1
|
Abstract
Tissue factor (TF), an initiator of extrinsic coagulation pathway, is positively correlated with venous thromboembolism (VTE) of tumor patients. Beyond thrombosis, TF plays a vital role in tumor progression. TF is highly expressed in cancer tissues and circulating tumor cell (CTC), and activates factor VIIa (FVIIa), which increases tumor cells proliferation, angiogenesis, epithelial-mesenchymal transition (EMT) and cancer stem cells(CSCs) activity. Furthermore, TF and TF-positive microvesicles (TF+MVs) activate the coagulation system to promote the clots formation with non-tumor cell components (e.g., platelets, leukocytes, fibrin), which makes tumor cells adhere to clots to form CTC clusters. Then, tumor cells utilize clots to cause its reducing fluid shear stress (FSS), anoikis resistance, immune escape, adhesion, extravasation and colonization. Herein, we review in detail that how TF signaling promotes tumor metastasis, and how TF-targeted therapeutic strategies are being in the preclinical and clinical trials.
Collapse
|
2
|
Yu H, Hua Y, He Y, Wang Y, Hu X, Chen S, Liu J, Yang J, Li H. Sustained Release of MiR-217 Inhibitor by Nanoparticles Facilitates MSC-Mediated Attenuation of Neointimal Hyperplasia After Vascular Injury. Front Cardiovasc Med 2021; 8:739107. [PMID: 34708092 PMCID: PMC8542691 DOI: 10.3389/fcvm.2021.739107] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Accepted: 09/13/2021] [Indexed: 01/05/2023] Open
Abstract
Mesenchymal stem cells (MSCs) have been proven capable of differentiating into endothelial cells (ECs) and increasing vascular density in mouse ischemia models. However, the therapeutic potential of MSCs in neointimal hyperplasia after vascular injury is still not fully understood. In this study, we proposed that sustained release of miR-217 inhibitor encapsulated by nanoparticles in MSCs can enhance the therapeutic effects of MSCs on alleviating neointimal hyperplasia in a standard mouse wire injury model. We intravenously administered MSCs to mice with injured arteries and examined neointimal proliferation, endothelial differentiation and senescence. We demonstrated that MSCs localized to the luminal surface of the injured artery within 24 h after injection and subsequently differentiated into endothelial cells, inhibited neointimal proliferation and migration of vascular smooth muscle cells. Transfection of MSCs with poly lactic-co-glycolic acid nanoparticles (PLGA-NP) encapsulating an miR-217 agomir abolished endothelial differentiation as well as the therapeutic effect of MSCs. On the contrary, silencing of endogenous miR-217 improved the therapeutic efficacy of MSCs. Our study provides a new strategy of augmenting the therapeutic potency of MSCs in treatment of vascular injury.
Collapse
Affiliation(s)
- Hong Yu
- Department of Otorhinolaryngology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Yutao Hua
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Yecheng He
- Department of Clinical Medicine, Suzhou Vocational Health College, Suzhou, China
| | - Yin Wang
- Department of Cardiovascular Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Xingjian Hu
- Department of Cardiovascular Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Si Chen
- Department of Cardiovascular Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Junwei Liu
- Department of Cardiovascular Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Junjie Yang
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Huadong Li
- Department of Cardiovascular Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
3
|
White blood cell count and clinical outcomes after left main coronary artery revascularization: insights from the EXCEL trial. Coron Artery Dis 2021; 31:45-51. [PMID: 34010180 DOI: 10.1097/mca.0000000000001052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Prior studies have reported an association between elevated white blood cell count (WBCc) and worse clinical outcomes after coronary artery bypass grafting (CABG) and percutaneous coronary intervention (PCI). We assessed the prognostic impact of WBCc in patients undergoing revascularization for left main coronary artery disease (LMCAD). METHODS In Evaluation of XIENCE Versus Coronary Artery Bypass Surgery for Effectiveness of Left Main Revascularization (EXCEL), 1905 patients with LMCAD and low or intermediate SYNTAX scores were randomized to PCI with everolimus-eluting stents versus CABG. The 1895 patients with baseline WBCc available were grouped in tertiles of WBCc (mean 5.6 ± 0.8, 7.5 ± 0.5, and 10.1 ± 1.6 × 109/L). RESULTS Five-year rates of the primary endpoint (death, myocardial infarction or stroke) were similar across increasing WBCc tertiles (21.2, 18.9, and 21.6%; P = 0.46). Individual components of the primary endpoint, Bleeding Academic Research Consortium (BARC) 3-5 bleeding, stent thrombosis or graft occlusion and ischemia-driven revascularization were all similar across WBCc tertiles. By multivariable analysis, WBCc as a continuous variable was not an independent predictor of adverse events (hazard radio per 1 × 109/L: 1.02; 95% CI, 0.97-1.08; P = 0.43). Results were consistent in the PCI and CABG arms individually. CONCLUSION There was no association between baseline WBCc and 30-day or 5-year clinical outcomes after PCI or CABG. The absence of a clear incremental increase in events with increasing WBCc in the current analysis indicates that WBCc should not routinely be used as a prognostic marker or to guide revascularization decisions in patients with LMCAD.
Collapse
|
4
|
FAK and Pyk2 activity promote TNF-α and IL-1β-mediated pro-inflammatory gene expression and vascular inflammation. Sci Rep 2019; 9:7617. [PMID: 31110200 PMCID: PMC6527705 DOI: 10.1038/s41598-019-44098-2] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 05/09/2019] [Indexed: 01/07/2023] Open
Abstract
Protein tyrosine kinase (PTK) activity has been implicated in pro-inflammatory gene expression following tumor necrosis factor-α (TNF-α) or interkeukin-1β (IL-1β) stimulation. However, the identity of responsible PTK(s) in cytokine signaling have not been elucidated. To evaluate which PTK is critical to promote the cytokine-induced inflammatory cell adhesion molecule (CAM) expression including VCAM-1, ICAM-1, and E-selectin in human aortic endothelial cells (HAoECs), we have tested pharmacological inhibitors of major PTKs: Src and the focal adhesion kinase (FAK) family kinases - FAK and proline-rich tyrosine kinase (Pyk2). We found that a dual inhibitor of FAK/Pyk2 (PF-271) most effectively reduced all three CAMs upon TNF-α or IL-1β stimulation compared to FAK or Src specific inhibitors (PF-228 or Dasatinib), which inhibited only VCAM-1 expression. In vitro inflammation assays showed PF-271 reduced monocyte attachment and transmigration on HAoECs. Furthermore, FAK/Pyk2 activity was not limited to CAM expression but was also required for expression of various pro-inflammatory molecules including MCP-1 and IP-10. Both TNF-α and IL-1β signaling requires FAK/Pyk2 activity to activate ERK and JNK MAPKs leading to inflammatory gene expression. Knockdown of either FAK or Pyk2 reduced TNF-α-stimulated ERK and JNK activation and CAM expression, suggesting that activation of ERK or JNK is specific through FAK and Pyk2. Finally, FAK/Pyk2 activity is required for VCAM-1 expression and macrophage recruitment to the vessel wall in a carotid ligation model in ApoE-/- mice. Our findings define critical roles of FAK/Pyk2 in mediating inflammatory cytokine signaling and implicate FAK/Pyk2 inhibitors as potential therapeutic agents to treat vascular inflammatory disease such as atherosclerosis.
