1
|
Liu W, Li G, Shi J, Gao Y, Fang P, Zhao Y, Zhong F, Guo X, Lyu Y, Da X, Li Z, Fa J, Hu L, Yuan A, Chen L, Liu J, Chen AF, Sheng B, Ji Y, Lu X, Pu J. NR4A1 Acts as a Novel Regulator of Platelet Activation and Thrombus Formation. Circ Res 2025; 136:809-826. [PMID: 40035146 PMCID: PMC11984555 DOI: 10.1161/circresaha.124.325645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 02/16/2025] [Accepted: 02/19/2025] [Indexed: 03/05/2025]
Abstract
BACKGROUND Mounting evidence indicates that nuclear receptors play a critical regulatory role in platelet pathophysiology and thrombotic disorders. Although NR4A (the nuclear receptor subfamily 4 group A) plays an important role in cardiovascular pathophysiology, the expression profile and biological function of NR4A member 1 (NR4A1) in platelets have never been reported. METHODS We evaluated the functions and the underlying mechanisms of NR4A1 in platelet activation and thrombus formation using platelet-specific NR4A1-deficient mice and NR4A1-specific agonists. Using a hyperlipidemic mouse model and platelets from patients with hypercholesterolemia, we explored the influence of hypercholesterolemia on NR4A1 expression and the effects of NR4A1-specific agonists on platelet hyperreactivity induced by hypercholesterolemia. RESULTS NR4A1 was expressed in both human and mouse platelets. Platelet-specific NR4A1 deletion accelerated FeCl3-induced carotid arterial occlusive thrombus formation, enhanced collagen/epinephrine-induced pulmonary thromboembolism, and exacerbated microvascular microthrombi obstruction and infarct expansion in an acute myocardial infarction model. NR4A1-deficient platelets exhibited enhanced agonist-induced aggregation responses, integrin αIIbβ3 activation, dense granule release, α-granule release, platelet spreading, and clot retraction. Consistently, pharmacological activation of NR4A1 by specific agonists decreased platelet activation in both mouse and human platelets. Mechanistically, CAP1 (adenylyl cyclase-associated protein 1) was identified as the direct downstream interacting protein of NR4A1. NR4A1 deletion decreased cAMP levels and phosphorylation of VASP (vasodilator-stimulated phosphoprotein), while NR4A1-specific agonists increased cAMP levels and phosphorylation of VASP in platelets. Importantly, NR4A1 expression in platelets was upregulated in the setting of hypercholesterolemia, which was derived from its upregulation in megakaryocytes in a reactive oxygen species-dependent manner. Platelets from hypercholesterolemic patients and mice exhibited hyperreactivity. However, NR4A1-specific agonists significantly inhibited the activation of hypercholesterolemic platelets to the levels of healthy control platelets. CONCLUSIONS We provide the first evidence that nuclear receptor NR4A1 negatively regulates platelet activation and thrombus formation. NR4A1 may serve as a novel therapeutic target for managing thrombosis-based cardiovascular diseases, especially with hypercholesterolemia.
Collapse
MESH Headings
- Animals
- Nuclear Receptor Subfamily 4, Group A, Member 1/genetics
- Nuclear Receptor Subfamily 4, Group A, Member 1/deficiency
- Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism
- Nuclear Receptor Subfamily 4, Group A, Member 1/agonists
- Nuclear Receptor Subfamily 4, Group A, Member 1/blood
- Platelet Activation/physiology
- Humans
- Thrombosis/metabolism
- Thrombosis/blood
- Thrombosis/genetics
- Blood Platelets/metabolism
- Mice
- Mice, Knockout
- Mice, Inbred C57BL
- Male
- Hypercholesterolemia/blood
- Hypercholesterolemia/genetics
- Female
- Disease Models, Animal
Collapse
Affiliation(s)
- Wenhua Liu
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute (W.L., G.L., J.S., Y.G., P.F., Y.Z., F.Z., X.G., Y.L., X.D., Z.L., J.F., L.H., A.Y., L.C., J.L., X.L., J.P.), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Gaoxiang Li
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute (W.L., G.L., J.S., Y.G., P.F., Y.Z., F.Z., X.G., Y.L., X.D., Z.L., J.F., L.H., A.Y., L.C., J.L., X.L., J.P.), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Jianfeng Shi
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute (W.L., G.L., J.S., Y.G., P.F., Y.Z., F.Z., X.G., Y.L., X.D., Z.L., J.F., L.H., A.Y., L.C., J.L., X.L., J.P.), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Yu Gao
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute (W.L., G.L., J.S., Y.G., P.F., Y.Z., F.Z., X.G., Y.L., X.D., Z.L., J.F., L.H., A.Y., L.C., J.L., X.L., J.P.), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Peiliang Fang
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute (W.L., G.L., J.S., Y.G., P.F., Y.Z., F.Z., X.G., Y.L., X.D., Z.L., J.F., L.H., A.Y., L.C., J.L., X.L., J.P.), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Yichao Zhao
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute (W.L., G.L., J.S., Y.G., P.F., Y.Z., F.Z., X.G., Y.L., X.D., Z.L., J.F., L.H., A.Y., L.C., J.L., X.L., J.P.), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Fangyuan Zhong
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute (W.L., G.L., J.S., Y.G., P.F., Y.Z., F.Z., X.G., Y.L., X.D., Z.L., J.F., L.H., A.Y., L.C., J.L., X.L., J.P.), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Xiao Guo
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute (W.L., G.L., J.S., Y.G., P.F., Y.Z., F.Z., X.G., Y.L., X.D., Z.L., J.F., L.H., A.Y., L.C., J.L., X.L., J.P.), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Yuyan Lyu
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute (W.L., G.L., J.S., Y.G., P.F., Y.Z., F.Z., X.G., Y.L., X.D., Z.L., J.F., L.H., A.Y., L.C., J.L., X.L., J.P.), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Xingwen Da
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute (W.L., G.L., J.S., Y.G., P.F., Y.Z., F.Z., X.G., Y.L., X.D., Z.L., J.F., L.H., A.Y., L.C., J.L., X.L., J.P.), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Zhaoyan Li
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute (W.L., G.L., J.S., Y.G., P.F., Y.Z., F.Z., X.G., Y.L., X.D., Z.L., J.F., L.H., A.Y., L.C., J.L., X.L., J.P.), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Jingjing Fa
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute (W.L., G.L., J.S., Y.G., P.F., Y.Z., F.Z., X.G., Y.L., X.D., Z.L., J.F., L.H., A.Y., L.C., J.L., X.L., J.P.), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
- Baoshan Branch (J.F.), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Liuhua Hu
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute (W.L., G.L., J.S., Y.G., P.F., Y.Z., F.Z., X.G., Y.L., X.D., Z.L., J.F., L.H., A.Y., L.C., J.L., X.L., J.P.), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Ancai Yuan
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute (W.L., G.L., J.S., Y.G., P.F., Y.Z., F.Z., X.G., Y.L., X.D., Z.L., J.F., L.H., A.Y., L.C., J.L., X.L., J.P.), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Lei Chen
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute (W.L., G.L., J.S., Y.G., P.F., Y.Z., F.Z., X.G., Y.L., X.D., Z.L., J.F., L.H., A.Y., L.C., J.L., X.L., J.P.), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Junling Liu
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute (W.L., G.L., J.S., Y.G., P.F., Y.Z., F.Z., X.G., Y.L., X.D., Z.L., J.F., L.H., A.Y., L.C., J.L., X.L., J.P.), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
- Department of Biochemistry and Molecular Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education (J.L.), Shanghai Jiao Tong University School of Medicine, China
| | - Alex F. Chen
- Department of Cardiology and Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital (A.F.C.), Shanghai Jiao Tong University School of Medicine, China
| | - Bin Sheng
- Department of Computer Science and Engineering, Shanghai Jiao Tong University, China (B.S.)
| | - Yong Ji
- Key Laboratory of Drug Targets and Translational Medicine for Cardio-cerebrovascular Diseases, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Jiangsu, China(Y.J.)
- State Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Heilongjiang, China (Y.J.)
| | - Xiyuan Lu
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute (W.L., G.L., J.S., Y.G., P.F., Y.Z., F.Z., X.G., Y.L., X.D., Z.L., J.F., L.H., A.Y., L.C., J.L., X.L., J.P.), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Jun Pu
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute (W.L., G.L., J.S., Y.G., P.F., Y.Z., F.Z., X.G., Y.L., X.D., Z.L., J.F., L.H., A.Y., L.C., J.L., X.L., J.P.), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| |
Collapse
|
2
|
Piazza A, Carlone R, Spencer GE. Non-canonical retinoid signaling in neural development, regeneration and synaptic function. Front Mol Neurosci 2024; 17:1371135. [PMID: 38516042 PMCID: PMC10954794 DOI: 10.3389/fnmol.2024.1371135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 02/21/2024] [Indexed: 03/23/2024] Open
Abstract
Canonical retinoid signaling via nuclear receptors and gene regulation is critical for the initiation of developmental processes such as cellular differentiation, patterning and neurite outgrowth, but also mediates nerve regeneration and synaptic functions in adult nervous systems. In addition to canonical transcriptional regulation, retinoids also exert rapid effects, and there are now multiple lines of evidence supporting non-canonical retinoid actions outside of the nucleus, including in dendrites and axons. Together, canonical and non-canonical retinoid signaling provide the precise temporal and spatial control necessary to achieve the fine cellular coordination required for proper nervous system function. Here, we examine and discuss the evidence supporting non-canonical actions of retinoids in neural development and regeneration as well as synaptic function, including a review of the proposed molecular mechanisms involved.
Collapse
Affiliation(s)
| | | | - Gaynor E. Spencer
- Department of Biological Sciences, Brock University, St. Catharines, ON, Canada
| |
Collapse
|
3
|
Lin XY, Chu Y, Zhang GS, Zhang HL, Kang K, Wu MX, Zhu J, Xu CS, Lin JX, Huang CK, Chai DJ. Retinoid X receptor agonists alleviate fibroblast activation and post-infarction cardiac remodeling via inhibition of TGF-β1/Smad pathway. Life Sci 2023; 329:121936. [PMID: 37453576 DOI: 10.1016/j.lfs.2023.121936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 07/10/2023] [Accepted: 07/10/2023] [Indexed: 07/18/2023]
Abstract
Retinoid X receptor (RXR), particularly RXRα, has been implicated in cardiovascular diseases. However, the functional role of RXR activation in myocardial infarction (MI) remains unclear. This study aimed to determine the effects of RXR agonists on MI and to dissect the underlying mechanisms. Sprague-Dawley (SD) rats were subjected to MI and then treated (once daily for 4 weeks) with either RXR agonist bexarotene (10 or 30 mg/kg body weight) or vehicle. Heart function was determined using echocardiography and cardiac hemodynamic measurements. Four weeks post MI, myocardial tissues were collected to evaluate cardiac remodeling. Primary cardiac fibroblasts (CFs) were treated with or without RXR ligand 9-cis-RA followed by stimulation with TGF-β1. Immunoblot, immunofluorescence, and co-immunoprecipitation were performed to elucidate the regulatory role of RXR agonists in TGF-β1/Smad signaling. In vivo treatment with Bexarotene moderately affects systemic inflammation and apoptosis and ameliorated left ventricular dysfunction after MI in rat model. In contrast, bexarotene significantly inhibited post-MI myocardial fibrosis. Immunoblot analysis of heart tissue homogenates from MI rats revealed that bexarotene regulated the activation of the TGF-β1/Smad signaling pathway. In vitro, 9-cis-RA inhibited the TGF-β1-induced proliferation and collagen production of CFs. Importantly, upon activation by 9-cis-RA, RXRα interacted with p-Smad2 in cytoplasm, inhibiting the TGF-β1-induced nuclear translocation of p-Smad2, thereby negatively regulating TGF-β1/Smad signaling and attenuating the fibrotic response of CFs. These findings suggest that RXR agonists ameliorate post-infarction myocardial fibrosis, maladaptive remodeling, and heart dysfunction via attenuation of fibrotic response in CFs through inhibition of the TGF-β1/Smad pathway activation.
