301
|
Badrnya S, Schrottmaier WC, Kral JB, Yaiw KC, Volf I, Schabbauer G, Söderberg-Nauclér C, Assinger A. Platelets mediate oxidized low-density lipoprotein-induced monocyte extravasation and foam cell formation. Arterioscler Thromb Vasc Biol 2014; 34:571-80. [PMID: 24371083 DOI: 10.1161/atvbaha.113.302919] [Citation(s) in RCA: 109] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
OBJECTIVE A growing body of evidence indicates that platelets contribute to the onset and progression of atherosclerosis by modulating immune responses. We aimed to elucidate the effects of oxidized low-density lipoprotein (OxLDL) on platelet-monocyte interactions and the consequences of these interactions on platelet phagocytosis, chemokine release, monocyte extravasation, and foam cell formation. APPROACH AND RESULTS Confocal microscopy and flow cytometric analysis revealed that in vitro and in vivo stimulation with OxLDL resulted in rapid formation of platelet-monocyte aggregates, with a preference for CD16+ monocyte subsets. This platelet-monocyte interaction facilitated OxLDL uptake by monocytes, in a process that involved platelet CD36-OxLDL interaction, release of chemokines, such as CXC motif ligand 4, direct platelet-monocyte interaction, and phagocytosis of platelets. Inhibition of cyclooxygenase with acetylsalicylic acid and antagonists of ADP receptors, P2Y1 and P2Y12, partly abrogated OxLDL-induced platelet-monocyte aggregates and platelet-mediated lipid uptake in monocytes. Platelets also enhanced OxLDL-induced monocyte transmigration across an endothelial monolayer via direct interaction with monocytes in a transwell assay. Importantly, in LDLR(-/-) mice, platelet depletion resulted in a significant decrease of peritoneal macrophage recruitment and foam cell formation in a thioglycollate-elicited peritonitis model. In platelet-depleted wild-type mice, transfusion of ex vivo OxLDL-stimulated platelets induced monocyte extravasation to a higher extent when compared with resting platelets. CONCLUSIONS Our results on OxLDL-mediated platelet-monocyte aggregate formation, which promoted phenotypic changes in monocytes, monocyte extravasation and enhanced foam cell formation in vitro and in vivo, provide a novel mechanism for how platelets potentiate key steps of atherosclerotic plaque development and plaque destabilization.
Collapse
Affiliation(s)
- Sigrun Badrnya
- From the Institute of Physiology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria (S.B., W.C.S., J.B.K., I.V., G.S., A.A.); and Department of Medicine, Solna, Center for Molecular Medicine, Karolinska Institute, Stockholm, Sweden (K.-C.Y., C.S.-N., A.A.)
| | | | | | | | | | | | | | | |
Collapse
|
302
|
Abstract
Despite their small size and anucleate status, platelets have diverse roles in vascular biology. Not only are platelets the cellular mediator of thrombosis, but platelets are also immune cells that initiate and accelerate many vascular inflammatory conditions. Platelets are linked to the pathogenesis of inflammatory diseases such as atherosclerosis, malaria infection, transplant rejection, and rheumatoid arthritis. In some contexts, platelet immune functions are protective, whereas in others platelets contribute to adverse inflammatory outcomes. In this review, we will discuss platelet and platelet-derived mediator interactions with the innate and acquired arms of the immune system and platelet-vessel wall interactions that drive inflammatory disease. There have been many recent publications indicating both important protective and adverse roles for platelets in infectious disease. Because of this new accumulating data, and the fact that infectious disease continues to be a leading cause of death globally, we will also focus on new and emerging concepts related to platelet immune and inflammatory functions in the context of infectious disease.
Collapse
|
303
|
Liu J, Li J, Deng X. Proteomic analysis of differential protein expression in platelets of septic patients. Mol Biol Rep 2014; 41:3179-85. [PMID: 24562620 DOI: 10.1007/s11033-014-3177-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Accepted: 01/16/2014] [Indexed: 12/26/2022]
Abstract
Sepsis is one of the major health problems all over the world. Early diagnostic of sepsis is an attractive strategy to decrease the mortality of septic patients. However, an effective biomarker that fulfills all the necessary requirements for the accurate characterization of sepsis is still unavailable until now. In this study, the 2-DE technique followed by mass spectrometry and a database search was used for searching and identifying the differential expressed proteins in platelets between septic patients and paired healthy controls. Platelet 2-DE profiles of septic patients and paired healthy controls with high resolution and reproducibility were obtained. Differential platelet 2-DE profiles between septic patients and paired healthy controls were established. Differential protein spots between normal healthy volunteers and septic patients from platelet 2-DE profiles were identified by 2-DE followed with mass spectrometry and a database search. Five proteins with increased expression were identified between septic patients and healthy controls from platelet samples. These up-expressed proteins were EF-hand calcium-binding domain-containing protein 7, actin, interleukin-1β, glycoprotein IX, and glycoprotein IIB. Sepsis induces a complex regulation of platelet protein changes. Our study highlights the important role of these differential expressed proteins in sepsis, which deserve further research as potential candidates for therapeutic strategies. Furthermore, our research is beneficial for the future developments of sepsis diagnosis and therapy.
Collapse
Affiliation(s)
- Ji Liu
- Department of Anesthesiology, 306 Hospital of PLA, Beijing, 100101, China
| | | | | |
Collapse
|
304
|
Murphy AJ, Sarrazy V, Wang N, Bijl N, Abramowicz S, Westerterp M, Welch CB, Schuetz JD, Yvan-Charvet L. Deficiency of ATP-binding cassette transporter B6 in megakaryocyte progenitors accelerates atherosclerosis in mice. Arterioscler Thromb Vasc Biol 2014; 34:751-8. [PMID: 24504733 DOI: 10.1161/atvbaha.113.302613] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE The ATP-binding cassette (ABC) transporter B6 (ABCB6) is highly expressed in megakaryocyte progenitors, but its role in platelet production and disease has not been elucidated. APPROACH AND RESULTS Among various ABC transporters, ABCB6 was highly expressed in megakaryocyte progenitors, exhibiting the same pattern of expression of genes involved in heme synthesis pathway. Transplantation of Abcb6 deficient (Abcb6(-/-)) bone marrow into low density lipoprotein receptor deficient recipient mice resulted in expansion and proliferation of megakaryocyte progenitors, attributable to increased reactive oxygen species production in response to porphyrin loading. The enhanced megakaryopoiesis in Abcb6(-/-) bone marrow-transplanted mice was further illustrated by increased platelet counts, mean platelet volume, and platelet activity. Platelets from Abcb6(-/-) bone marrow-transplanted mice had higher levels of chemokine (C-C motif) ligand 5, which was associated with increased plasma chemokine (C-C motif) ligand 5 levels. There were also increased platelet-leukocyte aggregates, which resulted in leukocyte activation. Abcb6(-/-) bone marrow-transplanted mice had accelerated atherosclerosis which was associated with deposition of the chemotactic agent, chemokine (C-C motif) ligand 5 in atherosclerotic plaques, resulting in increased macrophage accumulation. CONCLUSIONS Our findings identify a new role of ABCB6 in preventing atherosclerosis development by dampening platelet production, reactivity, and chemokine (C-C motif) ligand 5 deposition in atherosclerotic lesions.
Collapse
Affiliation(s)
- Andrew J Murphy
- From Haematopoiesis and Leukocyte Biology, Baker IDI Heart and Diabetes Institute, Melbourne, Australia (A.J.M.); Department of Immunology, Monash University, Melbourne, Australia (A.J.M.); Institut National de la Sante et de la Recherche Medicale U1065, Centre Mediterraneen de Medecine Molecuaire (C3M), Atip-Avenir, Nice, France (V.S., L.Y.-C.); Department of Medicine, Division of Molecular Medicine, Columbia University, New York, NY (N.W., N.B., S.A., M.W., C.B.W.); and Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN (J.D.S.)
| | | | | | | | | | | | | | | | | |
Collapse
|
305
|
Corken A, Russell S, Dent J, Post SR, Ware J. Platelet glycoprotein Ib-IX as a regulator of systemic inflammation. Arterioscler Thromb Vasc Biol 2014; 34:996-1001. [PMID: 24504734 DOI: 10.1161/atvbaha.113.303113] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OBJECTIVE The platelet glycoprotein Ib-IX (GP Ib-IX) receptor is a well-characterized adhesion receptor supporting hemostasis and thrombosis via interactions with von Willebrand factor. We examine the GP Ib-IX/von Willebrand factor axis in murine polymicrobial sepsis, as modeled by cecal ligation and puncture (CLP). APPROACH AND RESULTS Genetic absence of the GP Ib-IX ligand, von Willebrand factor, prolongs survival after CLP, but absence of the receptor, GP Ib-IX, does not. Because absence of either von Willebrand factor or GP Ib-IX significantly impairs hemostasis and thrombosis, we sought to define additional GP Ib-IX-dependent pathways impacting survival in the CLP model. We document that the absence of GP Ib-IX leads to reduced platelet-neutrophil and platelet-monocyte interactions. Twenty-four hours after CLP, absence of GP Ib-IX coincides with an alteration in cytokine levels, such as tumor necrosis factor-α secreted by monocytes, and increased macrophage-1 antigen expression by neutrophils. CONCLUSIONS In contrast to the well-characterized proinflammatory properties of platelets, we describe in the CLP model an anti-inflammatory property associated with platelet GP Ib-IX. Thus, a single platelet receptor displays a dual modulatory role in both the thrombotic and inflammatory pathways associated with polymicrobial sepsis. In sharing leucine-rich motifs with toll-like receptors, platelet GP Ib-IX can be considered a multifunctional participant in hemostasis, thrombosis, and the inflammatory cascade. The results highlight a dynamic role for platelets in systemic inflammation and add to the complex pathophysiologic events that occur during the dysregulated coagulation and inflammation associated with sepsis.
