1
|
Oh EB, Shin HJ, Yu H, Jang J, Park JW, Chang TS. NADPH oxidase 1/4 dual inhibitor setanaxib suppresses platelet activation and thrombus formation. Life Sci 2024; 357:123061. [PMID: 39293714 DOI: 10.1016/j.lfs.2024.123061] [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: 07/06/2024] [Revised: 09/07/2024] [Accepted: 09/12/2024] [Indexed: 09/20/2024]
Abstract
AIMS The production of reactive oxygen species (ROS) by NADPH oxidase (NOX) is able to induce platelet activation, making NOX a promising target for antiplatelet therapy. In this study, we examined the effects of setanaxib, a dual NOX1/4 inhibitor, on human platelet function and ROS-related signaling pathways. MATERIALS AND METHODS In collagen-stimulated human platelets, aggregometry, assessment of ROS and Ca2+, immunoblotting, ELISA, flow cytometry, platelet adhesion assay, and assessment of mouse arterial thrombosis were performed in this study. KEY FINDINGS Setanaxib inhibited both intracellular and extracellular ROS production in collagen-activated platelets. Additionally, setanaxib significantly inhibited collagen-induced platelet aggregation, P-selectin exposure from α-granule release, and ATP release from dense granules. Setanaxib blocked the specific tyrosine phosphorylation-mediated activation of Syk, LAT, Vav1, and Btk within collagen receptor signaling pathways, leading to reduced activation of PLCγ2, PKC, and Ca2+ mobilization. Setanaxib also inhibited collagen-induced activation of integrin αIIbβ3, which is linked to increased cGMP levels and VASP phosphorylation. Furthermore, setanaxib suppressed collagen-induced p38 MAPK activation, resulting in decreased phosphorylation of cytosolic PLA2 and reduced TXA2 generation. Setanaxib also inhibited ERK5 activation, affecting the exposure of procoagulant phosphatidylserine. Setanaxib reduced thrombus formation under shear conditions by preventing platelet adhesion to collagen. Finally, in vivo administration of setanaxib in animal models led to the inhibition of arterial thrombosis. SIGNIFICANCE This study is the first to show that setanaxib suppresses ROS generation, platelet activation, and collagen-induced thrombus formation, suggesting its potential use in treating thrombotic or cardiovascular diseases.
Collapse
Affiliation(s)
- Eun Bee Oh
- College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea.
| | - Hye Ji Shin
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea.
| | - Hyunseong Yu
- College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea.
| | - Joara Jang
- College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea.
| | - Ji Won Park
- College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea.
| | - Tong-Shin Chang
- College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea; Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea.
| |
Collapse
|
2
|
Cai N, Gao X, Li W, Yang L, Zhao J, Qu J, Zhou Y. Novel trifluoromethyl ketone derivatives as oral cPLA 2/COX-2 dual inhibitors for resolution of inflammation in rheumatoid arthritis. Bioorg Chem 2024; 148:107453. [PMID: 38761708 DOI: 10.1016/j.bioorg.2024.107453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 04/25/2024] [Accepted: 05/12/2024] [Indexed: 05/20/2024]
Abstract
Thirty-five trifluoromethyl hydrazones and seventeen trifluoromethyl oxime esters were designed and synthesized via molecular hybridization. All the target compounds were initially screened for in vitro anti-inflammatory activity by assessing their inhibitory effect on NO release in LPS-stimulated RAW264.7 cells, and the optimal compound was finally identified as 2-(3-Methoxyphenyl)-N'-((6Z,9Z,12Z,15Z)-1,1,1-trifluorohenicosa-6,9,12,15-tetraen-2-ylidene)acetohydrazide (F26, IC50 = 4.55 ± 0.92 μM) with no cytotoxicity. Moreover, F26 potently reduced the production of PGE2 in LPS-stimulated RAW264.7 cells compared to indomethacin. The interaction of F26 with COX-2 and cPLA2 was directly verified by the CETSA technique. F26 was found to modulate the phosphorylation levels of p38 MAPK and NF-κB p65, as well as the protein expression of IκB, cPLA2, COX-2, and iNOS in LPS-stimulated rat peritoneal macrophages. Additionally, F26 was observed to prevent the nuclear translocation of NF-κB p65 in LPS-stimulated rat peritoneal macrophages by immunofluorescence localization. Therefore, the aforementioned in vitro experiments demonstrated that F26 blocked the p38 MAPK and NF-κB pathways by binding to COX-2 and cPLA2. In the adjuvant-induced arthritis model, F26 demonstrated a significant effect in preventing arthritis symptoms and inflammatory status in rats, exerting an immunomodulatory role by regulating the homeostasis between Th17 and Treg through inhibition of the p38 MAPK/cPLA2/COX-2/PGE2 and NF-κB pathways. Encouragingly, F26 caused less acute ulcerogenicity in rats at a dose of 50 mg/kg compared to indomethacin. Overall, F26 is a promising candidate worthy of further investigation for treating inflammation and associated pain with lesser gastrointestinal irritation, as well as other symptoms in which cPLA2 and COX-2 are implicated in the pathophysiology.
Collapse
Affiliation(s)
- Nan Cai
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Engineering, School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, PR China
| | - Xiang Gao
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Engineering, School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, PR China
| | - Wenjing Li
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Engineering, School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, PR China
| | - Li Yang
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Engineering, School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, PR China.
| | - Jinfeng Zhao
- Instrumental Analysis Center, Dalian University of Technology, 2 Linggong Road, Dalian 116024, PR China.
| | - Jingping Qu
- State Key Laboratory of Fine Chemicals, School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, PR China.
| | - Yuhan Zhou
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Engineering, School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, PR China.
| |
Collapse
|
3
|
Cai N, Gao X, Yang L, Li W, Sun W, Zhang S, Zhao J, Qu J, Zhou Y. Discovery of novel NSAID hybrids as cPLA 2/COX-2 dual inhibitors alleviating rheumatoid arthritis via inhibiting p38 MAPK pathway. Eur J Med Chem 2024; 267:116176. [PMID: 38286094 DOI: 10.1016/j.ejmech.2024.116176] [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: 11/22/2023] [Revised: 01/20/2024] [Accepted: 01/22/2024] [Indexed: 01/31/2024]
Abstract
A series of NSAIDs hybrid molecules were synthesized and characterized, and their ability to inhibit NO release in LPS-induced RAW264.7 macrophages was evaluated. Most of the compounds showed significant anti-inflammatory activity in vitro, of which (2E,6Z,9Z,12Z,15Z)-1,1,1-trifluorohenicosa-2,6,9,12,15-pentaen-2-yl 2-(4-benzoylphenyl) propanoate (VI-60) was the most optimal (IC50 = 3.85 ± 0.25 μΜ) and had no cytotoxicity. In addition, VI-60 notably reduced the production of PGE2 in LPS-stimulated RAW264.7 cells compared to ketoprofen. Futhur more, VI-60 significantly inhibited the expression of iNOS, cPLA2, and COX-2 and the phosphorylation of p38 MAPK in LPS-stimulated RAW264.7 cells. The binding of VI-60 to cPLA2 and COX-2 was directly verified by the CETSA technique. In vivo studies illustrated that VI-60 exerted an excellent therapeutic effect on adjuvant-induced arthritis in rats by regulating the balance between Th17 and Treg through inhibiting the p38 MAPK/cPLA2/COX-2/PGE2 pathway. Encouragingly, VI-60 showed a lower ulcerative potential in rats at a dose of 50 mg/kg compared to ketoprofen. In conclusion, the hybrid molecules of NSAIDs and trifluoromethyl enols are promising candidates worthy of further investigation for the treatment of inflammation, pain, and other symptoms in which cPLA2 and COX-2 play a role in their etiology.
Collapse
Affiliation(s)
- Nan Cai
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Engineering, School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian, 116024, PR China.
| | - Xiang Gao
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Engineering, School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian, 116024, PR China.
| | - Li Yang
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Engineering, School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian, 116024, PR China.
| | - Wenjing Li
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Engineering, School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian, 116024, PR China.
| | - Wuding Sun
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Engineering, School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian, 116024, PR China.
| | - Shuaibo Zhang
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Engineering, School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian, 116024, PR China.
| | - Jinfeng Zhao
- Instrumental Analysis Center, Dalian University of Technology, 2 Linggong Road, Dalian, 116024, PR China.
| | - Jingping Qu
- State Key Laboratory of Fine Chemicals, School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian, 116024, PR China.
| | - Yuhan Zhou
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Engineering, School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian, 116024, PR China.
| |
Collapse
|
4
|
Manne BK, Campbell RA, Bhatlekar S, Ajanel A, Denorme F, Portier I, Middleton EA, Tolley ND, Kosaka Y, Montenont E, Guo L, Rowley JW, Bray PF, Jacob S, Fukanaga R, Proud C, Weyrich AS, Rondina MT. MAPK-interacting kinase 1 regulates platelet production, activation, and thrombosis. Blood 2022; 140:2477-2489. [PMID: 35930749 PMCID: PMC9918849 DOI: 10.1182/blood.2022015568] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 07/06/2022] [Accepted: 07/20/2022] [Indexed: 12/13/2022] Open
Abstract
The MAPK-interacting kinase (Mnk) family includes Mnk1 and Mnk2, which are phosphorylated and activated in response to extracellular stimuli. Mnk1 contributes to cellular responses by regulating messenger RNA (mRNA) translation, and mRNA translation influences platelet production and function. However, the role of Mnk1 in megakaryocytes and platelets has not previously been studied. The present study investigated Mnk1 in megakaryocytes and platelets using both pharmacological and genetic approaches. We demonstrate that Mnk1, but not Mnk2, is expressed and active in human and murine megakaryocytes and platelets. Stimulating human and murine megakaryocytes and platelets induced Mnk1 activation and phosphorylation of eIF4E, a downstream target of activated Mnk1 that triggers mRNA translation. Mnk1 inhibition or deletion significantly diminished protein synthesis in megakaryocytes as measured by polysome profiling and [35S]-methionine incorporation assays. Depletion of Mnk1 also reduced megakaryocyte ploidy and proplatelet forming megakaryocytes in vitro and resulted in thrombocytopenia. However, Mnk1 deletion did not affect the half-life of circulating platelets. Platelets from Mnk1 knockout mice exhibited reduced platelet aggregation, α granule secretion, and integrin αIIbβ3 activation. Ribosomal footprint sequencing indicated that Mnk1 regulates the translation of Pla2g4a mRNA (which encodes cPLA2) in megakaryocytes. Consistent with this, Mnk1 ablation reduced cPLA2 activity and thromboxane generation in platelets and megakaryocytes. In vivo, Mnk1 ablation protected against platelet-dependent thromboembolism. These results provide previously unrecognized evidence that Mnk1 regulates mRNA translation and cellular activation in platelets and megakaryocytes, endomitosis and thrombopoiesis, and thrombosis.
Collapse
Affiliation(s)
| | - Robert A. Campbell
- University of Utah Molecular Medicine Program, Salt Lake City, UT
- Department of Internal Medicine, University of Utah Health, Salt Lake City, UT
- Department of Pathology, University of Utah Health, Salt Lake City, UT
| | - Seema Bhatlekar
- University of Utah Molecular Medicine Program, Salt Lake City, UT
| | - Abigail Ajanel
- University of Utah Molecular Medicine Program, Salt Lake City, UT
- Department of Pathology, University of Utah Health, Salt Lake City, UT
| | - Frederik Denorme
- University of Utah Molecular Medicine Program, Salt Lake City, UT
| | - Irina Portier
- University of Utah Molecular Medicine Program, Salt Lake City, UT
| | - Elizabeth A. Middleton
- University of Utah Molecular Medicine Program, Salt Lake City, UT
- Department of Internal Medicine, University of Utah Health, Salt Lake City, UT
| | - Neal D. Tolley
- University of Utah Molecular Medicine Program, Salt Lake City, UT
| | - Yasuhiro Kosaka
- University of Utah Molecular Medicine Program, Salt Lake City, UT
| | - Emilie Montenont
- University of Utah Molecular Medicine Program, Salt Lake City, UT
| | - Li Guo
- University of Utah Molecular Medicine Program, Salt Lake City, UT
| | - Jesse W. Rowley
- University of Utah Molecular Medicine Program, Salt Lake City, UT
- Department of Internal Medicine, University of Utah Health, Salt Lake City, UT
| | - Paul F. Bray
- University of Utah Molecular Medicine Program, Salt Lake City, UT
- Department of Internal Medicine, University of Utah Health, Salt Lake City, UT
| | - Shancy Jacob
- University of Utah Molecular Medicine Program, Salt Lake City, UT
| | - Rikiro Fukanaga
- Department of Biochemistry, Osaka University of Pharmaceutical Sciences, Osaka, Japan
| | - Christopher Proud
- Lifelong Health, South Australian Health & Medical Research Institute, Adelaide, Australia
- Department of Biological Sciences, University of Adelaide, Adelaide, Australia
| | - Andrew S. Weyrich
- University of Utah Molecular Medicine Program, Salt Lake City, UT
- Department of Internal Medicine, University of Utah Health, Salt Lake City, UT
| | - Matthew T. Rondina
- University of Utah Molecular Medicine Program, Salt Lake City, UT
- Department of Internal Medicine, University of Utah Health, Salt Lake City, UT
- Department of Pathology, University of Utah Health, Salt Lake City, UT
- Department of Internal Medicine and the Geriatric Research, Education, and Clinical Center (GRECC), George E. Wahlen Veterans Affairs Medical Center (VAMC), Salt Lake City, UT
| |
Collapse
|
5
|
de Melo-Braga MN, Moreira RDS, Gervásio JHDB, Felicori LF. Overview of protein posttranslational modifications in Arthropoda venoms. J Venom Anim Toxins Incl Trop Dis 2022; 28:e20210047. [PMID: 35519418 PMCID: PMC9036706 DOI: 10.1590/1678-9199-jvatitd-2021-0047] [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: 04/12/2021] [Accepted: 08/27/2021] [Indexed: 11/22/2022] Open
Abstract
Accidents with venomous animals are a public health issue worldwide. Among the species involved in these accidents are scorpions, spiders, bees, wasps, and other members of the phylum Arthropoda. The knowledge of the function of proteins present in these venoms is important to guide diagnosis, therapeutics, besides being a source of a large variety of biotechnological active molecules. Although our understanding about the characteristics and function of arthropod venoms has been evolving in the last decades, a major aspect crucial for the function of these proteins remains poorly studied, the posttranslational modifications (PTMs). Comprehension of such modifications can contribute to better understanding the basis of envenomation, leading to improvements in the specificities of potential therapeutic toxins. Therefore, in this review, we bring to light protein/toxin PTMs in arthropod venoms by accessing the information present in the UniProtKB/Swiss-Prot database, including experimental and putative inferences. Then, we concentrate our discussion on the current knowledge on protein phosphorylation and glycosylation, highlighting the potential functionality of these modifications in arthropod venom. We also briefly describe general approaches to study "PTM-functional-venomics", herein referred to the integration of PTM-venomics with a functional investigation of PTM impact on venom biology. Furthermore, we discuss the bottlenecks in toxinology studies covering PTM investigation. In conclusion, through the mining of PTMs in arthropod venoms, we observed a large gap in this field that limits our understanding on the biology of these venoms, affecting the diagnosis and therapeutics development. Hence, we encourage community efforts to draw attention to a better understanding of PTM in arthropod venom toxins.
