed that PM2.5 induces the levels of PAI-1, type I collagen, fibronectin (Millipore), and sterol regulatory element binding protein-1 and 2 (SREBP-1 and SREBP-2), transcription factors that mediate profibrogenic signaling, in cardiac fibroblasts. TM5614 abrogated that stimulation, indicating that it may block PM2.5-induced PAI-1 and profibrogenic signaling through suppression of SREBP-1 and 2. Furthermore, TM5614 blocked PM2.5-mediated suppression of nuclear factor erythroid related factor 2 (Nrf2), a major antioxidant regulator, in cardiac fibroblasts. Pharmacological inhibition of PAI-1 with TM5614 is a promising therapeutic approach to control air pollutant PM2.5-induced cardiopulmonary and vascular pathologies.
Collapse
Affiliation(s)
- Asish K Ghosh
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| | - Saul Soberanes
- Pulmonary and Critical Care Division, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Elizabeth Lux
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Meng Shang
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Raul Piseaux Aillon
- Pulmonary and Critical Care Division, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Mesut Eren
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - G R Scott Budinger
- Pulmonary and Critical Care Division, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Toshio Miyata
- United Centers for Advanced Research and Translational Medicine, Tohoku University, Miyagi, Japan
| | - Douglas E Vaughan
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
13
|
Mahdi A, Cortese-Krott MM, Kelm M, Li N, Pernow J. Novel perspectives on redox signaling in red blood cells and platelets in cardiovascular disease. Free Radic Biol Med 2021; 168:95-109. [PMID: 33789125 DOI: 10.1016/j.freeradbiomed.2021.03.020] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 03/01/2021] [Accepted: 03/07/2021] [Indexed: 12/13/2022]
Abstract
The fundamental physiology of circulating red blood cells (RBCs) and platelets involving regulation of oxygen transport and hemostasis, respectively, are well-described in the literature. Their abundance in the circulation and their interaction with the vascular wall and each other have attracted the attention of other putative physiological and pathophysiological effects of these cells. RBCs and platelets are both important regulators of redox balance harboring powerful pro-oxidant and anti-oxidant (enzymatic and non-enzymatic) capacities. They are also involved in the regulation of vascular tone mainly via export of nitric oxide bioactivity and adenosine triphosphate. Of further importance are emerging observations that these cells undergo functional alterations when exposed to risk factors for cardiovascular disease and during developed cardiometabolic diseases. Under these conditions, the RBCs and platelets contribute to increased oxidative stress by their formation of reactive species including superoxide anion radical, hydrogen peroxide and peroxynitrite. These alterations trigger key changes in the vascular wall characterized by enhanced oxidative stress, reduced nitric oxide bioavailability and endothelial dysfunction. Additional pathophysiological effects are triggered in the heart resulting in increased susceptibility to ischemia-reperfusion injury with impairment in cardiac function. Pharmacological interventions aiming at restoring circulating cell function has been shown to exert marked beneficial effects on cardiovascular function. In this review, we summarize the current knowledge of RBC and platelet biology with special focus on redox biology, their roles in the development of cardiovascular disease and potential therapeutic strategies targeting RBC and platelet dysfunction. Finally, the complex and scarcely understood interaction between RBCs and platelets is discussed.
Collapse
Affiliation(s)
- Ali Mahdi
- Department of Medicine, Division of Cardiology, Karolinska Institutet, Stockholm, Sweden
| | - Miriam M Cortese-Krott
- Department of Cardiology, Pulmonology and Angiology Medical Faculty, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany; Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Malte Kelm
- Department of Cardiology, Pulmonology and Angiology Medical Faculty, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
| | - Nailin Li
- Department of Medicine, Division of Cardiovascular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - John Pernow
- Department of Medicine, Division of Cardiology, Karolinska Institutet, Stockholm, Sweden; Department of Cardiology, Heart and Vascular Division, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
14
|
Hu LL, Zou K, Chen Y, Wu LJ, Cao J, Xiong XY, Wang L, Cheng XS, Xiao QZ, Yang RQ. Functional role and molecular mechanisms underlying prohibitin 2 in platelet mitophagy and activation. Mol Med Rep 2021; 23:384. [PMID: 33760146 PMCID: PMC7986013 DOI: 10.3892/mmr.2021.12023] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 02/24/2021] [Indexed: 12/20/2022] Open
Abstract
Platelet mitophagy is a major pathway involved in the clearance of injured mitochondria during hemostasis and thrombosis. Prohibitin 2 (PHB2) has recently emerged as an inner mitochondrial membrane receptor involved in mitophagy. However, the mechanisms underlying PHB2-mediated platelet mitophagy and activation are not completely understood. PHB2 is a highly conserved inner mitochondrial membrane protein that regulates mitochondrial assembly and function due to its unique localization on the mitochondrial membrane. The present study aimed to investigate the role and mechanism underlying PHB2 in platelet mitophagy and activation. Phorbol-12-myristate-13-acetate (PMA) was used to induce MEG-01 cells maturation and differentiate into platelets following PHB2 knockdown. Cell Counting Kit-8 assays were performed to examine platelet viability. Flow cytometry was performed to assess platelet mitochondrial membrane potential. RT-qPCR and western blotting were conducted to measure mRNA and protein expression levels, respectively. Subsequently, platelets were exposed to CCCP and the role of PHB2 was assessed. The results of the present study identified a crucial role for PHB2 in platelet mitophagy and activation, suggesting that PHB2-mediated regulation of mitophagy may serve as a novel strategy for downregulating the expression of platelet activation genes. Although further research into mitophagy is required, the present study suggested that PHB2 may serve as a novel therapeutic target for thrombosis-related diseases due to its unique localization on the mitochondrial membrane.
Collapse
Affiliation(s)
- Long-Long Hu
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Kai Zou
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Yuan Chen
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Li-Juan Wu
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Jie Cao
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Xiao-Ying Xiong
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Ling Wang
- Medicine Lab, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Xiao-Shu Cheng
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Qing-Zhong Xiao
- Department of Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Ren-Qiang Yang
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
15
|
Hoppel F, Calabria E, Pesta DH, Kantner-Rumplmair W, Gnaiger E, Burtscher M. Effects of Ultramarathon Running on Mitochondrial Function of Platelets and Oxidative Stress Parameters: A Pilot Study. Front Physiol 2021; 12:632664. [PMID: 33679442 PMCID: PMC7935014 DOI: 10.3389/fphys.2021.632664] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 01/05/2021] [Indexed: 01/13/2023] Open
Abstract
Only a few studies have evaluated changes in mitochondrial function and oxidative stress associated with ultramarathon running. Invasive biopsies are needed to assess mitochondrial function of skeletal muscle, which may not be well tolerated by some individuals. Platelets (PLTs) as a metabolically highly active and homogenous cell population were suggested as a potentially valuable surrogate to investigate mitochondrial function. Thus, this study was aimed to evaluate mitochondrial function of PLTs and its association with individual race performance and markers of oxidative stress, muscle damage and renal dysfunction. Race performance and mitochondrial function (high-resolution respirometry, HRR) of PLTs using different substrates inducing ROUTINE, LEAK, N-pathway control state (Complex I linked oxidative phosphorylation; CI, OXPHOS), NS-pathway control state (CI + II linked OXPHOS and electron transfer pathway; ET), S-pathway control state (CII linked ET) as well as parameters of oxidative stress and antioxidant capacity, and markers of muscle and renal injury were assessed in eight male ultramarathon runners (26–45 years) before, immediately after and 24 h after an ultramarathon race (PRE, POST, and REC). Ultramarathon running induced an increase in LEAK O2 flux of PLT mitochondria and slight, largely non-significant changes in the oxidant/antioxidant balance. Levels of creatine kinase (CK), lactate dehydrogenase (LDH), blood urea nitrogen, and creatinine were all significantly elevated POST and remained high in REC. There were inverse correlations between race time and N-linked substrate state PRE-POST, and changes in CK and LDH levels were significantly related to PLT mitochondrial LEAK and N-linked respiration PRE. Although race-related changes in respirometry parameters of PLT mitochondria were rather small, a somewhat more pronounced increase in the relative N-linked respiration in faster runners might suggest PLT CI as indicator of physical fitness. The higher PLT LEAK PRE and diminished increase of CK during the race may represent a prophylactic preconditioning and the slight but non-significant elevation of the antioxidant potential post-race as a protective consequence of the race-related oxidative stress and potential threat to the kidney. Our findings point toward an interrelationship between mitochondrial function of PLTs, individual fitness levels and extreme physical and metal stresses, which stimulates further research.