Collapse
|
5
|
Absence of Nonclassical Monocytes in Hemolytic Patients: Free Hb and NO-Mediated Mechanism. J Immunol Res 2019; 2019:1409383. [PMID: 31032371 PMCID: PMC6458887 DOI: 10.1155/2019/1409383] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 12/20/2018] [Accepted: 01/22/2019] [Indexed: 12/25/2022] Open
Abstract
In a recent work, we have described the kinetics among the monocyte subsets in the peripheral blood of hemolytic patients including paroxysmal nocturnal hemoglobinuria (PNH) and sickle cell disease (SCD). After engulfing Hb-activated platelets, classical monocytes (CD14+CD16-) significantly transformed into highly inflammatory (CD14+CD16hi) subsets in vitro. An estimated 40% of total circulating monocytes in PNH and 70% in SCD patients existed as CD14+CD16hi subsets. In this study, we show that the nonclassical (CD14dimCD16+) monocyte subsets are nearly absent in patients with PNH or SCD, compared to 10-12% cells in healthy individuals. In mechanism, we have described the unique role of both free Hb and nitric oxide (NO) in reducing number of nonclassical subsets more than classical monocytes. After engulfing Hb-activated platelets, the monocytes including nonclassical subsets acquired rapid cell death within 12 h in vitro. Further, the treatment to monocytes either with the secretome of Hb-activated platelets containing NO and free Hb or purified free Hb along with GSNO (a physiological NO donor) enhanced rapid cell death. Besides, our data from both PNH and SCD patients exhibited a direct correlation between intracellular NO and cell death marker 7AAD in monocytes from the peripheral blood. Our data together suggest that due to the immune surveillance nature, the nonclassical or patrolling monocytes are encountered frequently by Hb-activated platelets, free Hb, and NO in the circulation of hemolytic patients and are predisposed to die rapidly.
Collapse
|
6
|
Brown RA, Lip GYH. Monocyte-platelet cross-talk in peripheral artery disease-how much does the pathogenesis of atherosclerosis depend on anatomical location? ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:S19. [PMID: 31032300 DOI: 10.21037/atm.2019.01.47] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Affiliation(s)
- Richard A Brown
- Barts Heart Centre, Barts Health NHS Trust, St Bartholomew's Hospital, London, UK
| | - Gregory Y H Lip
- Liverpool Centre for Cardiovascular Science, University of Liverpool and Liverpool Heart & Chest Hospital, Liverpool, UK.,Aalborg Thrombosis Research Unit, Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| |
Collapse
|
7
|
|
8
|
Aizik G, Grad E, Golomb G. Monocyte-mediated drug delivery systems for the treatment of cardiovascular diseases. Drug Deliv Transl Res 2018; 8:868-882. [PMID: 29058205 DOI: 10.1007/s13346-017-0431-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Major advances have been achieved in understanding the mechanisms and risk factors leading to cardiovascular disorders and consequently developing new therapies. A strong inflammatory response occurs with a substantial recruitment of innate immunity cells in atherosclerosis, myocardial infarction, and restenosis. Monocytes and macrophages are key players in the healing process that ensues following injury. In the inflamed arterial wall, monocytes, and monocyte-derived macrophages have specific functions in the initiation and resolution of inflammation, principally through phagocytosis, and the release of inflammatory cytokines and reactive oxygen species. In this review, we will focus on delivery systems, mainly nanoparticles, for modulating circulating monocytes/monocyte-derived macrophages. We review the different strategies of depletion or modulation of circulating monocytes and monocyte subtypes, using polymeric nanoparticles and liposomes for the therapy of myocardial infarction and restenosis. We will further discuss the strategies of exploiting circulating monocytes for biological targeting of nanocarrier-based drug delivery systems for therapeutic and diagnostic applications.
Collapse
Affiliation(s)
- Gil Aizik
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, 9112001, Jerusalem, Israel
| | - Etty Grad
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, 9112001, Jerusalem, Israel
| | - Gershon Golomb
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, 9112001, Jerusalem, Israel.
| |
Collapse
|
9
|
Brown RA, Lip GYH, Varma C, Shantsila E. Impact of Mon2 monocyte-platelet aggregates on human coronary artery disease. Eur J Clin Invest 2018; 48:e12911. [PMID: 29423944 DOI: 10.1111/eci.12911] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2017] [Accepted: 02/04/2018] [Indexed: 12/28/2022]
Abstract
BACKGROUND Monocyte-platelet aggregates (MPAs) form when Mon1, Mon2 or Mon3 monocyte subsets adhere to platelets. They are pathophysiologically linked to coronary artery disease (CAD). However, their individual roles in the occurrence of diffuse CAD remain unknown. MATERIALS AND METHODS Peripheral blood from 50 patients with diffuse CAD, 40 patients with focal CAD and 50 age-matched patients with normal coronary arteries was analysed by flow cytometry to quantify MPAs associated with individual monocyte subsets. Cutaneous forearm microcirculation was assessed using laser Doppler flowmetry at rest and after iontophoresis of acetylcholine (endothelium-dependent vasodilation) and sodium nitroprusside (endothelium-independent vasodilation) at 100 μA for 60 seconds. Patients with CAD had repeat assessment at 6 and 12 months. RESULTS Baseline counts of MPAs with Mon2 subset (CD14++CD16+CC2+ monocytes) were significantly higher in patients with diffuse CAD compared to focal CAD (P = .001) and patients without CAD (P = .006). On multivariate regression, MPAs with Mon2 independently predicted diffuse CAD (odds ratio 1.10, 95% confidence interval 1.02-1.19, P = .01) and correlated negatively with endothelium-dependent microvascular vasodilation (r = -.37, P = .008), an association which persisted after adjustment for covariates. Longitudinal observation confirmed the persistence of an inverse relationship between MPAs with Mon2 and endothelium-dependent microvascular function. CONCLUSION Monocyte-platelet aggregates with Mon2 are increased in patients with diffuse CAD and therefore could represent an important contributor to accelerated coronary atherosclerotic progression by a mechanism involving microvascular endothelial dysfunction.