Collapse
Affiliation(s)
- Marcella Nunes de Melo-Braga
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Raniele da Silva Moreira
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - João Henrique Diniz Brandão Gervásio
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Liza Figueiredo Felicori
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| |
Collapse
|
6
|
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
|
7
|
Duan X, Perveen R, Dandamudi A, Adili R, Johnson J, Funk K, Berryman M, Davis AK, Holinstat M, Zheng Y, Akbar H. Pharmacologic targeting of Cdc42 GTPase by a small molecule Cdc42 activity-specific inhibitor prevents platelet activation and thrombosis. Sci Rep 2021; 11:13170. [PMID: 34162972 PMCID: PMC8222210 DOI: 10.1038/s41598-021-92654-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 05/27/2021] [Indexed: 01/14/2023] Open
Abstract
Gene targeting of Cdc42 GTPase has been shown to inhibit platelet activation. In this study, we investigated a hypothesis that inhibition of Cdc42 activity by CASIN, a small molecule Cdc42 Activity-Specific INhibitor, may down regulate platelet activation and thrombus formation. We investigated the effects of CASIN on platelet activation in vitro and thrombosis in vivo. In human platelets, CASIN, but not its inactive analog Pirl7, blocked collagen induced activation of Cdc42 and inhibited phosphorylation of its downstream effector, PAK1/2. Moreover, addition of CASIN to washed human platelets inhibited platelet spreading on immobilized fibrinogen. Treatment of human platelets with CASIN inhibited collagen or thrombin induced: (a) ATP secretion and platelet aggregation; and (b) phosphorylation of Akt, ERK and p38-MAPK. Pre-incubation of platelets with Pirl7, an inactive analog of CASIN, failed to inhibit collagen induced aggregation. Washing of human platelets after incubation with CASIN eliminated its inhibitory effect on collagen induced aggregation. Intraperitoneal administration of CASIN to wild type mice inhibited ex vivo aggregation induced by collagen but did not affect the murine tail bleeding times. CASIN administration, prior to laser-induced injury in murine cremaster muscle arterioles, resulted in formation of smaller and unstable thrombi compared to control mice without CASIN treatment. These data suggest that pharmacologic targeting of Cdc42 by specific and reversible inhibitors may lead to the discovery of novel antithrombotic agents.
Collapse
Affiliation(s)
- Xin Duan
- Division of Experimental Hematology and Cancer Biology, Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, 45229, USA
| | - Rehana Perveen
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, 45701, USA
| | - Akhila Dandamudi
- Division of Experimental Hematology and Cancer Biology, Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, 45229, USA
| | - Reheman Adili
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - James Johnson
- Division of Experimental Hematology and Cancer Biology, Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, 45229, USA
| | - Kevin Funk
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, 45701, USA
| | - Mark Berryman
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, 45701, USA
| | - Ashley Kuenzi Davis
- Division of Experimental Hematology and Cancer Biology, Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, 45229, USA
| | - Michael Holinstat
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Yi Zheng
- Division of Experimental Hematology and Cancer Biology, Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, 45229, USA.
| | - Huzoor Akbar
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, 45701, USA.
| |
Collapse
|
8
|
Manjuprasanna VN, Urs AP, Rudresha GV, Milan Gowda MD, Jayachandra K, Hiremath V, Rajaiah R, Vishwanath BS. Drupin, a thrombin-like protease prompts platelet activation and aggregation through protease-activated receptors. J Cell Biochem 2021; 122:870-881. [PMID: 33748988 DOI: 10.1002/jcb.29917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 02/24/2021] [Accepted: 03/04/2021] [Indexed: 11/10/2022]
Abstract
Hemostasis is a proteolytically regulated process that requires activation of platelets and the blood coagulation cascade upon vascular injury. Activated platelets create a thrombogenic environment and amplify the coagulation process. Plant latex proteases (PLPs) have been used as therapeutic components to treat various ailments by folk healers. One of the main applications of plant latices is to stop bleeding from minor injuries and to enhance wound healing activity. Although many studies have reported the pro-coagulant activities of PLPs, an in-depth investigation is required to understand the mechanism of action of PLPs on platelets. Here, the effect of PLPs on platelet aggregation was studied systematically to validate the observed pharmacological effect by folk healers. Among 29 latices from the Ficus genus tested, Ficus drupacea exhibited potent pro-coagulant and thrombin-like activity. Drupin, a thrombin-like cysteine protease responsible for platelet aggregation was purified from F. drupacea latex. Drupin exhibits pro-coagulant activity and reduces the bleeding time in mice tail. It induces platelet aggregation by activating mitogen-activated protein kinases and the nuclear factor-κB and PI3K/Akt signalling cascade, which, in turn, phosphorylats, cytosolic phospholipase A2 leading to the release of thromboxane A2 from the granules to activate the nearby platelets to aggregate. Furthermore, we investigated the involvement of protease-activated receptors in drupin-induced platelet aggregation using specific protease activated receptor 1 (PAR1) and PAR4 receptor antagonists. The results confirmed that the drupin-induced platelet aggregation was mediated by both PAR1 and PAR4, synergistically. Overall, drupin reduces the bleeding time by exerting pro-coagulant activity and induces platelet aggregation by activating the intracellular signalling cascade.
Collapse
Affiliation(s)
| | - Amog P Urs
- Department of Studies in Biochemistry, University of Mysore, Manasagangothri, Mysuru, Karnataka, India
| | - Gotravalli V Rudresha
- Department of Studies in Biochemistry, University of Mysore, Manasagangothri, Mysuru, Karnataka, India
| | | | - Krishnegowda Jayachandra
- Department of Studies in Biochemistry, University of Mysore, Manasagangothri, Mysuru, Karnataka, India
| | - Vilas Hiremath
- Vijayashree Diagnostics, Specialized Coagulation Lab, Bengaluru, Karnataka, India
| | - Rajesh Rajaiah
- Department of Studies in Molecular Biology, University of Mysore, Manasagangothri, Mysuru, Karnataka, India
| | - Bannikuppe S Vishwanath
- Department of Studies in Biochemistry, University of Mysore, Manasagangothri, Mysuru, Karnataka, India.,Department of Studies in Molecular Biology, University of Mysore, Manasagangothri, Mysuru, Karnataka, India
| |
Collapse
|
9
|
Patel P, Shaik NF, Zhou Y, Golla K, McKenzie SE, Naik UP. Apoptosis signal-regulating kinase 1 regulates immune-mediated thrombocytopenia, thrombosis, and systemic shock. J Thromb Haemost 2020; 18:3013-3028. [PMID: 32767736 PMCID: PMC7831975 DOI: 10.1111/jth.15049] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 07/07/2020] [Accepted: 07/31/2020] [Indexed: 01/07/2023]
Abstract
BACKGROUND Immune complexes (ICs) bind to and activate platelets via FcγRIIA, causing patients to experience thrombocytopenia, as well as an increased risk of forming occlusive thrombi. Although platelets have been shown to mediate IC-induced pathologies, the mechanisms involved have yet to be fully elucidated. We identified that apoptosis signal-regulating kinase 1 (ASK1) is present in both human and mouse platelets and potentiates many platelet functions. OBJECTIVES Here we set out to study ASK1's role in regulating IC-mediated platelet functions in vitro and IC-induced pathologies using an in vivo mouse model. METHODS Using human platelets treated with an ASK1-specific inhibitor and platelets from FCGR2A/Ask1-/- transgenic mice, we examined various platelet functions induced by model ICs in vitro and in vivo. RESULTS We found that ASK1 was activated in human platelets following cross-linking of FcγRIIA using either anti-hCD9 or IV.3 + goat-anti-mouse. Although genetic deletion or inhibition of ASK1 significantly attenuated anti-CD9-induced platelet aggregation, activation of the canonical FcγRIIA signaling targets Syk and PLCγ2 was unaffected. We further found that anti-mCD9-induced cPla2 phosphorylation and TxA2 generation is delayed in Ask1 null transgenic mouse platelets leading to diminished δ-granule secretion. In vivo, absence of Ask1 protected FCGR2A transgenic mice from thrombocytopenia, thrombosis, and systemic shock following injection of anti-mCD9. In whole blood microfluidics, platelet adhesion and thrombus formation on fibrinogen was enhanced by Ask1. CONCLUSIONS These findings suggest that ASK1 inhibition may be a potential target for the treatment of IC-induced shock and other immune-mediated thrombotic disorders.
Collapse
Affiliation(s)
- Pravin Patel
- Cardeza Center for Hemostasis, Thrombosis, and Vascular Biology, Cardeza Foundation for Hematologic Research, Department of Medicine, Thomas Jefferson University, Philadelphia, PA
| | - Noor F. Shaik
- Cardeza Center for Hemostasis, Thrombosis, and Vascular Biology, Cardeza Foundation for Hematologic Research, Department of Medicine, Thomas Jefferson University, Philadelphia, PA
| | - Yuhang Zhou
- Cardeza Center for Hemostasis, Thrombosis, and Vascular Biology, Cardeza Foundation for Hematologic Research, Department of Medicine, Thomas Jefferson University, Philadelphia, PA
- Dell Children’s Hospital, University of Texas, Austin, TX
| | - Kalyan Golla
- Cardeza Center for Hemostasis, Thrombosis, and Vascular Biology, Cardeza Foundation for Hematologic Research, Department of Medicine, Thomas Jefferson University, Philadelphia, PA
- Center for Blood Research, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Steven E. McKenzie
- Cardeza Center for Hemostasis, Thrombosis, and Vascular Biology, Cardeza Foundation for Hematologic Research, Department of Medicine, Thomas Jefferson University, Philadelphia, PA
| | - Ulhas P. Naik
- Cardeza Center for Hemostasis, Thrombosis, and Vascular Biology, Cardeza Foundation for Hematologic Research, Department of Medicine, Thomas Jefferson University, Philadelphia, PA
| |
Collapse
|
10
|
Patel P, Naik UP. Platelet MAPKs-a 20+ year history: What do we really know? J Thromb Haemost 2020; 18:2087-2102. [PMID: 32574399 DOI: 10.1111/jth.14967] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 06/09/2020] [Accepted: 06/10/2020] [Indexed: 01/01/2023]
Abstract
The existence of mitogen activated protein kinases (MAPKs) in platelets has been known for more than 20 years. Since that time hundreds of reports have been published describing the conditions that cause MAPK activation in platelets and their role in regulating diverse platelet functions from the molecular to physiological level. However, this cacophony of reports, with inconsistent and sometimes contradictory findings, has muddied the waters leading to great confusion. Since the last review of platelet MAPKs was published more than a decade ago, there have been more than 50 reports, including the description of novel knockout mouse models, that have furthered our knowledge. Therefore, we undertook an extensive literature review to delineate what is known about platelet MAPKs. We specifically discuss what is currently known about how MAPKs are activated and what signaling cascades they regulate in platelets incorporating recent findings from knockout mouse models. In addition, we will discuss the role each MAPK plays in regulating distinct platelet functions. In doing so, we hope to clarify the role for MAPKs and identify knowledge gaps in this field that await future researchers. In addition, we discuss the limitations of current studies with a particular focus on the off-target effects of commonly used MAPK inhibitors. We conclude with a look at the clinical utility of MAPK inhibitors as potential antithrombotic therapies with an analysis of current clinical trial data.