Collapse
Affiliation(s)
- Adam Corken
- From the Departments of Physiology and Biophysics (A.C., S.R., J.D., J.W.) and Pathology (S.R.P.), University of Arkansas for Medical Sciences, Little Rock
| | | | | | | | | |
Collapse
|
306
|
Xu Y, Yu H, Sun H. Targeting the host hemostatic system function in bacterial infection for antimicrobial therapies. J Thromb Thrombolysis 2014; 37:66-73. [PMID: 24379143 DOI: 10.1007/s11239-013-0994-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The hemostatic system is an important player in host's response to infection. It has been shown that host hemostatic factors as well as platelets, interact with various proteins from bacteria and play important roles in host defense against infections. This review summarizes studies of function of host hemostatic system in host defense against bacterial infections and efforts to target hemostatic system interaction with pathogens to develop potential antimicrobial therapies.
Collapse
Affiliation(s)
- Yuanxi Xu
- Department of Internal Medicine, University of Missouri Hospital and Clinics, Columbia, MO, USA
| | | | | |
Collapse
|
307
|
Coimbra LS, Steffens JP, Muscará MN, Rossa C, Spolidorio LC. Antiplatelet drugs reduce the immunoinflammatory response in a rat model of periodontal disease. J Periodontal Res 2013; 49:729-35. [DOI: 10.1111/jre.12155] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/11/2013] [Indexed: 12/01/2022]
Affiliation(s)
- L. S. Coimbra
- Department of Physiology and Pathology; Faculdade de Odontologia de Araraquara; UNESP-Universidade Estadual Paulista; Araraquara SP Brazil
| | - J. P. Steffens
- Department of Physiology and Pathology; Faculdade de Odontologia de Araraquara; UNESP-Universidade Estadual Paulista; Araraquara SP Brazil
| | - M. N. Muscará
- Department of Pharmacology; Institute of Biomedical Sciences; Universidade de São Paulo São Paulo SP Brazil
| | - C. Rossa
- Department of Diagnosis and Surgery; Faculdade de Odontologia de Araraquara; UNESP-Universidade Estadual Paulista; Araraquara SP Brazil
| | - L. C. Spolidorio
- Department of Physiology and Pathology; Faculdade de Odontologia de Araraquara; UNESP-Universidade Estadual Paulista; Araraquara SP Brazil
| |
Collapse
|
308
|
Shifrin DA, Demory Beckler M, Coffey RJ, Tyska MJ. Extracellular vesicles: communication, coercion, and conditioning. Mol Biol Cell 2013; 24:1253-9. [PMID: 23630232 PMCID: PMC3639038 DOI: 10.1091/mbc.e12-08-0572] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Cells communicate with neighboring cells and condition their local environment by secreting soluble factors into the extracellular space. These well-studied facets of cell biology are essential for the establishment and maintenance of physiological homeostasis. However, accumulating evidence has revealed that specific ligands, enzymes, and macromolecules are distributed into the extracellular space by virtue of their association with small vesicles, which are released by a variety of cell types. Although the biological significance of such vesicles was initially debated, purification and subsequent functional studies have shown that these extracellular vesicles are bioactive organelles carrying a wide range of protein and nucleic acid cargoes. In many cases these vesicles are laden with molecules that are involved in cell signaling, although other diverse functions are being revealed at a rapid pace. In this Perspective, we discuss recent developments in the understanding of the major pathways of extracellular vesicle biogenesis and how these vesicles contribute to the maintenance of physiological homeostasis.
Collapse
Affiliation(s)
- David A Shifrin
- Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | | | | | | |
Collapse
|
309
|
Speth C, Löffler J, Krappmann S, Lass-Flörl C, Rambach G. Platelets as immune cells in infectious diseases. Future Microbiol 2013; 8:1431-51. [DOI: 10.2217/fmb.13.104] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Platelets have been shown to cover a broad range of functions. Besides their role in hemostasis, they have immunological functions and thus participate in the interaction between pathogens and host defense. Platelets have a broad repertoire of receptor molecules that enable them to sense invading pathogens and infection-induced inflammation. Consequently, platelets exert antimicrobial effector mechanisms, but also initiate an intense crosstalk with other arms of the innate and adaptive immunity, including neutrophils, monocytes/macrophages, dendritic cells, B cells and T cells. There is a fragile balance between beneficial antimicrobial effects and detrimental reactions that contribute to the pathogenesis, and many pathogens have developed mechanisms to influence these two outcomes. This review aims to highlight aspects of the interaction strategies between platelets and pathogenic bacteria, viruses, fungi and parasites, in addition to the subsequent networking between platelets and other immune cells, and the relevance of these processes for the pathogenesis of infections.
Collapse
Affiliation(s)
- Cornelia Speth
- Division of Hygiene & Medical Microbiology, Innsbruck Medical University Fritz-Pregl-Straße 3, A-6020 Innsbruck, Austria
| | - Jürgen Löffler
- Laboratory of Innate Immunity, Infection, Inflammation, University Hospital Würzburg, Würzburg, Germany
| | - Sven Krappmann
- Microbiology Institute – Clinical Microbiology, Immunology & Hygiene, University Hospital of Erlangen & Friedrich-Alexander-University Erlangen-Nürnberg, Germany
| | - Cornelia Lass-Flörl
- Division of Hygiene & Medical Microbiology, Innsbruck Medical University Fritz-Pregl-Straße 3, A-6020 Innsbruck, Austria
| | - Günter Rambach
- Division of Hygiene & Medical Microbiology, Innsbruck Medical University Fritz-Pregl-Straße 3, A-6020 Innsbruck, Austria
| |
Collapse
|
310
|
Jenne CN, Urrutia R, Kubes P. Platelets: bridging hemostasis, inflammation, and immunity. Int J Lab Hematol 2013; 35:254-61. [PMID: 23590652 DOI: 10.1111/ijlh.12084] [Citation(s) in RCA: 241] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2013] [Accepted: 02/06/2013] [Indexed: 01/08/2023]
Abstract
Although the function of platelets in the maintenance of hemostasis has been studied in great detail, more recent evidence has highlighted a central role for platelets in the host inflammatory and immune responses. Platelets by virtue of their large numbers and their ability to rapidly release a broad spectrum of immunomodulatory cytokines, chemokines, and other mediators act as circulating sentinels. Upon detection of a pathogen, platelets quickly activate and begin to drive the ensuing inflammatory response. Platelets have the ability to directly modulate the activity of neutrophils (phagocytosis, oxidative burst), endothelium (adhesion molecule and chemokine expression), and lymphocytes. Due to their diverse array of adhesion molecules and preformed chemokines, platelets are able to adhere to leukocytes and facilitate their recruitment to sites of tissue damage or infection. Furthermore, platelets directly participate in the capture and sequestration of pathogens within the vasculature. Platelet-neutrophil interactions are known to induce the release of neutrophil extracellular traps (NETs) in response to either bacterial or viral infection, and platelets have been shown to internalize pathogens, sequestering them in engulfment vacuoles. Finally, emerging data indicate that platelets also participate in the host immune response by directly killing infected cells. This review will highlight the central role platelets play in the initiation and modulation of the host inflammatory and immune responses.
Collapse
Affiliation(s)
- C N Jenne
- Calvin Phoebe & Joan Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | | | | |
Collapse
|
311
|
Gudbrandsdottir S, Hasselbalch HC, Nielsen CH. Activated Platelets Enhance IL-10 Secretion and Reduce TNF-α Secretion by Monocytes. THE JOURNAL OF IMMUNOLOGY 2013; 191:4059-67. [DOI: 10.4049/jimmunol.1201103] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
312
|
Aspirin attenuates platelet activation and immune activation in HIV-1-infected subjects on antiretroviral therapy: a pilot study. J Acquir Immune Defic Syndr 2013; 63:280-8. [PMID: 23406976 DOI: 10.1097/qai.0b013e31828a292c] [Citation(s) in RCA: 126] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BACKGROUND Mechanisms for increased cardiovascular risk in HIV-1-infected adults are incompletely understood, but platelet activation and immune activation leading to a prothrombotic state have been proposed as significant contributors. Aspirin has antiplatelet and immunomodulatory properties. We explored whether 1 week of low-dose aspirin attenuates platelet activation and immune activation in HIV-1-infected and virologically suppressed adults on antiretroviral therapy. METHODS Platelet activation and immune activation were measured in HIV-1-infected subjects virologically suppressed on antiretroviral therapy and controls before and after 1 week of low-dose aspirin. RESULTS Compared with control subjects, HIV-1-infected subjects had increased platelet activation, as measured by spontaneous platelet aggregation and aggregation in response to adenosine diphosphate, collagen, and arachidonic acid. After aspirin therapy, percent aggregation decreased similarly in both HIV-1-infected and control subjects to all platelet agonists tested except aggregation in response to arachidonic acid, which remained elevated in the HIV-1-infected group. HIV-1-infected subjects exhibited increased markers of T-cell activation (CD38 and HLA-DR) and monocyte activation (sCD14), which decreased after 1 week of aspirin therapy. Moreover, leukocyte responses to Toll-like receptor stimulation were enhanced after 1 week of aspirin therapy. In vitro studies showed that HIV-1 plasma could activate healthy platelets, which in turn activated monocytes, implicating a direct role for activated platelets in immune activation. CONCLUSIONS Our data demonstrate that heightened platelet activation and immune activation in treated HIV-1 disease are attenuated by 1 week of aspirin therapy. Aspirin should be further studied for its antithrombotic and immunomodulatory benefits in treated HIV-1 disease.