Collapse
Affiliation(s)
- Florian Hoppel
- Oroboros Instruments, Innsbruck, Austria.,Department of Sport Science, University of Innsbruck, Innsbruck, Austria
| | - Elisa Calabria
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Dominik H Pesta
- Department of Sport Science, University of Innsbruck, Innsbruck, Austria.,Institute for Clinical Diabetology, Leibniz Center for Diabetes Research at Heinrich-Heine University Düsseldorf, German Diabetes Center, Düsseldorf, Germany.,German Center for Diabetes Research (DZD e.V.), München-Neuherberg, Germany.,Institute of Aerospace Medicine, German Aerospace Center (DLR), Cologne, Germany
| | - Wilhelm Kantner-Rumplmair
- Department of Psychosomatic Pain Ambulance, University Hospital for Medical Psychology and Psychotherapy, Innsbruck, Austria
| | - Erich Gnaiger
- Oroboros Instruments, Innsbruck, Austria.,D. Swarovski Research Laboratory, Department of Visceral, Transplant Thoracic Surgery, Medical University Innsbruck, Innsbruck, Austria
| | - Martin Burtscher
- Department of Sport Science, University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
16
|
Ngo ATP, Parra-Izquierdo I, Aslan JE, McCarty OJT. Rho GTPase regulation of reactive oxygen species generation and signalling in platelet function and disease. Small GTPases 2021; 12:440-457. [PMID: 33459160 DOI: 10.1080/21541248.2021.1878001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
Platelets are master regulators and effectors of haemostasis with increasingly recognized functions as mediators of inflammation and immune responses. The Rho family of GTPase members Rac1, Cdc42 and RhoA are known to be major components of the intracellular signalling network critical to platelet shape change and morphological dynamics, thus playing a major role in platelet spreading, secretion and thrombus formation. Initially linked to the regulation of actomyosin contraction and lamellipodia formation, recent reports have uncovered non-canonical functions of platelet RhoGTPases in the regulation of reactive oxygen species (ROS), where intrinsically generated ROS modulate platelet function and contribute to thrombus formation. Platelet RhoGTPases orchestrate oxidative processes and cytoskeletal rearrangement in an interconnected manner to regulate intracellular signalling networks underlying platelet activity and thrombus formation. Herein we review our current knowledge of the regulation of platelet ROS generation by RhoGTPases and their relationship with platelet cytoskeletal reorganization, activation and function.
Collapse
Affiliation(s)
- Anh T P Ngo
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA
| | - Ivan Parra-Izquierdo
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA.,Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - Joseph E Aslan
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA.,Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon, USA.,Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, Oregon, USA
| | - Owen J T McCarty
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
17
|
Analysis of the mechanism of damage produced by thiazole orange photoinactivation in apheresis platelets. BLOOD TRANSFUSION = TRASFUSIONE DEL SANGUE 2020; 19:403-412. [PMID: 32955423 DOI: 10.2450/2020.0100-20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 08/06/2020] [Indexed: 11/21/2022]
Abstract
BACKGROUND Pathogen Reduction Technologies (PRTs) are broad spectrum nucleic acid replication-blocking antimicrobial treatments designed to mitigate risk of infection from blood product transfusions. Thiazole Orange (TO), a photosensitizing nucleic acid dye, was previously shown to photoinactivate several types of bacterial and viral pathogens in RBC suspensions without adverse effects on function. In this report we extended TO treatment to platelet concentrates (PCs) to see whether it is compatible with in vitro platelet functions also, and thus, could serve as a candidate technology for further evaluation. MATERIAL AND METHODS PCs were treated with TO, and an effective treatment dose for inactivation of Staphylococci was identified. Platelet function and physiology were then evaluated by various assays in vitro. RESULTS Phototreatment of PCs yielded significant reduction (≥4-log) in Staphylococci at TO concentrations ≥20 μM. However, treatment with TO reduced aggregation response to collagen over time, and platelets became unresponsive by 24 hours post-treatment (from >80% at 1 h to 0% at 24 h). TO treatment also significantly increased CD62P expression (<1% CD62P+ for untreated and >50% for TO treated at 1 h) and induced apoptosis in platelets (<1% Annexin V+ for untreated and >50% for TO treated at 1 h) and damaged mitochondrial DNA. A mitochondria-targeted antioxidant and reactive oxygen species (ROS) scavenger Mito-Tempo mitigated these adverse effects. DISCUSSION The results demonstrate that TO compromises mitochondria and perturbs internal signaling that activates platelets and triggers apoptosis. This study illustrates that protecting platelet mitochondria and its functions should be a fundamental consideration in selecting a PRT for transfusion units containing platelets, such as PCs.
Collapse
|
18
|
ROS in Platelet Biology: Functional Aspects and Methodological Insights. Int J Mol Sci 2020; 21:ijms21144866. [PMID: 32660144 PMCID: PMC7402354 DOI: 10.3390/ijms21144866] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 06/26/2020] [Accepted: 07/07/2020] [Indexed: 12/22/2022] Open
Abstract
Reactive oxygen species (ROS) and mitochondria play a pivotal role in regulating platelet functions. Platelet activation determines a drastic change in redox balance and in platelet metabolism. Indeed, several signaling pathways have been demonstrated to induce ROS production by NAPDH oxidase (NOX) and mitochondria, upon platelet activation. Platelet-derived ROS, in turn, boost further ROS production and consequent platelet activation, adhesion and recruitment in an auto-amplifying loop. This vicious circle results in a platelet procoagulant phenotype and apoptosis, both accounting for the high thrombotic risk in oxidative stress-related diseases. This review sought to elucidate molecular mechanisms underlying ROS production upon platelet activation and the effects of an altered redox balance on platelet function, focusing on the main advances that have been made in platelet redox biology. Furthermore, given the increasing interest in this field, we also describe the up-to-date methods for detecting platelets, ROS and the platelet bioenergetic profile, which have been proposed as potential disease biomarkers.
Collapse
|
19
|
Arachidonic Acid Attenuates Cell Proliferation, Migration and Viability by a Mechanism Independent on Calcium Entry. Int J Mol Sci 2020; 21:ijms21093315. [PMID: 32392840 PMCID: PMC7247542 DOI: 10.3390/ijms21093315] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 04/30/2020] [Accepted: 05/06/2020] [Indexed: 12/13/2022] Open
Abstract
Arachidonic acid (AA) is a phospholipase A2 metabolite that has been reported to mediate a plethora of cellular mechanisms involved in healthy and pathological states such as platelet aggregation, lymphocyte activation, and tissue inflammation. AA has been described to activate Ca2+ entry through the arachidonate-regulated Ca2+-selective channels (ARC channels). Here, the analysis of the changes in the intracellular Ca2+ homeostasis revealed that, despite MDA-MB-231 cells expressing the ARC channel components Orai1, Orai3, and STIM1, AA does not evoke Ca2+ entry in these cells. We observed that AA evokes Ca2+ entry in MDA-MB-231 cells transiently expressing ARC channels. Nevertheless, MDA-MB-231 cell treatment with AA reduces cell proliferation and migration while inducing cell death through apoptosis. The latter mostly likely occurs via mitochondria membrane depolarization and the activation of caspases-3, -8, and -9. Altogether, our results indicate that AA exerts anti-tumoral effects on MDA-MB-231 cells, without having any effect on non-tumoral breast epithelial cells, by a mechanism that is independent on the activation of Ca2+ influx via ARC channels.
Collapse
|
20
|
Caesalpinia crista coat extract protects red blood cell from sodium nitrite-induced oxidative stress and exhibits antiplatelet activity. Blood Coagul Fibrinolysis 2020; 31:293-302. [PMID: 32332278 DOI: 10.1097/mbc.0000000000000913] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
: To understand the RBC protecting efficiency and antiplatelet activity of methanolic extract of Caesalpinia crista coat (MECCC). RBC-protecting activity of MECCC was evaluated using assays, such as DPPH, level of lipid peroxidation, protein carbonyl content, superoxide dismutase and catalase as a marker of oxidative stress whereas, platelet aggregation inhibition was performed using human platelet-rich plasma (PRP). MECCC showed about 76% of DPPH-scavenging activity, with an IC50 value of 71.89 μg/ml. The MECCC reduced the level of lipid peroxidation and protein carboxylation in RBC caused by NaNO2 in a dose-dependent manner. In addition, MECCC normalized the levels of superoxide dismutase (SOD) and catalase (CAT) in oxidative stress-induced RBC in a dose-dependent manner. This suggested the protective effect of MECCC on RBC against oxidative stress. Furthermore, MECCC also exhibited mild antiplatelet activity by inhibiting both ADP and epinephrine agonists that induced platelet aggregation. The noticed inhibition percentage was found to be 28 and 23%, respectively at the concentration of 150 μg. Interestingly, MECCC did not hydrolyse the RBC suggesting its nontoxic properties. MECCC possesses protective effect of RBC against NaNO2 (10 mmol/l) induce oxidative stress and inhibits platelet aggregation.