Collapse
Affiliation(s)
- Pravin Patel
- Department of Medicine, Cardeza Center for Hemostasis, Thrombosis, and Vascular Biology, Cardeza Foundation for Hematologic Research, Thomas Jefferson University, Philadelphia, PA, USA
| | - Ulhas P Naik
- Department of Medicine, Cardeza Center for Hemostasis, Thrombosis, and Vascular Biology, Cardeza Foundation for Hematologic Research, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
11
|
Inamdar VV, Reddy H, Dangelmaier C, Kostyak JC, Kunapuli SP. The protein tyrosine phosphatase PTPN7 is a negative regulator of ERK activation and thromboxane generation in platelets. J Biol Chem 2019; 294:12547-12554. [PMID: 31266805 DOI: 10.1074/jbc.ra119.007735] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 06/20/2019] [Indexed: 11/06/2022] Open
Abstract
Protein tyrosine phosphatase nonreceptor type 7 (PTPN7), also called hematopoietic protein tyrosine phosphatase, controls extracellular signal-regulated protein kinase 1/2 (ERK1/2) and p38 mitogen-activated protein kinase in T lymphocytes. Because ERK1/2 plays an important role in regulating thromboxane A2 (TXA2) generation in platelets, we investigated the function of PTPN7 in these cells. Using immunoblot analysis, we detected PTPN7 in both human and mouse platelets but not in PTPN7-null mice. PTPN7 KO mouse platelets exhibited increased platelet functional responses, including aggregation, dense granule secretion, and TXA2 generation, compared with platelets from WT littermates, upon stimulation with both G protein-coupled receptor (GPCR) and glycoprotein VI (GPVI) agonists. Using the GPCR agonist AYPGKF in the presence of the COX inhibitor indomethacin, we found that PTPN7 KO mouse platelets aggregated and secreted to the same extent as WT platelets, suggesting that elevated TXA2 is responsible for the potentiation of platelet functional responses in PTPN7-KO platelets. Phosphorylation of ERK1/2 was also elevated in PTPN7 KO platelets. Stimulation of platelets with the GPVI agonist collagen-related peptide along with the COX inhibitor indomethacin did not result in phosphorylation of ERK1/2, indicating that GPVI-mediated ERK phosphorylation occurs through TXA2 Although bleeding times did not significantly differ between PTPN7-null and WT mice, time to death was significantly faster in PTPN7-null mice than in WT mice in a pulmonary thromboembolism model. We conclude that PTPN7 regulates platelet functional responses downstream of GPCR agonists, but not GPVI agonists, through inhibition of ERK activation and thromboxane generation.
Collapse
Affiliation(s)
- Vaishali V Inamdar
- Sol Sherry Thrombosis Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania 19140
| | - Haritha Reddy
- Sol Sherry Thrombosis Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania 19140
| | - Carol Dangelmaier
- Sol Sherry Thrombosis Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania 19140
| | - John C Kostyak
- Sol Sherry Thrombosis Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania 19140
| | - Satya P Kunapuli
- Sol Sherry Thrombosis Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania 19140.
| |
Collapse
|
12
|
Guidetti GF, Torti M, Canobbio I. Focal Adhesion Kinases in Platelet Function and Thrombosis. Arterioscler Thromb Vasc Biol 2019; 39:857-868. [DOI: 10.1161/atvbaha.118.311787] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The focal adhesion kinase family includes 2 homolog members, FAK and Pyk2 (proline-rich tyrosine kinase 2), primarily known for their roles in nucleated cells as regulators of cytoskeletal dynamics and cell adhesion. FAK and Pyk2 are also expressed in megakaryocytes and platelets and are activated by soluble agonists and on adhesion to the subendothelial matrix. Despite high sequence homology and similar molecular organization, FAK and Pyk2 play different roles in platelet function. Whereas FAK serves mostly as a traditional focal adhesion kinase activated downstream of integrins, Pyk2 coordinates multiple signals from different receptors. FAK, but not Pyk2, is involved in megakaryocyte maturation and platelet production. In circulating platelets, FAK is recruited by integrin αIIbβ3 to regulate hemostasis, whereas it plays minimal roles in thrombosis. By contrast, Pyk2 is implicated in platelet activation and is an important regulator of thrombosis. The direct activation of Pyk2 by calcium ions provides a connection between GPCRs (G-protein coupled receptors) and Src family kinases. In this review, we provide the comprehensive overview of >20 years of investigations on the role and regulation of focal adhesion kinases in blood platelets, highlighting common and distinctive features of FAK and Pyk2 in hemostasis and thrombosis.
Collapse
Affiliation(s)
| | - Mauro Torti
- From the Department of Biology and Biotechnology, University of Pavia, Italy
| | - Ilaria Canobbio
- From the Department of Biology and Biotechnology, University of Pavia, Italy
| |
Collapse
|
13
|
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: 13.0] [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
|
14
|
Zhu S, Li J, Bing Y, Yan W, Zhu Y, Xia B, Chen M. Diet-Induced Hyperhomocysteinaemia Increases Intestinal Inflammation in an Animal Model of Colitis. J Crohns Colitis 2015; 9:708-19. [PMID: 26071411 DOI: 10.1093/ecco-jcc/jjv094] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Accepted: 05/22/2015] [Indexed: 02/08/2023]
Abstract
BACKGROUND Hyperhomocysteinaemia [HHcy] is a common phenomenon observed in patients with inflammatory bowel disease [IBD]. Homocysteine is a pro-inflammatory molecule and has been identified as a risk factor for cardiovascular and cerebral diseases. Whether HHcy contributes to the chronic inflammation of the colon in IBD has rarely been explored. The aim of this study was to investigate the effect of HHcy on dextran sulphate sodium [DSS]-induced colitis. METHODS Wistar rats were randomly divided into eight groups: [1] Control; [2] HHcy; [3] p38 inhibitor; [4] DSS; [5] HHcy + DSS; [6] HHcy + DSS+p38 inhibitor; [7] HHcy + DSS [21 days]; and [8] HHcy + DSS + folate [21 days]. Colitis was induced by 5% DSS. HHcy was induced by the normal rodent diet containing 1.7% methionine. The mRNA expression of interleukin 17 [IL-17] was detected by qRT-PCR. The protein expressions of IL-17, retinoid-related orphan nuclear receptor-γt [RORγt], p38 MAPK, phosphorylated-p38 MAPK, cytosolic phospolipaseA2 [cPLA2], phosphorylated-cPLA2, and cyclooxygenase 2 [COX2] were detected by immunoblot analysis. RESULTS The rats of the HHcy + DSS group had significantly higher myeloperoxidase [MPO] activity, DAI score, and histological score. HHcy significantly increased the plasma concentration, the colonic mRNA, and the protein levels of IL-17. HHcy also activated p38 MAPK and cPLA2, and increased the protein levels of COX2 and RORγt as well as the plasma level of prostaglandin E2 [PGE2]. Folate supplementation down-regulated homocysteine-induced IL-17 and RORγt expressions. CONCLUSIONS HHcy aggravated DSS-induced colitis by stimulating IL-17 expression via the p38/cPLA2/COX2/PGE2 signalling pathway. The folate supplementation may represent a novel approach to treating the chronic intestinal inflammation of IBD exacerbated by HHcy.
Collapse
Affiliation(s)
- Siying Zhu
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, P.R. China Hubei Clinical Center & Key Laboratory of Intestinal & Colorectal Diseases, Wuhan, P.R. China
| | - Jin Li
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, P.R. China Hubei Clinical Center & Key Laboratory of Intestinal & Colorectal Diseases, Wuhan, P.R. China
| | - Yuntao Bing
- Hubei Clinical Center & Key Laboratory of Intestinal & Colorectal Diseases, Wuhan, P.R. China
| | - Wenfeng Yan
- Hubei Clinical Center & Key Laboratory of Intestinal & Colorectal Diseases, Wuhan, P.R. China
| | - Youqing Zhu
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, P.R. China Hubei Clinical Center & Key Laboratory of Intestinal & Colorectal Diseases, Wuhan, P.R. China
| | - Bing Xia
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, P.R. China Hubei Clinical Center & Key Laboratory of Intestinal & Colorectal Diseases, Wuhan, P.R. China
| | - Min Chen
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, P.R. China Hubei Clinical Center & Key Laboratory of Intestinal & Colorectal Diseases, Wuhan, P.R. China
| |
Collapse
|
15
|
Chen TH, Shih CY, Hsu WL, Chou TC. Mechanisms of Nifedipine-Downregulated CD40L/sCD40L Signaling in Collagen Stimulated Human Platelets. PLoS One 2015; 10:e0127054. [PMID: 25970603 PMCID: PMC4430314 DOI: 10.1371/journal.pone.0127054] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Accepted: 04/10/2015] [Indexed: 01/19/2023] Open
Abstract
The platelet-derived soluble CD40L (sCD40L) release plays a critical role in the development of atherosclerosis. Nifedipine, a dihydropyridine-based L-type calcium channel blocker (CCB), has been reported to have an anti-atherosclerotic effect beyond its blood pressure-lowering effect, but the molecular mechanisms remain unclear. The present study was designed to investigate whether nifedipine affects sCD40L release from collagen-stimulated human platelets and to determine the potential role of peroxisome proliferator-activated receptor-β/-γ (PPAR-β/-γ). We found that treatment with nifedipine significantly inhibited the platelet surface CD40L expression and sCD40L release in response to collagen, while the inhibition was markedly reversed by blocking PPAR-β/-γ activity with specific antagonist such as GSK0660 and GW9662. Meanwhile, nifedipine also enhanced nitric oxide (NO) and cyclic GMP formation in a PPAR-β/-γ-dependent manner. When the NO/cyclic GMP pathway was suppressed, nifedipine-mediated inhibition of sCD40L release was abolished significantly. Collagen-induced phosphorylation of p38MAPK, ERK1/2 and HSP27, matrix metalloproteinase-2 (MMP-2) expression/activity and reactive oxygen species (ROS) formation were significantly inhibited by nifedipine, whereas these alterations were all attenuated by co-treatment with PPAR-β/-γ antagonists. Collectively, these results demonstrate that PPAR-β/-γ-dependent pathways contribute to nifedipine-mediated downregulation of CD40L/sCD40L signaling in activated platelets through regulation of NO/ p38MAPK/ERK1/2/HSP27/MMP-2 signalings and provide a novel mechanism regarding the anti-atherosclerotic effect of nifedipine.
Collapse
Affiliation(s)
- Tso-Hsiao Chen
- Division of Nephrology, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Ching-Yu Shih
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Wen-Lin Hsu
- School of Medicine, Tzu Chi University; Department of Radiation Oncology, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
| | - Tz-Chong Chou
- Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan
- Department ofBiotechnology, Asia University, Taichung, Taiwan
- China Medical University Hospital, China Medical University, Taichung, Taiwan
- * E-mail:
| |
Collapse
|
16
|
Lê QH, El Alaoui M, Véricel E, Ségrestin B, Soulère L, Guichardant M, Lagarde M, Moulin P, Calzada C. Glycoxidized HDL, HDL enriched with oxidized phospholipids and HDL from diabetic patients inhibit platelet function. J Clin Endocrinol Metab 2015; 100:2006-14. [PMID: 25794249 PMCID: PMC4803888 DOI: 10.1210/jc.2014-4214] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
CONTEXT High-density lipoproteins (HDL) possess atheroprotective properties including anti-thrombotic and antioxidant effects. Very few studies relate to the functional effects of oxidized HDL on platelets in type 2 diabetes (T2D). OBJECTIVE The objective of our study was to investigate the effects of in vitro glycoxidized HDL and HDL from patients with T2D on platelet aggregation and arachidonic acid signaling cascade. At the same time, the contents of hydroxylated fatty acids were assessed in HDL. RESULTS Compared with control HDL, in vitro glycoxidized HDL had decreased proportions of linoleic (LA) and arachidonic (AA) acids in phospholipids and cholesteryl esters, and increased concentrations of hydroxy-octadecadienoic acids (9-HODE and 13-HODE) and 15-hydroxy-eicosatetraenoic acid (15-HETE), derived from LA and AA respectively, especially hydroxy derivatives esterified in phospholipids. Glycoxidized HDL dose-dependently decreased collagen-induced platelet aggregation by binding to scavenger receptor BI (SR-BI). Glycoxidized HDL prevented collagen-induced increased phosphorylation of platelet p38 MAPK and cytosolic phospholipase A2, as well as intracellular calcium mobilization. HDL enriched with oxidized phosphatidylcholine (PC), namely PC(16:0/13-HODE) dose-dependently inhibited platelet aggregation. Increased concentrations of 9-HODE, 13-HODE, and 15-HETE in phospholipids (2.1-, 2.1-, and 2.4-fold increase, respectively) were found in HDL from patients with T2D, and these HDL also inhibited platelet aggregation via SR-BI. CONCLUSIONS Our results suggest that in vitro glycoxidized HDL as well as HDL from patients with T2D inhibit platelet aggregation, and suggest that oxidized LA-containing phospholipids may contribute to the anti-aggregatory effects of glycoxidized HDL and HDL from patients with T2D.