Collapse
Affiliation(s)
- Richard A Brown
- Institute of Cardiovascular Sciences, City Hospital, University of Birmingham, Birmingham, UK.,Hull and East Yorkshire Hospitals NHS Trust, Castle Hill Hospital, Cottingham, UK
| | - Gregory Y H Lip
- Institute of Cardiovascular Sciences, City Hospital, University of Birmingham, Birmingham, UK.,Sandwell and West Birmingham Hospitals NHS Trust, City Hospital, Birmingham, UK
| | - Chetan Varma
- Sandwell and West Birmingham Hospitals NHS Trust, City Hospital, Birmingham, UK
| | - Eduard Shantsila
- Institute of Cardiovascular Sciences, City Hospital, University of Birmingham, Birmingham, UK
| |
Collapse
|
10
|
Vanags LZ, Tan JT, Galougahi KK, Schaefer A, Wise SG, Murphy A, Ali ZA, Bursill CA. Apolipoprotein A-I Reduces In-Stent Restenosis and Platelet Activation and Alters Neointimal Cellular Phenotype. JACC Basic Transl Sci 2018; 3:200-209. [PMID: 30062205 PMCID: PMC6060078 DOI: 10.1016/j.jacbts.2017.11.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 10/20/2017] [Accepted: 11/13/2017] [Indexed: 11/04/2022]
Abstract
Even the most advanced drug-eluting stents evoke unresolved issues, including chronic inflammation, late thrombosis, and neoatherosclerosis. This highlights the need for novel strategies that improve stent biocompatibility. Our studies show that apolipoprotein A-I (apoA-I) reduces in-stent restenosis and platelet activation, and enhances endothelialization. These findings have therapeutic implications for improving stent biocompatibility.
Collapse
Key Words
- ABCA1, ATP-binding cassette transporter A1
- CAD, coronary artery disease
- DES, drug-eluting stent(s)
- HDL, high-density lipoprotein
- PBS, phosphate-buffered saline
- PCI, percutaneous coronary intervention
- PPAR, peroxisome proliferator-activated receptor
- SMC, smooth muscle cell
- apoA-I, apolipoprotein A-I
- apoE−/−, apolipoprotein E deficient
- apolipoprotein A-I
- endothelialization
- neointimal hyperplasia
- platelet activation
- rHDL, reconstituted high- density lipoprotein
- stent biocompatibility
Collapse
Affiliation(s)
- Laura Z. Vanags
- Immunobiology Group, The Heart Research Institute, Sydney, Australia
- Sydney Medical School, University of Sydney, Sydney, Australia
| | - Joanne T.M. Tan
- Immunobiology Group, The Heart Research Institute, Sydney, Australia
- Sydney Medical School, University of Sydney, Sydney, Australia
| | - Keyvan K. Galougahi
- Center for Interventional Vascular Therapy, Columbia University, New York, New York
- Cardiovascular Research Foundation, New York, New York
| | - Andreas Schaefer
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Steven G. Wise
- Immunobiology Group, The Heart Research Institute, Sydney, Australia
- Sydney Medical School, University of Sydney, Sydney, Australia
| | - Andrew Murphy
- Haematopoiesis and Leukocyte Biology Group, Baker IDI Heart and Diabetes Institute, Melbourne, Australia
- Department of Immunology, Monash University, Melbourne, Australia
| | - Ziad A. Ali
- Center for Interventional Vascular Therapy, Columbia University, New York, New York
- Cardiovascular Research Foundation, New York, New York
| | - Christina A. Bursill
- Immunobiology Group, The Heart Research Institute, Sydney, Australia
- Sydney Medical School, University of Sydney, Sydney, Australia
| |
Collapse
|
11
|
Extracellular Matrix Metalloproteinase Inducer EMMPRIN (CD147) in Cardiovascular Disease. Int J Mol Sci 2018; 19:ijms19020507. [PMID: 29419744 PMCID: PMC5855729 DOI: 10.3390/ijms19020507] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 01/31/2018] [Accepted: 02/01/2018] [Indexed: 12/22/2022] Open
Abstract
The receptor EMMPRIN is involved in the development and progression of cardiovascular diseases and in the pathogenesis of myocardial infarction. There are several binding partners of EMMPRIN mediating the effects of EMMPRIN in cardiovascular diseases. EMMPRIN interaction with most binding partners leads to disease progression by mediating cytokine or chemokine release, the activation of platelets and monocytes, as well as the formation of monocyte-platelet aggregates (MPAs). EMMPRIN is also involved in atherosclerosis by mediating the infiltration of pro-inflammatory cells. There is also evidence that EMMPRIN controls energy metabolism of cells and that EMMPRIN binding partners modulate intracellular glycosylation and trafficking of EMMPRIN towards the cell membrane. In this review, we systematically discuss these multifaceted roles of EMMPRIN and its interaction partners, such as Cyclophilins, in cardiovascular disease.
Collapse
|
12
|
Shah B, Baber U, Pocock SJ, Krucoff MW, Ariti C, Gibson CM, Steg PG, Weisz G, Witzenbichler B, Henry TD, Kini AS, Stuckey T, Cohen DJ, Iakovou I, Dangas G, Aquino MB, Sartori S, Chieffo A, Moliterno DJ, Colombo A, Mehran R. White Blood Cell Count and Major Adverse Cardiovascular Events After Percutaneous Coronary Intervention in the Contemporary Era. Circ Cardiovasc Interv 2017; 10:CIRCINTERVENTIONS.117.004981. [DOI: 10.1161/circinterventions.117.004981] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 07/10/2017] [Indexed: 12/28/2022]
Abstract
Background—
Elevated white blood cell (WBC) count is associated with increased major adverse cardiovascular events (MACE) in the setting of acute coronary syndrome. The aim of this study was to evaluate whether similar associations persist in an all-comers population of patients undergoing percutaneous coronary intervention in the contemporary era.