Collapse
|
21
|
Cantonero C, Camello PJ, Abate C, Berardi F, Salido GM, Rosado JA, Redondo PC. NO1, a New Sigma 2 Receptor/TMEM97 Fluorescent Ligand, Downregulates SOCE and Promotes Apoptosis in the Triple Negative Breast Cancer Cell Lines. Cancers (Basel) 2020; 12:E257. [PMID: 31973006 PMCID: PMC7072710 DOI: 10.3390/cancers12020257] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 01/14/2020] [Accepted: 01/16/2020] [Indexed: 12/22/2022] Open
Abstract
(1) Background: The structure of the Sigma 2 receptor/TMEM97 (σ2RTMEM97) has recently been reported. (2, 3) Methods and results: We used genetic and biochemical approaches to identify the molecular mechanism downstream of σ2R/TMEM97. The novel σ2R/TMEM97 fluorescent ligand, NO1, reduced the proliferation and survival of the triple negative breast cancer cell lines (TNBC: MDA-MB-231 and MDA-MB-468 cell lines), due to NO1-induced apoptosis. Greater bioaccumulation and faster uptake of NO1 in MDA-MB-231 cells compared to MCF10A or MCF7 cell lines were also shown. Accordingly, elevated σ2R/TMEM97 expression was confirmed by Western blotting. In contrast to NO1, other σ2R/TMEM97 ligands, such as SM21 and PB28, enhanced MDA-MB-231 cell proliferation and migration. Store-operated calcium entry (SOCE) is crucial for different cancer hallmarks. Here, we show that NO1, but not other σ2R/TMEM97 ligands, reduced SOCE in MDA-MB-231 cells. Similarly, TMEM97 silencing in MDA-MB-231 cells also impaired SOCE. NO1 administration downregulated STIM1-Orai1 interaction, probably by impairing the positive regulatory effect of σ2R/TMEM97 on STIM1, as we were unable to detect interaction with Orai1. (4) Conclusion: σ2R/TMEM97 is a key protein for the survival of triple negative breast cancer cells by promoting SOCE; therefore, NO1 may become a good pharmacological tool to avoid their proliferation.
Collapse
Affiliation(s)
- Carlos Cantonero
- Department of Physiology, Phycell and FIMUL Groups, University of Extremadura, 10003 Caceres, Spain; (C.C.); (P.J.C.)
| | - Pedro Javier Camello
- Department of Physiology, Phycell and FIMUL Groups, University of Extremadura, 10003 Caceres, Spain; (C.C.); (P.J.C.)
| | - Carmen Abate
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari “Aldo Moro”, Via Orabona 4, I-70125 Bari, Italy; (C.A.); (F.B.)
| | - Francesco Berardi
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari “Aldo Moro”, Via Orabona 4, I-70125 Bari, Italy; (C.A.); (F.B.)
| | - Gines Maria Salido
- Institute of Molecular Pathology Biomarkers (IMPB) of University of Extremadura, 10003 Caceres, Spain; (G.M.S.); (J.A.R.)
| | - Juan Antonio Rosado
- Institute of Molecular Pathology Biomarkers (IMPB) of University of Extremadura, 10003 Caceres, Spain; (G.M.S.); (J.A.R.)
| | - Pedro C. Redondo
- Institute of Molecular Pathology Biomarkers (IMPB) of University of Extremadura, 10003 Caceres, Spain; (G.M.S.); (J.A.R.)
| |
Collapse
|
22
|
El Haouari M. Platelet Oxidative Stress and its Relationship with Cardiovascular Diseases in Type 2 Diabetes Mellitus Patients. Curr Med Chem 2019; 26:4145-4165. [PMID: 28982316 DOI: 10.2174/0929867324666171005114456] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 06/07/2017] [Accepted: 06/07/2017] [Indexed: 01/01/2023]
Abstract
Enhanced platelet activation and thrombosis are linked to various cardiovascular diseases (CVD). Among other mechanisms, oxidative stress seems to play a pivotal role in platelet hyperactivity. Indeed, upon stimulation by physiological agonists, human platelets generate and release several types of reactive oxygen species (ROS) such as O2 -, H2O2 or OH-, further amplifying the platelet activation response via various signalling pathways, including, formation of isoprostanes, Ca2+ mobilization and NO inactivation. Furthermore, excessive platelet ROS generation, incorporation of free radicals from environment and/or depletion of antioxidants induce pro-oxidant, pro-inflammatory and platelet hyperaggregability effects, leading to the incidence of cardiovascular events. Here, we review the current knowledge regarding the effect of oxidative stress on platelet signaling pathways and its implication in CVD such as type 2 diabetes mellitus. We also summarize the role of natural antioxidants included in vegetables, fruits and medicinal herbs in reducing platelet function via an oxidative stress-mediated mechanism.
Collapse
Affiliation(s)
- Mohammed El Haouari
- Centre Regional des Metiers de l'Education et de la Formation de Taza (CRMEF - Taza), B.P: 1178 - Taza Gare, Morocco
| |
Collapse
|
23
|
Melchinger H, Jain K, Tyagi T, Hwa J. Role of Platelet Mitochondria: Life in a Nucleus-Free Zone. Front Cardiovasc Med 2019; 6:153. [PMID: 31737646 PMCID: PMC6828734 DOI: 10.3389/fcvm.2019.00153] [Citation(s) in RCA: 125] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 10/08/2019] [Indexed: 12/19/2022] Open
Abstract
Platelets are abundant, small, anucleate circulating cells, serving many emerging pathophysiological roles beyond hemostasis; including active critical roles in thrombosis, injury response, and immunoregulation. In the absence of genomic DNA transcriptional regulation (no nucleus), platelets require strategic prepackaging of all the needed RNA and organelles from megakaryocytes, to sense stress (e.g., hyperglycemia), to protect themselves from stress (e.g., mitophagy), and to communicate a stress response to other cells (e.g., granule and microparticle release). Distinct from avian thrombocytes that have a nucleus, the absence of a nucleus allows the mammalian platelet to maintain its small size, permits morphological flexibility, and may improve speed and efficiency of protein expression in response to stress. In the absence of a nucleus, platelet lifespan of 7–10 days, is largely determined by the mitochondria. The packaging of 5–8 mitochondria is critical in aerobic respiration and yielding metabolic substrates needed for function and survival. Mitochondria damage or dysfunction, as observed with several disease processes, results in greatly attenuated platelet survival and increased risk for thrombovascular events. Here we provide insights into the emerging roles of platelets despite the lack of a nucleus, and the key role played by mitochondria in platelet function and survival both in health and disease.