Collapse
Affiliation(s)
- Xiao-Yan Lin
- Ultrasonography Department, The First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
| | - Yong Chu
- Cardiovascular Department, The First Affiliated Hospital, Fujian Medical University, Fujian Institute of Hypertension, Fuzhou 350005, China
| | - Guo-Shan Zhang
- Zhongshan Hospital (Xiamen), Fudan University, Xiamen 361015, China
| | - Hai-Lin Zhang
- Cardiovascular Department, The First Affiliated Hospital, Fujian Medical University, Fujian Institute of Hypertension, Fuzhou 350005, China
| | - Kai Kang
- Cardiovascular Department, The First Affiliated Hospital, Fujian Medical University, Fujian Institute of Hypertension, Fuzhou 350005, China
| | - Min-Xia Wu
- Electron Microscopy Laboratory of Public Technology Service Center, Fujian Medical University, Fuzhou 350004, China
| | - Jiang Zhu
- Cardiovascular Department, The First Affiliated Hospital, Fujian Medical University, Fujian Institute of Hypertension, Fuzhou 350005, China
| | - Chang-Sheng Xu
- Cardiovascular Department, The First Affiliated Hospital, Fujian Medical University, Fujian Institute of Hypertension, Fuzhou 350005, China
| | - Jin-Xiu Lin
- Cardiovascular Department, The First Affiliated Hospital, Fujian Medical University, Fujian Institute of Hypertension, Fuzhou 350005, China
| | - Chun-Kai Huang
- Cardiovascular Department, The First Affiliated Hospital, Fujian Medical University, Fujian Institute of Hypertension, Fuzhou 350005, China
| | - Da-Jun Chai
- Cardiovascular Department, The First Affiliated Hospital, Fujian Medical University, Fujian Institute of Hypertension, Fuzhou 350005, China; Cardiovascular Department, National Regional Medical Center, Binhai Branch of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China.
| |
Collapse
|
4
|
Karolczak K, Konieczna L, Soltysik B, Kostka T, Witas PJ, Kostanek J, Baczek T, Watala C. Plasma Concentration of Cortisol Negatively Associates with Platelet Reactivity in Older Subjects. Int J Mol Sci 2022; 24:ijms24010717. [PMID: 36614157 PMCID: PMC9820908 DOI: 10.3390/ijms24010717] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 12/06/2022] [Accepted: 12/06/2022] [Indexed: 01/03/2023] Open
Abstract
The interaction of platelets with steroid hormones is poorly investigated. Age is one of the factors that increase the risk of pathological platelet reactivity and thrombosis. The aim of this study was to assess whether there were associations between platelet reactivity and plasma cortisol levels in volunteers aged 60-65 years. For this purpose, impedance aggregometry in whole blood measured after arachidonic acid, collagen, or ADP stimulation was used to estimate platelet reactivity and mass spectrometry was used to measure peripheral plasma cortisol concentration. Statistically significant negative correlations were observed between cortisol concentration and platelet reactivity in response to arachidonic acid and ADP, but not to collagen. The presented results suggest for the very first time that cortisol is a new endogenous modulator of platelet reactivity in the elderly population.
Collapse
Affiliation(s)
- Kamil Karolczak
- Department of Haemostatic Disorders, Medical University of Lodz, ul. Mazowiecka 6/8, 92-215 Lodz, Poland
- Correspondence:
| | - Lucyna Konieczna
- Department of Pharmaceutical Chemistry, Medical University of Gdańsk, ul. Hallera 107, 80-416 Gdańsk, Poland
| | - Bartlomiej Soltysik
- Department of Geriatrics, Healthy Aging Research Center (HARC), Medical University of Lodz, pl. Hallera 1, 90-647 Lodz, Poland
| | - Tomasz Kostka
- Department of Geriatrics, Healthy Aging Research Center (HARC), Medical University of Lodz, pl. Hallera 1, 90-647 Lodz, Poland
| | - Piotr Jakub Witas
- Department of Haemostatic Disorders, Medical University of Lodz, ul. Mazowiecka 6/8, 92-215 Lodz, Poland
| | - Joanna Kostanek
- Department of Haemostatic Disorders, Medical University of Lodz, ul. Mazowiecka 6/8, 92-215 Lodz, Poland
| | - Tomasz Baczek
- Department of Pharmaceutical Chemistry, Medical University of Gdańsk, ul. Hallera 107, 80-416 Gdańsk, Poland
| | - Cezary Watala
- Department of Haemostatic Disorders, Medical University of Lodz, ul. Mazowiecka 6/8, 92-215 Lodz, Poland
| |
Collapse
|
5
|
Effects of Nutrients on Platelet Function: A Modifiable Link between Metabolic Syndrome and Neurodegeneration? Biomolecules 2021; 11:biom11101455. [PMID: 34680088 PMCID: PMC8533544 DOI: 10.3390/biom11101455] [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: 06/29/2021] [Revised: 09/27/2021] [Accepted: 09/30/2021] [Indexed: 11/17/2022] Open
Abstract
Metabolic syndrome increases the risk of vascular dementia and other neurodegenerative disorders. Recent studies underline that platelets play an important role in linking peripheral with central metabolic and inflammatory mechanisms. In this narrative review, we address the activation of platelets in metabolic syndrome, their effects on neuronal processes and the role of the mediators (e.g., serotonin, platelet-derived growth factor). Emerging evidence shows that nutritional compounds and their metabolites modulate these interactions-specifically, long chain fatty acids, endocannabinoids and phenolic compounds. We reviewed the role of activated platelets in neurovascular processes and nutritional compounds in platelet activation.
Collapse
|
6
|
El-Kadiry AEH, Merhi Y. The Role of the Proteasome in Platelet Function. Int J Mol Sci 2021; 22:3999. [PMID: 33924425 PMCID: PMC8069084 DOI: 10.3390/ijms22083999] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 04/06/2021] [Accepted: 04/08/2021] [Indexed: 02/06/2023] Open
Abstract
Platelets are megakaryocyte-derived acellular fragments prepped to maintain primary hemostasis and thrombosis by preserving vascular integrity. Although they lack nuclei, platelets harbor functional genomic mediators that bolster platelet activity in a signal-specific manner by performing limited de novo protein synthesis. Furthermore, despite their limited protein synthesis, platelets are equipped with multiple protein degradation mechanisms, such as the proteasome. In nucleated cells, the functions of the proteasome are well established and primarily include proteostasis among a myriad of other signaling processes. However, the role of proteasome-mediated protein degradation in platelets remains elusive. In this review article, we recapitulate the developing literature on the functions of the proteasome in platelets, discussing its emerging regulatory role in platelet viability and function and highlighting how its functional coupling with the transcription factor NF-κB constitutes a novel potential therapeutic target in atherothrombotic diseases.
Collapse
Affiliation(s)
- Abed El-Hakim El-Kadiry
- Laboratory of Thrombosis and Hemostasis, Montreal Heart Institute, Research Centre, Montreal, QC H1T 1C8, Canada;
- Biomedical Sciences Program, Faculty of Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada
| | - Yahye Merhi
- Laboratory of Thrombosis and Hemostasis, Montreal Heart Institute, Research Centre, Montreal, QC H1T 1C8, Canada;
- Biomedical Sciences Program, Faculty of Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada
| |
Collapse
|
7
|
Poizeau F, Balusson F, Jonville-Béra AP, Nowak E, Drici MD, Scarabin PY, Droitcourt C, Dupuy A, Oger E. The cardiovascular safety of oral alitretinoin: a population-based cohort study involving 19 513 patients exposed to oral alitretinoin. Br J Dermatol 2021; 185:764-771. [PMID: 33735442 DOI: 10.1111/bjd.20069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/13/2021] [Indexed: 12/01/2022]
Abstract
BACKGROUND Oral alitretinoin is a retinoid used for severe chronic hand eczema. Although caution is recommended for patients with uncontrolled dyslipidaemia or cardiovascular risk factors, the actual atherothrombotic risk has not been investigated thus far. OBJECTIVES To detect any excess of atherothrombotic events among patients exposed to alitretinoin, during treatment or in the 2 years following initiation. METHODS Using the French Health Insurance database, we compared the number of patients who had an atherothrombotic event (coronary artery disease, ischaemic stroke or peripheral artery disease requiring revascularization) in the population exposed to oral alitretinoin vs. the general population of the same age, sex and baseline cardiovascular risk, using standardized morbidity ratios (SMRs). RESULTS Between 2009 and 2017, 19 513 patients were exposed to oral alitretinoin in France. Sixty-four (0·3%) patients had an atherothrombotic event while on alitretinoin. Patients receiving alitretinoin experienced no more atherothrombotic events than the general population: patients without cardiovascular risk factors or previous atherothrombotic events had a SMR of 0·65 [95% confidence interval (CI) 0·26-1·34] during alitretinoin treatment, and 1·21 (95% CI 0·90-1·59) in the 2 years following initiation; patients with cardiovascular risk factors or previous atherothrombotic events had a SMR of 0·82 (95% CI 0·60-1·08) during alitretinoin treatment and 0·95 (95% CI 0·82-1·09) in the 2 years following initiation. Taken separately, SMRs for each outcome did not increase either. CONCLUSIONS These data from an exhaustive nationwide population-based study do not support an increase in the incidence of atherothrombotic events with alitretinoin use, regardless of the baseline cardiovascular risk of the patient.
Collapse
Affiliation(s)
- F Poizeau
- Univ Rennes, CHU Rennes, EA 7449 (Pharmacoepidemiology and Health Services Research) REPERES, Rennes, F 35043, France.,PEPS Research Consortium (Pharmacoepidemiology for Health Product Safety), Rennes, France.,Department of Dermatology, CHU Rennes, Rennes, France
| | - F Balusson
- Univ Rennes, CHU Rennes, EA 7449 (Pharmacoepidemiology and Health Services Research) REPERES, Rennes, F 35043, France
| | - A P Jonville-Béra
- Centre Régional de Pharmacovigilance, CHRU Tours, Tours, France.,Université de Tours, Université de Nantes, INSERM - U 1246, France
| | - E Nowak
- PEPS Research Consortium (Pharmacoepidemiology for Health Product Safety), Rennes, France.,University of Bretagne Occidentale, Brest University, Brest, France.,INSERM CIC 1412, CHRU Brest, Brest, France
| | - M D Drici
- Department of Clinical Pharmacology, University of Cote d'Azur Medical Center, Nice, France
| | - P Y Scarabin
- Centre de Recherche en Épidémiologie et Santé des Populations, INSERM UMRS1018, Paris-Saclay University, Villejuif, France
| | - C Droitcourt
- Univ Rennes, CHU Rennes, EA 7449 (Pharmacoepidemiology and Health Services Research) REPERES, Rennes, F 35043, France.,PEPS Research Consortium (Pharmacoepidemiology for Health Product Safety), Rennes, France.,Department of Dermatology, CHU Rennes, Rennes, France
| | - A Dupuy
- Univ Rennes, CHU Rennes, EA 7449 (Pharmacoepidemiology and Health Services Research) REPERES, Rennes, F 35043, France.,PEPS Research Consortium (Pharmacoepidemiology for Health Product Safety), Rennes, France.,Department of Dermatology, CHU Rennes, Rennes, France
| | - E Oger
- Univ Rennes, CHU Rennes, EA 7449 (Pharmacoepidemiology and Health Services Research) REPERES, Rennes, F 35043, France.,PEPS Research Consortium (Pharmacoepidemiology for Health Product Safety), Rennes, France
| |
Collapse
|
8
|
Upreti C, Woodruff CM, Zhang XL, Yim MJ, Zhou ZY, Pagano AM, Rehanian DS, Yin D, Kandel ER, Stanton PK, Nicholls RE. Loss of retinoid X receptor gamma subunit impairs group 1 mGluR mediated electrophysiological responses and group 1 mGluR dependent behaviors. Sci Rep 2021; 11:5552. [PMID: 33692389 PMCID: PMC7946894 DOI: 10.1038/s41598-021-84943-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 02/17/2021] [Indexed: 11/09/2022] Open
Abstract
Retinoid X receptors are members of the nuclear receptor family that regulate gene expression in response to retinoic acid and related ligands. Group 1 metabotropic glutamate receptors are G-protein coupled transmembrane receptors that activate intracellular signaling cascades in response to the neurotransmitter, glutamate. These two classes of molecules have been studied independently and found to play important roles in regulating neuronal physiology with potential clinical implications for disorders such as depression, schizophrenia, Parkinson's and Alzheimer's disease. Here we show that mice lacking the retinoid X receptor subunit, RXRγ, exhibit impairments in group 1 mGluR-mediated electrophysiological responses at hippocampal Schaffer collateral-CA1 pyramidal cell synapses, including impaired group 1 mGluR-dependent long-term synaptic depression (LTD), reduced group 1 mGluR-induced calcium release, and loss of group 1 mGluR-activated voltage-sensitive currents. These animals also exhibit impairments in a subset of group 1 mGluR-dependent behaviors, including motor performance, spatial object recognition, and prepulse inhibition. Together, these observations demonstrate convergence between the RXRγ and group 1 mGluR signaling pathways that may function to coordinate their regulation of neuronal activity. They also identify RXRγ as a potential target for the treatment of disorders in which group 1 mGluR signaling has been implicated.