Collapse
Affiliation(s)
- Quang Huy Lê
- CARMEN, Laboratoire de recherche en cardiovasculaire, métabolisme, diabétologie et nutrition
Université Claude Bernard Lyon 1Institut National des Sciences Appliquées LyonInstitut national de la recherche agronomique (INRA)INSERMHospices Civils de LyonFaculté de Médecine Lyon Sud - BP 12 - 165 Chemin du Grand Revoyet - 69921 Oullins cedex INSA, Bâtiment. IMBL, La Doua - 11 Avenue Jean Capelle - 69621 Villeurbanne Cedex
| | - Meddy El Alaoui
- ICBMS, Institut de Chimie et Biochimie Moléculaires et Supramoléculaires
Université Claude Bernard Lyon 1Institut National des Sciences Appliquées LyonÉcole Supérieure Chimie Physique Électronique de LyonCentre National de la Recherche ScientifiqueBâtiment CPE 43 Boulvard du 11 Novembre 1918 69622 Villeurbanne Cedex
| | - Evelyne Véricel
- CARMEN, Laboratoire de recherche en cardiovasculaire, métabolisme, diabétologie et nutrition
Université Claude Bernard Lyon 1Institut National des Sciences Appliquées LyonInstitut national de la recherche agronomique (INRA)INSERMHospices Civils de LyonFaculté de Médecine Lyon Sud - BP 12 - 165 Chemin du Grand Revoyet - 69921 Oullins cedex INSA, Bâtiment. IMBL, La Doua - 11 Avenue Jean Capelle - 69621 Villeurbanne Cedex
| | | | - Laurent Soulère
- ICBMS, Institut de Chimie et Biochimie Moléculaires et Supramoléculaires
Université Claude Bernard Lyon 1Institut National des Sciences Appliquées LyonÉcole Supérieure Chimie Physique Électronique de LyonCentre National de la Recherche ScientifiqueBâtiment CPE 43 Boulvard du 11 Novembre 1918 69622 Villeurbanne Cedex
| | - Michel Guichardant
- CARMEN, Laboratoire de recherche en cardiovasculaire, métabolisme, diabétologie et nutrition
Université Claude Bernard Lyon 1Institut National des Sciences Appliquées LyonInstitut national de la recherche agronomique (INRA)INSERMHospices Civils de LyonFaculté de Médecine Lyon Sud - BP 12 - 165 Chemin du Grand Revoyet - 69921 Oullins cedex INSA, Bâtiment. IMBL, La Doua - 11 Avenue Jean Capelle - 69621 Villeurbanne Cedex
| | - Michel Lagarde
- CARMEN, Laboratoire de recherche en cardiovasculaire, métabolisme, diabétologie et nutrition
Université Claude Bernard Lyon 1Institut National des Sciences Appliquées LyonInstitut national de la recherche agronomique (INRA)INSERMHospices Civils de LyonFaculté de Médecine Lyon Sud - BP 12 - 165 Chemin du Grand Revoyet - 69921 Oullins cedex INSA, Bâtiment. IMBL, La Doua - 11 Avenue Jean Capelle - 69621 Villeurbanne Cedex
| | - Philippe Moulin
- CARMEN, Laboratoire de recherche en cardiovasculaire, métabolisme, diabétologie et nutrition
Université Claude Bernard Lyon 1Institut National des Sciences Appliquées LyonInstitut national de la recherche agronomique (INRA)INSERMHospices Civils de LyonFaculté de Médecine Lyon Sud - BP 12 - 165 Chemin du Grand Revoyet - 69921 Oullins cedex INSA, Bâtiment. IMBL, La Doua - 11 Avenue Jean Capelle - 69621 Villeurbanne Cedex
- Fédération d'Endocrinologie
Hospices Civils de Lyon69677 Lyon Bron
| | - Catherine Calzada
- CARMEN, Laboratoire de recherche en cardiovasculaire, métabolisme, diabétologie et nutrition
Université Claude Bernard Lyon 1Institut National des Sciences Appliquées LyonInstitut national de la recherche agronomique (INRA)INSERMHospices Civils de LyonFaculté de Médecine Lyon Sud - BP 12 - 165 Chemin du Grand Revoyet - 69921 Oullins cedex INSA, Bâtiment. IMBL, La Doua - 11 Avenue Jean Capelle - 69621 Villeurbanne Cedex
- * Correspondence should be addressed to Catherine Calzada
| |
Collapse
|
17
|
Abstract
Multiple studies have now shown that various species of bacteria can stimulate platelets; many in a strain and donor-dependent manner. The signalling pathways underlying this platelet activation has been the subject of scrutiny for the last decade. The best-delineated pathway is that in response to Streptococcal species, such as Streptococcus sanguinis (S. sanguinis), Streptococcus gordonii (S. gordonii) and Streptococcus oralis (S. oralis), where a pathway is initiated by the engagement of the low affinity IgG receptor, FcγRIIA. This leads to and involves the tyrosine kinase Syk, the adaptor protein Linker of Activated T Cells (LAT) and subsequently both phospholipase Cγ2 (PLCγ2) and phosphatidylinositol-3-kinase (PI-3-K). Finally, this leads to the expression of the αIIbβ3 integrin, the synthesis and release of thromboxane A2 (T × A2) and the exocytosis of PF4, each of which plays a crucial role in secondary signalling and full platelet activation. Roles for other signalling pathways in Streptococcal-induced platelet activation are less clear, although an ADP-mediated inhibition of adenylyl cyclase, a glycoprotein Ib/IX/V-mediated pathway and perhaps a complement-induced pathway have each been proposed. Platelet activation by Porphyromonas gingivalis (P. gingivalis) at least partially shares the FcγRIIA/Syk/PLCγ2/PI-3-K mechanism utilised by Streptococcal species. However, it has also been suggested that P. gingivalis activates platelets by two additional methods; stimulation of the protease-activated receptors leading to activation of phospholipase Cβ (PLCβ), and the engagement of Toll-like receptors 2 and 4 by released lipopolysaccharide leading to an ill-defined pathway which may involve PI-3-K. Consequently, it appears that bacteria can stimulate platelets by eliciting multiple signalling pathways some of which are common, and some unique, to individual species.
Collapse
|
18
|
Phosphatidylinositol-3,4,5-trisphosphate stimulates Ca(2+) elevation and Akt phosphorylation to constitute a major mechanism of thromboxane A2 formation in human platelets. Cell Signal 2015; 27:1488-98. [PMID: 25797048 DOI: 10.1016/j.cellsig.2015.03.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Revised: 02/18/2015] [Accepted: 03/04/2015] [Indexed: 11/23/2022]
Abstract
Phosphatidylinositol trisphosphate (PIP3) has been implicated in many platelet functions however many of the mechanisms need clarification. We have used cell permeable analogues of PIP3,1-O-(1,2-di-palmitoyl-sn-glyero-3-O-phosphoryl)-D-myo-inositol-3,4,5-trisphosphate (DiC16-PIP3) or 1-O-(1,2-di-octanoyl-sn-glyero-3-O-phosphoryl)-D-myo-inositol-3,4,5-trisphosphate (DiC8-PIP3) to study their effects on activation on washed human platelets. Addition of either DiC8- or DiC16-PIP3 to human platelets induced aggregation in the presence of extracellular Ca(2+). This was reduced by the presence of indomethacin, the phospholipase C inhibitor U73122 and apyrase. DiC8-PIP3 induced the phosphorylation of Akt-Ser(473) which was reduced by the Akt inhibitor IV, wortmannin and EGTA (suggesting a dependence on Ca(2+) entry). In Fura2 loaded platelets DiC8-PIP3 was effective at increasing intracellular Ca(2+) in a distinct and transient manner that was reduced in the presence of indomethacin, U73122 and 2-aminoethyl diphenylborinate (2APB). Ca(2+) elevation was reduced by the non-SOCE inhibitor LOE908 and also by the SOCE inhibitor BTP2. DiC8-PIP3 induced the release of Ca(2+) from stores which was not affected by the proton dissipating agent bafilomycin A1 and was more potent than the two-pore channel agonist DiC8-PI[3,5]P2 suggesting release from an endoplasmic reticulum type store. DiC8-PIP3 weakly induced the tyrosine phosphorylation of Syk but not of PLCγ2. Finally like thrombin DiC8-PIP3 induced the formation of thromboxane B2 that was inhibited by the Akt inhibitor IV. These studies suggest that PIP3 via Ca(2+) elevation and Akt phosphorylation forms a central role in thromboxane A2 formation and the amplification of platelet activation.
Collapse
|
19
|
Jin M, Zhou Q, Lee E, Dan S, Duan HQ, Kong D. AS252424, a PI3Kγ Inhibitor, Downregulates Inflammatory Responsiveness in Mouse Bone Marrow-Derived Mast Cells. Inflammation 2014; 37:1254-60. [DOI: 10.1007/s10753-014-9852-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
20
|
Gaba M, Singh S, Mohan C. Benzimidazole: an emerging scaffold for analgesic and anti-inflammatory agents. Eur J Med Chem 2014; 76:494-505. [PMID: 24602792 DOI: 10.1016/j.ejmech.2014.01.030] [Citation(s) in RCA: 204] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Revised: 01/19/2014] [Accepted: 01/20/2014] [Indexed: 02/02/2023]
Abstract
Within the vast range of heterocycles, benzimidazole and its derivatives are found to be trendy structures employed for discovery of drugs in the field of pharmaceutical and medicinal chemistry. The unique structural features of benzimidazole and a wide range of biological activities of its derivatives made it privileged structure in drug discovery. Recently, benzimidazole scaffold has emerged as a pharmacophore of choice for designing analgesic and anti-inflammatory agents active on different clinically approved targets. To pave the way for future research, there is a need to collect the latest information in this promising area. In the present review we have collated published reports on this versatile core to provide an insight so that its full therapeutic potential can be utilized for the treatment of pain and inflammation.
Collapse
Affiliation(s)
- Monika Gaba
- Department of Pharmaceutical Sciences, ASBASJSM College of Pharmacy, Bela, Ropar, Punjab, India.
| | - Sarbjot Singh
- Drug Discovery Research, Panacea Biotec Pvt. Ltd., Mohali, Punjab, India
| | - Chander Mohan
- Rayat-Bahra Institute of Pharmacy, Hoshiarpur, Punjab, India
| |
Collapse
|
21
|
Rukoyatkina N, Mindukshev I, Walter U, Gambaryan S. Dual role of the p38 MAPK/cPLA2 pathway in the regulation of platelet apoptosis induced by ABT-737 and strong platelet agonists. Cell Death Dis 2013; 4:e931. [PMID: 24263105 PMCID: PMC3847335 DOI: 10.1038/cddis.2013.459] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Revised: 09/21/2013] [Accepted: 10/22/2013] [Indexed: 12/13/2022]
Abstract
p38 Mitogen-activated protein (MAP) kinase is involved in the apoptosis of nucleated cells. Although platelets are anucleated cells, apoptotic proteins have been shown to regulate platelet lifespan. However, the involvement of p38 MAP kinase in platelet apoptosis is not yet clearly defined. Therefore, we investigated the role of p38 MAP kinase in apoptosis induced by a mimetic of BH3-only proteins, ABT-737, and in apoptosis-like events induced by such strong platelet agonists as thrombin in combination with convulxin (Thr/Cvx), both of which result in p38 MAP kinase phosphorylation and activation. A p38 inhibitor (SB202190) inhibited the apoptotic events induced by ABT-737 but did not influence those induced by Thr/Cvx. The inhibitor also reduced the phosphorylation of cytosolic phospholipase A2 (cPLA2), an established p38 substrate, induced by ABT-737 or Thr/Cvx. ABT-737, but not Thr/Cvx, induced the caspase 3-dependent cleavage and inactivation of cPLA2. Thus, p38 MAPK promotes ABT-737-induced apoptosis by inhibiting the cPLA2/arachidonate pathway. We also show that arachidonic acid (AA) itself and in combination with Thr/Cvx or ABT-737 at low concentrations prevented apoptotic events, whereas at high concentrations it enhanced such events. Our data support the hypothesis that the p38 MAPK-triggered arachidonate pathway serves as a defense mechanism against apoptosis under physiological conditions.