Collapse
Affiliation(s)
- Hannah Melchinger
- Department of Internal Medicine, Yale Cardiovascular Research Center, Yale School of Medicine, New Haven, CT, United States
| | - Kanika Jain
- Department of Internal Medicine, Yale Cardiovascular Research Center, Yale School of Medicine, New Haven, CT, United States
| | - Tarun Tyagi
- Department of Internal Medicine, Yale Cardiovascular Research Center, Yale School of Medicine, New Haven, CT, United States
| | - John Hwa
- Department of Internal Medicine, Yale Cardiovascular Research Center, Yale School of Medicine, New Haven, CT, United States
| |
Collapse
|
24
|
Fuentes E, Araya-Maturana R, Urra FA. Regulation of mitochondrial function as a promising target in platelet activation-related diseases. Free Radic Biol Med 2019; 136:172-182. [PMID: 30625393 DOI: 10.1016/j.freeradbiomed.2019.01.007] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Revised: 12/22/2018] [Accepted: 01/04/2019] [Indexed: 12/13/2022]
Abstract
Platelets are anucleated cell elements produced by fragmentation of the cytoplasm of megakaryocytes and have a unique metabolic phenotype compared with circulating leukocytes, exhibiting a high coupling efficiency to mitochondrial adenosine triphosphate production with reduced respiratory reserve capacity. Platelet mitochondria are well suited for ex vivo analysis of different diseases. Even some diseases induce mitochondrial changes in platelets without reflecting them in other organs. During platelet activation, an integrated participation of glycolysis and oxidative phosphorylation is mediated by oxidative stress production-dependent signaling. The platelet activation-dependent procoagulant activity mediated by collagen, thrombin and hyperglycemia induce mitochondrial dysfunction to promote thrombosis in oxidative stress-associated pathological conditions. Interestingly, some compounds exhibit a protective action on platelet mitochondrial dysfunction through control of mitochondrial oxidative stress production or inhibition of respiratory complexes. They can be grouped in a) Natural source-derived compounds (e.g. Xanthohumol, Salvianoloc acid A and Sila-amide derivatives of NAC), b) TPP+-linked small molecules (e.g. mitoTEMPO and mitoQuinone) and c) FDA-approved drugs (e.g. metformin and statins), illustrating the wide range of molecular structures capable of effectively interacting with platelet mitochondria. The present review article aims to discuss the mechanisms of mitochondrial dysfunction and their association with platelet activation-related diseases.
Collapse
Affiliation(s)
- Eduardo Fuentes
- Thrombosis Research Center, Medical Technology School, Department of Clinical Biochemistry and Immunohaematology, Faculty of Health Sciences, Interdisciplinary Excellence Research Program on Healthy Aging (PIEI-ES), Universidad de Talca, Talca, Chile.
| | - Ramiro Araya-Maturana
- Instituto de Química de Recursos Naturales, Programa de Investigación Asociativa en Cáncer Gástrico (PIA-CG), Universidad de Talca, Talca, Chile
| | - Félix A Urra
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile.
| |
Collapse
|
25
|
Petrus AT, Lighezan DL, Danila MD, Duicu OM, Sturza A, Muntean DM, Ionita I. Assessment of platelet respiration as emerging biomarker of disease. Physiol Res 2019; 68:347-363. [PMID: 30904011 DOI: 10.33549/physiolres.934032] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Mitochondrial dysfunction is currently acknowledged as a central pathomechanism of most common diseases of the 21(st) century. Recently, the assessment of the bioenergetic profile of human peripheral blood cells has emerged as a novel research field with potential applications in the development of disease biomarkers. In particular, platelets have been successfully used for the ex vivo analysis of mitochondrial respiratory function in several acute and chronic pathologies. An increasing number of studies support the idea that evaluation of the bioenergetic function in circulating platelets may represent the peripheral signature of mitochondrial dysfunction in metabolically active tissues (brain, heart, liver, skeletal muscle). Accordingly, impairment of mitochondrial respiration in peripheral platelets might have potential clinical applicability as a diagnostic and prognostic tool as well as a biomarker in treatment monitoring. The aim of this minireview is to summarize current information in the field of platelet mitochondrial dysfunction in both acute and chronic diseases.
Collapse
Affiliation(s)
- A T Petrus
- Department of Anatomy, Physiology and Pathophysiology, Faculty of Pharmacy, "Victor Babes" University of Medicine and Pharmacy, Timisoara, Romania and Department of Functional Sciences - Pathophysiology, "Victor Babes" University of Medicine and Pharmacy of Timisoara, Timisoara, Romania.
| | | | | | | | | | | | | |
Collapse
|
26
|
Vishalakshi GJ, NaveenKumar SK, Hemshekhar M, Mahendra M, Kemparaju K, Girish KS. Para-tertiary butyl catechol (PTBC), an industrial antioxidant induces human platelet apoptosis. ENVIRONMENTAL TOXICOLOGY 2019; 34:262-270. [PMID: 30461186 DOI: 10.1002/tox.22681] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2018] [Revised: 10/25/2018] [Accepted: 11/03/2018] [Indexed: 06/09/2023]
Abstract
The catecholic derivative para-tertiary butyl catechol (PTBC) is a conventional antioxidant and polymerization inhibitor, which exhibits melanocytotoxic effects and contact dermatitis often leading to occupational leucoderma or vitiligo. Although numerous industrial workers will be in constant exposure to PTBC and its chances of getting entry into blood are most expected, its effect on blood components is still undisclosed. As platelets play a prominent role in dermatitis, inflammation, and immunity, in this study we have evaluated the effect of PTBC on human platelets in vitro. Exposure of platelets to PTBC showed increased reactive oxygen species (ROS), intracellular calcium, cardiolipin oxidation, mitochondrial permeability transition pore (MPTP) formation, activation of caspases, phosphatidylserine (PS) externalization and decreased mitochondrial membrane potential. In addition, there was a significant decrease in cellular glutathione level, increased γ-glutamyltransferase (GGT) activity and cell death. These findings demonstrate that PTBC could induce toxic effects on blood components, which is often ignored field of research. Since dermal exposure of humans to toxic chemicals covers an important issue in various industries, there is a need of such work to understand and update the long-term toxicities induced by PTBC usage in industrial sectors and public domain.
Collapse
Affiliation(s)
| | | | - Mahadevappa Hemshekhar
- Department of Internal Medicine, Manitoba Centre for Proteomics and Systems Biology, University of Manitoba, Winnipeg, Canada
| | | | - Kempaiah Kemparaju
- DOS in Biochemistry, University of Mysore, Manasagangothri, Mysuru, India
| | - Kesturu S Girish
- DOS in Biochemistry, University of Mysore, Manasagangothri, Mysuru, India
- Department of Studies and Research in Biochemistry, Tumkur University, Tumakuru, India
| |
Collapse
|
27
|
A new pyrrole based small molecule from Tinospora cordifolia induces apoptosis in MDA-MB-231 breast cancer cells via ROS mediated mitochondrial damage and restoration of p53 activity. Chem Biol Interact 2019; 299:120-130. [DOI: 10.1016/j.cbi.2018.12.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Revised: 11/26/2018] [Accepted: 12/07/2018] [Indexed: 12/17/2022]
|
28
|
|
29
|
Vara D, Cifuentes-Pagano E, Pagano PJ, Pula G. A novel combinatorial technique for simultaneous quantification of oxygen radicals and aggregation reveals unexpected redox patterns in the activation of platelets by different physiopathological stimuli. Haematologica 2019; 104:1879-1891. [PMID: 30679320 PMCID: PMC6717585 DOI: 10.3324/haematol.2018.208819] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 01/18/2019] [Indexed: 01/12/2023] Open
Abstract
The regulation of platelets by oxidants is critical for vascular health and may explain thrombotic complications in diseases such as diabetes and dementia, but remains poorly understood. Here, we describe a novel technique combining electron paramagnetic resonance spectroscopy and turbidimetry, which has been utilized to monitor simultaneously platelet activation and oxygen radical generation. This technique has been used to investigate the redox-dependence of human and mouse platelets. Using selective peptide inhibitors of NADPH oxidases (NOXs) on human platelets and genetically modified mouse platelets (NOX1−/− or NOX2−/−), we discovered that: 1) intracellular but not extracellular superoxide anion generated by NOX is critical for platelet activation by collagen; 2) superoxide dismutation to hydrogen peroxide is required for thrombin-dependent activation; 3) NOX1 is the main source of oxygen radicals in response to collagen, while NOX2 is critical for activation by thrombin; 4) two platelet modulators, namely oxidized low density lipoproteins (oxLDL) and amyloid peptide β (Aβ), require activation of both NOX1 and NOX2 to pre-activate platelets. This study provides new insights into the redox dependence of platelet activation. It suggests the possibility of selectively inhibiting platelet agonists by targeting either NOX1 (for collagen) or NOX2 (for thrombin). Selective inhibition of either NOX1 or NOX2 impairs the potentiatory effect of tested platelet modulators (oxLDL and Aβ), but does not completely abolish platelet hemostatic function. This information offers new opportunities for the development of disease-specific antiplatelet drugs with limited bleeding side effects by selectively targeting one NOX isoenzyme.