Collapse
|
313
|
Hoefer IE, Sels JW, Jukema JW, Bergheanu S, Biessen E, McClellan E, Daemen M, Doevendans P, de Groot P, Hillaert M, Horsman S, Ilhan M, Kuiper J, Pijls N, Redekop K, van der Spek P, Stubbs A, van de Veer E, Waltenberger J, van Zonneveld AJ, Pasterkamp G. Circulating cells as predictors of secondary manifestations of cardiovascular disease: design of the CIRCULATING CELLS study. Clin Res Cardiol 2013; 102:847-56. [PMID: 23975238 DOI: 10.1007/s00392-013-0607-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Accepted: 07/29/2013] [Indexed: 01/06/2023]
Abstract
Biomarkers for primary or secondary risk prediction of cardiovascular disease (CVD) are urgently needed to improve individual treatment and clinical trial design. The vast majority of biomarker discovery studies has concentrated on plasma/serum as an easily accessible source. Although numerous markers have been identified, their added predictive value on top of traditional risk factors has been limited, as the biological specimen does not specifically reflect expression profiles related with CVD progression and because the signal is often diluted by marker release from other organs. In contrast to serum markers, circulating cells serve as indicators of the actual disease state due to their active role in the pathogenesis of CVD and are responsible for the majority of secreted biomarkers. Therefore, the CIRCULATING CELLS study was initiated, focusing on the cellular effectors of atherosclerosis in the circulation. In total, 714 patients with coronary artery disease (CAD) symptoms were included. Blood cell fractions (monocytes, T-lymphocytes, platelets, granulocytes, PBMC) of all individual patients were isolated and stored for analysis. Concomitantly, extensive flow cytometric characterization of these populations was performed. From each patient, a detailed clinical profile together with extensive questionnaires about medical history and life style was obtained. Various high-throughput -omics approaches (protein, mRNA, miRNA) are currently being undertaken. Data will be integrated with advanced bioinformatics for discovery and validation of secondary risk markers for adverse events. Overall, the CIRCULATING CELLS study grants the interesting possibility that it will both identify novel biomarkers and provide useful insights into the pathophysiology of CAD in patients.
Collapse
Affiliation(s)
- Imo E Hoefer
- Laboratory of Experimental Cardiology, UMC Utrecht, Room G02.523, Heidelberglaan 100, 3584, Utrecht, CX, The Netherlands,
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
314
|
Du XJ, Kiriazis H. Pro-Inflammatory Role of Platelets in Hypertension-Mediated End-Organ Damage. Cardiovasc Drugs Ther 2013; 27:485-7. [DOI: 10.1007/s10557-013-6484-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
315
|
Gziut M, MacGregor HJ, Nevell TG, Mason T, Laight D, Shute JK. Anti-inflammatory effects of tobramycin and a copper-tobramycin complex with superoxide dismutase-like activity. Br J Pharmacol 2013; 168:1165-81. [PMID: 23072509 DOI: 10.1111/bph.12018] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2012] [Revised: 09/04/2012] [Accepted: 09/17/2012] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND AND PURPOSE Airway inflammation in cystic fibrosis (CF) patients is characterized by accumulations of neutrophils in the airway and T cells in bronchial tissue, with activation of platelets in the circulation. CF patients are routinely treated with systemic or inhaled tobramycin for airway infection with Pseudomonas aeruginosa. Clinical trials have indicated an anti-inflammatory effect of tobramycin beyond its bactericidal activity. Here, we investigate the anti-inflammatory properties of tobramycin in vitro and consider if these relate to the ability of tobramycin to bind copper, which is elevated in blood and sputum in CF. EXPERIMENTAL APPROACH A copper-tobramycin complex was synthesized. The effect of tobramycin and copper-tobramycin on neutrophil activation and migration of T cells and neutrophils across human lung microvascular endothelial cells in response to thrombin-activated platelets were investigated in vitro. Tobramycin uptake was detected by immunocytochemistry. Intracellular reactive oxygen species were detected using the fluorescent indicator, 2',7'-dichlorofluorescein diacetate (DCFDA). Neutrophil superoxide, hydrogen peroxide and neutrophil elastase activity were measured using specific substrates. Copper was measured using atomic absorption spectroscopy. KEY RESULTS Tobramycin and copper-tobramycin were taken up by endothelial cells via a heparan sulphate-dependent mechanism and significantly inhibited T-cell and neutrophil transendothelial migration respectively. Copper-tobramycin has intracellular and extracellular superoxide dismutase-like activity. Neutrophil elastase inhibition by α1-antitrypsin is enhanced in the presence of copper-tobramycin. Tobramycin and copper-tobramycin are equally effective anti-pseudomonal antibiotics. CONCLUSIONS AND IMPLICATIONS Anti-inflammatory effects of tobramycin in vivo may relate to the spontaneous formation of a copper-tobramycin complex, implying that copper-tobramycin may be more effective therapy.
Collapse
Affiliation(s)
- M Gziut
- Institute of Biomedical and Biomolecular Sciences, School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, UK
| | | | | | | | | | | |
Collapse
|
316
|
Buzgo M, Jakubova R, Mickova A, Rampichova M, Prosecka E, Kochova P, Lukas D, Amler E. Time-regulated drug delivery system based on coaxially incorporated platelet α-granules for biomedical use. Nanomedicine (Lond) 2013. [DOI: 10.2217/nnm.12.140] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Aim: Platelet derivatives serve as an efficient source of natural growth factors. In the current study, α-granules were incorporated into coaxial nanofibers. Materials & methods: A nanofiber scaffold containing α-granules was prepared by coaxial electrospinning. The biological potential of the nanofiber scaffold was evaluated in chondrocyte and mesenchymal stem cell cultivation studies. Additionally, the concentration of TGF-β1 was determined. Results: Microscopy studies showed that intact α-granules were incorporated into the coaxial nanofibers. The cultivation tests showed that the novel scaffold stimulated viability and extracellular matrix production of chondrocytes and mesenchymal stem cells. In addition, the concentration of growth factors necessary for the induction of cell proliferation significantly decreased. Conclusion: The system preserved α-granule bioactivity and stimulated cell viability and chondrogenic differentiation of mesenchymal stem cells. Core/shell nanofibers incorporating α-granules are a promising system for tissue engineering, particularly cartilage engineering. Original submitted 21 March 2012; Revised submitted 8 August 2012; Published online 2 December 2012
Collapse
Affiliation(s)
- Matej Buzgo
- Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, v.v.i, Vídeňská 1083, 142 20 Prague 4, Czech Republic
- Department of Biophysics, 2nd Faculty of Medicine, Charles University in Prague, V Úvalu 84, 150 06 Prague 5, Czech Republic.
| | - Radka Jakubova
- Department of Biophysics, 2nd Faculty of Medicine, Charles University in Prague, V Úvalu 84, 150 06 Prague 5, Czech Republic
- Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, v.v.i, Vídeňská 1083, 142 20 Prague 4, Czech Republic
| | - Andrea Mickova
- Department of Biophysics, 2nd Faculty of Medicine, Charles University in Prague, V Úvalu 84, 150 06 Prague 5, Czech Republic
- Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, v.v.i, Vídeňská 1083, 142 20 Prague 4, Czech Republic
| | - Michala Rampichova
- Department of Biophysics, 2nd Faculty of Medicine, Charles University in Prague, V Úvalu 84, 150 06 Prague 5, Czech Republic
- Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, v.v.i, Vídeňská 1083, 142 20 Prague 4, Czech Republic
| | - Eva Prosecka
- Department of Biophysics, 2nd Faculty of Medicine, Charles University in Prague, V Úvalu 84, 150 06 Prague 5, Czech Republic
- Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, v.v.i, Vídeňská 1083, 142 20 Prague 4, Czech Republic
| | - Petra Kochova
- Department of Mechanics, Faculty of Applied Sciences, University of West Bohemia, Univerzitni 8, 30614 Pilsen, Czech Republic
| | - David Lukas
- Department of Nonwovens, Technical University of Liberec, Studentska 2, 461 17 Liberec, Czech Republic
| | - Evžen Amler
- Department of Biophysics, 2nd Faculty of Medicine, Charles University in Prague, V Úvalu 84, 150 06 Prague 5, Czech Republic
- Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, v.v.i, Vídeňská 1083, 142 20 Prague 4, Czech Republic
| |
Collapse
|
317
|
Rahman M, Zhang S, Chew M, Syk I, Jeppsson B, Thorlacius H. Platelet shedding of CD40L is regulated by matrix metalloproteinase-9 in abdominal sepsis. J Thromb Haemost 2013; 11:1385-98. [PMID: 23617547 DOI: 10.1111/jth.12273] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2012] [Accepted: 04/12/2013] [Indexed: 01/05/2023]
Abstract
BACKGROUND AND OBJECTIVES Platelet-derived CD40L is known to regulate neutrophil recruitment and lung damage in sepsis. However, the mechanism regulating shedding of CD40L from activated platelets is not known. We hypothesized that matrix metalloproteinase (MMP)-9 might cleave surface-expressed CD40L and regulate pulmonary accumulation of neutrophils in sepsis. METHODS Abdominal sepsis was induced by cecal ligation and puncture (CLP) in wild-type and MMP-9-deficient mice. Edema formation, CXC chemokine levels, myeloperoxidase levels, neutrophils in the lung and plasma levels of CD40L and MMP-9 were quantified. RESULTS CLP increased plasma levels of MMP-9 but not MMP-2. The CLP-induced decrease in platelet surface CD40L and increase in soluble CD40L levels were significantly attenuated in MMP-9 gene-deficient mice. Moreover, pulmonary myeloperoxidase (MPO) activity and neutrophil infiltration in the alveolar space, as well as edema formation and lung injury, were markedly decreased in septic mice lacking MMP-9. In vitro studies revealed that inhibition of MMP-9 decreased platelet shedding of CD40L. Moreover, recombinant MMP-9 was capable of cleaving surface-expressed CD40L on activated platelets. In human studies, plasma levels of MMP-9 were significantly increased in patients with septic shock as compared with healthy controls, although MMP-9 levels did not correlate with organ injury score. CONCLUSIONS Our novel data propose a role of MMP-9 in regulating platelet-dependent infiltration of neutrophils and tissue damage in septic lung injury by controlling CD40L shedding from platelets. We conclude that targeting MMP-9 may be a useful strategy to limit acute lung injury in abdominal sepsis.