Collapse
Affiliation(s)
- Chirag Upreti
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, NY, 10595, USA
| | - Caitlin M Woodruff
- Department of Neuroscience, Columbia University, 3227 Broadway, New York, NY, 10027, USA
| | - Xiao-Lei Zhang
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, NY, 10595, USA
| | - Michael J Yim
- Department of Neuroscience, Columbia University, 3227 Broadway, New York, NY, 10027, USA
| | - Zhen-Yu Zhou
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, NY, 10595, USA.,Department of Neurology, New York Medical College, Valhalla, NY, 10595, USA
| | - Andrew M Pagano
- Department of Neuroscience, Columbia University, 3227 Broadway, New York, NY, 10027, USA
| | - Dina S Rehanian
- Department of Pathology and Cell Biology, Columbia University, 630 West 168thStreet, New York, NY, 10032, USA.,Taub Institute for Research on Alzheimer's Disease and Aging Brain, Columbia University, 630 West 168thStreet, New York, NY, 10032, USA
| | - Deqi Yin
- Department of Neuroscience, Columbia University, 3227 Broadway, New York, NY, 10027, USA.,Howard Hughes Medical Institute, Columbia University, 3227 Broadway, New York, NY, 10027, USA
| | - Eric R Kandel
- Department of Neuroscience, Columbia University, 3227 Broadway, New York, NY, 10027, USA.,Howard Hughes Medical Institute, Columbia University, 3227 Broadway, New York, NY, 10027, USA.,Kavli Institute for Brain Science, Columbia University, 3227 Broadway, New York, NY, 10027, USA.,Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, 3227 Broadway, New York, NY, 10027, USA
| | - Patric K Stanton
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, NY, 10595, USA.,Department of Neurology, New York Medical College, Valhalla, NY, 10595, USA
| | - Russell E Nicholls
- Department of Pathology and Cell Biology, Columbia University, 630 West 168thStreet, New York, NY, 10032, USA. .,Taub Institute for Research on Alzheimer's Disease and Aging Brain, Columbia University, 630 West 168thStreet, New York, NY, 10032, USA.
| |
Collapse
|
9
|
Shao M, Lu L, Wang Q, Ma L, Tian X, Li C, Li C, Guo D, Wang Q, Wang W, Wang Y. The multi-faceted role of retinoid X receptor in cardiovascular diseases. Biomed Pharmacother 2021; 137:111264. [PMID: 33761589 DOI: 10.1016/j.biopha.2021.111264] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/04/2021] [Accepted: 01/07/2021] [Indexed: 01/14/2023] Open
Abstract
Retinoid X receptors (RXRs) are members of ligand-dependent transcription factors whose effects on a diversity of cellular processes, including cellular proliferation, the immune response, and lipid and glucose metabolism. Knock out of RXRα causes a hypoplasia of the myocardium which is lethal during fetal life. In addition, the heart maintains a well-orchestrated balances in utilizing fatty acids (FAs) and other substrates to meet the high energy requirements. As the master transcriptional regulators of lipid metabolism, RXRs become particularly important for the energy needs of the heart. Accumulating evidence suggested that RXRs may exert direct beneficial effects in the heart both through heterodimerization with other nuclear receptors (NRs) and homodimerization, thus standing as suitable targets for treating in cardiovascular diseases. Although compounds that target RXRs are promising drugs, their use is limited by toxicity. A better understanding of the structural biology of RXRs in cardiovascular disease should enable the rational design of more selective nuclear receptor modulators to overcome these problems. Here, this review summarizes a brief overview of RXRs structure and versatility of RXR action in the control of cardiovascular diseases. And we also discussed the therapeutic potential of RXR ligand.
Collapse
Affiliation(s)
- Mingyan Shao
- School of Life Science, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Linghui Lu
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Qian Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Lin Ma
- School of Life Science, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xue Tian
- School of Life Science, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Changxiang Li
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Chun Li
- Modern Research Center of Traditional Chinese Medicine, School of Traditional Chinese Material Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Dongqing Guo
- School of Life Science, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Qiyan Wang
- School of Life Science, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Wei Wang
- School of Life Science, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Yong Wang
- School of Life Science, Beijing University of Chinese Medicine, Beijing, 100029, China; College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China.
| |
Collapse
|
10
|
Shatoor AS, Shati A, Humayed SA, Al-Qahtani S, Alkhateeb M. Opposite Modulatory Effects of Crataegus aronia Aqueous Extract on Platelet Aggregation in Rats. Chin J Integr Med 2020; 27:696-704. [PMID: 32418179 DOI: 10.1007/s11655-020-3187-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/13/2019] [Indexed: 11/29/2022]
Abstract
OBJECTIVES To reveal the mechanisms behind the dual effects of Crataegus aronia (C. aronia) aqueous extract on platelet aggregation by focusing on function, regulation, expression, and signaling of platelets P2Y12 receptors. METHODS Adult male Wistar rats (120 ± 10 g) were classified as control received the vehicle, C. aronia (200 mg/kg), and C. aronia (2,000 mg/kg)-treated rats. After treatments for consecutive 7 days, hematological and molecular experiments were conducted to detect alterations in platelet aggregation, thromboxane B2 (THXB2) and intracellular reactive oxygen species (ROS) content; protein levels of P2Y12, p-Akt, cyclic adenosine monophosphate (cAMP), phosphorylated vasodilator-stimulated-phosphoprotein (p-VASP), nuclear factor κB (NF-κB), P-selectin, and etc. in platelets were determined by Western blot; mRNA expressions of P2Y12 and some inflammatory markers were determined by real-time polymerase chain reaction. RESULTS At a concentration of 200 mg/kg, C. aronia inhibited platelet aggregation through multiple interconnected mechanisms including downregulation P2Y12 synthesis and expression, stimulating intracellular cAMP levels and protein levels of p-VASP, inhibiting platelets THXB2 release and protein levels of P-selectin. Also, it inhibited platelets level of ROS and of NF-κB, a major signaling pathway that stimulates the expression of P2Y12 and THXA2 synthesis. Opposite findings were seen in platelets of rats received C. aronia at a concentration of 2,000 mg/kg. Interestingly, co-administration of N-acetylcysteine prevented all hematological and molecular alterations exerted by the high dose of the extract and inhibited platelet aggregation. CONCLUSION Oral administration of C. aronia at low dose inhibits platelet aggregation by reducing THXB2 release, expression of P-selectin and activating cAMP and Akt signaling through two major mechanisms including downregulation of P2Y12 and inhibition of ROS-induced activation of NF-κB, an effect that is observed to be in the opposite direction with its high dose.
Collapse
Affiliation(s)
- Abdullah S Shatoor
- Department of Medicine, Cardiology Section, College of Medicine, King Khalid University, Abha, 64121, Saudi Arabia.
| | - Ali Shati
- Department of Biology, College of Science, College of Medicine, King Khalid University, Abha, 64121, Saudi Arabia
| | - S Al Humayed
- Department of Medicine, Cardiology Section, College of Medicine, King Khalid University, Abha, 64121, Saudi Arabia
| | - Sultan Al-Qahtani
- Department of Physiology, College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh, 11481, Saudi Arabia
| | - Mahmoud Alkhateeb
- Department of Physiology, College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh, 11481, Saudi Arabia
| |
Collapse
|
11
|
Abstract
Previous studies reported an inverse association between healthy dietary patterns (such as Mediterranean diet) and the incidence of cardiovascular events. As the mechanism accounting for cardiovascular disease is prevalently due to the atherothrombosis, where a pivotal role is played by platelet activation, it would be arguable that diets with protective effects against cardiovascular disease exert an anti-atherothrombotic effect via inhibition of platelet activation. There are several and sparse typologies of studies, which investigated if single nutrients by diets recognized as having cardiovascular protection may exert an antithrombotic effect. The most investigated nutrients are key components of the Mediterranean diets such as fruits and vegetables, fish, olive oil, and wine; other diets with protective effects include nuts and cocoa. Here we summarize experimental and human interventional studies which investigated the antithrombotic effects of such nutrients in experimental models of thrombosis or analyzed biomarkers of clotting, platelet, and fibrinolysis activation in human; furthermore in vitro studies explored the underlying mechanism at level of several cell lines such as platelets or endothelial cells. In this context, we analyzed if nutrients affect simultaneously or separately clotting, platelet, and fibrinolysis pathways giving special attention to the relationship between oxidative stress and thrombosis as most nutrients are believed to possess antioxidant properties.
Collapse
Affiliation(s)
- Francesco Violi
- From the Department of Clinical Internal, Anesthesiologic and Cardiovascular Sciences, Sapienza University of Rome, Italy (F.V., D.P., P.P.).,Mediterranea Cardiocentro, Napoli, Italy (F.V., P.P., R.C.)
| | - Daniele Pastori
- From the Department of Clinical Internal, Anesthesiologic and Cardiovascular Sciences, Sapienza University of Rome, Italy (F.V., D.P., P.P.)
| | - Pasquale Pignatelli
- From the Department of Clinical Internal, Anesthesiologic and Cardiovascular Sciences, Sapienza University of Rome, Italy (F.V., D.P., P.P.).,Mediterranea Cardiocentro, Napoli, Italy (F.V., P.P., R.C.)
| | - Roberto Carnevale
- Mediterranea Cardiocentro, Napoli, Italy (F.V., P.P., R.C.).,Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Italy (R.C.)
| |
Collapse
|
12
|
Flora GD, Sahli KA, Sasikumar P, Holbrook LM, Stainer AR, AlOuda SK, Crescente M, Sage T, Unsworth AJ, Gibbins JM. Non-genomic effects of the Pregnane X Receptor negatively regulate platelet functions, thrombosis and haemostasis. Sci Rep 2019; 9:17210. [PMID: 31748641 PMCID: PMC6868193 DOI: 10.1038/s41598-019-53218-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 10/29/2019] [Indexed: 01/30/2023] Open
Abstract
The pregnane X receptor (PXR) is a nuclear receptor (NR), involved in the detoxification of xenobiotic compounds. Recently, its presence was reported in the human vasculature and its ligands were proposed to exhibit anti-atherosclerotic effects. Since platelets contribute towards the development of atherosclerosis and possess numerous NRs, we investigated the expression of PXR in platelets along with the ability of its ligands to modulate platelet activation. The expression of PXR in human platelets was confirmed using immunoprecipitation analysis. Treatment with PXR ligands was found to inhibit platelet functions stimulated by a range of agonists, with platelet aggregation, granule secretion, adhesion and spreading on fibrinogen all attenuated along with a reduction in thrombus formation (both in vitro and in vivo). The effects of PXR ligands were observed in a species-specific manner, and the human-specific ligand, SR12813, was observed to attenuate thrombus formation in vivo in humanised PXR transgenic mice. PXR ligand-mediated inhibition of platelet function was found to be associated with the inhibition of Src-family kinases (SFKs). This study identifies acute, non-genomic regulatory effects of PXR ligands on platelet function and thrombus formation. In combination with the emerging anti-atherosclerotic properties of PXR ligands, these anti-thrombotic effects may provide additional cardio-protective benefits.
Collapse
Affiliation(s)
- Gagan D Flora
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, UK.,Department of Internal Medicine, University of Iowa, Iowa City, IA, USA
| | - Khaled A Sahli
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, UK.,General Directorate of Medical Services, Ministry of Interior, Riyadh, Kingdom of Saudi Arabia
| | - Parvathy Sasikumar
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, UK.,Centre for Haematology, Imperial College London, London, UK
| | - Lisa-Marie Holbrook
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, UK.,School of Cardiovascular Medicine and Sciences, King's College London, London, UK
| | - Alexander R Stainer
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, UK
| | - Sarah K AlOuda
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, UK
| | - Marilena Crescente
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, UK.,Centre for Immunobiology, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Tanya Sage
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, UK
| | - Amanda J Unsworth
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, UK.,School of Healthcare Science, Manchester Metropolitan University, Manchester, UK
| | - Jonathan M Gibbins
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, UK.
| |
Collapse
|
13
|
Lüsebrink E, Warm V, Pircher J, Ehrlich A, Zhang Z, Strecker J, Chambon P, Massberg S, Schulz C, Petzold T. Role of RXRβ in platelet function and arterial thrombosis. J Thromb Haemost 2019; 17:1489-1499. [PMID: 31172692 DOI: 10.1111/jth.14531] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 05/28/2019] [Accepted: 05/31/2019] [Indexed: 12/22/2022]
Abstract
OBJECTIVE Retinoid X receptors (RXR) are a family of nuclear receptors that play critical roles in the regulation of numerous fundamental biological processes including cell proliferation, differentiation, and death. Earlier studies suggested that treatment with RXR agonists attenuates platelet activation in all adults (male and femal) and mice; however, the underlying molecular mechanisms have remained insufficiently understood. To elaborate further on this issue, we characterized megakaryocyte and platelet-specific RXR knockout mice to study platelet function in vitro and arterial thrombosis in vivo. APPROACH AND RESULTS First, we identified RXRβ as the dominant RXR receptor in mouse platelets, prompting us to generate a megakaryocyte and platelet-specific PF4Cre ;RXRβflox/flox mouse. Second, we studied activation, spreading, and aggregation of platelets from C57Bl/6 wild-type mice (WT), PF4Cre+ ;RXRβflox/flox mice, and PF4Cre- ;RXRβflox/flox littermate controls in the presence or absence of RXR ligands, that is, 9-cis-retinoic acid (9cRA) and methoprene acid (MA). We found that in vitro treatment with RXR ligands attenuates spreading and aggregation of platelets and increases proplatelet particle formation from megakaryocytes (MK). However, these effects are also observed in RXRβ-deficient platelets and MKs and are thus independent of RXRβ. Third, we investigated arterial thrombus formation in an iron chloride (FeCl3)-induced vascular injury model in vivo, which is also not affected by the absence of RXRβ in platelets. CONCLUSIONS Absence of the most abundant RXR receptor in mouse platelets, RXRβ, does not affect platelet function in vitro and thrombus formation in vivo. Furthermore, RXR agonists' mediated effects on platelet function are independent of RXRβ expression. Hence, our data do not support a significant contribution of RXRβ to arterial thrombosis in mice.