Collapse
Affiliation(s)
- Dina Vara
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, UK
| | - Eugenia Cifuentes-Pagano
- Department of Pharmacology and Chemical Biology and Vascular Medicine Institute, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Patrick J Pagano
- Department of Pharmacology and Chemical Biology and Vascular Medicine Institute, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Giordano Pula
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, UK
| |
Collapse
|
30
|
Korjian S, Braunwald E, Daaboul Y, Mi M, Bhatt DL, Verheugt FW, Cohen M, Bode C, Burton P, Plotnikov AN, Gibson CM. Usefulness of Rivaroxaban for Secondary Prevention of Acute Coronary Syndrome in Patients With History of Congestive Heart Failure (from the ATLAS-ACS-2 TIMI-51 Trial). Am J Cardiol 2018; 122:1896-1901. [PMID: 30340765 DOI: 10.1016/j.amjcard.2018.08.034] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 08/15/2018] [Accepted: 08/21/2018] [Indexed: 12/25/2022]
Abstract
Patients with both acute coronary syndromes (ACS) and congestive heart failure are at an increased risk of recurrent cardiovascular (CV) events attributed in part to both excess thrombin generation and impaired fibrinolysis. We hypothesized that patients with the overlap of ACS and CHF would thus derive particular benefit from antithrombotic therapy with rivaroxaban. ATLAS-ACS-2 Thrombolysis in Myocardial Infarction-51 was a double-blind, multicenter, phase 3 clinical trial that randomized patients within 7 days of an ACS event to standard of care plus either rivaroxaban 2.5 mg BID, 5 mg BID, or placebo (n = 15,526). In this post hoc subgroup analysis, subjects with a history of CHF at randomization (n = 1,694) were evaluated. Among subjects with a history of CHF, both rivaroxaban doses reduced the primary composite end point of CV death, myocardial infarction, or stroke (2.5 mg BID vs placebo: hazard ratio [HR] 0.59, 95% confidence interval [CI] (0.42, 0.81), p = 0.001; 5 mg BID vs placebo: HR 0.61, 95% CI (0.44, 0.84), p = 0.002; p interaction = 0.006). Both doses of rivaroxaban reduced CV mortality (rivaroxaban 2.5 mg BID vs placebo: 4.1% vs 9.0%, HR 0.45, 95% CI [0.27, 0.74], p = 0.002; rivaroxaban 5 mg BID vs placebo: 5.8% vs 9.0%, HR 0.62, 95% CI [0.40, 0.96], p = 0.031) as well as all-cause mortality. There was no significant increase in noncoronary artery bypass graft-related Thrombolysis in Myocardial Infarction major bleeding with either dose of rivaroxaban as compared with placebo (rivaroxaban 2.5 mg BID = 0.4% vs rivaroxaban 5 mg BID = 1.1% vs placebo = 0.5%). Rivaroxaban also did not increase either intracranial hemorrhage or fatal bleeding. In conclusion, in ACS subjects with a history of CHF, secondary prevention with rivaroxaban reduced the composite of CV death, myocardial infarction, or stroke without an increase in noncoronary artery bypass graft-related major bleeding. These findings require further prospective evaluation in an adequately powered phase 3 study.
Collapse
|
31
|
Platelet mitochondrial dysfunction and mitochondria-targeted quinone-and hydroquinone-derivatives: Review on new strategy of antiplatelet activity. Biochem Pharmacol 2018; 156:215-222. [DOI: 10.1016/j.bcp.2018.08.035] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 08/21/2018] [Indexed: 01/03/2023]
|
32
|
Ghasemzadeh M, Hosseini E, Roudsari ZO, Zadkhak P. Intraplatelet reactive oxygen species (ROS) correlate with the shedding of adhesive receptors, microvesiculation and platelet adhesion to collagen during storage: Does endogenous ROS generation downregulate platelet adhesive function? Thromb Res 2018; 163:153-161. [DOI: 10.1016/j.thromres.2018.01.048] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2017] [Revised: 01/02/2018] [Accepted: 01/26/2018] [Indexed: 01/01/2023]
|
33
|
De Silva E, Kim H. Drug-induced thrombocytopenia: Focus on platelet apoptosis. Chem Biol Interact 2018; 284:1-11. [PMID: 29410286 DOI: 10.1016/j.cbi.2018.01.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 12/23/2017] [Accepted: 01/18/2018] [Indexed: 12/16/2022]
Abstract
Thrombocytopenia is a serious and potentially fatal complication of drug therapy that results either from a decrease in bone marrow platelet production or the excessive destruction of circulating platelets. Although multiple mechanisms are responsible for deregulated platelet clearance, the role of programmed platelet death (apoptosis) in drug-induced thrombocytopenia has been relatively under-investigated until recently. Here we review apoptotic signaling pathways in platelets, with a focus on current data that provide mechanistic insights into drug-induced apoptosis and thrombocytopenia.
Collapse
Affiliation(s)
- Enoli De Silva
- Centre for Blood Research, University of British Columbia, Vancouver, Canada; Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, Canada
| | - Hugh Kim
- Centre for Blood Research, University of British Columbia, Vancouver, Canada; Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, Canada; Faculty of Dentistry, University of British Columbia, Vancouver, Canada.
| |
Collapse
|
34
|
Platelet subpopulations remain despite strong dual agonist stimulation and can be characterised using a novel six-colour flow cytometry protocol. Sci Rep 2018; 8:1441. [PMID: 29362366 PMCID: PMC5780418 DOI: 10.1038/s41598-017-19126-8] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 12/19/2017] [Indexed: 12/16/2022] Open
Abstract
It is recognised that platelets respond differently to activation, where a subpopulation of platelets adopt a procoagulant phenotype while others are aggregatory. However, it has not been thoroughly tested whether these subpopulations will remain in maximally activated samples, or if they are merely a result of different platelet sensitivities to agonist activation. Here platelets were activated with gradually increasing concentrations of thrombin and/or the GPVI agonist cross-linked collagen-related peptide (CRP-XL). Platelet activation was investigated using a novel six-colour flow cytometry protocol evaluating exposure of phosphatidylserine, active conformation of the fibrinogen receptor αIIbβ3, α-granule and lysosomal release (P-selectin and LAMP-1 exposure), mitochondrial membrane integrity and platelet fragmentation. Upon activation by CRP-XL or thrombin+CRP-XL, platelets formed three differently sized subpopulations. Normal-sized platelets showed high exposure of aggregatory active αIIbβ3 and intact mitochondria, while the smaller platelets and platelet fragments showed high exposure of procoagulant phosphatidylserine. The distribution of platelets between the differently sized subpopulations remained stable despite high agonist concentrations. All three were still present after 30 and 60 min of activation, showing that all platelets will not have the same characteristics even after maximal stimulation. This suggests that platelet subpopulations with distinct activation patterns exist within the total platelet population.
Collapse
|
35
|
Mondal NK, Li T, Chen Z, Chen HH, Sorensen EN, Pham SM, Sobieski MA, Koenig SC, Slaughter MS, Griffith BP, Wu ZJ. Mechanistic insight of platelet apoptosis leading to non-surgical bleeding among heart failure patients supported by continuous-flow left ventricular assist devices. Mol Cell Biochem 2017; 433:125-137. [PMID: 28343311 DOI: 10.1007/s11010-017-3021-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 03/21/2017] [Indexed: 11/24/2022]
Abstract
Non-surgical bleeding (NSB) is the most common clinical complication in heart failure (HF) patients supported by continuous-flow left ventricular assist devices (CF-LVADs). In this study, oxidative stress and alteration of signal pathways leading to platelet apoptosis were investigated. Thirty-one HF patients supported by CF-LVADs were divided into bleeder (n = 12) and non-bleeder (n = 19) groups. Multiple blood samples were collected at pre-implant (baseline) and weekly up to 1-month post-implant. A single blood sample was collected from healthy subjects (reference). Production of reactive oxygen species (ROS) in platelets, total antioxidant capacity (TAC), oxidized low-density lipoproteins (oxLDL), expression of Bcl-2 and Bcl-xL, Bax and release of cytochrome c (Cyt.c), platelet mitochondrial membrane potential (Δψ m), activation of caspases, gelsolin cleavage and platelet apoptosis were examined. Significantly elevated ROS, oxLDL and depleted TAC were evident in the bleeder group compared to non-bleeder group (p < 0.05). Platelet pro-survival proteins (Bcl-2, Bcl-xL) were significantly reduced in the bleeder group in comparison to the non-bleeder group (p < 0.05). Translocation of Bax into platelet mitochondria membrane and subsequent release of Cyt.c were more prevalent in the bleeder group. Platelet mitochondrial damage, activation of caspases, gelsolin cleavage, and ultimate platelet apoptosis in the bleeder group were observed. Oxidative stress and activation of both intrinsic and extrinsic pathways of platelet apoptosis may be linked to NSB in CF-LVAD patients. Additionally, biomarkers of oxidative stress, examination of pro-survivals and pro-apoptotic proteins in platelets, mitochondrial damage, caspase activation, and platelet apoptosis may be used to help identify HF patients at high risk of NSB post-implant.