Collapse
Affiliation(s)
- M Rahman
- Department of Clinical Sciences, Section for Surgery, Lund University, Malmö, Sweden
| | | | | | | | | | | |
Collapse
|
318
|
The expression and concentration of CD40 ligand in normal pregnancy, preeclampsia, and hemolytic anemia, elevated liver enzymes and low platelet count (HELLP) syndrome. Blood Coagul Fibrinolysis 2013; 24:71-5. [PMID: 23241952 DOI: 10.1097/mbc.0b013e32835a8aca] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Preeclampsia has been associated with increased platelet activation detected before disease onset. Inappropriate activation of platelets may be involved in pathogenesis in preeclampsia by promoting coagulation and thrombosis and also as a mediator of inflammation. The exaggerated platelet activation and inflammation leading to endothelial damage in preeclampsia can be explained by the CD40-CD40 ligand (CD40L) system. Expression of CD40L on platelets was determined by whole-blood flow cytometry, and serum levels of soluble CD40L (sCD40L) were measured by enzyme-linked immunosorbent assay in 11 women with mild preeclampsia, 11 women with severe preeclampsia, and six women with hemolytic anemia, elevated liver enzymes and low platelet count (HELLP) syndrome compared with 13 normotensive pregnant women as a control group. The platelet surface expression of CD40L was significantly higher in women with mild and severe preeclampsia and HELLP compared with normal pregnancy group (P = 0.001; P ≤ 0.001; P = 0.003, respectively), with no significant difference being found between women with mild preeclampsia compared with HELLP and severe preeclampsia compared with HELLP (P = 0.2; P = 0.8, respectively). The serum concentration of sCD40L was significantly higher in women with mild and severe preeclampsia and HELLP compared with the normal pregnancy group (P = 0.001; P ≤ 0.001; P = 0.022, respectively), with no significant difference being found between women with mild compared with severe preeclampsia or HELLP and severe preeclampsia compared with HELLP (P = 0.7; P = 0.6; P = 0.6, respectively). In conclusion, the higher expression and concentration of CD40L in women with preeclampsia and HELLP syndrome compared with normal pregnant women may indicate an exaggerated activation of platelets and endothelial cells in the disorder.
Collapse
|
319
|
Langer HF, Chavakis T. Platelets and neurovascular inflammation. Thromb Haemost 2013; 110:888-93. [PMID: 23636306 DOI: 10.1160/th13-02-0096] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Accepted: 03/25/2013] [Indexed: 01/24/2023]
Abstract
Platelets participate in haemostasis and in thrombus formation in health and disease. Moreover, they contribute to inflammation and cooperate with immune cells in a magnitude of inflammatory/immune responses. Although the inflammatory response has been recognised to be critical in neuronal diseases such as Alzheimer's disease or multiple sclerosis and its mouse counterpart, experimental autoimmune encephalomyelitis, the participation of platelets in these diseases is poorly investigated so far. Emerging studies, however, point to an interesting crosstalk between platelets and neuroinflammation. For instance, when the integrity of the blood brain barrier is compromised, platelets may be relevant for endothelial inflammation, as well as recruitment and activation of inflammatory cells, thereby potentially contributing to central nervous tissue pathogenesis. This review summarises recent insights in the role of platelets for neurovascular inflammation and addresses potential underlying mechanisms, by which platelets may affect the pathophysiology of neurovascular diseases.
Collapse
Affiliation(s)
- H F Langer
- Harald F. Langer, MD, Department of Cardiology and Cardiovascular Medicine, University Clinic of Tuebingen, Tuebingen, Germany, E-mail:
| | | |
Collapse
|
320
|
Solomon Tsegaye T, Gnirß K, Rahe-Meyer N, Kiene M, Krämer-Kühl A, Behrens G, Münch J, Pöhlmann S. Platelet activation suppresses HIV-1 infection of T cells. Retrovirology 2013; 10:48. [PMID: 23634812 PMCID: PMC3660175 DOI: 10.1186/1742-4690-10-48] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2012] [Accepted: 04/22/2013] [Indexed: 11/10/2022] Open
Abstract
Background Platelets, anucleate cell fragments abundant in human blood, can capture HIV-1 and platelet counts have been associated with viral load and disease progression. However, the impact of platelets on HIV-1 infection of T cells is unclear. Results We found that platelets suppress HIV-1 spread in co-cultured T cells in a concentration-dependent manner. Platelets containing granules inhibited HIV-1 spread in T cells more efficiently than degranulated platelets, indicating that the granule content might exert antiviral activity. Indeed, supernatants from activated and thus degranulated platelets suppressed HIV-1 infection. Infection was inhibited at the stage of host cell entry and inhibition was independent of the viral strain or coreceptor tropism. In contrast, blockade of HIV-2 and SIV entry was less efficient. The chemokine CXCL4, a major component of platelet granules, blocked HIV-1 entry and neutralization of CXCL4 in platelet supernatants largely abrogated their anti-HIV-1 activity. Conclusions Release of CXCL4 by activated platelets inhibits HIV-1 infection of adjacent T cells at the stage of virus entry. The inhibitory activity of platelet-derived CXCL4 suggests a role of platelets in the defense against infection by HIV-1 and potentially other pathogens.
Collapse
|
321
|
Wasiluk A, Kemona H, Mantur M, Polewko A, Ozimirski A, Milewski R. Expression of P-selectin (CD62P) on platelets after thrombin and ADP in hypotrophic and healthy, full-term newborns. J Matern Fetal Neonatal Med 2013; 26:1321-4. [PMID: 23484959 DOI: 10.3109/14767058.2013.784251] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND Hypotrophic newborns in comparison with eutrophic newborns demonstrate a reduced blood platelet count and therefore may have haemostasis disorders. Expression of P-selectin (CD62P) on the surface of platelets is a marker of stimulated, activated blood platelets. The ability of platelets to react can be determined after the addition of the following activators: strong (thrombin) and weak (ADP). MATERIALS AND METHODS We studied 48 hypotrophic newborns, 25 females and 23 males, weighing less than the 10th centile and 55 healthy, full-term newborns, 25 females and 30 males. Expression of CD62P on the surface of platelets was examined in the native state, after the addition of thrombin or ADP. RESULTS Hypotrophic newborns exhibited almost double the percentage of platelets expressing CD62P compared with the control group, 4.21% versus 2.88%. After the addition of thrombin, the percentage was 31.5% versus 12.5%, p < 0.001, whereas after the addition of ADP, the percentage was 9.54% versus 4.5%, p = 0.002. CONCLUSIONS Hypotrophic newborns are capable of greater platelet activation in comparison with healthy term newborns. However, gender does not affect the expression of P-selectin.
Collapse
Affiliation(s)
- Alicja Wasiluk
- Department of Neonatology, Medical University of Bialystok, 15-276 Bialystok, ul. MC. Sklodowskiej-Curie 24a, Poland.
| | | | | | | | | | | |
Collapse
|
322
|
Chen KB, Chang SS, Tseng YL, Chiu TH, Liao CC, Ho M, Huang GS, Li CY. Amniotic fluid induces platelet-neutrophil aggregation and neutrophil activation. Am J Obstet Gynecol 2013; 208:318.e1-7. [PMID: 23313310 DOI: 10.1016/j.ajog.2013.01.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2012] [Revised: 11/02/2012] [Accepted: 01/07/2013] [Indexed: 01/20/2023]
Abstract
OBJECTIVE Amniotic fluid embolism syndrome is a fatal disease in pregnant women. The exact role of platelets and neutrophils in amniotic fluid embolism syndrome is not clear. We examined whether amniotic fluid could affect platelet-neutrophil aggregation and activation and the possible mechanisms. STUDY DESIGN Blood samples from the pregnant women were pretreated ex vivo with their own amniotic fluid. Flow cytometry was used to measure platelet-neutrophil aggregation and activation. Neutrophil-mediated activity of p38 mitogen-activated protein kinase and extracellular signal-regulated protein kinases 1 and 2 was analyzed by Western blotting. RESULTS Amniotic fluid significantly induced platelet-neutrophil aggregation, neutrophil CD11b expression, and reactive oxygen species production. Amniotic fluid induced minimal platelet P-selectin expression. The increase of intracellular calcium level of neutrophils and the activity of p38 mitogen-activated protein kinase were enhanced by amniotic fluid stimulation. CONCLUSION Amniotic fluid was able to induce neutrophil activation and platelet-neutrophil aggregation with minimal effect on platelet activation. These findings may provide a new insight in the understanding of the pathophysiologic condition of amniotic fluid embolism syndrome.
Collapse
|
323
|
Experimental and Clinical Pharmacology of Andrographis paniculata and Its Major Bioactive Phytoconstituent Andrographolide. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2013; 2013:846740. [PMID: 23634174 PMCID: PMC3619690 DOI: 10.1155/2013/846740] [Citation(s) in RCA: 140] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2012] [Revised: 02/12/2013] [Accepted: 02/12/2013] [Indexed: 12/11/2022]
Abstract
Andrographis paniculata (Burm. F) Nees, generally known as “king of bitters,” is an herbaceous plant in the family Acanthaceae. In China, India, Thailand, and Malaysia, this plant has been widely used for treating sore throat, flu, and upper respiratory tract infections. Andrographolide, a major bioactive chemical constituent of the plant, has shown anticancer potential in various investigations. Andrographolide and its derivatives have anti-inflammatory effects in experimental models asthma, stroke, and arthritis. In recent years, pharmaceutical chemists have synthesized numerous andrographolide derivatives, which exhibit essential pharmacological activities such as those that are anti-inflammatory, antibacterial, antitumor, antidiabetic, anti-HIV, antifeedant, and antiviral. However, what is noteworthy about this paper is summarizing the effects of andrographolide against cardiovascular disease, platelet activation, infertility, and NF-κB activation. Therefore, this paper is intended to provide evidence reported in relevant literature on qualitative research to assist scientists in isolating and characterizing bioactive compounds.