Collapse
Affiliation(s)
- Enzo Lüsebrink
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Verena Warm
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Joachim Pircher
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Andreas Ehrlich
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Zhe Zhang
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Jan Strecker
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Pierre Chambon
- Département de Biologie, Cellulaire and Développement, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Strasbourg, France
| | - Steffen Massberg
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Christian Schulz
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Tobias Petzold
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| |
Collapse
|
14
|
Kojok K, El-Kadiry AEH, Merhi Y. Role of NF-κB in Platelet Function. Int J Mol Sci 2019; 20:E4185. [PMID: 31461836 PMCID: PMC6747346 DOI: 10.3390/ijms20174185] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 08/25/2019] [Accepted: 08/26/2019] [Indexed: 01/04/2023] Open
Abstract
Platelets are megakaryocyte-derived fragments lacking nuclei and prepped to maintain primary hemostasis by initiating blood clots on injured vascular endothelia. Pathologically, platelets undergo the same physiological processes of activation, secretion, and aggregation yet with such pronouncedness that they orchestrate and make headway the progression of atherothrombotic diseases not only through clot formation but also via forcing a pro-inflammatory state. Indeed, nuclear factor-κB (NF-κB) is largely implicated in atherosclerosis and its pathological complication in atherothrombotic diseases due to its transcriptional role in maintaining pro-survival and pro-inflammatory states in vascular and blood cells. On the other hand, we know little on the functions of platelet NF-κB, which seems to function in other non-genomic ways to modulate atherothrombosis. Therein, this review will resemble a rich portfolio for NF-κB in platelets, specifically showing its implications at the levels of platelet survival and function. We will also share the knowledge thus far on the effects of active ingredients on NF-κB in general, as an extrapolative method to highlight the potential therapeutic targeting of NF-κB in coronary diseases. Finally, we will unzip a new horizon on a possible extra-platelet role of platelet NF-κB, which will better expand our knowledge on the etiology and pathophysiology of atherothrombosis.
Collapse
Affiliation(s)
- Kevin Kojok
- The Laboratory of Thrombosis and Hemostasis, Montreal Heart Institute, Research Centre, 5000 Belanger Street, Montreal, H1T 1C8, QC, Canada
- Faculty of Medicine, Université de Montréal, Montreal, H3T 1J4, QC, Canada
| | - Abed El-Hakim El-Kadiry
- The Laboratory of Thrombosis and Hemostasis, Montreal Heart Institute, Research Centre, 5000 Belanger Street, Montreal, H1T 1C8, QC, Canada
- Faculty of Medicine, Université de Montréal, Montreal, H3T 1J4, QC, Canada
| | - Yahye Merhi
- The Laboratory of Thrombosis and Hemostasis, Montreal Heart Institute, Research Centre, 5000 Belanger Street, Montreal, H1T 1C8, QC, Canada.
- Faculty of Medicine, Université de Montréal, Montreal, H3T 1J4, QC, Canada.
| |
Collapse
|
15
|
Abstract
Some vitamins have beneficial effects on cardiovascular diseases, normalizing platelet function and preventing their excess activation. Anti-platelet vitamins can act directly through inhibitory biochemical pathways in platelets or indirectly by preventing damage to the endothelium or low-density lipoprotein from oxidation. As a rule, each vitamin alone is a weak inhibitor of platelet aggregation. However, in combination, they may act synergistically or enhance the effects of endogenous anti-platelet compounds, such as prostacyclin or nitric oxide, and appear to have a sufficient anti-thrombotic effect. This review will focus on vitamins, which inhibit platelet activation and the mechanisms of their action. The relationship between the vitamins that inhibit platelet aggregation and vascular diseases is examined.
Collapse
Affiliation(s)
- Gennadi Kobzar
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia
| |
Collapse
|
16
|
Inyushin M, Zayas-Santiago A, Rojas L, Kucheryavykh Y, Kucheryavykh L. Platelet-generated amyloid beta peptides in Alzheimer's disease and glaucoma. Histol Histopathol 2019; 34:843-856. [PMID: 30945258 PMCID: PMC6667289 DOI: 10.14670/hh-18-111] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Amyloid beta (Aβ) peptides have been implicated in both Alzheimer's disease (AD) and glaucoma and have been shown to be the key etiological factor in these dangerous health complications. On the other hand, it is well known that Aβ peptide can be generated from its precursor protein and massively released from the blood to nearby tissue upon the activation of platelets due to their involvement in innate immunity and inflammation processes. Here we review evidence about the development of AD and glaucoma neuronal damage showing their dependence on platelet count and activation. The correlation between the effect on platelet count and the effectiveness of anti-AD and anti-glaucoma therapies suggest that platelets may be an important player in these diseases.
Collapse
Affiliation(s)
- Mikhail Inyushin
- School of Medicine, Universidad Central del Caribe (UCC), PR, USA.
| | | | - Legier Rojas
- School of Medicine, Universidad Central del Caribe (UCC), PR, USA
| | | | | |
Collapse
|
17
|
Unsworth AJ, Flora GD, Gibbins JM. Non-genomic effects of nuclear receptors: insights from the anucleate platelet. Cardiovasc Res 2019; 114:645-655. [PMID: 29452349 PMCID: PMC5915957 DOI: 10.1093/cvr/cvy044] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 02/13/2018] [Indexed: 12/12/2022] Open
Abstract
Nuclear receptors (NRs) have the ability to elicit two different kinds of responses, genomic and non-genomic. Although genomic responses control gene expression by influencing the rate of transcription, non-genomic effects occur rapidly and independently of transcriptional regulation. Due to their anucleate nature and mechanistically well-characterized and rapid responses, platelets provide a model system for the study of any non-genomic effects of the NRs. Several NRs have been found to be present in human platelets, and multiple NR agonists have been shown to elicit anti-platelet effects by a variety of mechanisms. The non-genomic functions of NRs vary, including the regulation of kinase and phosphatase activity, ion channel function, intracellular calcium levels, and production of second messengers. Recently, the characterization of mechanisms and identification of novel binding partners of NRs have further strengthened the prospects of developing their ligands into potential therapeutics that offer cardio-protective properties in addition to their other defined genomic effects.
Collapse
Affiliation(s)
- Amanda J Unsworth
- School of Biological Sciences, Institute of Cardiovascular and Metabolic Research, Harborne Building, Whiteknights, Reading RG6 6AS, Berkshire, UK
| | - Gagan D Flora
- School of Biological Sciences, Institute of Cardiovascular and Metabolic Research, Harborne Building, Whiteknights, Reading RG6 6AS, Berkshire, UK
| | - Jonathan M Gibbins
- School of Biological Sciences, Institute of Cardiovascular and Metabolic Research, Harborne Building, Whiteknights, Reading RG6 6AS, Berkshire, UK
| |
Collapse
|
18
|
de Hoog E, Lukewich MK, Spencer GE. Retinoid receptor-based signaling plays a role in voltage-dependent inhibition of invertebrate voltage-gated Ca 2+ channels. J Biol Chem 2019; 294:10076-10093. [PMID: 31048374 DOI: 10.1074/jbc.ra118.006444] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 04/30/2019] [Indexed: 01/04/2023] Open
Abstract
The retinoic acid receptor (RAR) and retinoid X receptor (RXR) mediate the cellular effects of retinoids (derivatives of vitamin A). Both RAR and RXR signaling events are implicated in hippocampal synaptic plasticity. Furthermore, retinoids can interact with calcium signaling during homeostatic plasticity. We recently provided evidence that retinoids attenuate calcium current (I Ca) through neuronal voltage-gated calcium channels (VGCCs). We now examined the possibility that constitutive activity of neuronal RXR and/or RAR alters calcium influx via the VGCCs. We found that in neurons of the mollusk Lymnaea stagnalis, two different RXR antagonists (PA452 and HX531) had independent and opposing effects on I Ca that were also time-dependent; whereas the RXR pan-antagonist PA452 enhanced I Ca, HX531 reduced I Ca Interestingly, this effect of HX531 occurred through voltage-dependent inhibition of VGCCs, a phenomenon known to influence neurotransmitter release from neurons. This inhibition appeared to be independent of G proteins and was largely restricted to Cav2 Ca2+ channels. Of note, an RAR pan-antagonist, LE540, also inhibited I Ca but produced G protein-dependent, voltage-dependent inhibition of VGCCs. These findings provide evidence that retinoid receptors interact with G proteins in neurons and suggest mechanisms by which retinoids might affect synaptic calcium signaling.
Collapse
Affiliation(s)
- Eric de Hoog
- From the Department of Biological Sciences, Brock University, St. Catharines, Ontario L2S 3A1, Canada
| | - Mark K Lukewich
- From the Department of Biological Sciences, Brock University, St. Catharines, Ontario L2S 3A1, Canada
| | - Gaynor E Spencer
- From the Department of Biological Sciences, Brock University, St. Catharines, Ontario L2S 3A1, Canada
| |
Collapse
|
19
|
|
20
|
Unsworth AJ, Bye AP, Kriek N, Sage T, Osborne AA, Donaghy D, Gibbins JM. Cobimetinib and trametinib inhibit platelet MEK but do not cause platelet dysfunction. Platelets 2018; 30:762-772. [PMID: 30252580 PMCID: PMC6594423 DOI: 10.1080/09537104.2018.1514107] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The MEK inhibitors cobimetinib and trametinib are used in combination with BRAF inhibitors to treat metastatic melanoma but increase rates of hemorrhage relative to BRAF inhibitors alone. Platelets express several members of the MAPK signalling cascade including MEK1 and MEK2 and ERK1 and ERK2 but their role in platelet function and haemostasis is ambiguous as previous reports have been contradictory. It is therefore unclear if MEK inhibitors might be causing platelet dysfunction and contributing to increased hemorrhage. In the present study we performed pharmacological characterisation of cobimetinib and trametinib in vitro to investigate potential for MEK inhibitors to cause platelet dysfunction. We report that whilst both cobimetinib and trametinib are potent inhibitors of platelet MEK activity, treatment with trametinib did not alter platelet function. Treatment with cobimetinib results in inhibition of platelet aggregation, integrin activation, alpha-granule secretion and adhesion but only at suprapharmacological concentrations. We identified that the inhibitory effects of high concentrations of cobimetinib are associated with off-target inhibition on Akt and PKC. Neither inhibitor caused any alteration in thrombus formation on collagen under flow conditions in vitro. Our findings demonstrate that platelets are able to function normally when MEK activity is fully inhibited, indicating MEK activity is dispensable for normal platelet function. We conclude that the MEK inhibitors cobimetinib and trametinib do not induce platelet dysfunction and are therefore unlikely to contribute to increased incidence of bleeding reported during MEK inhibitor therapy.
Collapse
Affiliation(s)
- Amanda J Unsworth
- a Institute for Cardiovascular and Metabolic Research , University of Reading, School of Biological Sciences , Reading , UK
| | - Alexander P Bye
- a Institute for Cardiovascular and Metabolic Research , University of Reading, School of Biological Sciences , Reading , UK
| | - Neline Kriek
- a Institute for Cardiovascular and Metabolic Research , University of Reading, School of Biological Sciences , Reading , UK
| | - Tanya Sage
- a Institute for Cardiovascular and Metabolic Research , University of Reading, School of Biological Sciences , Reading , UK
| | - Ashley A Osborne
- a Institute for Cardiovascular and Metabolic Research , University of Reading, School of Biological Sciences , Reading , UK
| | - Dillon Donaghy
- b Department of Microbiology Immunology and Pathology , Colorado State University , Fort Collins , CO , USA
| | - Jonathan M Gibbins
- a Institute for Cardiovascular and Metabolic Research , University of Reading, School of Biological Sciences , Reading , UK
| |
Collapse
|
21
|
Schwertz H, Rowley JW, Zimmerman GA, Weyrich AS, Rondina MT. Retinoic acid receptor-α regulates synthetic events in human platelets. J Thromb Haemost 2017; 15:2408-2418. [PMID: 28981191 DOI: 10.1111/jth.13861] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Indexed: 12/01/2022]
Abstract
Essentials Platelets express retinoic acid receptor (RAR)α protein, specifically binding target mRNAs. mRNAs under RARα control include MAP1LC3B2, SLAIN2, and ANGPT1. All-trans retinoic acid (atRA) releases RARα from its target mRNA. RARα expressed in human platelets exerts translational control via direct mRNA binding. SUMMARY Background Translational control mechanisms in platelets are incompletely defined. Here, we determined whether the nuclear transcription factor RARα controls protein translational events in human platelets. Methods Isolated human platelets were treated with the pan-RAR agonist all-trans-retinoic acid (atRA). Global and targeted translational events were examined. Results Stimulation of platelets with atRA significantly increased global protein expression. RARα protein bound to a subset of platelet mRNAs, as measured by next-generation RNA-sequencing. In-depth analyses of 5' and 3'-untranslated regions of the RARα-bound mRNAs revealed consensus RARα binding sites in microtubule-associated protein 1 light chain 3 beta 2 (MAP1LC3B2), SLAIN motif-containing protein 2 (SLAIN2) and angiopoietin-1 (ANGPT1) transcripts. When platelets were treated with atRA, binding interactions between RARα protein and mRNA for MAP1LC3B2, SLAIN2 and ANGPT1 were significantly decreased. Consistent with the release of bound RARα protein from MAP1LCB2mRNA, we observed an increase in the synthesis of MAP1LC3B2 protein. Conclusions These findings provide the first evidence that RARα, a nuclear transcriptional factor, regulates synthetic events in anucleate human platelets. They also reveal an additional non-genomic role for RARα in platelets that may have implications for the vitamin A-dependent signaling in humans.