Collapse
Affiliation(s)
- Nandan K Mondal
- Department of Cardiovascular and Thoracic Surgery, Cardiovascular Innovation Institute, University of Louisville School of Medicine, Louisville, KY, USA.,Department of Surgery, Artificial Organs Laboratory, University of Maryland School of Medicine, 10 South Pine Street, MSTF 434A, Baltimore, MD, 21201, USA
| | - Tieluo Li
- Department of Surgery, Artificial Organs Laboratory, University of Maryland School of Medicine, 10 South Pine Street, MSTF 434A, Baltimore, MD, 21201, USA
| | - Zengsheng Chen
- Department of Cardiovascular and Thoracic Surgery, Cardiovascular Innovation Institute, University of Louisville School of Medicine, Louisville, KY, USA
| | - Hegang H Chen
- Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Erik N Sorensen
- Department of Clinical Engineering, University of Maryland Medical Center, Baltimore, MD, USA
| | - Si M Pham
- Department of Surgery, Artificial Organs Laboratory, University of Maryland School of Medicine, 10 South Pine Street, MSTF 434A, Baltimore, MD, 21201, USA
| | - Michael A Sobieski
- Department of Cardiovascular and Thoracic Surgery, Cardiovascular Innovation Institute, University of Louisville School of Medicine, Louisville, KY, USA
| | - Steven C Koenig
- Department of Cardiovascular and Thoracic Surgery, Cardiovascular Innovation Institute, University of Louisville School of Medicine, Louisville, KY, USA
| | - Mark S Slaughter
- Department of Cardiovascular and Thoracic Surgery, Cardiovascular Innovation Institute, University of Louisville School of Medicine, Louisville, KY, USA
| | - Bartley P Griffith
- Department of Surgery, Artificial Organs Laboratory, University of Maryland School of Medicine, 10 South Pine Street, MSTF 434A, Baltimore, MD, 21201, USA
| | - Zhongjun J Wu
- Department of Cardiovascular and Thoracic Surgery, Cardiovascular Innovation Institute, University of Louisville School of Medicine, Louisville, KY, USA. .,Department of Surgery, Artificial Organs Laboratory, University of Maryland School of Medicine, 10 South Pine Street, MSTF 434A, Baltimore, MD, 21201, USA.
| |
Collapse
|
36
|
Chen Z, Schubert P, Bakkour S, Culibrk B, Busch MP, Devine DV. p38 mitogen-activated protein kinase regulates mitochondrial function and microvesicle release in riboflavin- and ultraviolet light-treated apheresis platelet concentrates. Transfusion 2017; 57:1199-1207. [DOI: 10.1111/trf.14035] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 12/03/2016] [Accepted: 12/31/2016] [Indexed: 12/11/2022]
Affiliation(s)
- Zhongming Chen
- Centre for Innovation; Canadian Blood Services; Vancouver British Columbia Canada
- Centre for Blood Research; University of British Columbia; Vancouver British Columbia Canada
| | - Peter Schubert
- Centre for Innovation; Canadian Blood Services; Vancouver British Columbia Canada
- Centre for Blood Research; University of British Columbia; Vancouver British Columbia Canada
- Department of Pathology and Laboratory Medicine; University of British Columbia; Vancouver British Columbia Canada
| | - Sonia Bakkour
- Blood Systems Research Institute; University of California; San Francisco California
| | - Brankica Culibrk
- Centre for Innovation; Canadian Blood Services; Vancouver British Columbia Canada
- Centre for Blood Research; University of British Columbia; Vancouver British Columbia Canada
| | - Michael P. Busch
- Blood Systems Research Institute; University of California; San Francisco California
- Department of Laboratory Medicine; University of California; San Francisco California
| | - Dana V. Devine
- Centre for Innovation; Canadian Blood Services; Vancouver British Columbia Canada
- Centre for Blood Research; University of British Columbia; Vancouver British Columbia Canada
- Department of Pathology and Laboratory Medicine; University of British Columbia; Vancouver British Columbia Canada
| |
Collapse
|
37
|
Zhu J, Wang Q, Nie Y, Yan R, Dai K, Zhou B. Platelet Integrin αIIbβ3 Inhibitor Rescues Progression of Apoptosis in Human Platelets. Med Sci Monit 2016; 22:4261-4270. [PMID: 27827357 PMCID: PMC5108368 DOI: 10.12659/msm.900820] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Apoptosis plays an important role in the physiology of platelet function. We aimed to detect the effect of the platelet integrin αIIbβ3 inhibitor, tirofiban, on apoptotic events, including mitochondrial inner-membrane potential (ΔΨm), phosphatidylserine (PS) exposure on platelet surface, and the generation of reactive oxygen species (ROS), when washed platelets were stimulated with thrombin. MATERIAL AND METHODS The study included washed platelets from healthy humans, divided into 4 groups: vehicle, and tirofiban (0.05 μg/ml, 0.25 μg/ml, and 0.5 μg/ml). Platelets were pretreated with vehicle or tirofiban and incubated at 37°C with agitation for 6 h and 24 h. Before thrombin addition, the vehicle group divided into 2 equal groups. Except one vehicle group, the other 4 groups were all stimulated with thrombin (1 U/ml) for 30 min at 37°C. Using flow cytometry, we studied the DYm and PS exposure on platelet surfaces, and the generation of ROS in platelets. RESULTS We observed that at the time of 6 h and 24 h, thrombin-stimulated vehicle platelets induced significant depo-larization of ΔΨm, higher PS exposure, and increased ROS production compared with the vehicle group (P<0.01). However, the tirofiban group had significantly more recovery of DYm, PS exposure, and ROS production compared with the thrombin group (P<0.01). CONCLUSIONS The platelet integrin αIIbβ3 inhibitor, tirofiban, inhibits the depolarization of DYm, PS exposure on platelet surface, and ROS production when stimulated with thrombin. These results suggest that αIIbβ3 inhibitor inhibits the initiation of apoptosis in platelets, showing a potential clinical application of tirofiban as an apoptosis inhibitor.