Collapse
|
324
|
Vara DS, Campanella M, Canobbio I, Dunn WB, Pizzorno G, Hirano M, Pula G. Autocrine amplification of integrin αIIbβ3 activation and platelet adhesive responses by deoxyribose-1-phosphate. Thromb Haemost 2013; 109:1108-19. [PMID: 23494007 DOI: 10.1160/th12-10-0751] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2012] [Accepted: 02/13/2013] [Indexed: 12/21/2022]
Abstract
Using direct injection mass spectrometry (DIMS) we discovered that deoxyribose-1-phosphate (dRP) is released by platelets upon activation. Interestingly, the addition of exogenous dRP to human platelets significantly increased platelet aggregation and integrin αIIbβ3 activation in response to thrombin. In parallel, genetically modified platelets with double genetic deletion of thymidine phosphorylase and uridine phosphorylase were characterised by reduced release of dRP, impaired aggregation and decreased integrin αIIbβ3 activation in response to thrombin. In vitro platelet adhesion onto fibrinogen and collagen under physiological flow conditions was potentiated by treatment of human platelets with exogenous dRP and impaired in transgenic platelets with reduced dRP release. Human and mouse platelets responded to dRP treatment with a sizeable increase in reactive oxygen species (ROS) generation and the pre-treament with the antioxidant apocynin abolished the effect of dRP on aggregation and integrin activation. Experiments directly assessing the activation of the small G protein Rap1b and protein kinase C suggested that dRP increases the basal levels of activity of these two pivotal platelet-activating pathways in a redox-dependent manner. Taken together, we present evidence that dRP is a novel autocrine amplifier of platelet activity, which acts on platelet redox levels and modulates integrin αIIbβ3.
Collapse
Affiliation(s)
- Dina S Vara
- Department of Pharmacy and Pharmacology, University of Bath, Claverton Campus, BA2 7AY, Bath, UK.
| | | | | | | | | | | | | |
Collapse
|
325
|
Gachet C. Les mécanismes moléculaires de l’activation plaquettaire. BULLETIN DE L ACADEMIE NATIONALE DE MEDECINE 2013. [DOI: 10.1016/s0001-4079(19)31591-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
326
|
Nagasawa A, Matsuno K, Tamura S, Hayasaka K, Shimizu C, Moriyama T. The basis examination of leukocyte-platelet aggregates with CD45 gating as a novel platelet activation marker. Int J Lab Hematol 2013; 35:534-41. [DOI: 10.1111/ijlh.12051] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2012] [Accepted: 11/28/2012] [Indexed: 11/30/2022]
Affiliation(s)
- A. Nagasawa
- Graduate School of Health Sciences; Hokkaido University; Sapporo Hokkaido Japan
| | - K. Matsuno
- Division of Laboratory and Transfusion Medicine; Hokkaido University Hospital; Sapporo Japan
| | - S. Tamura
- Graduate School of Health Sciences; Hokkaido University; Sapporo Hokkaido Japan
- Research Fellow of the Japan Society for the Promotion of Science; Tokyo Japan
| | - K. Hayasaka
- Division of Laboratory and Transfusion Medicine; Hokkaido University Hospital; Sapporo Japan
| | - C. Shimizu
- Division of Laboratory and Transfusion Medicine; Hokkaido University Hospital; Sapporo Japan
| | - T. Moriyama
- Medical Laboratory Science; Faculty of Health Sciences; Hokkaido University; Sapporo Japan
| |
Collapse
|
327
|
|
328
|
Langer HF, Geisler T, Gawaz M. Atherothrombosis and Coronary Artery Disease. Platelets 2013. [DOI: 10.1016/b978-0-12-387837-3.00032-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
329
|
Jayakumar T, Chen WF, Lu WJ, Chou DS, Hsiao G, Hsu CY, Sheu JR, Hsieh CY. A novel antithrombotic effect of sulforaphane via activation of platelet adenylate cyclase: ex vivo and in vivo studies. J Nutr Biochem 2012; 24:1086-95. [PMID: 23246160 DOI: 10.1016/j.jnutbio.2012.08.007] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2012] [Revised: 07/12/2012] [Accepted: 08/13/2012] [Indexed: 01/07/2023]
Abstract
Sulforaphane is a naturally occurring isothiocyanate, which can be found in cruciferous vegetables such as broccoli and cabbage. Sulforaphane was found to have very potent inhibitory effects on tumor growth through regulation of diverse mechanisms. However, no data are available concerning the effects of sulforaphane on platelet activation and its relative issues. Activation of platelets caused by arterial thrombosis is relevant to a variety of cardiovascular diseases. Hence, the aim of this study was to examine the in vivo antithrombotic effects of sulforaphane and its possible mechanisms in platelet activation. Sulforaphane (0.125 and 0.25 mg/kg) was effective in reducing the mortality of ADP-induced acute pulmonary thromboembolism in mice. Other in vivo studies also revealed that sulforaphane (0.25 mg/kg) significantly prolonged platelet plug formation in mice. In addition, sulforaphane (15-75 μM) exhibited more-potent activity of inhibiting platelet aggregation stimulated by collagen. Sulforaphane inhibited platelet activation accompanied by inhibiting relative Ca(2+) mobilization; phosphorylation of phospholipase C (PLC)γ2, protein kinase C (PKC), mitogen-activated protein kinases (MAPKs) and Akt; and hydroxyl radical (OH(●)) formation. Sulforaphane markedly increased cyclic (c)AMP, but not cyclic (c)GMP levels, and stimulated vasodilator-stimulated phosphoprotein (VASP) phosphorylation. SQ22536, an inhibitor of adenylate cyclase, but not ODQ (1H-[1,2,4]Oxadiazolo[4,3-a]quinoxal in-1-one), an inhibitor of guanylate cyclase, obviously reversed the sulforaphane-mediated effects on platelet aggregation; PKC activation, p38 MAPK, Akt and VASP phosphorylation; and OH(●) formation. Furthermore, a PI3-kinase inhibitor (LY294002) and a p38 MAPK inhibitor (SB203580) both significantly diminished PKC activation and p38 MAPK and Akt phosphorylation; in contrast, a PKC inhibitor (RO318220) did not diminish p38 MAPK or Akt phosphorylation stimulated by collagen. This study demonstrates for the first time that in addition to it originally being considered as an agent for prevention of tumor growth, sulforaphane possesses potent antiplatelet activity which may initially activate adenylate cyclase/cAMP, followed by inhibiting intracellular signals (such as the PI3-kinase/Akt and PLCγ2-PKC-p47 cascades) and ultimately inhibiting platelet activation. Therefore, this novel role of sulforaphane may represent a high therapeutic potential for treatment or prevention of cardiovascular diseases.
Collapse
Affiliation(s)
- Thanasekaran Jayakumar
- Department of Pharmacology, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
330
|
Speth C, Hagleitner M, Ott HW, Würzner R, Lass-Flörl C, Rambach G. Aspergillus
fumigatus Activates Thrombocytes by Secretion of Soluble Compounds. J Infect Dis 2012; 207:823-33. [DOI: 10.1093/infdis/jis743] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
331
|
Abstract
Inflammation is an underlying feature of a variety of human diseases. Because inflammatory diseases are a major cause of morbidity and mortality in developed countries, understanding the interaction of the most important factors involved is an important challenge. Although platelets are widely recognized as having a critical role in primary hemostasis and thrombosis, basic and clinical evidence increasingly identifies these enucleated cells as relevant modulators, as both effector and target cells, of the inflammatory response. The cross-talk between platelets, endothelial cells and leukocytes in the inflammatory milieu mat be seen as a double-edged sword which functions not only as an effective first-line defense mechanism but may also lead to organ failure and death in the absence of counter-regulation systems. The molecular mechanisms involved in the reciprocal activation of platelets, endothelial cells and leukocytes are beginning to be elucidated. In the light of the existing data from experimental and clinical studies it is conceivable that platelet adhesion molecules and platelet mediators provide promising targets for novel therapeutic strategies in inflammatory diseases. The potentially adverse effects of these approaches need to be carefully addressed and monitored, including alterations in hemostasis and coagulation and particularly the impairment of host defense mechanisms, given the recently identified pivotal role of platelets in pathogen recognition and bacterial trapping. In this review we discuss the most important recent advances in research into the cross-talk between platelets and vascular cells during inflammation and the clinical consequences of these interactions.
Collapse
|
332
|
Bonten TN, Snoep JD, Roest M, Rosendaal FR, van der Bom JG. Platelet reactivity is not associated with recurrent cardiovascular events in men with a history of myocardial infarction: a cohort study. J Thromb Haemost 2012; 10:2616-8. [PMID: 23072487 DOI: 10.1111/jth.12027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
333
|
Abstract
Abstract
Review on platelet function in inflammation and atherosclerosis.
Platelets play a crucial role in the physiology of the primary hemostasis and in the pathophysiological activity of arterial thrombosis, provide rapid protection against bleeding, and catalyze the formation of stable blood clots via the coagulation cascade. Over the past years, it has become clear that platelets are important, not only in hemostasis and thrombosis but also in inflammation and in distinct aspects of atherosclerosis. Nowadays, platelets are known to have a large variety of functions. Platelets are able to interact with a large variety of cell types, such as leukocytes, endothelial cells, and SMCs, and these interactions have been implicated in the pathophysiology of vascular inflammation. In addition, platelets carry a highly inflammatory payload and are able to transport, synthesize, and deposit cytokines, chemokines, and lipid mediators, thereby initiating and propagating atherosclerotic disease. In this review, the current state of the art of the proinflammatory functions in the context of atherosclerotic cardiovascular disease will be outlined.