Collapse
Affiliation(s)
- H Schwertz
- Molecular Medicine Program, University of Utah, Salt Lake City, UT, USA
- Departments of Surgery, University of Utah, Salt Lake City, UT, USA
| | - J W Rowley
- Molecular Medicine Program, University of Utah, Salt Lake City, UT, USA
- Internal Medicine, University of Utah, Salt Lake City, UT, USA
| | - G A Zimmerman
- Molecular Medicine Program, University of Utah, Salt Lake City, UT, USA
- Internal Medicine, University of Utah, Salt Lake City, UT, USA
| | - A S Weyrich
- Molecular Medicine Program, University of Utah, Salt Lake City, UT, USA
- Internal Medicine, University of Utah, Salt Lake City, UT, USA
| | - M T Rondina
- Molecular Medicine Program, University of Utah, Salt Lake City, UT, USA
- Internal Medicine, University of Utah, Salt Lake City, UT, USA
- The Geriatric Research Education and Clinical Center (GRECC), University of Utah, Salt Lake City, UT, USA
- Department of Internal Medicine at the George E. Wahlen Salt Lake City VAMC, Salt Lake City, Utah, USA
| |
Collapse
|
22
|
A β Peptide Originated from Platelets Promises New Strategy in Anti-Alzheimer's Drug Development. BIOMED RESEARCH INTERNATIONAL 2017; 2017:3948360. [PMID: 29018812 PMCID: PMC5605787 DOI: 10.1155/2017/3948360] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 07/10/2017] [Indexed: 12/20/2022]
Abstract
The amyloid beta (Aβ) peptide and its deposits in the brain are known to be implicated in the neurodegeneration that occurs during Alzheimer's disease (AD). Recently, alternative theories views concerning both the source of this peptide and its functions have been developed. It has been shown that, as in all other known types of amyloidosis, the production of Aβ originates in blood cells or cells related to blood plasma, from which it can then spread from the blood to inside the brain, with the greatest concentration around brain blood vessels. In this review, we summarize research progress in this new area and outline some future perspectives. While it is still unclear whether the main source of Aβ deposits in AD is the blood, the possibility of blocking the chain of reactions that lead to constant Aβ release from the blood to the brain may be exploited in an attempt to reduce the amyloid burden in AD. Solving the problem of Aβ accumulation in this way may provide an alternative strategy for developing anti-AD drugs.
Collapse
|
23
|
Cheng B, Al-Shammari FH, Ghader IA, Sequeira F, Thakkar J, Mathew TC. Fundamental studies of adrenal retinoid-X-receptor: Protein isoform, tissue expression, subcellular distribution, and ligand availability. J Steroid Biochem Mol Biol 2017; 171:110-120. [PMID: 28267642 DOI: 10.1016/j.jsbmb.2017.03.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 02/14/2017] [Accepted: 03/02/2017] [Indexed: 11/21/2022]
Abstract
Adrenal gland reportedly expresses many nuclear receptors that are known to heterodimerize with retinoid-X-receptor (RXR) for functions, but the information regarding the glandular RXR is not adequate. Studies of rat adrenal homogenate by Western blotting revealed three RXR proteins: RXRα (55kDa), RXRβ (47kDa) and RXR (56kDa). RXRγ was not detectable. After fractionation, RXRα was almost exclusively localized in the nuclear fraction. In comparison, substantial portions of RXRβ and RXR were found in both nuclear and post-nuclear particle fractions, suggesting genomic and non-genomic functions. Cells immunostained for RXRα were primarily localized in zona fasciculata (ZF) and medulla, although some stained cells were found in zona glomerulosa (ZG) and zona reticularis (ZR). In contrast, cells immunostained for RXRβ were concentrated principally in ZG, although some stained cells were seen in ZR, ZF, and medulla (in descending order, qualitatively). Analysis of adrenal lipid extracts by LC/MS did not detect 9-cis-retinoic acid (a potent RXR-ligand) but identified all-trans retinoic acid. Since C20 and C22 polyunsaturated fatty acids (PUFAs) can also activate RXR, subcellular availabilities of unesterified fatty acids were investigated by GC/MS. As results, arachidonic acid (C20:4), adrenic acid (C22:4), docosapentaenoic acid (C22:5), and cervonic acid (C22:6) were detected in the lipids extracted from each subcellular fraction. Thus, the RXR-agonizing PUFAs are available in all the main subcellular compartments considerably. The present findings not only shed light on the adrenal network of RXRs but also provide baseline information for further investigations of RXR heterodimers in the regulation of adrenal steroidogenesis.
Collapse
Affiliation(s)
- Behling Cheng
- Department of Biochemistry, Faculty of Medicine, Kuwait University Health Science Center, P. O. Box 24923, Safat 13110, Kuwait.
| | - Fatema H Al-Shammari
- Department of Biochemistry, Faculty of Medicine, Kuwait University Health Science Center, P. O. Box 24923, Safat 13110, Kuwait
| | - Isra'a A Ghader
- Department of Biochemistry, Faculty of Medicine, Kuwait University Health Science Center, P. O. Box 24923, Safat 13110, Kuwait
| | - Fatima Sequeira
- Department of Biochemistry, Faculty of Medicine, Kuwait University Health Science Center, P. O. Box 24923, Safat 13110, Kuwait
| | - Jitendra Thakkar
- Department of Biochemistry, Faculty of Medicine, Kuwait University Health Science Center, P. O. Box 24923, Safat 13110, Kuwait
| | - Thazhumpal C Mathew
- Department of Medical Laboratory Science, Faculty of Allied Health, Kuwait University Health Science Center, P.O. Box 31470, Sulaibekhat 90805, Kuwait
| |
Collapse
|
24
|
Unsworth AJ, Flora GD, Sasikumar P, Bye AP, Sage T, Kriek N, Crescente M, Gibbins JM. RXR Ligands Negatively Regulate Thrombosis and Hemostasis. Arterioscler Thromb Vasc Biol 2017; 37:812-822. [PMID: 28254816 PMCID: PMC5405776 DOI: 10.1161/atvbaha.117.309207] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 02/13/2017] [Indexed: 12/17/2022]
Abstract
Supplemental Digital Content is available in the text. Objective— Platelets have been found to express intracellular nuclear receptors including the retinoid X receptors (RXRα and RXRβ). Treatment of platelets with ligands of RXR has been shown to inhibit platelet responses to ADP and thromboxane A2; however, the effects on responses to other platelet agonists and the underlying mechanism have not been fully characterized. Approach and Results— The effect of 9-cis-retinoic acid, docosahexaenoic acid and methoprene acid on collagen receptor (glycoprotein VI [GPVI]) agonists and thrombin-stimulated platelet function; including aggregation, granule secretion, integrin activation, calcium mobilization, integrin αIIbβ3 outside-in signaling and thrombus formation in vitro and in vivo were determined. Treatment of platelets with RXR ligands resulted in attenuation of platelet functional responses after stimulation by GPVI agonists or thrombin and inhibition of integrin αIIbβ3 outside-in signaling. Treatment with 9-cis-retinoic acid caused inhibition of thrombus formation in vitro and an impairment of thrombosis and hemostasis in vivo. Both RXR ligands stimulated protein kinase A activation, measured by VASP S157 phosphorylation, that was found to be dependent on both cAMP and nuclear factor κ-light-chain-enhancer of activated B cell activity. Conclusions— This study identifies a widespread, negative regulatory role for RXR in the regulation of platelet functional responses and thrombus formation and describes novel events that lead to the upregulation of protein kinase A, a known negative regulator of many aspects of platelet function. This mechanism may offer a possible explanation for the cardioprotective effects described in vivo after treatment with RXR ligands.
Collapse
Affiliation(s)
- Amanda J Unsworth
- From the Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, United Kingdom
| | - Gagan D Flora
- From the Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, United Kingdom
| | - Parvathy Sasikumar
- From the Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, United Kingdom
| | - Alexander P Bye
- From the Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, United Kingdom
| | - Tanya Sage
- From the Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, United Kingdom
| | - Neline Kriek
- From the Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, United Kingdom
| | - Marilena Crescente
- From the Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, United Kingdom
| | - Jonathan M Gibbins
- From the Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, United Kingdom.
| |
Collapse
|
25
|
Unsworth AJ, Kriek N, Bye AP, Naran K, Sage T, Flora GD, Gibbins JM. PPARγ agonists negatively regulate αIIbβ3 integrin outside-in signaling and platelet function through up-regulation of protein kinase A activity. J Thromb Haemost 2017; 15:356-369. [PMID: 27896950 PMCID: PMC5396324 DOI: 10.1111/jth.13578] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Indexed: 12/31/2022]
Abstract
Essentials peroxisome proliferator-activated receptor γ (PPARγ) agonists inhibit platelet function. PPARγ agonists negatively regulate outside-in signaling via integrin αIIbβ3. PPARγ agonists disrupt the interaction of Gα13 with integrin β3. This is attributed to an upregulation of protein kinase A activity. SUMMARY Background Agonists for the peroxisome proliferator-activated receptor (PPARγ) have been shown to have inhibitory effects on platelet activity following stimulation by GPVI and GPCR agonists. Objectives Profound effects on thrombus formation led us to suspect a role for PPARγ agonists in the regulation of integrin αIIbβ3 mediated signaling. Both GPVI and GPCR signaling pathways lead to αIIbβ3 activation, and signaling through αIIbβ3 plays a critical role in platelet function and normal hemostasis. Methods The effects of PPARγ agonists on the regulation of αIIbβ3 outside-in signaling was determined by monitoring the ability of platelets to adhere and spread on fibrinogen and undergo clot retraction. Effects on signaling components downstream of αIIbβ3 activation were also determined following adhesion to fibrinogen by Western blotting. Results Treatment of platelets with PPARγ agonists inhibited platelet adhesion and spreading on fibrinogen and diminished clot retraction. A reduction in phosphorylation of several components of αIIbβ3 signaling, including the integrin β3 subunit, Syk, PLCγ2, focal adhesion kinase (FAK) and Akt, was also observed as a result of reduced interaction of the integrin β3 subunit with Gα13. Studies of VASP phosphorylation revealed that this was because of an increase in PKA activity following treatment with PPARγ receptor agonists. Conclusions This study provides further evidence for antiplatelet actions of PPARγ agonists, identifies a negative regulatory role for PPARγ agonists in the control of integrin αIIbβ3 outside-in signaling, and provides a molecular basis by which the PPARγ agonists negatively regulate platelet activation and thrombus formation.
Collapse
Affiliation(s)
- A. J. Unsworth
- Institute for Cardiovascular and Metabolic ResearchSchool of Biological SciencesUniversity of ReadingReadingUK
| | - N. Kriek
- Institute for Cardiovascular and Metabolic ResearchSchool of Biological SciencesUniversity of ReadingReadingUK
| | - A. P. Bye
- Institute for Cardiovascular and Metabolic ResearchSchool of Biological SciencesUniversity of ReadingReadingUK
| | - K. Naran
- Institute for Cardiovascular and Metabolic ResearchSchool of Biological SciencesUniversity of ReadingReadingUK
| | - T. Sage
- Institute for Cardiovascular and Metabolic ResearchSchool of Biological SciencesUniversity of ReadingReadingUK
| | - G. D. Flora
- Institute for Cardiovascular and Metabolic ResearchSchool of Biological SciencesUniversity of ReadingReadingUK
| | - J. M. Gibbins
- Institute for Cardiovascular and Metabolic ResearchSchool of Biological SciencesUniversity of ReadingReadingUK
| |
Collapse
|
26
|
Downstream Regulatory Element Antagonist Modulator (DREAM), a target for anti-thrombotic agents. Pharmacol Res 2017; 117:283-287. [PMID: 28065857 DOI: 10.1016/j.phrs.2017.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 01/03/2017] [Indexed: 11/21/2022]
Abstract
Circulating platelets participate in the process of numerous diseases including thrombosis, inflammation, and cancer. Thus, it is of great importance to understand the underlying mechanisms mediating platelet activation under disease conditions. Emerging evidence indicates that despite the lack of a nucleus, platelets possess molecules that are involved in gene transcription in nucleated cells. This review will summarize downstream regulatory element antagonist modulator (DREAM), a transcriptional repressor, and highlight recent findings suggesting its novel non-transcriptional role in hemostasis and thrombosis.
Collapse
|
27
|
Unsworth AJ, Bye AP, Gibbins JM. Platelet-Derived Inhibitors of Platelet Activation. PLATELETS IN THROMBOTIC AND NON-THROMBOTIC DISORDERS 2017. [PMCID: PMC7123044 DOI: 10.1007/978-3-319-47462-5_37] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
28
|
Toral M, Romero M, Pérez-Vizcaíno F, Duarte J, Jiménez R. Antihypertensive effects of peroxisome proliferator-activated receptor-β/δ activation. Am J Physiol Heart Circ Physiol 2016; 312:H189-H200. [PMID: 27881385 DOI: 10.1152/ajpheart.00155.2016] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Revised: 09/12/2016] [Accepted: 11/21/2016] [Indexed: 01/16/2023]
Abstract
Peroxisome proliferator-activated receptors (PPARs) are members of the nuclear hormone receptor superfamily of ligand-activated transcription factors, which is composed of three members encoded by distinct genes: PPARα, PPARβ/δ, and PPARγ. The biological actions of PPARα and PPARγ and their potential as a cardiovascular therapeutic target have been extensively reviewed, whereas the biological actions of PPARβ/δ and its effectiveness as a therapeutic target in the treatment of hypertension remain less investigated. Preclinical studies suggest that pharmacological PPARβ/δ activation induces antihypertensive effects in direct [spontaneously hypertensive rat (SHR), ANG II, and DOCA-salt] and indirect (dyslipemic and gestational) models of hypertension, associated with end-organ damage protection. This review summarizes mechanistic insights into the antihypertensive effects of PPARβ/δ activators, including molecular and functional mechanisms. Pharmacological PPARβ/δ activation induces genomic actions including the increase of regulators of G protein-coupled signaling (RGS), acute nongenomic vasodilator effects, as well as the ability to improve the endothelial dysfunction, reduce vascular inflammation, vasoconstrictor responses, and sympathetic outflow from central nervous system. Evidence from clinical trials is also examined. These preclinical and clinical outcomes of PPARβ/δ ligands may provide a basis for the development of therapies in combating hypertension.