Methods and Results—
In the multicenter, prospective, observational PARIS study (Patterns of Non-Adherence to Anti-Platelet Regimens in Stented Patients Registry), 4222 patients who underwent percutaneous coronary intervention in the United States and Europe between July 1, 2009, and December 2, 2010, were evaluated. The associations between baseline WBC and MACE (composite of cardiac death, stent thrombosis, spontaneous myocardial infarction, or target lesion revascularization) at 24-month follow-up were analyzed using multivariable Cox regression. Patients with higher WBC were more often younger, smokers, and with less comorbid risk factors compared with those with lower WBC. After adjustment for baseline and procedural characteristics, WBC remained independently associated with MACE (hazard ratio [HR] per 10
3
cells/μL increase, 1.05 [95% confidence intervals (CI), 1.02–1.09];
P
=0.001), cardiac death (HR, 1.10 [95% CI, 1.05–1.17];
P
<0.001), and clinically indicated target revascularization (HR, 1.04 [95% CI, 1.00–1.09];
P
=0.03) but not stent thrombosis (HR, 1.07 [95% CI, 0.99–1.16];
P
=0.10) or spontaneous myocardial infarction (HR, 1.03 [95% CI, 0.97–1.09];
P
=0.29). The association between WBC and MACE was consistent in acute coronary syndrome and non–acute coronary syndrome presentations (interaction
P
=0.15).
Conclusions—
Increased WBC is an independent predictor of MACE after percutaneous coronary intervention in a contemporary all-comers cohort. Further studies to delineate the underlying pathophysiologic role of elevated WBC across a spectrum of coronary artery disease presentations are warranted.
Clinical Trial Registration—
URL:
http://www.clinicaltrials.gov
. Unique identifier: NCT00998127.
Collapse
Affiliation(s)
- Binita Shah
- From the Department of Medicine (Cardiology), New York Harbor Health Care System, Manhattan VA Hospital (B.S.); Department of Medicine (Cardiology), New York University School of Medicine (B.S.); Department of Medicine (Cardiology), Icahn School of Medicine at Mount Sinai, New York, NY (U.B., A.S.K., G.D., M.B.A., S.S., R.M.); Medical Statistics, London School of Hygiene and Tropical Medicine, United Kingdom (S.J.P., C.A.); Department of Medicine (Cardiology), Duke University School of Medicine,
| | - Usman Baber
- From the Department of Medicine (Cardiology), New York Harbor Health Care System, Manhattan VA Hospital (B.S.); Department of Medicine (Cardiology), New York University School of Medicine (B.S.); Department of Medicine (Cardiology), Icahn School of Medicine at Mount Sinai, New York, NY (U.B., A.S.K., G.D., M.B.A., S.S., R.M.); Medical Statistics, London School of Hygiene and Tropical Medicine, United Kingdom (S.J.P., C.A.); Department of Medicine (Cardiology), Duke University School of Medicine,
| | - Stuart J. Pocock
- From the Department of Medicine (Cardiology), New York Harbor Health Care System, Manhattan VA Hospital (B.S.); Department of Medicine (Cardiology), New York University School of Medicine (B.S.); Department of Medicine (Cardiology), Icahn School of Medicine at Mount Sinai, New York, NY (U.B., A.S.K., G.D., M.B.A., S.S., R.M.); Medical Statistics, London School of Hygiene and Tropical Medicine, United Kingdom (S.J.P., C.A.); Department of Medicine (Cardiology), Duke University School of Medicine,
| | - Mitchell W. Krucoff
- From the Department of Medicine (Cardiology), New York Harbor Health Care System, Manhattan VA Hospital (B.S.); Department of Medicine (Cardiology), New York University School of Medicine (B.S.); Department of Medicine (Cardiology), Icahn School of Medicine at Mount Sinai, New York, NY (U.B., A.S.K., G.D., M.B.A., S.S., R.M.); Medical Statistics, London School of Hygiene and Tropical Medicine, United Kingdom (S.J.P., C.A.); Department of Medicine (Cardiology), Duke University School of Medicine,
| | - Cono Ariti
- From the Department of Medicine (Cardiology), New York Harbor Health Care System, Manhattan VA Hospital (B.S.); Department of Medicine (Cardiology), New York University School of Medicine (B.S.); Department of Medicine (Cardiology), Icahn School of Medicine at Mount Sinai, New York, NY (U.B., A.S.K., G.D., M.B.A., S.S., R.M.); Medical Statistics, London School of Hygiene and Tropical Medicine, United Kingdom (S.J.P., C.A.); Department of Medicine (Cardiology), Duke University School of Medicine,
| | - C. Michael Gibson
- From the Department of Medicine (Cardiology), New York Harbor Health Care System, Manhattan VA Hospital (B.S.); Department of Medicine (Cardiology), New York University School of Medicine (B.S.); Department of Medicine (Cardiology), Icahn School of Medicine at Mount Sinai, New York, NY (U.B., A.S.K., G.D., M.B.A., S.S., R.M.); Medical Statistics, London School of Hygiene and Tropical Medicine, United Kingdom (S.J.P., C.A.); Department of Medicine (Cardiology), Duke University School of Medicine,
| | - Philippe Gabriel Steg
- From the Department of Medicine (Cardiology), New York Harbor Health Care System, Manhattan VA Hospital (B.S.); Department of Medicine (Cardiology), New York University School of Medicine (B.S.); Department of Medicine (Cardiology), Icahn School of Medicine at Mount Sinai, New York, NY (U.B., A.S.K., G.D., M.B.A., S.S., R.M.); Medical Statistics, London School of Hygiene and Tropical Medicine, United Kingdom (S.J.P., C.A.); Department of Medicine (Cardiology), Duke University School of Medicine,
| | - Giora Weisz
- From the Department of Medicine (Cardiology), New York Harbor Health Care System, Manhattan VA Hospital (B.S.); Department of Medicine (Cardiology), New York University School of Medicine (B.S.); Department of Medicine (Cardiology), Icahn School of Medicine at Mount Sinai, New York, NY (U.B., A.S.K., G.D., M.B.A., S.S., R.M.); Medical Statistics, London School of Hygiene and Tropical Medicine, United Kingdom (S.J.P., C.A.); Department of Medicine (Cardiology), Duke University School of Medicine,
| | - Bernhard Witzenbichler
- From the Department of Medicine (Cardiology), New York Harbor Health Care System, Manhattan VA Hospital (B.S.); Department of Medicine (Cardiology), New York University School of Medicine (B.S.); Department of Medicine (Cardiology), Icahn School of Medicine at Mount Sinai, New York, NY (U.B., A.S.K., G.D., M.B.A., S.S., R.M.); Medical Statistics, London School of Hygiene and Tropical Medicine, United Kingdom (S.J.P., C.A.); Department of Medicine (Cardiology), Duke University School of Medicine,