Collapse
Affiliation(s)
- Jie Zhu
- Department of Cardiology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China (mainland)
| | - Qinghang Wang
- Department of Cardiology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China (mainland)
| | - Yumei Nie
- Department of Cardiology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China (mainland)
| | - Rong Yan
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Collaborative Innovation Center of Hematology, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, Suzhou, Jiangsu, China (mainland)
| | - Kesheng Dai
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Collaborative Innovation Center of Hematology, Key Laboratory of Thrombosis and Hemostasis, Ministry of Healt, Suzhou, Jiangsu, China (mainland)
| | - Birong Zhou
- Department of Cardiology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China (mainland)
| |
Collapse
|
38
|
El Haouari M, Rosado JA. Medicinal Plants with Antiplatelet Activity. Phytother Res 2016; 30:1059-71. [PMID: 27062716 DOI: 10.1002/ptr.5619] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 02/29/2016] [Accepted: 03/12/2016] [Indexed: 12/25/2022]
Abstract
Blood platelets play an essential role in the hemostasis and wound-healing processes. However, platelet hyperactivity is associated to the development and the complications of several cardiovascular diseases. In this sense, the search for potent and safer antiplatelet agents is of great interest. This article provides an overview of experimental studies performed on medicinal plants with antiplatelet activity available through literature with particular emphasis on the bioactive constituents, the parts used, and the various platelet signaling pathways modulated by medicinal plants. From this review, it was suggested that medicinal plants with antiplatelet activity mainly belong to the family of Asteraceae, Rutaceae, Fabaceae, Lamiaceae, Zygophyllaceae, Rhamnaceae, Liliaceae, and Zingiberaceae. The antiplatelet effect is attributed to the presence of bioactive compounds such as polyphenols, flavonoids, coumarins, terpenoids, and other substances which correct platelet abnormalities by interfering with different platelet signalization pathways including inhibition of the ADP pathway, suppression of TXA2 formation, reduction of intracellular Ca(2+) mobilization, and phosphoinositide breakdown, among others. The identification and/or structure modification of the plant constituents and the understanding of their action mechanisms will be helpful in the development of new antiplatelet agents based on medicinal plants which could contribute to the prevention of thromboembolic-related disorders by inhibiting platelet aggregation. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Mohammed El Haouari
- Centre Régional des Métiers de l'Education et de la Formation de Taza (CRMEF - Taza), B.P. 1178, Taza Gare, Morocco.,Faculté Polydisciplinaire de Taza, Laboratoire des Matériaux, Substances Naturelles, Environnement et Modélisation (LMSNEM), Université Sidi Mohamed Ben Abdellah, B.P. 1223, Taza Gare, Morocco
| | - Juan A Rosado
- Department of Physiology (Cell Physiology Research Group), University of Extremadura, 10003, Cáceres, Spain
| |
Collapse
|
39
|
Bynum JA, Adam Meledeo M, Getz TM, Rodriguez AC, Aden JK, Cap AP, Pidcoke HF. Bioenergetic profiling of platelet mitochondria during storage: 4°C storage extends platelet mitochondrial function and viability. Transfusion 2016; 56 Suppl 1:S76-84. [DOI: 10.1111/trf.13337] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- James A. Bynum
- US Army Institute of Surgical Research; JBSA-Fort Sam Houston Texas
| | - M. Adam Meledeo
- US Army Institute of Surgical Research; JBSA-Fort Sam Houston Texas
| | - Todd M. Getz
- US Army Institute of Surgical Research; JBSA-Fort Sam Houston Texas
| | | | - James K. Aden
- US Army Institute of Surgical Research; JBSA-Fort Sam Houston Texas
| | - Andrew P. Cap
- US Army Institute of Surgical Research; JBSA-Fort Sam Houston Texas
| | | |
Collapse
|
40
|
Platelet protective efficacy of 3,4,5 trisubstituted isoxazole analogue by inhibiting ROS-mediated apoptosis and platelet aggregation. Mol Cell Biochem 2016; 414:137-51. [DOI: 10.1007/s11010-016-2667-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 02/11/2016] [Indexed: 12/23/2022]
|
41
|
Comparison of intraplatelet reactive oxygen species, mitochondrial damage, and platelet apoptosis after implantation of three continuous flow left ventricular assist devices: HeartMate II, Jarvik 2000, and HeartWare. ASAIO J 2016; 61:244-52. [PMID: 25757140 DOI: 10.1097/mat.0000000000000208] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Differences in device design may have an effect on platelet damage and associated clinical complications. We aimed to compare device-specific platelet functionality in 26 heart failure patients supported with three continuous-flow left ventricular assist devices: HeartMate II (n = 8), Jarvik 2000 (n = 9), and HeartWare (n = 9). Intraplatelet reactive oxygen species (ROS) generation, mitochondrial damage, and platelet apoptosis were compared between device types before and after the implantation at every week up to 1 month. Overall, the baseline characteristics, demographics, routine laboratory values were comparable between the three device groups. Intraplatelet ROS, mitochondrial damage, and platelet apoptosis significantly elevated in the HeartWare group in comparison with the other two device groups after implantation. The major bleeding, infections, systemic inflammatory response syndrome, and right ventricular failure were found to be more common among the HeartWare group than others. Intraplatelet ROS and platelet damage levels were returned to baseline in both the HeartMate II and the Jarvik groups, whereas in HeartWare group they remained elevated. The patients with the Jarvik and the HeartMate II experienced less clinical complications and the platelet functionality is not compromised by these devices. Data from this study suggests that the continuous-flow left ventricular assist devices design may exert different effects on platelet function.
Collapse
|
42
|
Knez J, Winckelmans E, Plusquin M, Thijs L, Cauwenberghs N, Gu Y, Staessen JA, Nawrot TS, Kuznetsova T. Correlates of Peripheral Blood Mitochondrial DNA Content in a General Population. Am J Epidemiol 2016; 183:138-46. [PMID: 26702630 PMCID: PMC4706678 DOI: 10.1093/aje/kwv175] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 06/24/2015] [Indexed: 12/19/2022] Open
Abstract
Accumulation of mitochondrial DNA (mtDNA) mutations leads to alterations of mitochondrial biogenesis and function that might produce a decrease in mtDNA content within cells. This implies that mtDNA content might be a potential biomarker associated with oxidative stress and inflammation. However, data on correlates of mtDNA content in a general population are sparse. Our goal in the present study was to describe in a randomly recruited population sample the distribution and determinants of peripheral blood mtDNA content. From 2009 to 2013, we examined 689 persons (50.4% women; mean age = 54.4 years) randomly selected from a Flemish population (Flemish Study on Environment, Genes, and Health Outcomes). Relative mtDNA copy number as compared with nuclear DNA was measured by quantitative real-time polymerase chain reaction in peripheral blood. There was a curvilinear relationship between relative mtDNA copy number and age. mtDNA content slightly increased until the fifth decade of life and declined in older subjects (Page2 = 0.0002). mtDNA content was significantly higher in women (P = 0.007) and increased with platelet count (P < 0.0001), whereas it was inversely associated with white blood cell count (P < 0.0001). We also observed lower mtDNA content in women using estroprogestogens (P = 0.044). This study demonstrated in a general population that peripheral blood mtDNA content is significantly associated with sex and age. Blood mtDNA content is also influenced by platelet and white blood cell counts and estroprogestogen intake. Further studies are required to clarify the impact of chronic inflammation and hormone therapy on mitochondrial function.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Tatiana Kuznetsova
- Correspondence to Dr. Tatiana Kuznetsova, Hypertension and Cardiovascular Epidemiology Research Unit, Department of Cardiovascular Sciences, Biomedical Sciences Group, University of Leuven, Campus Sint Rafaël, Kapucijnenvoer 35, Box 7001, B 3000 Leuven, Belgium (e-mail: )
| |
Collapse
|
43
|
Hargrave B, Li F. Nanosecond Pulse Electric Field Activated-Platelet Rich Plasma Enhances the Return of Blood Flow to Large and Ischemic Wounds in a Rabbit Model. Physiol Rep 2015. [PMID: 26197934 PMCID: PMC4552537 DOI: 10.14814/phy2.12461] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Platelet-rich plasma is a therapeutic strategy used for accelerating wound healing of a wide range of tissues through the release of platelet growth factors. Here, we describe a nonchemical, safe method for preparing platelet-rich plasma using nanosecond-pulsed electric fields (nsPEFs) and investigated the effect of this platelet-rich plasma on reperfusion of blood in large skin flap or ischemic hind limb wounds in New Zealand White rabbits. Laser Doppler images of blood flow to the dorsal surface of skin flap wounds or to ischemic hind limb wounds were obtained from wounds treated with 0.9% saline or nanosecond-pulsed electric field prepared platelet-rich plasma (nsPRP). Reperfusion in the skin flap wounds was greater in the nsPRP-treated wounds than in the wounds treated with saline on postoperative days 3 (P < 0.001) and 21 (P < 0.03). Reperfusion in the ischemic hind-limb treated with nsPRP was greater than in the saline-treated limb on post-operative Day 3 (P < 0.001), post-operative week 1 (P < 0.025) and post-operative week 4 (P < 0.015). In the hind limb ischemic tissue, the number of endothelial cells, collagen, and cells containing vascular endothelial growth factor (VEGF) was greater in the nsPRP-treated tissue. These results demonstrate that nsPRP improves blood flow in large surgical skin wounds and in ischemic wounds.
Collapse
Affiliation(s)
- Barbara Hargrave
- Department of Medical Diagnostics and Translational Science, Old Dominion University, Norfolk, Virginia, USA Frank Reidy Center for Bioelectrics, Old Dominion University, Norfolk, Virginia, USA
| | - Francis Li
- Frank Reidy Center for Bioelectrics, Old Dominion University, Norfolk, Virginia, USA
| |
Collapse
|
44
|
Methotrexate Promotes Platelet Apoptosis via JNK-Mediated Mitochondrial Damage: Alleviation by N-Acetylcysteine and N-Acetylcysteine Amide. PLoS One 2015; 10:e0127558. [PMID: 26083398 PMCID: PMC4471342 DOI: 10.1371/journal.pone.0127558] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2015] [Accepted: 04/16/2015] [Indexed: 12/26/2022] Open
Abstract
Thrombocytopenia in methotrexate (MTX)-treated cancer and rheumatoid arthritis (RA) patients connotes the interference of MTX with platelets. Hence, it seemed appealing to appraise the effect of MTX on platelets. Thereby, the mechanism of action of MTX on platelets was dissected. MTX (10 μM) induced activation of pro-apoptotic proteins Bid, Bax and Bad through JNK phosphorylation leading to ΔΨm dissipation, cytochrome c release and caspase activation, culminating in apoptosis. The use of specific inhibitor for JNK abrogates the MTX-induced activation of pro-apoptotic proteins and downstream events confirming JNK phosphorylation by MTX as a key event. We also demonstrate that platelet mitochondria as prime sources of ROS which plays a central role in MTX-induced apoptosis. Further, MTX induces oxidative stress by altering the levels of ROS and glutathione cycle. In parallel, the clinically approved thiol antioxidant N-acetylcysteine (NAC) and its derivative N-acetylcysteine amide (NACA) proficiently alleviate MTX-induced platelet apoptosis and oxidative damage. These findings underpin the dearth of research on interference of therapeutic drugs with platelets, despite their importance in human health and disease. Therefore, the use of antioxidants as supplementary therapy seems to be a safe bet in pathologies associated with altered platelet functions.