Collapse
Affiliation(s)
- Marta Toral
- Department of Pharmacology, School of Pharmacy, University of Granada, Granada, Spain
| | - Miguel Romero
- Department of Pharmacology, School of Pharmacy, University of Granada, Granada, Spain.,Instituto de Investigación Biosanitaria de Granada, ibs.GRANADA, Granada, Spain
| | - Francisco Pérez-Vizcaíno
- Department of Pharmacology, School of Medicine, University Complutense of Madrid, Madrid, Spain.,Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid. Spain; and.,Ciber Enfermedades Respiratorias (Ciberes). Madrid. Spain
| | - Juan Duarte
- Department of Pharmacology, School of Pharmacy, University of Granada, Granada, Spain.,Instituto de Investigación Biosanitaria de Granada, ibs.GRANADA, Granada, Spain
| | - Rosario Jiménez
- Department of Pharmacology, School of Pharmacy, University of Granada, Granada, Spain; .,Instituto de Investigación Biosanitaria de Granada, ibs.GRANADA, Granada, Spain
| |
Collapse
|
29
|
Moraes LA, Unsworth AJ, Vaiyapuri S, Ali MS, Sasikumar P, Sage T, Flora GD, Bye AP, Kriek N, Dorchies E, Molendi-Coste O, Dombrowicz D, Staels B, Bishop-Bailey D, Gibbins JM. Farnesoid X Receptor and Its Ligands Inhibit the Function of Platelets. Arterioscler Thromb Vasc Biol 2016; 36:2324-2333. [PMID: 27758768 DOI: 10.1161/atvbaha.116.308093] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 09/20/2016] [Indexed: 12/16/2022]
Abstract
OBJECTIVE Although initially seemingly paradoxical because of the lack of nucleus, platelets possess many transcription factors that regulate their function through DNA-independent mechanisms. These include the farnesoid X receptor (FXR), a member of the superfamily of ligand-activated transcription factors, that has been identified as a bile acid receptor. In this study, we show that FXR is present in human platelets and FXR ligands, GW4064 and 6α-ethyl-chenodeoxycholic acid, modulate platelet activation nongenomically. APPROACH AND RESULTS FXR ligands inhibited the activation of platelets in response to stimulation of collagen or thrombin receptors, resulting in diminished intracellular calcium mobilization, secretion, fibrinogen binding, and aggregation. Exposure to FXR ligands also reduced integrin αIIbβ3 outside-in signaling and thereby reduced the ability of platelets to spread and to stimulate clot retraction. FXR function in platelets was found to be associated with the modulation of cyclic guanosine monophosphate levels in platelets and associated downstream inhibitory signaling. Platelets from FXR-deficient mice were refractory to the actions of FXR agonists on platelet function and cyclic nucleotide signaling, firmly linking the nongenomic actions of these ligands to the FXR. CONCLUSIONS This study provides support for the ability of FXR ligands to modulate platelet activation. The atheroprotective effects of GW4064, with its novel antiplatelet effects, indicate FXR as a potential target for the prevention of atherothrombotic disease.
Collapse
Affiliation(s)
- Leonardo A Moraes
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, Harborne Building, University of Reading, Reading, Berkshire, RG6 6AS, UK.,Department of Physiology & NUS Immunology Program, Centre for Life Sciences, Yong Loo Lin School of Medicine, National University of Singapore, 117456, Singapore
| | - Amanda J Unsworth
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, Harborne Building, University of Reading, Reading, Berkshire, RG6 6AS, UK
| | | | - Marfoua S Ali
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, Harborne Building, University of Reading, Reading, Berkshire, RG6 6AS, UK
| | - Parvathy Sasikumar
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, Harborne Building, University of Reading, Reading, Berkshire, RG6 6AS, UK
| | - Tanya Sage
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, Harborne Building, University of Reading, Reading, Berkshire, RG6 6AS, UK
| | - Gagan D Flora
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, Harborne Building, University of Reading, Reading, Berkshire, RG6 6AS, UK
| | - Alex P Bye
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, Harborne Building, University of Reading, Reading, Berkshire, RG6 6AS, UK
| | - Neline Kriek
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, Harborne Building, University of Reading, Reading, Berkshire, RG6 6AS, UK
| | - Emilie Dorchies
- European Genomic Institute for Diabetes (EGID), F-59000, Lille, France; INSERM UMR1011, F-59000 Lille, France, University of Lille, F-59000 Lille, France; Institut Pasteur de Lille, F-59019 Lille, France
| | - Olivier Molendi-Coste
- European Genomic Institute for Diabetes (EGID), F-59000, Lille, France; INSERM UMR1011, F-59000 Lille, France, University of Lille, F-59000 Lille, France; Institut Pasteur de Lille, F-59019 Lille, France
| | - David Dombrowicz
- European Genomic Institute for Diabetes (EGID), F-59000, Lille, France; INSERM UMR1011, F-59000 Lille, France, University of Lille, F-59000 Lille, France; Institut Pasteur de Lille, F-59019 Lille, France
| | - Bart Staels
- European Genomic Institute for Diabetes (EGID), F-59000, Lille, France; INSERM UMR1011, F-59000 Lille, France, University of Lille, F-59000 Lille, France; Institut Pasteur de Lille, F-59019 Lille, France
| | - David Bishop-Bailey
- Comparative Biomedical Sciences, Royal Veterinary College, University of London, London, NW1 OTU, UK
| | - Jonathan M Gibbins
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, Harborne Building, University of Reading, Reading, Berkshire, RG6 6AS, UK
| |
Collapse
|
30
|
Bye AP, Unsworth AJ, Gibbins JM. Platelet signaling: a complex interplay between inhibitory and activatory networks. J Thromb Haemost 2016; 14:918-30. [PMID: 26929147 PMCID: PMC4879507 DOI: 10.1111/jth.13302] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 02/11/2016] [Indexed: 01/22/2023]
Abstract
The role of platelets in hemostasis and thrombosis is dependent on a complex balance of activatory and inhibitory signaling pathways. Inhibitory signals released from the healthy vasculature suppress platelet activation in the absence of platelet receptor agonists. Activatory signals present at a site of injury initiate platelet activation and thrombus formation; subsequently, endogenous negative signaling regulators dampen activatory signals to control thrombus growth. Understanding the complex interplay between activatory and inhibitory signaling networks is an emerging challenge in the study of platelet biology, and necessitates a systematic approach to utilize experimental data effectively. In this review, we will explore the key points of platelet regulation and signaling that maintain platelets in a resting state, mediate activation to elicit thrombus formation, or provide negative feedback. Platelet signaling will be described in terms of key signaling molecules that are common to the pathways activated by platelet agonists and can be described as regulatory nodes for both positive and negative regulators.
Collapse
Affiliation(s)
- A P Bye
- Institute of Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, UK
| | - A J Unsworth
- Institute of Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, UK
| | - J M Gibbins
- Institute of Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, UK
| |
Collapse
|
31
|
Rondina MT, Freitag M, Pluthero FG, Kahr WHA, Rowley JW, Kraiss LW, Franks Z, Zimmerman GA, Weyrich AS, Schwertz H. Non-genomic activities of retinoic acid receptor alpha control actin cytoskeletal events in human platelets. J Thromb Haemost 2016; 14:1082-94. [PMID: 26848712 PMCID: PMC5497578 DOI: 10.1111/jth.13281] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Indexed: 12/29/2022]
Abstract
UNLABELLED Essentials Platelets employ proteins/signaling pathways traditionally thought reserved for nuclear niche. We determined retinoic-acid-receptor alpha (RARα) expression and function in human platelets. RARα/actin-related protein-2/3 complex (Arp2/3) interact via non-genomic signaling in platelets. RARα regulates Arp2/3-mediated actin cytoskeletal dynamics and platelet spreading. SUMMARY Background Platelets utilize proteins and pathways classically reserved for the nuclear niche. Methods We determined whether human platelets express retinoic-acid-receptor family members, traditionally thought of as nuclear transcription factors, and deciphered the function of RARα. Results We found that RARα is robustly expressed in human platelets and megakaryocytes and interacts directly with actin-related protein-2/3 complex (Arp2/3) subunit 5 (Arp2/3s5). Arp2/3s5 co-localized with RARα in situ and regulated platelet cytoskeletal processes. The RARα ligand all-trans retinoic acid (atRA) disrupted RARα-Arp2/3 interactions. When isolated human platelets were treated with atRA, rapid cytoskeletal events (e.g. platelet spreading) were inhibited. In addition, when platelets were cultured for 18 h in the presence of atRA, actin-dependent morphological changes (e.g. extended cell body formation) were similarly inhibited. Using in vitro actin branching assays, RARα and Arp2/3-regulated complex actin branch formation was demonstrated. Consistent with inhibition of cytoskeletal processes in platelets, atRA, when added to this branching assay, resulted in dysregulated actin branching. Conclusion Our findings identify a previously unknown mechanism by which RARα regulates Arp2/3-mediated actin cytoskeletal dynamics through a non-genomic signaling pathway. These findings have broad implications in both nucleated and anucleate cells, where actin cytoskeletal events regulate cell morphology, movement and division.
Collapse
Affiliation(s)
- M T Rondina
- Molecular Medicine Program, University of Utah, Salt Lake City, UT, USA
- Department of Internal Medicine, University of Utah, Salt Lake City, UT, USA
- Department of Internal Medicine, George E. Wahlen Salt Lake City VAMC, Salt Lake City, UT, USA
| | - M Freitag
- Department of Immunology and Transfusion Medicine, University of Greifswald, Greifswald, Germany
| | - F G Pluthero
- Program in Cell Biology, Research Institute, Hospital for Sick Children, Toronto, ON, Canada
| | - W H A Kahr
- Program in Cell Biology, Research Institute, Hospital for Sick Children, Toronto, ON, Canada
- Departments of Paediatrics and Biochemistry, University of Toronto, Toronto, ON, Canada
| | - J W Rowley
- Molecular Medicine Program, University of Utah, Salt Lake City, UT, USA
| | - L W Kraiss
- Molecular Medicine Program, University of Utah, Salt Lake City, UT, USA
- Division of Vascular Surgery, Department of Surgery, University of Utah, Salt Lake City, UT, USA
| | - Z Franks
- Molecular Medicine Program, University of Utah, Salt Lake City, UT, USA
| | - G A Zimmerman
- Molecular Medicine Program, University of Utah, Salt Lake City, UT, USA
- Department of Internal Medicine, University of Utah, Salt Lake City, UT, USA
| | - A S Weyrich
- Molecular Medicine Program, University of Utah, Salt Lake City, UT, USA
- Department of Internal Medicine, University of Utah, Salt Lake City, UT, USA
| | - H Schwertz
- Molecular Medicine Program, University of Utah, Salt Lake City, UT, USA
- Department of Immunology and Transfusion Medicine, University of Greifswald, Greifswald, Germany
- Division of Vascular Surgery, Department of Surgery, University of Utah, Salt Lake City, UT, USA
- Lichtenberg-Professor for Experimental Hemostasis, University of Greifswald, Greifswald, Germany
| |
Collapse
|
32
|
da Motta NAV, de Brito FCF. Cilostazol exerts antiplatelet and anti-inflammatory effects through AMPK activation and NF-kB inhibition on hypercholesterolemic rats. Fundam Clin Pharmacol 2016; 30:327-37. [PMID: 26950185 DOI: 10.1111/fcp.12195] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Revised: 03/01/2016] [Accepted: 03/04/2016] [Indexed: 12/16/2022]
Abstract
This work presents a model of rats fed a high-cholesterol diet, receiving a long-term oral administration of cilostazol, a PDE3-inhibitor. The aim of this study was to evaluate the molecular mechanisms by which cilostazol interferes with platelets signaling pathways to avoid atherosclerosis early development. Male Wistar rats were divided into 3 groups: Control group received standard rat chow (C), hypercholesterolemic group (HCD), and HCD+CIL (cilostazol group) received hypercholesterolemic diet for 45 days. HCD+CIL group received cilostazol (30 mg/kg/p.o.) once daily in the last 15 days. Platelet aggregation, lipid profile, lipid peroxidation, and cytokine serum levels were assessed. Expression of P-selectin, CD40L, PKC-α, IkB-α, and iNOS and activation of AMPK, NF-κB, and eNOS in the platelets were assessed using Western blot analysis. Cilostazol reduced the levels of total cholesterol (361.0 ± 12.8 vs. 111.5 ± 1.6 mg/dL), triglycerides (186.9 ± 17.7 vs. 55.4 ±3.1 mg/dL), cLDL (330.9 ± 9.7 vs. 61.5 ± 3.5 mg/dL), cVLDL (45.0 ± 4.6 vs. 11.1 ± 0.6 mg/dL), and malondialdehyde (9.4 ± 0.5 vs. 3.2 ± 0.3 nmol/mL) compared to the HCD group. Cilostazol presented antiplatelet properties and decreased inflammatory markers levels. These effects seem to be related to AMPK activation, NF-kB inhibition, and eNOS activation.