Collapse
Affiliation(s)
- Delia Projahn
- Institute for Cardiovascular Prevention, University Clinic of the Ludwig-Maximilians-University of Munich , Munich, Germany
- Institute for Molecular Cardiovascular Research, Medical Faculty, Rheinisch-Westfälische Technische Hochschule Aachen University , Aachen, Germany
| | - Rory R Koenen
- Institute for Cardiovascular Prevention, University Clinic of the Ludwig-Maximilians-University of Munich , Munich, Germany
- Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht , The Netherlands
| |
Collapse
|
334
|
Ferroni P, Vazzana N, Riondino S, Cuccurullo C, Guadagni F, Davì G. Platelet function in health and disease: from molecular mechanisms, redox considerations to novel therapeutic opportunities. Antioxid Redox Signal 2012; 17:1447-85. [PMID: 22458931 DOI: 10.1089/ars.2011.4324] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Increased oxidative stress appears to be of fundamental importance in the pathogenesis and development of several disease processes. Indeed, it is well known that reactive oxygen species (ROS) exert critical regulatory functions within the vascular wall, and it is, therefore, plausible that platelets represent a relevant target for their action. Platelet activation cascade (including receptor-mediated tethering to the endothelium, rolling, firm adhesion, aggregation, and thrombus formation) is tightly regulated. In addition to already well-defined platelet regulatory factors, ROS may participate in the regulation of platelet activation. It is already established that enhanced ROS release from the vascular wall can indirectly affect platelet activity by scavenging nitric oxide (NO), thereby decreasing the antiplatelet properties of endothelium. On the other hand, recent data suggest that platelets themselves generate ROS, which may evoke pro-thrombotic responses, triggering many biological processes participating in atherosclerosis initiation, progression, and complication. That oxidative stress may alter platelet function is conceivable when considering that antioxidants play a role in the prevention of cardiovascular disease, although the precise mechanism accounting for changes attributable to antioxidants in atherosclerosis remains unknown. It is possible that the effects of antioxidants may be a consequence of their enhancing or promoting the antiplatelet effects of NO derived from both endothelial cells and platelets. This review focuses on current knowledge regarding ROS-dependent regulation of platelet function in health and disease, and summarizes in vitro and in vivo evidence for their physiological and potential therapeutic relevance.
Collapse
Affiliation(s)
- Patrizia Ferroni
- Department of Laboratory Medicine and Advanced Biotechnologies, IRCCS San Raffaele Pisana, Rome, Italy
| | | | | | | | | | | |
Collapse
|
335
|
Berthet J, Damien P, Hamzeh-Cognasse H, Arthaud CA, Eyraud MA, Zéni F, Pozzetto B, McNicol A, Garraud O, Cognasse F. Human platelets can discriminate between various bacterial LPS isoforms via TLR4 signaling and differential cytokine secretion. Clin Immunol 2012; 145:189-200. [PMID: 23108090 DOI: 10.1016/j.clim.2012.09.004] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Revised: 09/07/2012] [Accepted: 09/12/2012] [Indexed: 12/15/2022]
Abstract
Platelets are currently acknowledged as cells of innate immunity and inflammation and play a complex role in sepsis. We examined whether different types of LPS have different effects on the release of soluble signaling/effective molecules from platelets. We used platelet-rich plasma from healthy volunteers and LPS from two strains of gram-negative bacteria with disparate LPS structures. We combined LPS-stimulated platelet supernatants with reporter cells and measured the PBMC cytokine secretion profiles. Upon stimulation of platelets with both Escherichia coli O111 and Salmonella minnesota LPS, the platelet LPS::TLR4 interaction activated pathways to trigger the production of a large number of molecules. The different platelet supernatants caused differential PBMC secretion of IL-6, TNFα, and IL-8. Our data demonstrate that platelets have the capacity to sense external signals differentially through a single type of pathogen recognition receptor and adjust the innate immune response appropriately for pathogens exhibiting different types of 'danger' signals.
Collapse
Affiliation(s)
- Julien Berthet
- Université de Lyon, F-42023, GIMAP, EA3064, Saint-Etienne, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
336
|
Crosstalk between Platelets and the Immune System: Old Systems with New Discoveries. Adv Hematol 2012; 2012:384685. [PMID: 23008717 PMCID: PMC3447344 DOI: 10.1155/2012/384685] [Citation(s) in RCA: 170] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2012] [Accepted: 08/15/2012] [Indexed: 11/17/2022] Open
Abstract
Platelets are small anucleate cells circulating in the blood. It has been recognized for more than 100 years that platelet adhesion and aggregation at the site of vascular injury are critical events in hemostasis and thrombosis; however, recent studies demonstrated that, in addition to these classic roles, platelets also have important functions in inflammation and the immune response. Platelets contain many proinflammatory molecules and cytokines (e.g., P-selectin, CD40L, IL-1β, etc.), which support leukocyte trafficking, modulate immunoglobulin class switch, and germinal center formation. Platelets express several functional Toll-like receptors (TLRs), such as TLR-2, TLR-4, and TLR-9, which may potentially link innate immunity with thrombosis. Interestingly, platelets also contain multiple anti-inflammatory molecules and cytokines (e.g., transforming growth factor-β and thrombospondin-1). Emerging evidence also suggests that platelets are involved in lymphatic vessel development by directly interacting with lymphatic endothelial cells through C-type lectin-like receptor 2. Besides the active contributions of platelets to the immune system, platelets are passively targeted in several immune-mediated diseases, such as autoimmune thrombocytopenia, infection-associated thrombocytopenia, and fetal and neonatal alloimmune thrombocytopenia. These data suggest that platelets are important immune cells and may contribute to innate and adaptive immunity under both physiological and pathological conditions.
Collapse
|
337
|
Patel S, Huang YW, Reheman A, Pluthero FG, Chaturvedi S, Mukovozov IM, Tole S, Liu GY, Li L, Durocher Y, Ni H, Kahr WHA, Robinson LA. The cell motility modulator Slit2 is a potent inhibitor of platelet function. Circulation 2012; 126:1385-95. [PMID: 22865890 DOI: 10.1161/circulationaha.112.105452] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Vascular injury and atherothrombosis involve vessel infiltration by inflammatory leukocytes, migration of medial vascular smooth muscle cells to the intimal layer, and ultimately acute thrombosis. A strategy to simultaneously target these pathological processes has yet to be identified. The secreted protein, Slit2, and its transmembrane receptor, Robo-1, repel neuronal migration in the developing central nervous system. More recently, it has been appreciated that Slit2 impairs chemotaxis of leukocytes and vascular smooth muscle cells toward diverse inflammatory attractants. The effects of Slit2 on platelet function and thrombus formation have never been explored. METHODS AND RESULTS We detected Robo-1 expression in human and murine platelets and megakaryocytes and confirmed its presence via immunofluorescence microscopy and flow cytometry. In both static and shear microfluidic assays, Slit2 impaired platelet adhesion and spreading on diverse extracellular matrix substrates by suppressing activation of Akt. Slit2 also prevented platelet activation on exposure to ADP. In in vivo studies, Slit2 prolonged bleeding times in murine tail bleeding assays. Using intravital microscopy, we found that after mesenteric arteriolar and carotid artery injury, Slit2 delayed vessel occlusion time and prevented the stable formation of occlusive arteriolar thrombi. CONCLUSIONS These data demonstrate that Slit2 is a powerful negative regulator of platelet function and thrombus formation. The ability to simultaneously block multiple events in vascular injury may allow Slit2 to effectively prevent and treat thrombotic disorders such as myocardial infarction and stroke.
Collapse
Affiliation(s)
- Sajedabanu Patel
- The Hospital for Sick Children, 555 University Ave, Toronto, ON, Canada M5G 1X8
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
338
|
Huang HS, Chang HH. Platelets in inflammation and immune modulations: functions beyond hemostasis. Arch Immunol Ther Exp (Warsz) 2012; 60:443-51. [PMID: 22940877 DOI: 10.1007/s00005-012-0193-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2011] [Accepted: 02/29/2012] [Indexed: 12/13/2022]
Abstract
Platelets play central roles for maintaining the homeostasis of the blood coagulation. As they are also involved in immune responses and host defenses, increasing evidences have suggested that platelets exert other roles beyond their well-recognized function in preventing bleeding. This review is focused on inflammation, allergy and immune modulations of platelets. Platelets conduct immunoregulation through secretion of functional mediators, interaction with various immune cells, endothelial cells and beneficial for the leukocyte infiltration to inflamed/allergic tissues. In these regulations, the leukocytes are influenced by and receiving the signals from platelets. In contrast, rare attentions were focused on platelet regulations by immune system. An intriguingly example in the intravenous immunoglobulin (IVIg) treatment is discussed, in which dendritic cells exert anti-inflammatory effect through platelets. This further suggests that coagulant and immune systems are tightly associated rather than separate entities. The cross-talks between these two systems implicate that platelet therapy may have application beyond thrombosis, and immune interventions may have potentials to treat thrombosis diseases.
Collapse
Affiliation(s)
- Hsuan-Shun Huang
- Department of Molecular Biology and Human Genetics, Tzu Chi University, 701 Sec. 3, Chung Yang Rd, Hualien, 970, Taiwan, ROC
| | | |
Collapse
|
339
|
Riccioni G, Sblendorio V. Atherosclerosis: from biology to pharmacological treatment. J Geriatr Cardiol 2012; 9:305-17. [PMID: 23097661 PMCID: PMC3470030 DOI: 10.3724/sp.j.1263.2012.02132] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2012] [Revised: 05/23/2012] [Accepted: 06/01/2012] [Indexed: 12/13/2022] Open
Abstract
A recent explosion in the amount of cardiovascular risk has swept across the globe. Primary prevention is the preferred method to lower cardiovascular risk. Lowering the prevalence of obesity is the most urgent matter, and is pleiotropic since it affects blood pressure, lipid profiles, glucose metabolism, inflammation, and atherothrombotic disease progression. Given the current obstacles, success of primary prevention remains uncertain. At the same time, the consequences of delay and inaction will inevitably be disastrous, and the sense of urgency mounts. Pathological and epidemiological data confirm that atherosclerosis begins in early childhood, and advances seamlessly and inexorably throughout life. Risk factors in childhood are similar to those in adults, and track between stages of life. When indicated, aggressive treatment should begin at the earliest indication, and be continued for many years. For those patients at intermediate risk according to global risk scores, C-reactive protein, coronary artery calcium, and carotid intima-media thickness are available for further stratification. Using statins for primary prevention is recommended by guidelines, is prevalent, but remains under prescribed. Statin drugs are unrivaled, evidence-based, major weapons to lower cardiovascular risk. Even when low density lipoprotein cholesterol targets are attained, over half of patients continue to have disease progression and clinical events. Though clinical evidence is incomplete, altering or raising the blood high density lipoprotein cholesterol level continues to be pursued. The aim of this review is to point out the attention of key aspects of vulnerable plaques regarding their pathogenesis and treatment.