Collapse
|
45
|
Kumari S, Chaurasia SN, Nayak MK, Mallick RL, Dash D. Sirtuin Inhibition Induces Apoptosis-like Changes in Platelets and Thrombocytopenia. J Biol Chem 2015; 290:12290-9. [PMID: 25829495 DOI: 10.1074/jbc.m114.615948] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Indexed: 12/16/2022] Open
Abstract
Sirtuins are evolutionarily conserved NAD(+)-dependent acetyl-lysine deacetylases that belong to class III type histone deacetylases. In humans, seven sirtuin isoforms (Sirt1 to Sirt7) have been identified. Sirtinol, a cell-permeable lactone ring derived from naphthol, is a dual Sirt1/Sirt2 inhibitor of low potency, whereas EX-527 is a potent and selective Sirt1 inhibitor. Here we demonstrate that Sirt1, Sirt2, and Sirt3 are expressed in enucleate platelets. Both sirtinol and EX-527 induced apoptosis-like changes in platelets, as revealed by enhanced annexin V binding, reactive oxygen species production, and drop in mitochondrial transmembrane potential. These changes were associated with increased phagocytic clearance of the platelets by macrophages. Expression of acetylated p53 and the conformationally active form of Bax were found to be significantly higher in both sirtinol- and EX-527-treated platelets, implicating the p53-Bax axis in apoptosis induced by sirtuin inhibitors. Administration of either sirtinol or EX-527 in mice led to a reduction in both platelet count and the number of reticulated platelets. Our results, for the first time, implicate sirtuins as a central player in the determination of platelet aging. Because sirtuin inhibitors are being evaluated for their antitumor activity, this study refocuses attention on the potential side effect of sirtuin inhibition in delimiting platelet life span and management of thrombosis.
Collapse
Affiliation(s)
- Sharda Kumari
- From the Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| | - Susheel N Chaurasia
- From the Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| | - Manasa K Nayak
- From the Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| | - Ram L Mallick
- From the Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| | - Debabrata Dash
- From the Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| |
Collapse
|
46
|
Wachowicz B. Blood Platelet as a Peripheral Cell in Oxidative Stress in Psychiatric Disorders. OXIDATIVE STRESS IN APPLIED BASIC RESEARCH AND CLINICAL PRACTICE 2015. [DOI: 10.1007/978-1-4939-0440-2_16] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
47
|
Signaling mechanism of protease activated receptor 1-induced proliferation of astrocytes: Stabilization of hypoxia inducible factor-1α triggers glucose metabolism and accumulation of cyclin D1. Neurochem Int 2014; 79:20-32. [DOI: 10.1016/j.neuint.2014.09.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Revised: 09/24/2014] [Accepted: 09/25/2014] [Indexed: 02/07/2023]
|
48
|
Kumari S, Chaurasia SN, Kumar K, Dash D. Anti-apoptotic role of sonic hedgehog on blood platelets. Thromb Res 2014; 134:1311-5. [DOI: 10.1016/j.thromres.2014.09.022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Revised: 08/23/2014] [Accepted: 09/20/2014] [Indexed: 01/12/2023]
|
49
|
A new ibuprofen derivative inhibits platelet aggregation and ROS mediated platelet apoptosis. PLoS One 2014; 9:e107182. [PMID: 25238069 PMCID: PMC4169656 DOI: 10.1371/journal.pone.0107182] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Accepted: 08/06/2014] [Indexed: 12/17/2022] Open
Abstract
Thrombocytopenia is a serious issue connected with the pathogenesis of several human diseases including chronic inflammation, arthritis, Alzheimer's disease, cardiovascular diseases (CVDs) and other oxidative stress-associated pathologies. The indiscriminate use of antibiotics and other biological drugs are reported to result in thrombocytopenia, which is often neglected during the treatment regime. In addition, augmented oxidative stress induced by drugs and pathological conditions has also been shown to induce thrombocytopenia, which seems to be the most obvious consequence of elevated rate of platelet apoptosis. Thus, blocking oxidative stress-induced platelet apoptosis would be of prime importance in order to negotiate thrombocytopenia and associated human pathologies. The current study presents the synthesis and platelet protective nature of novel ibuprofen derivatives. The potent anti-oxidant ibuprofen derivative 4f was selected for the study and the platelet protective efficacy and platelet aggregation inhibitory property has been demonstrated. The compound 4f dose dependently mitigates the oxidative stress-induced platelet apoptosis in both platelet rich plasma and washed platelets. The platelet protective nature of compound 4f was determined by assessing various apoptotic markers such as ROS generation, cytosolic Ca2+ levels, PS externalization, cytochrome C translocation, Caspase activation, mitochondrial membrane depolarization, cytotoxicity, LDH leakage and tyrosine phosphorylation of cytosolic proteins. Furthermore, compound 4f dose dependently ameliorated agonist induced platelet aggregation. Therefore, compound 4f can be estimated as a potential candidate in the treatment regime of pathological disorders associated with platelet activation and apoptosis. In addition, compound 4f can be used as an auxiliary therapeutic agent in pathologies associated with thrombocytopenia.
Collapse
|
50
|
Mondal NK, Sorensen EN, Hiivala NJ, Feller ED, Pham SM, Griffith BP, Wu ZJ. Intraplatelet reactive oxygen species, mitochondrial damage and platelet apoptosis augment non-surgical bleeding in heart failure patients supported by continuous-flow left ventricular assist device. Platelets 2014; 26:536-44. [PMID: 25167344 DOI: 10.3109/09537104.2014.948840] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Non-surgical bleeding (NSB) is the most common clinical complication among heart failure (HF) patients supported by continuous-flow left ventricular assist devices (CF-LVADs). Understanding the role of platelet functionality contributing to NSB after CF-LVAD implantation is crucial for prevention and management of this adverse event. The aim of this study was to examine the role of intraplatelet reactive oxygen species (ROS) and platelet damage on the incidence of bleeding events after CF-LVAD implantation in HF patients. We recruited 25 HF patients implanted with CF-LVADs and 11 healthy volunteers as the control. Intraplatelet ROS generation, platelet mitochondrial damage and platelet apoptosis were quantified by flow cytometry. Among 25 patients, 8 patients developed non-surgical bleeding within one month after CF-LVAD implantation. Intraplatelet ROS, depolarized and apoptotic platelet were found to be pre-existing conditions in all baseline samples of the 25 HF patients when compared to the healthy volunteers. There was no significant difference in the levels of ROS between the non-bleeder and the bleeder groups prior to CF-LVAD implantation, although we noticed 2-fold and 1.5-fold rise in depolarized and apoptotic platelets, respectively, in the bleeder group compared to those in the non-bleeder group. Post implant levels of intraplatelet ROS, depolarized and apoptotic platelets increased and remained elevated in the bleeder group, whereas periodic decreases were noticed in the non-bleeder group, suggesting the potential role of platelet damage on bleeding incidence. ROS generation after CF-LVAD implantation positively associated with platelet apoptosis (ρ = 0.4263, p = 0.0023) and depolarized platelets (ρ = 0.4774, p = 0.0002), especially the latter. In conclusion, elevated intraplatelet ROS and platelet damage may be linked to the NSB among HF patients supported by CF-LVAD. These results provide mechanistic insights into the bleeding complication in patients with CF-LVAD support.
Collapse
Affiliation(s)
- Nandan K Mondal
- Department of Surgery, Artificial Organs Laboratory, University of Maryland School of Medicine , Baltimore, Maryland , USA
| | | | | | | | | | | | | |
Collapse
|