Collapse
Affiliation(s)
- N Rukoyatkina
- 1] Institute of Clinical Biochemistry and Pathobiochemistry, University of Würzburg, Würzburg D-97080, Germany [2] Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St Petersburg 194223, Russia
| | | | | | | |
Collapse
|
22
|
Jackson ECG, Ortar G, McNicol A. The Effects of an Inhibitor of Diglyceride Lipase on Collagen-Induced Platelet Activation. J Pharmacol Exp Ther 2013; 347:582-8. [DOI: 10.1124/jpet.113.205591] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
|
23
|
Carestia A, Rivadeneyra L, Romaniuk MA, Fondevila C, Negrotto S, Schattner M. Functional responses and molecular mechanisms involved in histone-mediated platelet activation. Thromb Haemost 2013; 110:1035-45. [PMID: 23965842 DOI: 10.1160/th13-02-0174] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2013] [Accepted: 07/24/2013] [Indexed: 01/09/2023]
Abstract
Histones are highly alkaline proteins found in cell nuclei and they can be released by either dying or inflammatory cells. The recent observations that histones are major components of neutrophil extracellular traps and promote platelet aggregation and platelet-dependent thrombin generation have shown that these proteins are potent prothrombotic molecules. Because the mechanism(s) of platelet activation by histones are not completely understood, we explored the ability of individual recombinant human histones H1, H2A, H2B, H3 and H4 to induce platelet activation as well as the possible molecular mechanisms involved. All histones were substrates for platelet adhesion and spreading and triggered fibrinogen binding, aggregation, von Willebrand factor release, P-selectin and phosphatidylserine (PS) exposure and the formation of platelet-leukocyte aggregates; however, H4 was the most potent. Histone-mediated fibrinogen binding, P-selectin and PS exposure and the formation of mixed aggregates were potentiated by thrombin. Histones induced the activation of ERK, Akt, p38 and NFκB. Accordingly, histone-induced platelet activation was significantly impaired by pretreatment of platelets with inhibitors of ERK (U 0126), PI3K/Akt (Ly 294002), p38 (SB 203580) and NFκB (BAY 11-7082 and Ro 106-9920). Preincubation of platelets with either aspirin or dexamethasone markedly decreased fibrinogen binding and the adhesion mediated by histones without affecting P-selectin exposure. Functional platelet responses induced by H3 and H4, but not H1, H2A and H2B, were partially mediated through interaction with Toll-like receptors -2 and -4. Our data identify histones as important triggers of haemostatic and proinflammatory platelet responses, and only haemostatic responses are partially inhibited by anti-inflammatory drugs.
Collapse
Affiliation(s)
- A Carestia
- Mirta Schattner, Instituto de Medicina Experimental, CONICET-ANM, Pacheco de Melo 3081, Buenos Aires 1425, Argentina, Tel.: +54 11 4805 5759 ext. 301, Fax: +54 11 4805 0712, E-mail:
| | | | | | | | | | | |
Collapse
|
24
|
Young PR. Perspective on the Discovery and Scientific Impact of p38 MAP Kinase. ACTA ACUST UNITED AC 2013; 18:1156-63. [DOI: 10.1177/1087057113497401] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
It has now been almost 20 years since the discovery of p38 MAP kinase and its role in inflammatory cytokine synthesis through reverse pharmacology and its subsequent exploration as a potential target for autoimmune and other diseases. At the time of its discovery, the use of cell-based phenotypic screens to identify new molecular targets was at its infancy, and while p38 MAP kinase was not the first target to be identified this way, it provides a useful model for reviewing the pros and cons of this approach and the subsequent impact it can have on discovering new medicines.
Collapse
|
25
|
Hughes CE, Radhakrishnan UP, Lordkipanidzé M, Egginton S, Dijkstra JM, Jagadeeswaran P, Watson SP. G6f-like is an ITAM-containing collagen receptor in thrombocytes. PLoS One 2012; 7:e52622. [PMID: 23285115 PMCID: PMC3528668 DOI: 10.1371/journal.pone.0052622] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2012] [Accepted: 11/20/2012] [Indexed: 01/22/2023] Open
Abstract
Collagen activates mammalian platelets through a complex of the immunoglobulin (Ig) receptor GPVI and the Fc receptor γ-chain, which has an immunoreceptor tyrosine-based activation motif (ITAM). Cross-linking of GPVI mediates activation through the sequential activation of Src and Syk family kinases and activation of PLCγ2. Nucleated thrombocytes in fish are activated by collagen but lack an ortholog of GPVI. In this study we show that collagen activates trout thrombocytes in whole blood and under flow conditions through a Src kinase driven pathway. We identify the Ig receptor G6f-like as a collagen receptor and demonstrate in a cell line assay that it signals through its cytoplasmic ITAM. Using a morpholino for in vivo knock-down of G6f-like levels in zebrafish, we observed a marked delay or absence of occlusion of the venous and arterial systems in response to laser injury. Thus, G6f-like is a physiologically relevant collagen receptor in fish thrombocytes which signals through the same ITAM-based signalling pathway as mammalian GPVI, providing a novel example of convergent evolution.
Collapse
Affiliation(s)
- Craig E Hughes
- Centre for Cardiovascular Sciences, Institute for Biomedical Research, The College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, United Kingdom.
| | | | | | | | | | | | | |
Collapse
|
26
|
Impaired thrombin-induced platelet activation and thrombus formation in mice lacking the Ca(2+)-dependent tyrosine kinase Pyk2. Blood 2012; 121:648-57. [PMID: 23175689 DOI: 10.1182/blood-2012-06-438762] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In the present study, we used a knockout murine model to analyze the contribution of the Ca(2+)-dependent focal adhesion kinase Pyk2 in platelet activation and thrombus formation in vivo. We found that Pyk2-knockout mice had a tail bleeding time that was slightly increased compared with their wild-type littermates. Moreover, in an in vivo model of femoral artery thrombosis, the time to arterial occlusion was significantly prolonged in mice lacking Pyk2. Pyk2-deficient mice were also significantly protected from collagen plus epinephrine-induced pulmonary thromboembolism. Ex vivo aggregation of Pyk2-deficient platelets was normal on stimulation of glycoprotein VI, but was significantly reduced in response to PAR4-activating peptide, low doses of thrombin, or U46619. Defective platelet aggregation was accompanied by impaired inside-out activation of integrin α(IIb)β(3) and fibrinogen binding. Granule secretion was only slightly reduced in the absence of Pyk2, whereas a marked inhibition of thrombin-induced thromboxane A(2) production was observed, which was found to be responsible for the defective aggregation. Moreover, we have demonstrated that Pyk2 is implicated in the signaling pathway for cPLA(2) phosphorylation through p38 MAPK. The results of the present study show the importance of the focal adhesion kinase Pyk2 downstream of G-protein-coupled receptors in supporting platelet aggregation and thrombus formation.
Collapse
|
27
|
Abdulrehman AY, Jackson ECG, McNicol A. Platelet activation by Streptococcus sanguinis is accompanied by MAP kinase phosphorylation. Platelets 2012; 24:6-14. [PMID: 22372533 DOI: 10.3109/09537104.2012.661105] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
There is increasing interest in the role of infections in atherothrombotic conditions. In particular, bacteria, notably those of oral origin, have been shown to activate platelets using a variety of mechanisms. Previous studies have shown that S. sanguinis strain 2017-78 induces platelet aggregation which requires the presence of both vWF and IgG. This aggregation is accompanied by the consecutive phosphorylation/desphosphorylation/rephosphorylation of several signalling proteins. The first two phases are thromboxane-dependent whereas the rephosphorylation phase is mediated by engagement of the αIIbβ3 integrin. Here signalling events, specifically the potential role of MAP kinases, associated with S. sanguinis strain 2017-78-induced platelet activation have been further examined using an immunoblotting approach. The addition of S. sanguinis strain 2017-78 caused a similar triphasic phosphorylation profile of the platelet MAP kinase Erk2 to that seen with other phosphoproteins. Pretreatment with aspirin or RGDS did not affect 2017-78-induced Erk2 phosphorylation or desphosphorylation but both inhibited the rephosphorylation phase. In contrast the level of 2017-78-induced platelet MAP kinase p38 phosphorylation remained at an elevated level, and this was unaffected by aspirin. Similarly, 2017-78-induced cPLA(2) phosphorylation remained above basal levels during the aggregation process. The p38 inhibitor SB203580 inhibited S. sanguinis-induced aggregation with no effect on the phosphorylation of either p38 or cPLA(2). Thus the current study demonstrates the activation of both the Erk2 and p38 forms of MAP kinases, and of cPLA(2), in platelets stimulated with S. sanguinis strain 2017-78, and is consistent with a role for Erk2, but not for p38, in the cPLA(2) phosphorylation in response to S. sanguinis.
Collapse
Affiliation(s)
- Ahmed Y Abdulrehman
- Departments of Oral Biology, University of Manitoba, Winnipeg, Manitoba, Canada
| | | | | |
Collapse
|
28
|
Colas R, Sassolas A, Guichardant M, Cugnet-Anceau C, Moret M, Moulin P, Lagarde M, Calzada C. LDL from obese patients with the metabolic syndrome show increased lipid peroxidation and activate platelets. Diabetologia 2011; 54:2931-40. [PMID: 21847583 PMCID: PMC3367234 DOI: 10.1007/s00125-011-2272-8] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2011] [Accepted: 07/14/2011] [Indexed: 10/17/2022]
Abstract
AIMS/HYPOTHESIS This study assessed oxidative stress in LDL from obese patients with the metabolic syndrome and compared it with that in LDL from type 2 diabetic patients or control volunteers. It also determined the effect on platelets of LDL from the three groups. METHODS The profiles of lipids, fatty acids and fatty acid oxidation products were determined in LDL isolated from plasma of patients with the metabolic syndrome, patients with type 2 diabetes and volunteers (n = 10 per group). The effects of LDL from the participant groups on the platelet arachidonic acid signalling cascade and aggregation were investigated. RESULTS Compared with LDL from control volunteers, LDL from obese metabolic syndrome and type 2 diabetic patients had lower cholesteryl ester, higher triacylglycerol and lower ethanolamine plasmalogen levels. Proportions of linoleic acid were decreased in phosphatidylcholine and cholesteryl esters in LDL from both patient groups. Among the markers of lipid peroxidation, oxidation products of linoleic acid (hydroxy-octadecadienoic acids) and malondialdehyde were increased by 59% and twofold, respectively in LDL from metabolic syndrome and type 2 diabetic patients. LDL from metabolic syndrome and type 2 diabetic patients were equally potent in activating the platelet arachidonic acid signalling cascade through increased phosphorylation of p38 mitogen-activated protein kinase and cytosolic phospholipase A(2), and through increased thromboxane B(2) formation. LDL from patients with the metabolic syndrome and type 2 diabetes potentiated platelet aggregation by threefold and 3.5-fold respectively, whereas control LDL had no activating effects on platelets. CONCLUSIONS/INTERPRETATION The metabolic syndrome in obese patients, without or with diabetes, is associated with increased oxidative stress in LDL, which triggers platelet activation.