Collapse
Affiliation(s)
- Graziano Riccioni
- Cardiology Unit, San Camillo de Lellis Hospital, Manfredonia, Via Isonzo 71043 Manfredonia (FG), Italy
| | - Valeriana Sblendorio
- Department of General Surgery and Surgical Specialties, University of Modena and Reggio Emilia Medical School, Surgical Clinic, Via Università, 41121 Modena, Italy
| |
Collapse
|
340
|
Chang YL, Pei C, Pei D, Tang SH, Hsu CH, Chen YL, Hsia TL, Lin JD, Wu CZ, Chang JB. Association Between Platelet Count and Components of Metabolic Syndrome in Geriatric Taiwanese Males. INT J GERONTOL 2012. [DOI: 10.1016/j.ijge.2012.01.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
341
|
Hachem A, Yacoub D, Zaid Y, Mourad W, Merhi Y. Involvement of nuclear factor κB in platelet CD40 signaling. Biochem Biophys Res Commun 2012; 425:58-63. [PMID: 22820189 DOI: 10.1016/j.bbrc.2012.07.049] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2012] [Accepted: 07/11/2012] [Indexed: 12/21/2022]
Abstract
CD40 ligand (CD40L) is a thrombo-inflammatory molecule that predicts cardiovascular events. Platelets constitute the major source of soluble CD40L (sCD40L), which has been shown to potentiate platelet activation and aggregation, in a CD40-dependent manner, via p38 mitogen activated protein kinase (MAPK) and Rac1 signaling. In many cells, the CD40L/CD40 dyad also induces activation of nuclear factor kappa B (NF-κB). Given that platelets contain NF-κB, we hypothesized that it may be involved in platelet CD40 signaling and function. In human platelets, sCD40L induces association of CD40 with its adaptor protein the tumor necrosis factor receptor associated factor 2 and triggers phosphorylation of IκBα, which are abolished by CD40L blockade. Inhibition of IκBα phosphorylation reverses sCD40L-induced IκBα phosphorylation without affecting p38 MAPK phosphorylation. On the other hand, inhibition of p38 MAPK phosphorylation has no effect on IκBα phosphorylation, indicating a divergence in the signaling pathway originating from CD40 upon its ligation. In functional studies, inhibition of IκBα phosphorylation reverses sCD40L-induced platelet activation and potentiation of platelet aggregation in response to a sub-threshold concentration of collagen. This study demonstrates that the sCD40L/CD40 axis triggers NF-κB activation in platelets. This signaling pathway plays a critical role in platelet activation and aggregation upon sCD40L stimulation and may represent an important target against thrombo-inflammatory disorders.
Collapse
Affiliation(s)
- Ahmed Hachem
- Laboratory of Thrombosis and Hemostasis, Montreal Heart Institute, 5000 Belanger, Montréal, Québec, Canada H1T 1C8
| | | | | | | | | |
Collapse
|
342
|
Tzakos AG, Kontogianni VG, Tsoumani M, Kyriakou E, Hwa J, Rodrigues FA, Tselepis AD. Exploration of the antiplatelet activity profile of betulinic acid on human platelets. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2012; 60:6977-83. [PMID: 22720759 PMCID: PMC3676635 DOI: 10.1021/jf3006728] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Betulinic acid, a natural pentacyclic triterpene acid, presents a diverse mode of biological actions including antiretroviral, antibacterial, antimalarial, and anti-inflammatory activities. The potency of betulinic acid as an inhibitor of human platelet activation was evaluated, and its antiplatelet profile against in vitro platelet aggregation, induced by several platelet agonists (adenosine diphosphate, thrombin receptor activator peptide-14, and arachidonic acid), was explored. Flow cytometric analysis was performed to examine the effect of betulinic acid on P-selectin membrane expression and PAC-1 binding to activated platelets. Betulinic acid potently inhibits platelet aggregation and also reduced PAC-1 binding and the membrane expression of P-selectin. Principal component analysis was used to screen, on the chemical property space, for potential common pharmacophores of betulinic acid with approved antithrombotic drugs. A common pharmacophore was defined between the NMR-derived structure of betulinic acid and prostacyclin agonists (PGI2), and the importance of its carboxylate group in its antiplatelet activity was determined. The present results indicate that betulinic acid has potential use as an antithrombotic compound and suggest that the mechanism underlying the antiplatelet effects of betulinic acid is similar to that of the PGI2 receptor agonists, a hypothesis that deserves further investigation.
Collapse
Affiliation(s)
- Andreas G. Tzakos
- Section of Organic Chemistry and Biochemistry, Department of Chemistry, University of Ioannina, GR-45110, Ioannina, Greece
| | - Vassiliki G. Kontogianni
- Section of Organic Chemistry and Biochemistry, Department of Chemistry, University of Ioannina, GR-45110, Ioannina, Greece
- Yale School of Medicine, Section of Cardiovascular Medicine, 300 George St, Rm 759 New Haven, CT 06511
| | - Maria Tsoumani
- Section of Organic Chemistry and Biochemistry, Department of Chemistry, University of Ioannina, GR-45110, Ioannina, Greece
- Yale School of Medicine, Section of Cardiovascular Medicine, 300 George St, Rm 759 New Haven, CT 06511
| | - Eleni Kyriakou
- Section of Organic Chemistry and Biochemistry, Department of Chemistry, University of Ioannina, GR-45110, Ioannina, Greece
| | - John Hwa
- Yale School of Medicine, Section of Cardiovascular Medicine, 300 George St, Rm 759 New Haven, CT 06511
| | - Francisco A. Rodrigues
- Departamento de Matemática Aplicada e Estatística, Instituto de Ciências Matemáticas e de Computação, Universidade de São Paulo-Campus de São Carlos, Caixa Postal 668, 13560-970 São Carlos, SP, Brazil
| | - Alexandros D. Tselepis
- Section of Organic Chemistry and Biochemistry, Department of Chemistry, University of Ioannina, GR-45110, Ioannina, Greece
| |
Collapse
|
343
|
Bhatnagar P, Lu X, Evans MK, Laveist TA, Zonderman AB, Carter DL, Arking DE, Fletcher CA. Genetic variants in platelet factor 4 modulate inflammatory and platelet activation biomarkers. ACTA ACUST UNITED AC 2012; 5:412-21. [PMID: 22763266 DOI: 10.1161/circgenetics.111.961813] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
BACKGROUND African Americans suffer from higher prevalence and severity of atherosclerosis compared with whites, highlighting racial and ethnic disparities in cardiovascular disease. Previous studies have pointed to the role of vascular inflammation and platelet activation in the formation of atherosclerotic lesions. METHODS AND RESULTS We explored the role of genetic variation in 4 chemokine/chemokine receptor genes (CX3CR1, CX3CL1, CXCR3, and PF4) on systemic inflammation and platelet activation serum biomarkers (fractalkine, platelet P-selectin, platelet factor 4 [PF4], and tumor necrosis factor-α). In total, 110 single nucleotide polymorphisms were tested among 1042 African Americans and 763 whites. The strongest association with serum PF4 levels was observed for rs168449, which was significant in both racial groups (P value: African Americans=0.0017, whites=0.014, combined=1.2 × 10(-4)), and remained significant after permutation-based multiple corrections (P(c) value: combined=0.0013). After accounting for the effect of rs168449, we identified another significant single nucleotide polymorphism (rs1435520), suggesting a second independent signal regulating serum PF4 levels (conditional P value: African Americans=0.02, whites=0.02). Together, these single nucleotide polymorphisms explained 0.98% and 1.23% of serum PF4 variance in African Americans and whites, respectively. Additionally, in African Americans, we found an additional PF4 variant (rs8180167), uncorrelated with rs168449 and rs1435520, associated with serum tumor necrosis factor-α levels (P=0.008, P(c)=0.048). CONCLUSIONS Our study highlights the importance of PF4 variants in the regulation of platelet activation (PF4) and systemic inflammation (tumor necrosis factor-α) serum biomarkers.
Collapse
Affiliation(s)
- Pallav Bhatnagar
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | | | | | | | | | | | | | | |
Collapse
|
344
|
O'Loughlin A, Lynn DJ, McGee M, Doyle S, McCabe M, Earley B. Transcriptomic analysis of the stress response to weaning at housing in bovine leukocytes using RNA-seq technology. BMC Genomics 2012; 13:250. [PMID: 22708644 PMCID: PMC3583219 DOI: 10.1186/1471-2164-13-250] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2011] [Accepted: 06/18/2012] [Indexed: 01/01/2023] Open
Abstract
Background Weaning of beef calves is a necessary husbandry practice and involves separating the calf from its mother, resulting in numerous stressful events including dietary change, social reorganisation and the cessation of the maternal-offspring bond and is often accompanied by housing. While much recent research has focused on the physiological response of the bovine immune system to stress in recent years, little is known about the molecular mechanisms modulating the immune response. Therefore, the objective of this study was to provide new insights into the molecular mechanisms underlying the physiological response to weaning at housing in beef calves using Illumina RNA-seq. Results The leukocyte transcriptome was significantly altered for at least 7 days following either housing or weaning at housing. Analysis of differentially expressed genes revealed that four main pathways, cytokine signalling, transmembrane transport, haemostasis and G-protein-coupled receptor (GPRC) signalling were differentially regulated between control and weaned calves and underwent significant transcriptomic alterations in response to weaning stress on day 1, 2 and 7. Of particular note, chemokines, cytokines and integrins were consistently found to be up-regulated on each day following weaning. Evidence for alternative splicing of genes was also detected, indicating a number of genes involved in the innate and adaptive immune response may be alternatively transcribed, including those responsible for toll receptor cascades and T cell receptor signalling. Conclusions This study represents the first application of RNA-Seq technology for genomic studies in bovine leukocytes in response to weaning stress. Weaning stress induces the activation of a number of cytokine, chemokine and integrin transcripts and may alter the immune system whereby the ability of a number of cells of the innate and adaptive immune system to locate and destroy pathogens is transcriptionally enhanced. Stress alters the homeostasis of the transcriptomic environment of leukocytes for at least 7 days following weaning, indicating long term effects of stress exposure in the bovine. The identification of gene signature networks that are stress activated provides a mechanistic framework to characterise the multifaceted nature of weaning stress adaptation in beef calves. Thus, capturing subtle transcriptomic changes provides insight into the molecular mechanisms that underlie the physiological response to weaning stress.