| | - Timothy D. Henry
- From the Department of Medicine (Cardiology), New York Harbor Health Care System, Manhattan VA Hospital (B.S.); Department of Medicine (Cardiology), New York University School of Medicine (B.S.); Department of Medicine (Cardiology), Icahn School of Medicine at Mount Sinai, New York, NY (U.B., A.S.K., G.D., M.B.A., S.S., R.M.); Medical Statistics, London School of Hygiene and Tropical Medicine, United Kingdom (S.J.P., C.A.); Department of Medicine (Cardiology), Duke University School of Medicine,
| | - Annapoorna S. Kini
- From the Department of Medicine (Cardiology), New York Harbor Health Care System, Manhattan VA Hospital (B.S.); Department of Medicine (Cardiology), New York University School of Medicine (B.S.); Department of Medicine (Cardiology), Icahn School of Medicine at Mount Sinai, New York, NY (U.B., A.S.K., G.D., M.B.A., S.S., R.M.); Medical Statistics, London School of Hygiene and Tropical Medicine, United Kingdom (S.J.P., C.A.); Department of Medicine (Cardiology), Duke University School of Medicine,
| | - Thomas Stuckey
- From the Department of Medicine (Cardiology), New York Harbor Health Care System, Manhattan VA Hospital (B.S.); Department of Medicine (Cardiology), New York University School of Medicine (B.S.); Department of Medicine (Cardiology), Icahn School of Medicine at Mount Sinai, New York, NY (U.B., A.S.K., G.D., M.B.A., S.S., R.M.); Medical Statistics, London School of Hygiene and Tropical Medicine, United Kingdom (S.J.P., C.A.); Department of Medicine (Cardiology), Duke University School of Medicine,
| | - David J. Cohen
- From the Department of Medicine (Cardiology), New York Harbor Health Care System, Manhattan VA Hospital (B.S.); Department of Medicine (Cardiology), New York University School of Medicine (B.S.); Department of Medicine (Cardiology), Icahn School of Medicine at Mount Sinai, New York, NY (U.B., A.S.K., G.D., M.B.A., S.S., R.M.); Medical Statistics, London School of Hygiene and Tropical Medicine, United Kingdom (S.J.P., C.A.); Department of Medicine (Cardiology), Duke University School of Medicine,
| | - Ioannis Iakovou
- From the Department of Medicine (Cardiology), New York Harbor Health Care System, Manhattan VA Hospital (B.S.); Department of Medicine (Cardiology), New York University School of Medicine (B.S.); Department of Medicine (Cardiology), Icahn School of Medicine at Mount Sinai, New York, NY (U.B., A.S.K., G.D., M.B.A., S.S., R.M.); Medical Statistics, London School of Hygiene and Tropical Medicine, United Kingdom (S.J.P., C.A.); Department of Medicine (Cardiology), Duke University School of Medicine,
| | - George Dangas
- From the Department of Medicine (Cardiology), New York Harbor Health Care System, Manhattan VA Hospital (B.S.); Department of Medicine (Cardiology), New York University School of Medicine (B.S.); Department of Medicine (Cardiology), Icahn School of Medicine at Mount Sinai, New York, NY (U.B., A.S.K., G.D., M.B.A., S.S., R.M.); Medical Statistics, London School of Hygiene and Tropical Medicine, United Kingdom (S.J.P., C.A.); Department of Medicine (Cardiology), Duke University School of Medicine,
| | - Melissa B. Aquino
- From the Department of Medicine (Cardiology), New York Harbor Health Care System, Manhattan VA Hospital (B.S.); Department of Medicine (Cardiology), New York University School of Medicine (B.S.); Department of Medicine (Cardiology), Icahn School of Medicine at Mount Sinai, New York, NY (U.B., A.S.K., G.D., M.B.A., S.S., R.M.); Medical Statistics, London School of Hygiene and Tropical Medicine, United Kingdom (S.J.P., C.A.); Department of Medicine (Cardiology), Duke University School of Medicine,
| | - Samantha Sartori
- From the Department of Medicine (Cardiology), New York Harbor Health Care System, Manhattan VA Hospital (B.S.); Department of Medicine (Cardiology), New York University School of Medicine (B.S.); Department of Medicine (Cardiology), Icahn School of Medicine at Mount Sinai, New York, NY (U.B., A.S.K., G.D., M.B.A., S.S., R.M.); Medical Statistics, London School of Hygiene and Tropical Medicine, United Kingdom (S.J.P., C.A.); Department of Medicine (Cardiology), Duke University School of Medicine,
| | - Alaide Chieffo
- From the Department of Medicine (Cardiology), New York Harbor Health Care System, Manhattan VA Hospital (B.S.); Department of Medicine (Cardiology), New York University School of Medicine (B.S.); Department of Medicine (Cardiology), Icahn School of Medicine at Mount Sinai, New York, NY (U.B., A.S.K., G.D., M.B.A., S.S., R.M.); Medical Statistics, London School of Hygiene and Tropical Medicine, United Kingdom (S.J.P., C.A.); Department of Medicine (Cardiology), Duke University School of Medicine,
| | - David J. Moliterno
- From the Department of Medicine (Cardiology), New York Harbor Health Care System, Manhattan VA Hospital (B.S.); Department of Medicine (Cardiology), New York University School of Medicine (B.S.); Department of Medicine (Cardiology), Icahn School of Medicine at Mount Sinai, New York, NY (U.B., A.S.K., G.D., M.B.A., S.S., R.M.); Medical Statistics, London School of Hygiene and Tropical Medicine, United Kingdom (S.J.P., C.A.); Department of Medicine (Cardiology), Duke University School of Medicine,
| | - Antonio Colombo
- From the Department of Medicine (Cardiology), New York Harbor Health Care System, Manhattan VA Hospital (B.S.); Department of Medicine (Cardiology), New York University School of Medicine (B.S.); Department of Medicine (Cardiology), Icahn School of Medicine at Mount Sinai, New York, NY (U.B., A.S.K., G.D., M.B.A., S.S., R.M.); Medical Statistics, London School of Hygiene and Tropical Medicine, United Kingdom (S.J.P., C.A.); Department of Medicine (Cardiology), Duke University School of Medicine,
| | - Roxana Mehran
- From the Department of Medicine (Cardiology), New York Harbor Health Care System, Manhattan VA Hospital (B.S.); Department of Medicine (Cardiology), New York University School of Medicine (B.S.); Department of Medicine (Cardiology), Icahn School of Medicine at Mount Sinai, New York, NY (U.B., A.S.K., G.D., M.B.A., S.S., R.M.); Medical Statistics, London School of Hygiene and Tropical Medicine, United Kingdom (S.J.P., C.A.); Department of Medicine (Cardiology), Duke University School of Medicine,
| |
Collapse
|
13
|