Collapse
Affiliation(s)
- Nadia Alice Vieira da Motta
- Departamento de Fisiologia e Farmacologia, Laboratório de Farmacologia Experimental (LAFE), Instituto Biomédico, Universidade Federal Fluminense (UFF), Sala 204-A, 24230-210, Niterói, Rio de Janeiro, Brazil
| | - Fernanda Carla Ferreira de Brito
- Departamento de Fisiologia e Farmacologia, Laboratório de Farmacologia Experimental (LAFE), Instituto Biomédico, Universidade Federal Fluminense (UFF), Sala 204-A, 24230-210, Niterói, Rio de Janeiro, Brazil
| |
Collapse
|
33
|
Barlaka E, Galatou E, Mellidis K, Ravingerova T, Lazou A. Role of Pleiotropic Properties of Peroxisome Proliferator-Activated Receptors in the Heart: Focus on the Nonmetabolic Effects in Cardiac Protection. Cardiovasc Ther 2016; 34:37-48. [DOI: 10.1111/1755-5922.12166] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
- Eleftheria Barlaka
- School of Biology; Aristotle University of Thessaloniki; Thessaloniki Greece
| | - Eleftheria Galatou
- School of Biology; Aristotle University of Thessaloniki; Thessaloniki Greece
| | - Kyriakos Mellidis
- School of Biology; Aristotle University of Thessaloniki; Thessaloniki Greece
| | - Tanya Ravingerova
- Institute for Heart Research; Slovak Academy of Sciences; Bratislava Slovak Republic
| | - Antigone Lazou
- School of Biology; Aristotle University of Thessaloniki; Thessaloniki Greece
| |
Collapse
|
34
|
Abstract
Nuclear receptors sense a wide range of steroids and hormones (estrogens, progesterone, androgens, glucocorticoid, and mineralocorticoid), vitamins (A and D), lipid metabolites, carbohydrates, and xenobiotics. In response to these diverse but critically important mediators, nuclear receptors regulate the homeostatic control of lipids, carbohydrate, cholesterol, and xenobiotic drug metabolism, inflammation, cell differentiation and development, including vascular development. The nuclear receptor family is one of the most important groups of signaling molecules in the body and as such represent some of the most important established and emerging clinical and therapeutic targets. This review will highlight some of the recent trends in nuclear receptor biology related to vascular biology.
Collapse
Affiliation(s)
- David Bishop-Bailey
- Comparative Biomedical Sciences, The Royal Veterinary College, Royal College Street, London, NW1 0TU, UK,
| |
Collapse
|
35
|
Dynamic Trk and G Protein Signalings Regulate Dopaminergic Neurodifferentiation in Human Trophoblast Stem Cells. PLoS One 2015; 10:e0143852. [PMID: 26606046 PMCID: PMC4659658 DOI: 10.1371/journal.pone.0143852] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 11/09/2015] [Indexed: 12/18/2022] Open
Abstract
Understanding the mechanisms in the generation of neural stem cells from pluripotent stem cells is a fundamental step towards successful management of neurodegenerative diseases in translational medicine. Albeit all-trans retinoic acid (RA) has been associated with axon outgrowth and nerve regeneration, the maintenance of differentiated neurons, the association with degenerative disease like Parkinson's disease, and its regulatory molecular mechanism from pluripotent stem cells to neural stem cells remain fragmented. We have previously reported that RA is capable of differentiation of human trophoblast stem cells to dopamine (DA) committed progenitor cells. Intracranial implantation of such neural progenitor cells into the 6-OHDA-lesioned substantia nigra pars compacta successfully regenerates dopaminergic neurons and integrity of the nigrostriatal pathway, ameliorating the behavioral deficits in the Parkinson’s disease rat model. Here, we demonstrated a dynamic molecular network in systematic analysis by addressing spatiotemporal molecular expression, intracellular protein-protein interaction and inhibition, imaging study, and genetic expression to explore the regulatory mechanisms of RA induction in the differentiation of human trophoblast stem cells to DA committed progenitor cells. We focused on the tyrosine receptor kinase (Trk), G proteins, canonical Wnt2B/β-catenin, genomic and non-genomic RA signaling transductions with Tyrosine hydroxylase (TH) gene expression as the differentiation endpoint. We found that at the early stage, integration of TrkA and G protein signalings aims for axonogenesis and morphogenesis, involving the novel RXRα/Gαq/11 and RARβ/Gβ signaling pathways. While at the later stage, five distinct signaling pathways together with epigenetic histone modifications emerged to regulate expression of TH, a precursor of dopamine. RA induction generated DA committed progenitor cells in one day. Our results provided substantial mechanistic evidence that human trophoblast stem cell-derived neural stem cells can potentially be used for neurobiological study, drug discovery, and as an alternative source of cell-based therapy in neurodegenerative diseases like Parkinson’s disease.
Collapse
|
36
|
Kapur R, Zufferey A, Boilard E, Semple JW. Nouvelle cuisine: platelets served with inflammation. THE JOURNAL OF IMMUNOLOGY 2015; 194:5579-87. [PMID: 26048965 DOI: 10.4049/jimmunol.1500259] [Citation(s) in RCA: 214] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Platelets are small cellular fragments with the primary physiological role of maintaining hemostasis. In addition to this well-described classical function, it is becoming increasingly clear that platelets have an intimate connection with infection and inflammation. This stems from several platelet characteristics, including their ability to bind infectious agents and secrete many immunomodulatory cytokines and chemokines, as well as their expression of receptors for various immune effector and regulatory functions, such as TLRs, which allow them to sense pathogen-associated molecular patterns. Furthermore, platelets contain RNA that can be nascently translated under different environmental stresses, and they are able to release membrane microparticles that can transport inflammatory cargo to inflammatory cells. Interestingly, acute infections can also result in platelet breakdown and thrombocytopenia. This report highlights these relatively new aspects of platelets and, thus, their nonhemostatic nature in an inflammatory setting.
Collapse
Affiliation(s)
- Rick Kapur
- Toronto Platelet Immunobiology Group, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario M5B 1W8, Canada; Canadian Blood Services, Toronto, Ontario M5B 1W8, Canada
| | - Anne Zufferey
- Toronto Platelet Immunobiology Group, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario M5B 1W8, Canada
| | - Eric Boilard
- Centre de Recherche en Rhumatologie et Immunologie, Centre de Recherche du Centre Hospitalier Universitaire de Québec, Faculté de Médecine de l'Université Laval, Quebec City, Quebec G1V 4G2, Canada
| | - John W Semple
- Toronto Platelet Immunobiology Group, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario M5B 1W8, Canada; Canadian Blood Services, Toronto, Ontario M5B 1W8, Canada; Department of Pharmacology, University of Toronto, Toronto, Ontario M5B 1W8, Canada; Department of Medicine, University of Toronto, Toronto, Ontario M5B 1W8, Canada; and Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario M5B 1W8, Canada
| |
Collapse
|
37
|
Zeng Z, Sun Z, Huang M, Zhang W, Liu J, Chen L, Chen F, Zhou Y, Lin J, Huang F, Xu L, Zhuang Z, Guo S, Alitongbieke G, Xie G, Xu Y, Lin B, Cao X, Su Y, Zhang XK, Zhou H. Nitrostyrene Derivatives Act as RXRα Ligands to Inhibit TNFα Activation of NF-κB. Cancer Res 2015; 75:2049-60. [PMID: 25795708 DOI: 10.1158/0008-5472.can-14-2435] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2014] [Accepted: 01/21/2015] [Indexed: 11/16/2022]
Abstract
Retinoid X receptor alpha (RXRα) and its N-terminally truncated version, tRXRα, are widely implicated in cancer development and represent intriguing targets for cancer prevention and treatment. Successful manipulation of RXRα and tRXRα requires the identification of their modulators that could produce therapeutic effects. Here, we report that a class of nitrostyrene derivatives bind to RXRα by a unique mechanism, of which the nitro group of nitrostyrene derivatives and Cys432 of RXRα are required for binding. The binding results in the potent activation of Gal4-DBD-RXRα-LBD transactivation. However, the binding inhibits the transactivation of RXRα homodimer, which might be due to the distinct conformation of RXRα homodimer induced by these nitrostyrene derivatives. Two RXRα point mutants with Cys432 substituted with Tyr and Trp, respectively, could mimic the bindings of two nitrostyrene derivatives and have the ability of autotransactivation. In studying the functional consequences of the binding, we show that these nitrostyrene derivatives could potently inhibit the TNFα/NFκB signaling pathway in a tRXRα-dependent manner. tRXRα promotes TNFα-induced NF-κB activation through its interaction with TRAF2 and enhances TNFα-induced ubiquitination of RIP1, which is strongly inhibited by nitrostyrene derivatives. The inhibition of TNFα-induced NF-κB activation results in the synergistic effect of the combination of nitrostyrene derivatives and TNFα on the induction of cancer cell apoptosis. Together, our results show a new class of RXRα modulators that induce apoptosis of cancer cells through their unique binding mode and new mechanism of action.
Collapse
Affiliation(s)
- Zhiping Zeng
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China
| | - Zhe Sun
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China
| | - Mingfeng Huang
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China
| | - Weidong Zhang
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China
| | - Jie Liu
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China
| | - Liqun Chen
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China
| | - Fan Chen
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China
| | - Yuqi Zhou
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China
| | - Jiacheng Lin
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China
| | - Fengyu Huang
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China
| | - Lin Xu
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China
| | - Zixing Zhuang
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China
| | - Shangjie Guo
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China
| | | | - Guobin Xie
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China
| | - Yang Xu
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China
| | - Bingzhen Lin
- Cancer Center, Sanford-Burnham Medical Research Institute, La Jolla, California
| | - Xihua Cao
- Cancer Center, Sanford-Burnham Medical Research Institute, La Jolla, California
| | - Ying Su
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China. Cancer Center, Sanford-Burnham Medical Research Institute, La Jolla, California
| | - Xiao-Kun Zhang
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China. Cancer Center, Sanford-Burnham Medical Research Institute, La Jolla, California.
| | - Hu Zhou
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China.
| |
Collapse
|
38
|
Lannan KL, Sahler J, Kim N, Spinelli SL, Maggirwar SB, Garraud O, Cognasse F, Blumberg N, Phipps RP. Breaking the mold: transcription factors in the anucleate platelet and platelet-derived microparticles. Front Immunol 2015; 6:48. [PMID: 25762994 PMCID: PMC4327621 DOI: 10.3389/fimmu.2015.00048] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 01/26/2015] [Indexed: 01/15/2023] Open
Abstract
Platelets are small anucleate blood cells derived from megakaryocytes. In addition to their pivotal roles in hemostasis, platelets are the smallest, yet most abundant, immune cells and regulate inflammation, immunity, and disease progression. Although platelets lack DNA, and thus no functional transcriptional activities, they are nonetheless rich sources of RNAs, possess an intact spliceosome, and are thus capable of synthesizing proteins. Previously, it was thought that platelet RNAs and translational machinery were remnants from the megakaryocyte. We now know that the initial description of platelets as "cellular fragments" is an antiquated notion, as mounting evidence suggests otherwise. Therefore, it is reasonable to hypothesize that platelet transcription factors are not vestigial remnants from megakaryocytes, but have important, if only partly understood functions. Proteins play multiple cellular roles to minimize energy expenditure for maximum cellular function; thus, the same can be expected for transcription factors. In fact, numerous transcription factors have non-genomic roles, both in platelets and in nucleated cells. Our lab and others have discovered the presence and non-genomic roles of transcription factors in platelets, such as the nuclear factor kappa β (NFκB) family of proteins and peroxisome proliferator-activated receptor gamma (PPARγ). In addition to numerous roles in regulating platelet activation, functional transcription factors can be transferred to vascular and immune cells through platelet microparticles. This method of transcellular delivery of key immune molecules may be a vital mechanism by which platelet transcription factors regulate inflammation and immunity. At the very least, platelets are an ideal model cell to dissect out the non-genomic roles of transcription factors in nucleated cells. There is abundant evidence to suggest that transcription factors in platelets play key roles in regulating inflammatory and hemostatic functions.
Collapse
Affiliation(s)
- Katie L Lannan
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry , Rochester, NY , USA
| | - Julie Sahler
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry , Rochester, NY , USA ; Department of Biological and Environmental Engineering, Cornell University , Ithaca, NY , USA
| | - Nina Kim
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry , Rochester, NY , USA
| | - Sherry L Spinelli
- Department of Pathology and Laboratory Medicine, University of Rochester School of Medicine and Dentistry , Rochester, NY , USA
| | - Sanjay B Maggirwar
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry , Rochester, NY , USA
| | - Olivier Garraud
- Faculté de Médecine, Université de Lyon , Saint-Etienne , France
| | - Fabrice Cognasse
- Faculté de Médecine, Université de Lyon , Saint-Etienne , France ; Etablissement Français du Sang Auvergne-Loire , Saint-Etienne , France
| | - Neil Blumberg
- Department of Pathology and Laboratory Medicine, University of Rochester School of Medicine and Dentistry , Rochester, NY , USA
| | - Richard P Phipps
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry , Rochester, NY , USA ; Department of Pathology and Laboratory Medicine, University of Rochester School of Medicine and Dentistry , Rochester, NY , USA ; Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry , Rochester, NY , USA
| |
Collapse
|
39
|
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]
|
40
|
Fuentes E, Palomo I. Mechanism of antiplatelet action of hypolipidemic, antidiabetic and antihypertensive drugs by PPAR activation. Vascul Pharmacol 2014; 62:162-6. [PMID: 24874279 DOI: 10.1016/j.vph.2014.05.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Revised: 05/08/2014] [Accepted: 05/15/2014] [Indexed: 01/08/2023]
|
41
|
Fuentes E, Rojas A, Palomo I. Role of multiligand/RAGE axis in platelet activation. Thromb Res 2014; 133:308-314. [PMID: 24296115 DOI: 10.1016/j.thromres.2013.11.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Revised: 11/08/2013] [Accepted: 11/12/2013] [Indexed: 02/07/2023]
Abstract
In the context of plaque progression, platelet hyperactivity associated with hyperlipidemia contributes to the development of a pro-thrombotic state. In this context, it has been demonstrated that advanced glycation end products (AGEs) significantly increases platelet activation and receptor for AGEs (RAGE) expression at the platelet surface membrane. In addition to AGEs, other ligands (S100, HMGB1 and amyloid β, among others) of RAGE have raised particular attention in platelet activation. Therefore, in this article we describe platelet hyperactivity by AGEs via RAGE-independent and RAGE-dependent pathways.