Collapse
Affiliation(s)
- Romain Colas
- CARMEN, Laboratoire de recherche en cardiovasculaire, métabolisme, diabétologie et nutrition
INSERM : U1060INRAInstitut National des Sciences Appliquées de LyonUniversité Claude Bernard - Lyon IHospices Civils de LyonFaculté de Médecine Lyon Sud - BP 12 - 165 Chemin du Grand Revoyet - 69921 Oullins cedex INSA, Bât. IMBL, La Doua - 11 Avenue Jean Capelle - 69621 Villeurbanne Cedex,FR
| | - Agnès Sassolas
- CARMEN, Laboratoire de recherche en cardiovasculaire, métabolisme, diabétologie et nutrition
INSERM : U1060INRAInstitut National des Sciences Appliquées de LyonUniversité Claude Bernard - Lyon IHospices Civils de LyonFaculté de Médecine Lyon Sud - BP 12 - 165 Chemin du Grand Revoyet - 69921 Oullins cedex INSA, Bât. IMBL, La Doua - 11 Avenue Jean Capelle - 69621 Villeurbanne Cedex,FR
| | - Michel Guichardant
- CARMEN, Laboratoire de recherche en cardiovasculaire, métabolisme, diabétologie et nutrition
INSERM : U1060INRAInstitut National des Sciences Appliquées de LyonUniversité Claude Bernard - Lyon IHospices Civils de LyonFaculté de Médecine Lyon Sud - BP 12 - 165 Chemin du Grand Revoyet - 69921 Oullins cedex INSA, Bât. IMBL, La Doua - 11 Avenue Jean Capelle - 69621 Villeurbanne Cedex,FR
| | | | - Myriam Moret
- Fédération d'endocrinologie
Hospices Civils de LyonBron,FR
| | - Philippe Moulin
- CARMEN, Laboratoire de recherche en cardiovasculaire, métabolisme, diabétologie et nutrition
INSERM : U1060INRAInstitut National des Sciences Appliquées de LyonUniversité Claude Bernard - Lyon IHospices Civils de LyonFaculté de Médecine Lyon Sud - BP 12 - 165 Chemin du Grand Revoyet - 69921 Oullins cedex INSA, Bât. IMBL, La Doua - 11 Avenue Jean Capelle - 69621 Villeurbanne Cedex,FR
- Fédération d'endocrinologie
Hospices Civils de LyonBron,FR
| | - Michel Lagarde
- CARMEN, Laboratoire de recherche en cardiovasculaire, métabolisme, diabétologie et nutrition
INSERM : U1060INRAInstitut National des Sciences Appliquées de LyonUniversité Claude Bernard - Lyon IHospices Civils de LyonFaculté de Médecine Lyon Sud - BP 12 - 165 Chemin du Grand Revoyet - 69921 Oullins cedex INSA, Bât. IMBL, La Doua - 11 Avenue Jean Capelle - 69621 Villeurbanne Cedex,FR
| | - Catherine Calzada
- CARMEN, Laboratoire de recherche en cardiovasculaire, métabolisme, diabétologie et nutrition
INSERM : U1060INRAInstitut National des Sciences Appliquées de LyonUniversité Claude Bernard - Lyon IHospices Civils de LyonFaculté de Médecine Lyon Sud - BP 12 - 165 Chemin du Grand Revoyet - 69921 Oullins cedex INSA, Bât. IMBL, La Doua - 11 Avenue Jean Capelle - 69621 Villeurbanne Cedex,FR
- * Correspondence should be adressed to: Catherine Calzada
| |
Collapse
|
29
|
Signorello MG, Giacobbe E, Leoncini G. Activation by 2-arachidonoylglycerol of platelet p38MAPK/cPLA2 pathway. J Cell Biochem 2011; 112:2794-802. [DOI: 10.1002/jcb.23194] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
30
|
Walters JN, Bickford JS, Beachy DE, Newsom KJ, Herlihy JDH, Peck MV, Qiu X, Nick HS. cPLA(2)α gene activation by IL-1β is dependent on an upstream kinase pathway, enzymatic activation and downstream 15-lipoxygenase activity: a positive feedback loop. Cell Signal 2011; 23:1944-51. [PMID: 21771656 DOI: 10.1016/j.cellsig.2011.07.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2011] [Accepted: 07/04/2011] [Indexed: 01/01/2023]
Abstract
Cytosolic phospholipase A(2)α (cPLA(2)α) is the most widely studied member of the Group IV PLA(2) family. The enzyme is Ca(2+)-dependent with specificity for phospholipid substrates containing arachidonic acid. As the pinnacle of the arachidonic acid pathway, cPLA(2)α has a primary role in the biosynthesis of a diverse family of eicosanoid metabolites, with potent physiological, inflammatory and pathological consequences. cPLA(2)α activity is regulated by pro-inflammatory stimuli through pathways involving increased intracellular Ca(2+) levels, phosphorylation coupled to increased enzymatic activity and de novo gene transcription. This study addresses the signal transduction pathways for protein phosphorylation and gene induction following IL-1β stimulation in human fetal lung fibroblasts. Our results utilizing both inhibitors and kinase-deficient cells demonstrate that cPLA(2)α is phosphorylated within 10min of IL-1β treatment, an event requiring p38 MAPK as well as the upstream kinase, MKK3/MKK6. Inhibition of p38 MAPK also blocks the phosphorylation of a downstream, nuclear kinase, MSK-1. Our results further demonstrate that the activities of both cPLA(2)α and a downstream lipoxygenase (15-LOX2) are required for IL-1β-dependent induction of cPLA(2)α mRNA expression. Overall, these data support an MKK3/MKK6→p38 MAPK→MSK-1→cPLA(2)α→15-LOX2-dependent, positive feedback loop where a protein's enzymatic activity is required to regulate its own gene induction by a pro-inflammatory stimulus.
Collapse
Affiliation(s)
- Jewell N Walters
- Department of Neuroscience, University of Florida, Gainesville, FL 32610, USA
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Matsui T, Svensson CI, Hirata Y, Mizobata K, Hua XY, Yaksh TL. Release of prostaglandin E(2) and nitric oxide from spinal microglia is dependent on activation of p38 mitogen-activated protein kinase. Anesth Analg 2010; 111:554-60. [PMID: 20610553 DOI: 10.1213/ane.0b013e3181e3a2a2] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
BACKGROUND The spinal release of prostaglandins (PGs), nitric oxide (NO), and cytokines has been implicated in spinal nociceptive processing. Microglia represent a possible cell of origin for these proexcitatory mediators. Spinal microglia possess Toll-like receptor 4 (TLR4) and neurokinin 1 (NK1) receptors, and both receptors play a significant role in peripheral nerve injury- and inflammation-induced spinal sensitization. Accordingly, we examined the properties of the cascades activated by the respective targets, which led to the release of PGE(2) and an increase in nitrite (NO(2)(-)) (a marker of NO) from cultured rat spinal microglia. METHODS Spinal microglia isolated from Sprague-Dawley neonatal rats were cultured with lipopolysaccharide (LPS) or substance P (SP) alone, with LPS in combination with SP, and with LPS in the presence of each inhibitor of cyclooxygenase (COX), NO synthase 2 (NOS2) or p38 mitogen-activated protein kinase (p38), or minocycline for 24 hours and 48 hours. Concentrations of PGE(2) and NO(2)(-) in culture supernatants were measured using an enzyme immunoassay and a colorimetric assay, respectively. RESULTS Application of LPS (a TLR4 ligand, 0.1 to 10 ng/mL) to cultured microglia produced a dose- and time-dependent increase in PGE(2) and NO(2)(-) production, whereas no effects were observed after incubation with SP (an NK1 agonist, up to 10(-5) M) alone or in combination with LPS. Antagonist studies with SC-560 (COX-1 inhibitor) and SC-236 (COX-2 inhibitor) showed that LPS-induced PGE(2) release was generated from both COX-1 and COX-2. LPS-induced NO release was suppressed by 1400W, an inhibitor of NOS2. Minocycline, an agent blocking microglial activation, and SB203580, an inhibitor of p38, both attenuated the LPS-induced PGE(2) and NO release. The 1400W, at the doses that suppressed NO release, also blocked increased PGE(2) release. CONCLUSIONS Our findings suggest that (a) activation of spinal microglia via TLR4 but not NK1 receptors produces PGE(2) and NO release from these cells; (b) the evoked PGE(2) release is generated by both COX-1 and COX-2, and (c) the COX-PGE(2) pathway is regulated by p38 and NOS2. Taken together with our previous in vivo work, the current findings emphasize that p38 in spinal microglia is a key player in regulating production of pronociceptive molecules, such as PGE(2) and NO.
Collapse
Affiliation(s)
- Tomohiro Matsui
- Department of Laboratory Sciences, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan
| | | | | | | | | | | |
Collapse
|
32
|
Harper MT, Poole AW. Diverse functions of protein kinase C isoforms in platelet activation and thrombus formation. J Thromb Haemost 2010; 8:454-62. [PMID: 20002545 DOI: 10.1111/j.1538-7836.2009.03722.x] [Citation(s) in RCA: 123] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Platelet activation is a complex balance of positive and negative signaling pathways. The protein kinase C (PKC) family is a major regulator of platelet granule secretion, integrin activation, aggregation, spreading and procoagulant activity. As broad-spectrum PKC inhibitors reduce secretion and aggregation, the PKC family is generally considered to be a positive regulator of platelet activation. However, the individual members of the PKC family that are expressed in platelets are regulated in different ways, and an increasing body of evidence indicates that they have distinct, and often opposing, roles. Many of the recent advances in understanding the contributions of individual PKC isoforms have come from mouse gene knockout studies. PKCalpha, a classic isoform, is an essential positive regulator of granule secretion and thrombus formation, both in vitro and in vivo. Mice lacking PKCalpha show much reduced thrombus formation in vivo but do not have a bleeding defect, suggesting that PKCalpha could be an attractive antithrombotic target. Important, apparently non-redundant, roles, both positive and negative, for the novel PKC isoforms delta, theta and epsilon in granule secretion have also been proposed, indicating highly complex regulation of this essential process. Similarly, PKCbeta, PKCdelta and PKCtheta have non-redundant roles in platelet spreading, as absence of either PKCbeta or PKCtheta reduces spreading, whereas PKCdelta negatively regulates filopodial formation. This negative signaling by PKCdelta may reduce platelet aggregation and so restrict thrombus formation. In this review, we discuss the current understanding of the regulation and functions of individual PKC isoforms in platelet activation and thrombus formation.
Collapse
Affiliation(s)
- M T Harper
- Department of Physiology and Pharmacology, University of Bristol, Bristol, UK
| | | |
Collapse
|
33
|
Lv XJ, Li YY, Zhang YJ, Mao M, Qian GS. Over-expression of caveolin-1 aggravate LPS-induced inflammatory response in AT-1 cells via up-regulation of cPLA2/p38 MAPK. Inflamm Res 2010; 59:531-41. [PMID: 20099006 DOI: 10.1007/s00011-010-0157-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2009] [Revised: 12/08/2009] [Accepted: 01/07/2010] [Indexed: 11/26/2022] Open
Abstract
OBJECTIVE AND DESIGN The aim of this study was to study the effect of caveolin-1 on the cytosolic phospholipase A2 (cPLA2), p38 mitogen-activated protein kinase (p38 MAPK) and nuclear factor kappaB (NF-kappaB) in mouse lung alveolar type-1 cells' (AT-1 cells) inflammatory response induced by LPS. MATERIALS AND METHODS Gene clone technique was used to over-express caveolin-1 in AT-1 cells by lentivirus vector. The level of tumor necrosis factor alpha (TNF-alpha), interleukin 6 (IL-6), cPLA2, p38 MAPK and NF-kappaB was measured by ELISA, western blotting and EMSA. TREATMENT AT-1 cells were treated with LPS. RESULTS Over-expression of caveolin-1 not only increased the production of pro-inflammatory cytokine TNF-alpha and IL-6, but also enhanced the expression of the cPLA2, p38 MAPK, and NF-kappaB. CONCLUSIONS Our data demonstrated that over-expression of caveolin-1 aggravates the AT-1 injury induced by LPS, involving in modulation of the cPLA2 mediated by the cPLA2/p38 MAPK pathway.
Collapse
Affiliation(s)
- Xue-Jun Lv
- Institute of Respiratory Disease, The Second Affiliated Hospital, Third Military Medical University, Chongqing, China
| | | | | | | | | |
Collapse
|
34
|
p38 mitogen-activated protein kinase activation during platelet storage: consequences for platelet recovery and hemostatic function in vivo. Blood 2009; 115:1835-42. [PMID: 19965619 DOI: 10.1182/blood-2009-03-211706] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Platelets undergo several modifications during storage that reduce their posttransfusion survival and functionality. One important feature of these changes, which are known as platelet storage lesion, is the shedding of the surface glycoproteins GPIb-alpha and GPV. We recently demonstrated that tumor necrosis factor-alpha converting enzyme (TACE/ADAM17) mediates mitochondrial injury-induced shedding of adhesion receptors and that TACE activity correlates with reduced posttransfusion survival of these cells. We now confirm that TACE mediates receptor shedding and clearance of platelets stored for 16 hours at 37 degrees C or 22 degrees C. We further demonstrate that both storage and mitochondrial injury lead to the phosphorylation of p38 mitogen-activated kinase (MAPK) in platelets and that TACE-mediated receptor shedding from mouse and human platelets requires p38 MAP kinase signaling. Protein kinase C, extracellular regulated-signal kinase MAPK, and caspases were not involved in TACE activation. Both inhibition of p38 MAPK and inactivation of TACE during platelet storage led to a markedly improved posttransfusion recovery and hemostatic function of platelets in mice. p38 MAPK inhibitors had only minor effects on the aggregation of fresh platelets under static or flow conditions in vitro. In summary, our data suggest that inhibition of p38 MAPK or TACE during storage may significantly improve the quality of stored platelets.
Collapse
|
35
|
Malaver E, Romaniuk MA, D'Atri LP, Pozner RG, Negrotto S, Benzadón R, Schattner M. NF-kappaB inhibitors impair platelet activation responses. J Thromb Haemost 2009; 7:1333-43. [PMID: 19566544 DOI: 10.1111/j.1538-7836.2009.03492.x] [Citation(s) in RCA: 98] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
BACKGROUND Although platelets are anucleated cells, they express several transcription factors that exert non-genomic functions, including the positive and negative regulation of platelet activation. NF-kappaB is a major transcriptional regulator of genes involved in survival, proliferation and inflammation. OBJECTIVE Because platelets play a critical role not only in hemostasis, but also in inflammation and tumor progression, we evaluated the role of NF-kappaB in platelet physiology. RESULTS Immunofluorescence, Western blotting and ELISA studies revealed that platelets express IkappaBalpha and NF-kappaB, and that stimulation with thrombin triggers IkappaBalpha phosphorylation and degradation and the binding of platelet NF-kappaB p65 subunit to synthetic oligonucleotides containing the consensus sequence for NF-kappaB. Two specific unrelated inhibitors of NF-kappaB activation, BAY 11-7082 and Ro 106-9920, reduced PAC-1 and fibrinogen binding to integrin alpha(IIb)beta3 and restricted platelet spreading on immobilized fibrinogen. Both inhibitors impaired aggregation mediated by ADP, epinephrine, collagen or thrombin, but not arachidonic acid. ATP release, TXB2 formation, P-selectin expression, ERK phosphorylation and cPLA2 activity stimulated by thrombin were reduced in BAY 11-7082- or Ro 106-9920-treated platelets. Although bleeding time was not affected, ADP-induced platelet aggregation was impaired in mice treated with BAY 11-7082. CONCLUSIONS Our results suggest that NF-kappaB may be a novel mediator of platelet responses. The blockade of platelet function by NF-kappaB inhibitors might be relevant in those clinical situations where these drugs are being considered for anti-tumor and/or anti-inflammatory therapy.