Cocco G, Jerie P, Amiet P, Pandolfi S. Inflammation in Heart Failure: known knowns and unknown unknowns. Expert Opin Pharmacother 2017; 18:1225-1233. [DOI: 10.1080/14656566.2017.1351948] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
| | - Paul Jerie
- Cardiology Office, Rheinfelden, Switzerland
| | | | | |
Collapse
|
14
|
Brockman KS, Lai BFL, Kizhakkedathu JN, Santerre JP. Hemocompatibility of Degrading Polymeric Biomaterials: Degradable Polar Hydrophobic Ionic Polyurethane versus Poly(lactic-co-glycolic) Acid. Biomacromolecules 2017. [PMID: 28621927 DOI: 10.1021/acs.biomac.7b00456] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The use of degradable polymers in vascular tissue regeneration has sparked the need to characterize polymer biocompatibility during degradation. While tissue compatibility has been frequently addressed, studies on polymer hemocompatibility during degradation are limited. The current study evaluated the differences in hemocompatibility (platelet response, complement activation, and coagulation cascade initiation) between as-made and hydrolyzed poly(lactic-co-glycolic) acid (PLGA) and degradable polar hydrophobic ionic polyurethane (D-PHI). Platelet activation decreased (in whole blood) and platelet adhesion decreased (in blood without leukocytes) for degraded versus as-made PLGA. D-PHI showed minimal hemocompatibility changes over degradation. Leukocytes played a major role in mediating platelet activation for samples and controls, as well as influencing platelet-polymer adhesion on the degraded materials. This study demonstrates the importance of assessing the blood compatibility of biomaterials over the course of degradation since the associated changes in surface chemistry and physical state could significantly change biomaterial hemocompatibility.
Collapse
Affiliation(s)
- Kathryne S Brockman
- Department of Chemical Engineering and Applied Chemistry, University of Toronto , Toronto, Ontario M5S 3R5, Canada.,Institute of Biomaterials and Biomedical Engineering, Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, University of Toronto , Toronto, Ontario M5G 1M1, Canada
| | - Benjamin F L Lai
- Department of Pathology and Laboratory Medicine and Centre for Blood Research, University of British Columbia , Vancouver, British Columbia V6T 1Z3, Canada
| | - Jayachandran N Kizhakkedathu
- Department of Pathology and Laboratory Medicine and Centre for Blood Research, University of British Columbia , Vancouver, British Columbia V6T 1Z3, Canada.,Department of Chemistry, University of British Columbia , Vancouver, British Columbia V6T 1Z1, Canada
| | - J Paul Santerre
- Department of Chemical Engineering and Applied Chemistry, University of Toronto , Toronto, Ontario M5S 3R5, Canada.,Institute of Biomaterials and Biomedical Engineering, Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, University of Toronto , Toronto, Ontario M5G 1M1, Canada.,Faculty of Dentistry, University of Toronto , Toronto, Ontario M5G 1G6, Canada
| |
Collapse
|
15
|
Singhal R, Chawla S, Rathore DK, Bhasym A, Annarapu GK, Sharma V, Seth T, Guchhait P. Development of pro-inflammatory phenotype in monocytes after engulfing Hb-activated platelets in hemolytic disorders. Clin Immunol 2016; 175:133-142. [PMID: 28039017 DOI: 10.1016/j.clim.2016.12.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 10/31/2016] [Accepted: 12/03/2016] [Indexed: 01/31/2023]
Abstract
Monocytes and macrophage combat infections and maintain homeostatic balance by engulfing microbes and apoptotic cells, and releasing inflammatory cytokines. Studies have described that these cells develop anti-inflammatory properties upon recycling the free-hemoglobin (Hb) in hemolytic conditions. While investigating the phenotype of monocytes in two hemolytic disorders-paroxysmal nocturnal hemoglobinuria (PNH) and sickle cell disease (SCD), we observed a high number of pro-inflammatory (CD14+CD16hi) monocytes in these patients. We further investigated in vitro the phenotype of these monocytes and found an estimated 55% of CD14+ cells were transformed into the CD14+CD16hi subset after engulfing Hb-activated platelets. The CD14+CD16hi monocytes, which were positive for both intracellular Hb and CD42b (platelet marker), secreted significant amounts of TNF-α and IL-1β, unlike monocytes treated with only free Hb, which secreted more IL-10. We have shown recently the presence of a high number of Hb-bound hyperactive platelets in patients with both diseases, and further investigated if the monocytes engulfed these activated platelets in vivo. As expected, we found 95% of CD14+CD16hi monocytes with both intracellular Hb and CD42b in both diseases, and they expressed high TNF-α. Furthermore our data showed that these monocytes whether from patients or developed in vitro after treatment with Hb-activated platelets, secreted significant amounts of tissue factor. Besides, these CD14+CD16hi monocytes displayed significantly decreased phagocytosis of E. coli. Our study therefore suggests that this alteration of monocyte phenotype may play a role in the increased propensity to pro-inflammatory/coagulant complications observed in these hemolytic disorders-PNH and SCD.
Collapse
Affiliation(s)
- Rashi Singhal
- Regional Centre for Biotechnology, National Capital Region Biotech Science Cluster, Faridabad, India; Department of Biotechnology, Manipal University, Karnataka, India
| | - Sheetal Chawla
- Regional Centre for Biotechnology, National Capital Region Biotech Science Cluster, Faridabad, India
| | - Deepak K Rathore
- Translational Health Science & Technology Institute, National Capital Region Biotech Science Cluster, Faridabad, India
| | - Angika Bhasym
- Regional Centre for Biotechnology, National Capital Region Biotech Science Cluster, Faridabad, India; Department of Biotechnology, Manipal University, Karnataka, India
| | - Gowtham K Annarapu
- Regional Centre for Biotechnology, National Capital Region Biotech Science Cluster, Faridabad, India; Department of Biotechnology, Manipal University, Karnataka, India
| | - Vandana Sharma
- Department of Hematology, All India Institute of Medical Sciences, New Delhi, India
| | - Tulika Seth
- Department of Hematology, All India Institute of Medical Sciences, New Delhi, India
| | - Prasenjit Guchhait
- Regional Centre for Biotechnology, National Capital Region Biotech Science Cluster, Faridabad, India.