Collapse
Affiliation(s)
- Eduardo Fuentes
- Department of Clinical Biochemistry and Immunohematology, Faculty of Health Sciences, Interdisciplinary Excellence Research Program on Healthy Aging (PIEI-ES), Universidad de Talca, Talca, Chile; Centro de Estudios en Alimentos Procesados (CEAP), CONICYT-Regional, Gore Maule, R09I2001, Talca, Chile
| | - Armando Rojas
- Biomedical Research Laboratories, Medicine Faculty, Catholic University of Maule, Talca, Chile.
| | - Iván Palomo
- Department of Clinical Biochemistry and Immunohematology, Faculty of Health Sciences, Interdisciplinary Excellence Research Program on Healthy Aging (PIEI-ES), Universidad de Talca, Talca, Chile; Centro de Estudios en Alimentos Procesados (CEAP), CONICYT-Regional, Gore Maule, R09I2001, Talca, Chile.
| |
Collapse
|
42
|
Abstract
INTRODUCTION Retinoid X receptors (RXRs) are nuclear receptors that act as ligand-dependent transcription factors. RXRs function as homodimers or as heterodimers with other nuclear receptors, such as retinoic acid receptors, PPARs, liver X receptors, farnesoid X receptor, vitamin D receptor or thyroid hormone receptors. RXR ligands (agonists or antagonists) show various physiological effects, depending on their partner receptors. RXR agonist bexarotene (Targretin®) is used for the treatment of cutaneous T-cell lymphoma in clinical practice. RXR agonists were also reported to be useful for treatment of type 2 diabetes, autoimmune disease and Alzheimer's disease. RXR antagonists were also reported to be effective in type 2 diabetes treatment. AREAS COVERED Here patent applications (2007 - 2013) concerning RXR ligands are summarized, and the usefulness of RXR ligands as pharmaceutical agents is discussed. EXPERT OPINION RXR agonists show a wide variety of biological effects. However, they cause serious side effects, such as blood triglyceride elevation, hypothyroidism and others. Thus, for clinical application of RXR agonists, abrogation of these side effects is required. RXR heterodimer-selective agonists and RXR partial agonists exhibiting desired effects without side effects are expected to find clinical application.
Collapse
Affiliation(s)
- Shoya Yamada
- Okayama University Graduate School of Medicine, Division of Pharmaceutical Sciences, Dentistry and Pharmaceutical Sciences , 1-1-1, Tsushima-Naka, Kita-Ku, Okayama 700-8530 , Japan +81 086 251 7963 ; +81 086 251 7963 ;
| | | |
Collapse
|
43
|
Rivadeneyra L, Carestia A, Etulain J, Pozner RG, Fondevila C, Negrotto S, Schattner M. Regulation of platelet responses triggered by Toll-like receptor 2 and 4 ligands is another non-genomic role of nuclear factor-kappaB. Thromb Res 2013; 133:235-43. [PMID: 24331207 DOI: 10.1016/j.thromres.2013.11.028] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Revised: 11/06/2013] [Accepted: 11/26/2013] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Platelets express Toll-like receptors (TLRs) that recognise molecular components of pathogens and, in nucleated cells, elicit immune responses through nuclear factor-kappaB (NF-κB) activation. We have shown that NF-κB mediates platelet activation in response to classical agonists, suggesting that this transcription factor exerts non-genomic functions in platelets. The aim of this study was to determine whether NF-κB activation is a downstream signal involved in TLR2 and 4-mediated platelet responses. MATERIAL AND METHODS Aggregation and ATP release were measured with a Lumi-aggregometer. Fibrinogen binding, P-selectin and CD40 ligand (CD40L) levels and platelet-neutrophil aggregates were measured by cytometry. I kappa B alpha (IκBα) degradation and p65 phosphorylation were determined by Western blot and von Willebrand factor (vWF) by ELISA. RESULTS Platelet stimulation with Pam3CSK4 or LPS resulted in IκBα degradation and p65 phosphorylation. These responses were suppressed by TLR2 and 4 blocking and synergised by thrombin. Aggregation, fibrinogen binding and ATP and vWF release were triggered by Pam3CSK4. LPS did not induce platelet responses per se, except for vWF release, but it did potentiate thrombin-induced aggregation, fibrinogen binding and ATP secretion. Pam3CSK4, but not LPS, induced P-selectin and CD40L expression and mixed aggregate formation. All of these responses, except for CD40L expression, were inhibited in platelets treated with the NF-κB inhibitors BAY 11-7082 or Ro 106-9920. CONCLUSION TLR2 and 4 agonists trigger platelet activation responses through NF-κB. These data show another non-genomic function of NF-κB in platelets and highlight this molecule as a potential target to prevent platelet activation in inflammatory or infectious diseases.
Collapse
Affiliation(s)
- Leonardo Rivadeneyra
- Laboratory of Experimental Thrombosis, Institute of Experimental Medicine (CONICET), National Academy of Medicine, Buenos Aires, Argentina
| | - Agostina Carestia
- Laboratory of Experimental Thrombosis, Institute of Experimental Medicine (CONICET), National Academy of Medicine, Buenos Aires, Argentina
| | - Julia Etulain
- Laboratory of Experimental Thrombosis, Institute of Experimental Medicine (CONICET), National Academy of Medicine, Buenos Aires, Argentina
| | - Roberto G Pozner
- Laboratory of Experimental Thrombosis, Institute of Experimental Medicine (CONICET), National Academy of Medicine, Buenos Aires, Argentina
| | - Carlos Fondevila
- Service of Hematology, Bazterrica Clinic, Buenos Aires, Argentina
| | - Soledad Negrotto
- Laboratory of Experimental Thrombosis, Institute of Experimental Medicine (CONICET), National Academy of Medicine, Buenos Aires, Argentina
| | - Mirta Schattner
- Laboratory of Experimental Thrombosis, Institute of Experimental Medicine (CONICET), National Academy of Medicine, Buenos Aires, Argentina.
| |
Collapse
|
44
|
Fuentes E, Fuentes F, Palomo I. Mechanism of the anti-platelet effect of natural bioactive compounds: Role of peroxisome proliferator-activated receptors activation. Platelets 2013; 25:471-9. [DOI: 10.3109/09537104.2013.849334] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
45
|
Kasimanickam VR, Kasimanickam RK, Rogers HA. Immunolocalization of retinoic acid receptor-alpha, -beta, and -gamma, in bovine and canine sperm. Theriogenology 2013; 79:1010-8. [PMID: 23465288 DOI: 10.1016/j.theriogenology.2013.01.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2012] [Revised: 01/19/2013] [Accepted: 01/19/2013] [Indexed: 01/17/2023]
Abstract
Retinoic acid is an important regulator of cellular proliferation and differentiation. The action of retinoic acid is mediated by retinoic acid receptors (RARs) and the retinoid X receptors. The objective was to elucidate the protein localization and expression of RARα, RARβ, and RARγ in bull and dog sperm. Bull and dog sperm were subjected to an immunostaining procedure to determine presence of RARα, RARβ, and RARγ. We concluded that all three receptors were present in different regions of bull and dog sperm at varying levels. Protein expression in bull and dog sperm lysates was investigated using protein dot-blot analyses. The protein levels of RARα and RARγ were higher than the protein level of RARβ in bull and dog sperm. Protein sequences of RARα, RARβ, and RARγ for bull and dog were 98%, 89%, and 98%, respectively, on similarity alignment. In conclusion, the presence of RARα, RARβ, and RARγ receptors supported their role in sperm structure and function.
Collapse
Affiliation(s)
- Vanmathy R Kasimanickam
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Washington State University, Pullman, Washington, USA
| | | | | |
Collapse
|
46
|
|
47
|
Mitchell HL, Gibbins JM, Griffin BA, Lovegrove JA, Stowell JD, Foot E. Food and Health Forum meeting - nutritional approaches to cardiovascular health: workshop report. NUTR BULL 2012. [DOI: 10.1111/j.1467-3010.2012.01984.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
48
|
Jones CI, Barrett NE, Moraes LA, Gibbins JM, Jackson DE. Endogenous inhibitory mechanisms and the regulation of platelet function. Methods Mol Biol 2012; 788:341-66. [PMID: 22130718 DOI: 10.1007/978-1-61779-307-3_23] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The response of platelets to changes in the immediate environment is always a balance between activatory and inhibitory signals, the cumulative effect of which is either activation or quiescence. This is true of platelets in free flowing blood and of their regulation of haemostasis and thrombosis. In this review, we consider the endogenous inhibitory mechanisms that combine to regulate platelet activation. These include those derived from the endothelium (nitric oxide, prostacyclin, CD39), inhibitory receptors on the surface of platelets (platelet endothelial cell adhesion molecule-1, carcinoembryonic antigen cell adhesion molecule 1, G6b-B - including evidence for the role of Ig-ITIM superfamily members in the negative regulation of ITAM-associated GPVI platelet-collagen interactions and GPCR-mediated signalling and in positive regulation of "outside-in" integrin α(IIb)β(3)-mediated signalling), intracellular inhibitory receptors (retinoic X receptor, glucocorticoid receptor, peroxisome proliferator-activated receptors, liver X receptor), and emerging inhibitory pathways (canonical Wnt signalling, Semaphorin 3A, endothelial cell specific adhesion molecule, and junctional adhesion molecule-A).
Collapse
Affiliation(s)
- Chris I Jones
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, The University of Reading, Reading, UK
| | | | | | | | | |
Collapse
|
49
|
Dawson MI, Xia Z. The retinoid X receptors and their ligands. Biochim Biophys Acta Mol Cell Biol Lipids 2011; 1821:21-56. [PMID: 22020178 DOI: 10.1016/j.bbalip.2011.09.014] [Citation(s) in RCA: 269] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2011] [Revised: 08/23/2011] [Accepted: 09/23/2011] [Indexed: 12/12/2022]
Abstract
This chapter presents an overview of the current status of studies on the structural and molecular biology of the retinoid X receptor subtypes α, β, and γ (RXRs, NR2B1-3), their nuclear and cytoplasmic functions, post-transcriptional processing, and recently reported ligands. Points of interest are the different changes in the ligand-binding pocket induced by variously shaped agonists, the communication of the ligand-bound pocket with the coactivator binding surface and the heterodimerization interface, and recently identified ligands that are natural products, those that function as environmental toxins or drugs that had been originally designed to interact with other targets, as well as those that were deliberately designed as RXR-selective transcriptional agonists, synergists, or antagonists. Of these synthetic ligands, the general trend in design appears to be away from fully aromatic rigid structures to those containing partial elements of the flexible tetraene side chain of 9-cis-retinoic acid. This article is part of a Special Issue entitled Advances in High Density Lipoprotein Formation and Metabolism: A Tribute to John F. Oram (1945-2010).
Collapse
Affiliation(s)
- Marcia I Dawson
- Cancer Center, Sanford-Burn Medical Research Institute, 10901 North Torrey Pines Rd., La Jolla, CA 93207, USA.
| | | |
Collapse
|
50
|
The PPAR-Platelet Connection: Modulators of Inflammation and Potential Cardiovascular Effects. PPAR Res 2011; 2008:328172. [PMID: 18288284 PMCID: PMC2233896 DOI: 10.1155/2008/328172] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2007] [Accepted: 11/06/2007] [Indexed: 01/08/2023] Open
Abstract
Historically, platelets were viewed as simple anucleate cells responsible for initiating thrombosis and maintaining
hemostasis, but clearly they are also key mediators of inflammation and immune cell activation. An emerging body of
evidence links platelet function and thrombosis to vascular inflammation. peroxisome proliferator-activated receptors
(PPARs) play a major role in modulating inflammation and, interestingly, PPARs (PPARβ/δ and PPARγ) were recently
identified in platelets. Additionally, PPAR agonists attenuate platelet activation; an important discovery for two reasons.
First, activated platelets are formidable antagonists that initiate and prolong a cascade of events that contribute to
cardiovascular disease (CVD) progression. Dampening platelet release of proinflammatory mediators, including
CD40 ligand (CD40L, CD154), is essential to hinder this cascade. Second, understanding the biologic importance
of platelet PPARs and the mechanism(s) by which PPARs regulate platelet activation will be imperative in designing
therapeutic strategies lacking the deleterious or unwanted side effects of current treatment options.
Collapse
|