Collapse
Affiliation(s)
- E Malaver
- Hematological Research Institute, National Academy of Medicine, National Research Council (CONICET), Buenos Aires, Argentina
| | | | | | | | | | | | | |
Collapse
|
36
|
Hsu MF, Chang LC, Huang LJ, Kuo SC, Lee HY, Lu MC, Wang JP. The influence of acetylshikonin, a natural naphthoquinone, on the production of leukotriene B4 and thromboxane A2 in rat neutrophils. Eur J Pharmacol 2009; 607:234-43. [PMID: 19232341 DOI: 10.1016/j.ejphar.2009.02.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2008] [Revised: 01/20/2009] [Accepted: 02/09/2009] [Indexed: 12/11/2022]
Abstract
Both A23187 and formyl-Met-Leu-Phe (fMLP) induced the release of arachidonic acid and the production of thromboxane B(2) and leukotriene B(4) from rat neutrophils that were inhibited by acetylshikonin in a concentration-dependent manner. Acetylshikonin blocked exogenous arachidonic acid-induced leukotriene B(4) and thromboxane B(2) production in neutrophils and inhibited the enzymatic activity of ram seminal vesicles cyclooxygenase and human recombinant 5-lipoxygenase, whereas it had no effect on cytosolic phospholipase A(2) activity, in cell-free systems. 3-Morpholinosydnonimine- and 13S-hydroperoxy-9Z,11E-octadecadienoic acid (13-HpODE)-mediated dihydrorhodamine 123 oxidation (to assess the lipid peroxide and peroxynitrite scavenging activity) was reduced by acetylshikonin. The membrane recruitment of cytosolic phospholipase A(2) was inhibited, but the phosphorylation of cytosolic phospholipase A(2) was enhanced, by acetylshikonin in the A23187-induced response. Acetylshikonin alone stimulated extracellular signal regulated kinase (ERK) phosphorylation and enhanced this response in cells stimulated with A23187 and fMLP. The phosphorylation of ERKs and cytosolic phospholipase A(2) was attenuated by U0126, a mitogen-activated protein kinase (MAPK)/ERK kinase (MEK) inhibitor. Acetylshikonin facilitated both A23187- and fMLP-mediated translocation of 5-lipoxygenase to the membrane. Acetylshikonin attenuated both fMLP- and ionomycin-mediated [Ca(2+)](i) elevation. These results indicate that the inhibition of eicosanoid production by acetylshikonin is due to the attenuation of cytosolic phospholipase A(2) membrane recruitment via the decrease in [Ca(2+)](i) and to the blockade of cyclooxygenase and 5-lipoxygenase activity.
Collapse
Affiliation(s)
- Mei-Feng Hsu
- Department of Biochemistry, China Medical University, Taichung 404, Taiwan, Republic of China
| | | | | | | | | | | | | |
Collapse
|
37
|
Adam F, Kauskot A, Rosa JP, Bryckaert M. Mitogen-activated protein kinases in hemostasis and thrombosis. J Thromb Haemost 2008; 6:2007-16. [PMID: 18826389 DOI: 10.1111/j.1538-7836.2008.03169.x] [Citation(s) in RCA: 124] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The mitogen-activated protein (MAP) kinases ERK2, p38 and JNK1 are present in platelets and are activated by various stimuli, such as thrombin, collagen, von Willebrand factor (VWF) and ADP. Until recently, MAP kinases were only studied in the conventional model of agonist-induced platelet aggregation mediated by fibrinogen and integrin alphaIIbbeta3. However, this approach is likely to be too limited for a physiological understanding of platelet MAP kinases and their signaling pathways. Recent studies with varying blood-flow conditions and animal models of thrombosis have provided deeper insight into the role of MAP kinases in thrombus formation and the dependence of these kinases on shear conditions. This review summarizes and discusses the physiological functions of these kinases in hemostasis and thrombosis as revealed by various technical approaches.
Collapse
Affiliation(s)
- F Adam
- Centre de Recherche Cardiovasculaire INSERM Lariboisiére U689, Hôpital Lariboisiére, Paris, France
| | | | | | | |
Collapse
|
38
|
Two distinct roles of mitogen-activated protein kinases in platelets and a novel Rac1-MAPK-dependent integrin outside-in retractile signaling pathway. Blood 2008; 113:893-901. [PMID: 18957688 DOI: 10.1182/blood-2008-05-155978] [Citation(s) in RCA: 161] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Mitogen-activated protein kinases (MAPK), p38, and extracellular stimuli-responsive kinase (ERK), are acutely but transiently activated in platelets by platelet agonists, and the agonist-induced platelet MAPK activation is inhibited by ligand binding to the integrin alpha(IIb)beta(3). Here we show that, although the activation of MAPK, as indicated by MAPK phosphorylation, is initially inhibited after ligand binding to integrin alpha(IIb)beta(3), integrin outside-insignaling results in a late but sustained activation of MAPKs in platelets. Furthermore, we show that the early agonist-induced MAPK activation and the late integrin-mediated MAPK activation play distinct roles in different stages of platelet activation. Agonist-induced MAPK activation primarily plays an important role in stimulating secretion of platelet granules, while integrin-mediated MAPK activation is important in facilitating clot retraction. The stimulatory role of MAPK in clot retraction is mediated by stimulating myosin light chain (MLC) phosphorylation. Importantly, integrin-dependent MAPK activation, MAPK-dependent MLC phosphorylation, and clot retraction are inhibited by a Rac1 inhibitor and in Rac1 knockout platelets, indicating that integrin-induced activation of MAPK and MLC and subsequent clot retraction is Rac1-dependent. Thus, our results reveal 2 different activation mechanisms of MAPKs that are involved in distinct aspects of platelet function and a novel Rac1-MAPK-dependent cell retractile signaling pathway.
Collapse
|
39
|
Role of the p38 mitogen-activated protein kinase/cytosolic phospholipase A2 signaling pathway in blood-brain barrier disruption after focal cerebral ischemia and reperfusion. J Cereb Blood Flow Metab 2008; 28:1686-96. [PMID: 18545259 PMCID: PMC2562626 DOI: 10.1038/jcbfm.2008.60] [Citation(s) in RCA: 104] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Cytosolic phospholipase A(2) (cPLA(2)) is a key enzyme that mediates arachidonic acid metabolism, which causes cerebral ischemia-induced oxidative injury, blood-brain barrier (BBB) dysfunction, and edema. Recent reports have shown that p38 mitogen-activated protein kinase (MAPK) is related to phosphorylation and activation of cPLA(2) and release of arachidonic acid. However, involvement of the p38 MAPK pathway in cPLA(2) activation and of reactive oxygen species in expression of p38 MAPK/cPLA(2) after ischemia-reperfusion injury in the brain remains unclear. To address these issues, we used a model of transient focal cerebral ischemia (tFCI) in rats. Western blot analysis showed a significant increase in expression of phospho-p38 MAPK and phospho-cPLA(2) in rat brain cortex after tFCI. Activity assays showed that both p38 MAPK and cPLA(2) activation markedly increased 1 day after reperfusion. Intraventricular administration of SB203580 significantly suppressed activation and phosphorylation of cPLA(2) and attenuated BBB extravasation and subsequent edema. Moreover, overexpression of copper/zinc-superoxide dismutase remarkably diminished activation and phosphorylation of both p38 MAPK and cPLA(2) after reperfusion. These findings suggest that the p38 MAPK/cPLA(2) pathway may promote BBB disruption with secondary vasogenic edema and that superoxide anions can stimulate this pathway after ischemia-reperfusion injury.
Collapse
|
40
|
Garcia A, Shankar H, Murugappan S, Kim S, Kunapuli S. Regulation and functional consequences of ADP receptor-mediated ERK2 activation in platelets. Biochem J 2007; 404:299-308. [PMID: 17298299 PMCID: PMC1868805 DOI: 10.1042/bj20061584] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
We have previously shown that ADP-induced thromboxane generation in platelets requires signalling events from the G(q)-coupled P2Y1 receptor (platelet ADP receptor coupled to stimulation of phospholipase C) and the G(i)-coupled P2Y12 receptor (platelet ADP receptor coupled to inhibition of adenylate cyclase) in addition to outside-in signalling. While it is also known that extracellular calcium negatively regulates ADP-induced thromboxane A2 generation, the underlying mechanism remains unclear. In the present study we sought to elucidate the signalling mechanisms and regulation by extracellular calcium of ADP-induced thromboxane A2 generation in platelets. ERK (extracllular-signal-regulated kinase) 2 activation occurred when outside-in signalling was blocked, indicating that it is a downstream event from the P2Y receptors. However, blockade of either P2Y1 or the P2Y12 receptors with corresponding antagonists completely abolished ERK phosphorylation, indicating that both P2Y receptors are required for ADP-induced ERK activation. Inhibitors of Src family kinases or the ERK upstream kinase MEK [MAPK (mitogen-activated protein kinase)/ERK kinase] abrogated ADP-induced ERK phosphorylation and thromboxane A2 generation. Finally ADP- or G(i)+G(z)-induced ERK phosphorylation was blocked in the presence of extracellular calcium. The present studies show that ERK2 is activated downstream of P2Y receptors through a complex mechanism involving Src kinases and this plays an important role in ADP-induced thromboxane A2 generation. We also conclude that extracellular calcium blocks ADP-induced thromboxane A2 generation through the inhibition of ERK activation.
Collapse
Affiliation(s)
- Analia Garcia
- *Department of Physiology, Temple University School of Medicine, Philadelphia, PA 19140, U.S.A
- †Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, PA 19140, U.S.A
| | - Haripriya Shankar
- *Department of Physiology, Temple University School of Medicine, Philadelphia, PA 19140, U.S.A
| | - Swaminathan Murugappan
- *Department of Physiology, Temple University School of Medicine, Philadelphia, PA 19140, U.S.A
- †Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, PA 19140, U.S.A
| | - Soochong Kim
- *Department of Physiology, Temple University School of Medicine, Philadelphia, PA 19140, U.S.A
- †Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, PA 19140, U.S.A
| | - Satya P. Kunapuli
- *Department of Physiology, Temple University School of Medicine, Philadelphia, PA 19140, U.S.A
- †Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, PA 19140, U.S.A
- ‡Department of Pharmacology, Temple University School of Medicine, Philadelphia, PA 19140, U.S.A
- To whom correspondence should be addressed (email )
| |
Collapse
|
41
|
Pöschel B, Stanton PK. Comparison of cellular mechanisms of long-term depression of synaptic strength at perforant path-granule cell and Schaffer collateral-CA1 synapses. PROGRESS IN BRAIN RESEARCH 2007; 163:473-500. [PMID: 17765734 DOI: 10.1016/s0079-6123(07)63026-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
This chapter compares the cellular mechanisms that have been implicated in the induction and expression of long-term depression (LTD) at Schaffer collateral-CA1 synapses to perforant path-dentate gyrus (DG) synapses. In general, Schaffer collateral LTD and long-term potentiation (LTP) both appear to be a complex combination of many alterations in synaptic transmission that occur at both presynaptic and postsynaptic sites, while at perforant path synapses, most evidence has focused on postsynaptic long-term alterations. Within the DG, the medial perforant path is far more studied than lateral perforant path synapses, where most evidence relates to the induction of heterosynaptic LTD at lateral perforant path synapses when LTP is induced in the medial perforant path. Of course, there remain many other classes of synapses in the DG where synaptic plasticity, including LTD, have been largely neglected. It is clear that a better understanding of the range of DG loci where long-lasting activity-dependent plasticity, both LTD and LTP, are expressed will be essential to improve our understanding of the cognitive roles of such DG plasticity.
Collapse
Affiliation(s)
- Beatrice Pöschel
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, NY 10595, USA
| | | |
Collapse
|
42
|
García A, Senis YA, Antrobus R, Hughes CE, Dwek RA, Watson SP, Zitzmann N. A global proteomics approach identifies novel phosphorylated signaling proteins in GPVI-activated platelets: involvement of G6f, a novel platelet Grb2-binding membrane adapter. Proteomics 2006; 6:5332-43. [PMID: 16941570 PMCID: PMC1869047 DOI: 10.1002/pmic.200600299] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Collagen-related peptide (CRP) stimulates powerful activation of platelets through the glycoprotein VI (GPVI)-FcR gamma-chain complex. We have combined proteomics and traditional biochemistry approaches to study the proteome of CRP-activated platelets, focusing in detail on tyrosine phosphorylation. In two separate approaches, phosphotyrosine immunoprecipitations followed by 1-D-PAGE, and 2-DE, were used for protein separation. Proteins were identified by MS. By following these approaches, 96 proteins were found to undergo PTM in response to CRP in human platelets, including 11 novel platelet proteins such as Dok-1, SPIN90, osteoclast stimulating factor 1, and beta-Pix. Interestingly, the type I transmembrane protein G6f was found to be specifically phosphorylated on Tyr-281 in response to platelet activation by CRP, providing a docking site for the adapter Grb2. G6f tyrosine phoshporylation was also found to take place in response to collagen, although not in response to the G protein-coupled receptor agonists, thrombin and ADP. Further, we also demonstrate for the first time that Grb2 and its homolog Gads are tyrosine-phosphorylated in CRP-stimulated platelets. This study provides new insights into the mechanism of platelet activation through the GPVI collagen receptor, helping to build the basis for the development of new drug targets for thrombotic disease.