| |
Collapse
|
16
|
Stähli BE, Gebhard C, Duchatelle V, Cournoyer D, Petroni T, Tanguay JF, Robb S, Mann J, Guertin MC, Wright RS, L L'Allier P, Tardif JC. Effects of the P-Selectin Antagonist Inclacumab on Myocardial Damage After Percutaneous Coronary Intervention According to Timing of Infusion: Insights From the SELECT-ACS Trial. J Am Heart Assoc 2016; 5:e004255. [PMID: 27852589 PMCID: PMC5210344 DOI: 10.1161/jaha.116.004255] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 10/07/2016] [Indexed: 11/16/2022]
Abstract
BACKGROUND The Effects of the P-Selectin Antagonist Inclacumab on Myocardial Damage After Percutaneous Coronary Intervention for Non-ST-Segment Elevation Myocardial Infarction (SELECT-ACS) trial suggested beneficial effects of inclacumab, a monoclonal antibody directed against P-selectin, on periprocedural myocardial damage. This study evaluated the effect of inclacumab on myocardial damage according to varying time intervals between study drug infusion and percutaneous coronary intervention (PCI). METHODS AND RESULTS Patients (n=544) enrolled in the SELECT-ACS trial and randomized to receive 1 infusion of placebo or inclacumab (5 or 20 mg/kg, administered between 1 and 24 hours before PCI) were divided according to the time interval between study drug infusion and PCI. The primary end point was the change in troponin I from baseline at 16 and 24 hours after PCI. In patients receiving inclacumab 20 mg/kg with a short (less than median) time interval between infusion and PCI, placebo-adjusted geometric mean percent changes in troponin I, creatine kinase-myocardial band, and peak troponin I at 24 hours were -45.6% (P=0.005), -30.7% (P=0.01), and -37.3% (P=0.02), respectively. No significant changes were observed in patients with a long (greater than median) time interval between infusion and PCI. Placebo-adjusted geometric mean percent changes in troponin I and creatine kinase-myocardial band were -43.5% (P=0.02) and -26.0% (P=0.07), respectively, when inclacumab 20 mg/kg was administered between 1 and 3 hours before PCI, whereas the drug had no effect with longer intervals. CONCLUSIONS Inclacumab 20 mg/kg significantly reduces myocardial damage after PCI in patients with non-ST-segment elevation myocardial infarction, and benefits are larger when the infusion is administered <3 hours before PCI. CLINICAL TRIAL REGISTRATION URL: http://www.clinicaltrials.gov. Unique identifier: NCT01327183.
Collapse
Affiliation(s)
- Barbara E Stähli
- Montreal Heart Institute, Université de Montréal, Montreal, Canada
| | | | | | - Daniel Cournoyer
- Montreal Health Innovations Coordinating Center (MHICC), Montreal, Canada
| | - Thibaut Petroni
- Montreal Heart Institute, Université de Montréal, Montreal, Canada
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Percutaneous Transluminal Angioplasty in Patients with Peripheral Arterial Disease Does Not Affect Circulating Monocyte Subpopulations. BIOMED RESEARCH INTERNATIONAL 2016; 2016:2708957. [PMID: 27818999 PMCID: PMC5081453 DOI: 10.1155/2016/2708957] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 07/30/2016] [Accepted: 09/20/2016] [Indexed: 11/29/2022]
Abstract
Monocytes are mononuclear cells characterized by distinct morphology and expression of CD14 and CD16 surface receptors. Classical, quiescent monocytes are positive for CD14 (lipopolysaccharide receptor) but do not express Fc gamma receptor III (CD16). Intermediate monocytes coexpress CD16 and CD14. Nonclassical monocytes with low expression of CD14 represent mature macrophage-like monocytes. Monocyte behavior in peripheral arterial disease (PAD) and during vessel wall directed treatment is not well defined. This observation study aimed at monitoring of acute changes in monocyte subpopulations during percutaneous transluminal angioplasty (PTA) in PAD patients. Patients with Rutherford 3 and 4 PAD with no signs of inflammatory process underwent PTA of iliac, femoral, or popliteal segments. Flow cytometry for CD14, CD16, HLA-DR, CD11b, CD11c, and CD45RA antigens allowed characterization of monocyte subpopulations in blood sampled before and after PTA (direct angioplasty catheter sampling). Patients were clinically followed up for 12 months. All 61 enrolled patients completed 12-month follow-up. Target vessel failure occurred in 12 patients. While absolute counts of monocyte were significantly lower after PTA, only subtle monocyte activation after PTA (CD45RA and β-integrins) occurred. None of the monocyte parameters correlated with long-term adverse clinical outcome. Changes in absolute monocyte counts and subtle changes towards an activation phenotype after PTA may reflect local cell adhesion phenomenon in patients with Rutherford 3 or 4 peripheral arterial disease.
Collapse
|
18
|
Gerrits AJ, Frelinger AL, Michelson AD. Whole Blood Analysis of Leukocyte-Platelet Aggregates. CURRENT PROTOCOLS IN CYTOMETRY 2016; 78:6.15.1-6.15.10. [PMID: 27723089 DOI: 10.1002/cpcy.8] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
In inflammatory and thrombotic syndromes, platelets aggregate with circulating leukocytes, especially monocytes and neutrophils. This leukocyte-platelet aggregate formation is initiated primarily through platelet surface expression of P-selectin (CD62P), following activation-dependent degranulation of α-granules, binding to its constitutively expressed counter-receptor, P-selectin glycoprotein ligand 1 (PSGL-1), on leukocytes. Monocyte-platelet aggregates are a more sensitive marker of platelet activation than platelet surface P-selectin. Detection of leukocyte-platelet aggregates is relatively simple by whole-blood flow cytometry. Light scatter and at least one leukocyte-specific antibody are used to gate the desired population, and the presence of associated platelets is detected by immunostaining for abundant platelet-specific markers. © 2016 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Anja J Gerrits
- Center for Platelet Research Studies, Division of Hematology/Oncology, Boston Children's Hospital, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Andrew L Frelinger
- Center for Platelet Research Studies, Division of Hematology/Oncology, Boston Children's Hospital, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Alan D Michelson
- Center for Platelet Research Studies, Division of Hematology/Oncology, Boston Children's Hospital, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
19
|
Żmigrodzka M, Guzera M, Miśkiewicz A, Jagielski D, Winnicka A. The biology of extracellular vesicles with focus on platelet microparticles and their role in cancer development and progression. Tumour Biol 2016; 37:14391-14401. [PMID: 27629289 PMCID: PMC5126185 DOI: 10.1007/s13277-016-5358-6] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2016] [Accepted: 09/07/2016] [Indexed: 12/21/2022] Open
Abstract
Extracellular vesicles (EVs) are a heterogeneous group of structures which can be classified into smaller in size and relatively homogenous exosomes (EXSMs)—spherical fragments of lipid bilayers from inner cell compartments—and bigger in size ectosomes (ECSMs)—a direct consequence of cell-membrane blebbing. EVs can be found in body fluids of healthy individuals. Their number increases in cancer and other pathological conditions. EVs can originate from various cell types, including leukocytes, erythrocytes, thrombocytes, and neoplastic cells. Platelet microparticles (PMPs) are the most abundant population of EVs in blood. It is well documented that PMPs, being a crucial element of EVs signaling, are involved in tumor growth, metastasis, and angiogenesis and may participate in the development of multidrug resistance by tumor cells. The aim of this review is to present the role of PMPs in carcinogenesis. The biology and functions of PMPs with a particular emphasis on the most recent scientific reports on EV properties are also characterized.