Collapse
Affiliation(s)
- Aran O'Loughlin
- Animal and Bioscience Research Department, Animal & Grassland Research and Innovation Centre, Grange, Dunsany, Co. Meath, Ireland
| | | | | | | | | | | |
Collapse
|
345
|
Fuentes Q. E, Fuentes Q. F, Andrés V, Pello OM, de Mora JF, Palomo G. I. Role of platelets as mediators that link inflammation and thrombosis in atherosclerosis. Platelets 2012; 24:255-62. [DOI: 10.3109/09537104.2012.690113] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
346
|
Furuya M, Tanaka R, Miyagi E, Kami D, Nagahama K, Miyagi Y, Nagashima Y, Hirahara F, Inayama Y, Aoki I. Impaired CXCL4 expression in tumor-associated macrophages (TAMs) of ovarian cancers arising in endometriosis. Cancer Biol Ther 2012; 13:671-80. [PMID: 22555803 PMCID: PMC3408972 DOI: 10.4161/cbt.20084] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Inflammatory cells play important roles in progression of solid neoplasms including ovarian cancers. Tumor-associated macrophages (TAMs) contribute to angiogenesis and immune suppression by modulating microenvironment. Ovarian cancer develops occasionally on the bases of endometriosis, a chronic inflammatory disease. We have recently demonstrated differential expressions of CXCR3 variants in endometriosis and ovarian cancers. In this study, we showed impaired CXCL4 expression in TAMs of ovarian cancers arising in endometriosis. The expressions of CXCL4 and its variant CXCL4L1 were investigated among normal ovaries (n = 26), endometriosis (n = 18) and endometriosis-associated ovarian cancers (EAOCs) composed of clear cell (n = 13) and endometrioid (n = 11) types. In addition, four cases of EAOCs that contained both benign and cancer lesions contiguously in single cysts were investigated in the study. Western blot and quantitative RT-PCR analyses revealed significant downregulation of CXCL4 and CXCL4L1 in EAOCs compared with those in endometriosis. In all EAOCs coexisting with endometriosis in the single cyst, the expression levels of CXCL4 and CXCL4L1 were significantly lower in cancer lesions than in corresponding endometriosis. Histopathological study revealed that CXCL4 was strongly expressed in CD68+ infiltrating macrophages of endometriosis. In microscopically transitional zone between endometriosis and EAOC, CD68+ macrophages often demonstrated CXCL4− pattern. The majority of CD68+ TAMs in overt cancer lesions were negative for CXCL4. Collective data indicate that that CXCL4 insufficiency may be involved in specific inflammatory microenvironment of ovarian cancers arising in endometriosis. Suppression of CXCL4 in cancer lesions is likely to be attributable to TAMs in part.
Collapse
Affiliation(s)
- Mitsuko Furuya
- Department of Molecular Pathology, Yokohama City University Graduate School of Medicine, Yokohama, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
347
|
Reny JL, Berdagué P, Poncet A, Barazer I, Nolli S, Fabbro-Peray P, Schved JF, Bounameaux H, Mach F, de Moerloose P, Fontana P. Antiplatelet drug response status does not predict recurrent ischemic events in stable cardiovascular patients: results of the Antiplatelet Drug Resistances and Ischemic Events study. Circulation 2012; 125:3201-10. [PMID: 22615340 DOI: 10.1161/circulationaha.111.085464] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND The biological response to antiplatelet drugs has repeatedly been shown to predict the recurrence of major adverse cardiovascular events (MACEs). However, most studies involved coronary artery disease patients with recent vessel injury shortly after the initiation of antiplatelet therapy. Data on stable cardiovascular patients are scarce, and the added predictive value of specific assays (the vasodilator phosphoprotein assay for the clopidogrel response and serum thromboxane B2 for the aspirin response) and aggregation-based assays relative to common predictors has rarely been addressed. METHODS AND RESULTS Stable cardiovascular outpatients participating in the Antiplatelet Drug Resistances and Ischemic Events (ADRIE) study (n=771) were tested twice, at 2 separate visits, with specific and aggregation-based assays. Follow-up lasted 3 years, and <1% of patients were lost to follow-up. MACEs were adjudicated by an independent committee. Multivariate survival analyses included relevant variables identified in univariate analysis and platelet function test results. The C-index was used to express the prognostic value of various multivariate models. MACEs, the primary end point, occurred in 16% of patients. Hypertension, smoking, older age, and elevated low-density lipoprotein cholesterol were predictive of MACE recurrence, with a C-index of 0.63 (P<0.001). Neither the specific nor the aggregation-based assays added significant predictive value for the primary end point. CONCLUSIONS Biological antiplatelet drug responsiveness, measured with specific or aggregation-based assays, has no incremental predictive value over common cardiovascular risk factors for MACE recurrence in stable cardiovascular outpatients. These results do not support platelet function testing for MACE risk evaluation in stable cardiovascular patients. CLINICAL TRIAL REGISTRATION URL: http://www.clinicaltrials.gov. Unique identifier: NCT00501423.
Collapse
Affiliation(s)
- Jean-Luc Reny
- Division of General Internal Medicine, University Hospitals of Geneva, 4, Rue Gabrielle-Perret-Gentil, CH-1211 Geneva 14, Switzerland
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
348
|
Abstract
The role of platelets in hemostasis and thrombosis is clearly established; however, the mechanisms by which platelets mediate inflammatory and immune pathways are less well understood. Platelets interact and modulate the function of blood and vascular cells by releasing bioactive molecules. Although the platelet is anucleate, it contains transcripts that may mirror disease. Platelet mRNA is only associated with low-level protein translation; however, platelets have a unique membrane structure allowing for the passage of small molecules, leading to the possibility that its cytoplasmic RNA may be passed to nucleated cells. To examine this question, platelet-like particles with labeled RNA were cocultured with vascular cells. Coculture of platelet-like particles with activated THP-1, monocytic, and endothelial cells led to visual and functional RNA transfer. Posttransfer microarray gene expression analysis of THP-1 cells showed an increase in HBG1/HBG2 and HBA1/HBA2 expression that was directly related to the transfer. Infusion of wild-type platelets into a TLR2-deficient mouse model established in vivo confirmation of select platelet RNA transfer to leukocytes. By specifically transferring green fluorescent protein, we also observed external RNA was functional in the recipient cells. The observation that platelets possess the capacity to transfer cytosolic RNA suggests a new function for platelets in the regulation of vascular homeostasis.
Collapse
|
349
|
Gao XM, White DA, Dart AM, Du XJ. Post-infarct cardiac rupture: Recent insights on pathogenesis and therapeutic interventions. Pharmacol Ther 2012; 134:156-79. [DOI: 10.1016/j.pharmthera.2011.12.010] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2011] [Accepted: 12/20/2011] [Indexed: 01/15/2023]
|
350
|
de Mel A, Chaloupka K, Malam Y, Darbyshire A, Cousins B, Seifalian AM. A silver nanocomposite biomaterial for blood-contacting implants. J Biomed Mater Res A 2012; 100:2348-57. [PMID: 22528182 DOI: 10.1002/jbm.a.34177] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2011] [Revised: 01/22/2012] [Accepted: 02/09/2012] [Indexed: 11/12/2022]
Abstract
Cardiovascular implants must resist infection and thrombosis. A nanocomposite polymeric material [polyhedral-oligomeric-silsesquioxane-poly(carbonate-urea)urethane; POSS-PCU] demonstrates ideal properties for cardiovascular applications. Silver nanoparticles or nanosilver (NS) are recognized for efficient antibacterial properties. This study aims to determine the influence of NS integrated POSS-PCU on thrombogenicity. Silver nitrate was reduced with dimethylformamide and stabilized by the inclusion of fumed silica nanoparticles to prevent aggregation of NS and were incorporated into POSS-PCU to form a range of POSS-PCU-NS concentrations (by weight); 0.20% (NS16), 0.40% (NS32), 0.75% (NS64), and 1.50% (NS128). Surface wettability was determined with sessile-drop water contact angles. Platelets were introduced onto test samples and Alamar Blue (AB), mitochondrial-activity assay, quantified the degree of platelet adhesion whilst platelet-factor-4 (PF4) ELISA quantified the degree of platelet activation. Thromboelastography (TEG) determined the profiles of whole blood kinetics while hemolysis assay demonstrated the degree of blood compatibility. Increasing levels of NS induced greater hydrophilicity. A concentration dependant decrease in platelet adhesion and activation was observed with AB and PF4 readings, respectively. TEG demonstrated that the antithrombogenic properties of POSS-PCU were retained with POSS-PCU-NS16, and enhanced with POSS-PCU-NS32, but was reduced with POSS-PCU-NS64 and POSS-PCU-NS128. POSS-PCU-NS64 and POSS-PCU-NS128 demonstrated a hemolytic tendency, but no hemolysis was observed with POSS-PCU-NS16 and POSS-PCU-NS32. Overall, POSS-PCU-NS32 rendered potent antithrombogenic properties.
Collapse
Affiliation(s)
- Achala de Mel
- UCL Centre for Nanotechnology and Regenerative Medicine, Division of Surgery and Interventional Science, University College London, London, United Kingdom
| | | | | | | | | | | |
Collapse
|