Collapse
Affiliation(s)
- Angel García
- Department of Biochemistry, Oxford Glycobiology Institute, University of Oxford, Oxford, UK.
| | | | | | | | | | | | | |
Collapse
|
43
|
Shankar H, Garcia A, Prabhakar J, Kim S, Kunapuli SP. P2Y12 receptor-mediated potentiation of thrombin-induced thromboxane A2 generation in platelets occurs through regulation of Erk1/2 activation. J Thromb Haemost 2006; 4:638-47. [PMID: 16460446 DOI: 10.1111/j.1538-7836.2006.01789.x] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
BACKGROUND Thromboxane A2 (TXA2) is a positive feedback lipid mediator that is generated upon stimulation of platelets with various agonists. Aspirin works as an antithrombotic drug by blocking the generation of TXA2. The aim of this study was to evaluate the role of the purinergic P2Y receptors in thrombin-induced TXA2 generation. RESULTS PAR1-activating peptide (SFLLRN), PAR4-activating peptide (AYPGKF), and thrombin, induced the activation of cytosolic phospholipase A2 (cPLA2), release of arachidonic acid (AA) from membrane-bound phospholipids, and subsequent TXA2 generation in human platelets. The actions of these agonists were significantly inhibited in the presence of the P2Y12 receptor antagonist, AR-C69931MX, but not the P2Y1 receptor antagonist, MRS2179. In addition, AYPGKF- and thrombin-induced TXA2 generation was significantly reduced in platelets from mice dosed with clopidogrel, confirming the results obtained with the human platelets. Also, Pearl mouse platelets that lack releasable nucleotides generated significantly less TXA2 when compared with the wild-type littermates in response to PAR stimulation. Inhibition of extracellular signal-regulated protein kinase 1/2 (Erk 1/2) activation using U0126, an inhibitor of MAP kinase kinase (MEK), suppressed PAR-mediated cPLA2 phosphorylation and TXA2 generation. Further, platelets that were pretreated with AR-C69931MX, as well as Pearl mouse platelets, displayed the reduced levels of Erk1/2 phosphorylation upon stimulation with the PAR agonists. CONCLUSIONS Based on these findings, we conclude that thrombin-induced Erk1/2 activation is essential for PAR-mediated TXA2 generation, which is potentiated by the P2Y12 receptor-mediated signaling pathway but not the P2Y1 receptor-mediated signaling pathway. Finally, using selective inhibitors of Src kinases, we show that PAR-mediated Src activation precedes Erk1/2 activation.
Collapse
Affiliation(s)
- H Shankar
- Department of Physiology, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | | | | | | | | |
Collapse
|
44
|
Leoncini G, Bruzzese D, Signorello MG. Activation of p38 MAPKinase/cPLA2 pathway in homocysteine-treated platelets. J Thromb Haemost 2006; 4:209-16. [PMID: 16409471 DOI: 10.1111/j.1538-7836.2005.01708.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Hyperhomocysteinemia is considered a risk factor in arterial and venous thrombosis. The mechanism by which homocysteine (HCy) supports atherothrombosis is still unknown and may be multifactorial. Earlier in vitro studies demonstrated that HCy induced arachidonic acid release and increased thromboxane B2 (TXB2) formation. In this work, we found that HCy stimulated the rapid and sustained phosphorylation of platelet p38 mitogen-activated protein kinase (p38 MAPK). The effect was time- and dose-dependent. The HCy effect on p38 MAPK phosphorylation was prevented by N-acetyl-L-cysteine and iloprost and was partially inhibited by nordihydroguaiaretic acid. Moreover, the incubation of platelets with HCy led to the phosphorylation of cytosolic phospholipase A2 (cPLA2). In addition HCy promoted cPLA2 activation, assessed as arachidonic acid release. The cPLA2 phosphorylation and activation were both impaired by the inhibition of p38 MAPK through SB203580. This effect was not complete, reaching at the most the 50% of the total. In FURA 2-loaded platelets, HCy induced a dose-dependent intracellular calcium rise suggesting that the calcium elevation promoted by HCy could participate in the cPLA2 activation, leading to arachidonic acid release and TXB2 formation. In conclusion, our data provide insight into the mechanisms of platelet activation induced by HCy, suggesting that the p38 MAPK/cPLA2 pathway could play a relevant role in platelet hyperactivity described in hyperhomocysteinemia.
Collapse
Affiliation(s)
- G Leoncini
- Department of Experimental Medicine, Biochemistry Section, University of Genoa, Genova, Italy.
| | | | | |
Collapse
|
45
|
Svensson CI, Hua XY, Powell HC, Lai J, Porreca F, Yaksh TL. Prostaglandin E2 release evoked by intrathecal dynorphin is dependent on spinal p38 mitogen activated protein kinase. Neuropeptides 2005; 39:485-94. [PMID: 16176831 DOI: 10.1016/j.npep.2005.08.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2005] [Accepted: 07/29/2005] [Indexed: 12/30/2022]
Abstract
Spinal dynorphin has been hypothesized to play a pivotal role in spinal sensitization. Although the mechanism of this action is not clear, several lines of evidence suggest that spinal dynorphin-induced hyperalgesia is mediated through an increase in spinal cyclooxygenase products via an enhanced N-methyl-D-aspartate (NMDA) receptor function. Spinal NMDA-evoked prostaglandin release and nociception has been linked to the activation of p38 mitogen activated protein kinase (p38). In the present work, we show that intrathecal delivery of an N-truncated fragment of dynorphin A, dynorphin A 2-17 (dyn2-17), which has no activity at opioid receptors, induced a 8-10-fold increase in phosphorylation of p38 in the spinal cord. The increase in phosphorylated p38 was detected in laminae I-IV of the dorsal horn. Moreover, confocal microscopy showed that the activation of p38 occurred in microglia, but not in neurons or astrocytes. In awake rats, prepared with chronically placed intrathecal loop dialysis catheters, the concentration of prostaglandin E2 in lumbar cerebrospinal fluid was increased 5-fold by intrathecal administration of dyn2-17. Injection of SD-282, a selective p38 inhibitor, but not PD98059, an ERK1/2 inhibitor, attenuated the prostaglanin E2 release. These data, taken together, support the hypothesis that dynorphin, independent of effects mediated by opioid receptors, has properties that can induce spinal sensitization and indicates that dyn2-17 effects may be mediated through activation of the p38 pathway. These studies provide an important downstream linkage where by dynorphin may act through a non-neuronal link to induce a facilitation of spinal nociceptive processing.
Collapse
Affiliation(s)
- Camilla I Svensson
- Department of Anesthesiology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0818, USA.
| | | | | | | | | | | |
Collapse
|
46
|
Svensson CI, Fitzsimmons B, Azizi S, Powell HC, Hua XY, Yaksh TL. Spinal p38beta isoform mediates tissue injury-induced hyperalgesia and spinal sensitization. J Neurochem 2005; 92:1508-20. [PMID: 15748168 DOI: 10.1111/j.1471-4159.2004.02996.x] [Citation(s) in RCA: 104] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Antagonist studies show that spinal p38 mitogen-activated protein kinase plays a crucial role in spinal sensitization. However, there are two p38 isoforms found in spinal cord and the relative contribution of these two to hyperalgesia is not known. Here we demonstrate that the isoforms are distinctly expressed in spinal dorsal horn: p38alpha in neurons and p38beta in microglia. In lieu of isoform selective inhibitors, we examined the functional role of these two individual isoforms in nociception by using intrathecal isoform-specific antisense oligonucleotides to selectively block the expression of the respective isoform. In these rats, down-regulation of spinal p38beta, but not p38alpha, prevented nocifensive flinching evoked by intraplantar injection of formalin and hyperalgesia induced by activation of spinal neurokinin-1 receptors through intrathecal injection of substance P. Both intraplantar formalin and intrathecal substance P produced an increase in spinal p38 phosphorylation and this phosphorylation (activation) was prevented when spinal p38beta, but not p38alpha, was down-regulated. Thus, spinal p38beta, probably in microglia, plays a significant role in spinal nociceptive processing and represents a potential target for pain therapy.
Collapse
Affiliation(s)
- Camilla I Svensson
- Department of Anesthesiology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | | | | | | | | | | |
Collapse
|
47
|
|
48
|
Cowan MJ, Yao XL, Pawliczak R, Huang X, Logun C, Madara P, Alsaaty S, Wu T, Shelhamer JH. The role of TFIID, the initiator element and a novel 5' TFIID binding site in the transcriptional control of the TATA-less human cytosolic phospholipase A2-alpha promoter. ACTA ACUST UNITED AC 2004; 1680:145-57. [PMID: 15507318 DOI: 10.1016/j.bbaexp.2004.09.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2004] [Revised: 09/20/2004] [Accepted: 09/20/2004] [Indexed: 12/17/2022]
Abstract
Human cytosolic phospholipase A2-alpha (cPLA2-alpha) is a critical enzyme in the liberation of arachidonic acid (AA) from cellular membranes and the subsequent formation of prostaglandins (PGs), leukotrienes (LTs), hydroxyeicosatetraenoic acids (HETEs) and platelet activating factor in many different cell types. Much is known of the effect of posttranslational phosphorylation and calcium binding events on the enzymatic activity of cPLA2-alpha, but to date little is known about its specific transcriptional control. Through the use of reporter gene constructs and eletrophoretic mobility shift assays (EMSAs), this study determined the minimal promoter required for basal transcriptional activity of the human cPLA2-alpha promoter to include base pairs -40 through the transcription start site (TSS). In addition, it confirms the importance of an initiator (Inr) element at the TSS by deletion reporter gene analysis, and further identifies bases -3 (C) and -2 (T) as critical bases in the Inr function by mutation reporter gene analysis. Finally, this study describes a novel AAGGAG motif at -30 to -35 which is bound by TATA-box binding protein (TBP) and is critical for basal transcriptional activity.
Collapse
Affiliation(s)
- Mark J Cowan
- Division of Pulmonary and Critical Care Medicine, The University of Maryland, 10 North Greene Street, Room 3D-127, Baltimore, MD 21201, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Canobbio I, Balduini C, Torti M. Signalling through the platelet glycoprotein Ib-V–IX complex. Cell Signal 2004; 16:1329-44. [PMID: 15381249 DOI: 10.1016/j.cellsig.2004.05.008] [Citation(s) in RCA: 96] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2004] [Accepted: 05/12/2004] [Indexed: 11/16/2022]
Abstract
The glycoprotein Ib-V-IX is one of the major adhesive receptors expressed on the surface of circulating platelets. It is composed of four different polypeptides-GPIbalpha, GPIbbeta, GPIX, and GPV-and represents a multifunctional receptor able to interact with a number of ligands, including the adhesive protein von Willebrand factor, the coagulation factors thrombin, factors XI and XII, and the membrane glycoproteins P-selectin and Mac-1. Interaction of GPIb-V-IX with the subendothelial von Willebrand factor is essential for primary haemostasis, as it initiates platelet adhesion to the subendothelial matrix at the sites of vascular injury even under high flow conditions. Upon interaction with von Willebrand factor, GPIb-V-IX initiates transmembrane signalling events for platelet activation, which eventually result in integrin alpha(IIb)beta(3) stimulation and platelet aggregation. The investigation of the biochemical mechanisms for platelet activation by GPIb-V-IX has attracted increasing attention during the last years. This review will describe and discuss recent findings that have provided new insights into the events underlying GPIb-V-IX transmembrane signalling. In particular, it will summarise basic concepts on the structure of this receptor, extracellular ligands, and intracellular interactors potentially involved in transmembrane signalling. The recently suggested role of membrane Fc receptors in GPIb-V-IX-initiated platelet activation will also be discussed, along with the involvement of lipid metabolising enzymes, tyrosine kinases, and the cytoskeleton in the crosstalk between GPIb-V-IX and integrin alpha(IIb)beta(3).
Collapse
Affiliation(s)
- Ilaria Canobbio
- Center of Excellence for Applied Biology, Department of Biochemistry, University of Pavia, via Bassi 21, Pavia 27100, Italy
| | | | | |
Collapse
|
50
|
Abstract
Hypercholesterolemia is the dominant risk factor associated with atherothrombotic disorders in the western world. Consequently, much attention has been devoted to defining its role in the pathogenesis of atherosclerosis. It is currently recognized that hypercholesterolemia induces phenotypic changes in the microcirculation that are consistent with oxidative and nitrosative stresses. Superoxide is generated via several cellular systems and, once formed, participates in a number of reactions, yielding various free radicals, such as hydrogen peroxide, peroxynitrite, or oxidized low-density lipoproteins. Once oxidant stress is invoked, characteristic pathophysiologic features ensue, such as platelet activation and lipid peroxidation, which are both involved in the initiation and progression of the atherosclerotic lesions. Thus, therapeutic strategies that act to maintain the normal balance in the oxidant status of the vascular bed may prove effective in reducing the deleterious consequences of hypercholesterolemia.
Collapse
Affiliation(s)
- Patrizia Ferroni
- Department of Experimental Medicine and Pathology, University La Sapienza, 00161 Rome, Italy
| | | | | | | |
Collapse
|