Collapse
Affiliation(s)
- M Żmigrodzka
- Department of Pathology and Veterinary Diagnostics, Faculty of Veterinary Medicine, Warsaw University of Life Sciences (WULS-SGGW), Nowoursynowska 159c, Warsaw, Poland
| | - M Guzera
- Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge, UK
| | - A Miśkiewicz
- Department of Pathology and Veterinary Diagnostics, Faculty of Veterinary Medicine, Warsaw University of Life Sciences (WULS-SGGW), Nowoursynowska 159c, Warsaw, Poland.
| | - D Jagielski
- Veterinary Clinic BIALOBRZESKA, Częstochowska 20, Warsaw, Poland
| | - A Winnicka
- Department of Pathology and Veterinary Diagnostics, Faculty of Veterinary Medicine, Warsaw University of Life Sciences (WULS-SGGW), Nowoursynowska 159c, Warsaw, Poland
| |
Collapse
|
20
|
Brown RA, Shantsila E, Varma C, Lip GYH. Epidemiology and pathogenesis of diffuse obstructive coronary artery disease: the role of arterial stiffness, shear stress, monocyte subsets and circulating microparticles. Ann Med 2016; 48:444-455. [PMID: 27282244 DOI: 10.1080/07853890.2016.1190861] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Despite falling age-adjusted mortality rates coronary artery disease (CAD) remains the leading cause of death worldwide. Advanced diffuse CAD is becoming an important entity of modern cardiology as more patients with historical revascularisation no longer have suitable anatomy for additional procedures. Advances in the treatment of diffuse obstructive CAD are hampered by a poor understanding of its development. Although the likelihood of developing clinically significant (obstructive) CAD is linked to traditional risk factors, the morphology of obstructive CAD among individuals is highly variable - some patients have diffuse stenotic disease, while others have a focal stenosis. This is challenging to explain in mechanistic terms as vascular endothelium is equally exposed to injury stimulants. Patients with diffuse disease are at high risk of adverse outcomes, particularly if unsuitable for revascularisation. We searched multiple electronic databases (MEDLINE, EMBASE and the Cochrane Database) and reviewed the epidemiology, pathogenesis and prognosis relating to advanced diffuse CAD with particular focus on the role of endothelial shear stress, large artery stiffness, monocyte subsets and circulating microparticles. Key messages Although traditional CAD risk factors correlate strongly with disease severity, significant individual variation in disease morphology exists. Advanced, diffuse CAD is difficult to treat effectively and can significantly impair quality of life and increases mortality. The pathophysiology associated with the progression of CAD is the result of complex maladaptive interaction between the endothelium, cells of the immune system and patterns of blood flow.
Collapse
Affiliation(s)
- Richard A Brown
- a University of Birmingham Institute of Cardiovascular Sciences, City Hospital , Birmingham , UK
| | - Eduard Shantsila
- a University of Birmingham Institute of Cardiovascular Sciences, City Hospital , Birmingham , UK.,b Cardiology Department, City Hospital , Birmingham , UK
| | - Chetan Varma
- b Cardiology Department, City Hospital , Birmingham , UK
| | - Gregory Y H Lip
- a University of Birmingham Institute of Cardiovascular Sciences, City Hospital , Birmingham , UK.,b Cardiology Department, City Hospital , Birmingham , UK
| |
Collapse
|
21
|
Wun T. The Role of Inflammation and Leukocytes in the Pathogenesis of Sickle Cell Disease. Hematology 2016; 5:403-12. [DOI: 10.1080/10245332.2000.11746536] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Affiliation(s)
- Ted Wun
- Division of Hematology and Oncology, Department of Internal Medicine, UC Davis School of Medicine and the VA Northern California Health Care System, Sacramento, CA, USA
| |
Collapse
|
22
|
Fibronectin adsorption on surface-modified polyetherurethanes and their differentiated effect on specific blood elements related to inflammatory and clotting processes. Biointerphases 2016; 11:029809. [PMID: 27246517 DOI: 10.1116/1.4950887] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
After the introduction of a medical device into the body, adhesive proteins such as fibronectin (Fn) will adsorb to the surface of the biomaterial. Monocytes (MCs) will interact with these adsorbed proteins, and adopt either a proinflammatory and/or prowound healing phenotype, thereby influencing many blood interaction events including thrombogenesis. In this work, Fn adsorption as well as subsequent MC response and thrombus formation were investigated on two surfaces-modified polyetherurethanes (PEUs) using different surface modifiers: an anionic/dihydroxyl oligomeric (ADO) additive, known to enable cell adhesion, and a fluorinated polypropylene oxide oligomer (PPO), known to reduce platelet adhesion. Results indicated that at 24 h of MC culture, PEU-ADO and PEU-PPO promoted an anti-inflammatory character relative to the base PEU. Longer clotting times, based on a free hemoglobin assay, were also found on the two surface-modified PEUs relative to the native one, suggesting their potential for the reduction of thrombus formation. In presence of a Fn monolayer, the surface-modified PEUs conserved a lower thrombogenic character than the base PEU, and was however significantly decreased when compared to prior protein adsorption. Furthermore, Fn coatings increased the MC production levels of tumor necrosis factor-α and interleukin-10 at 24 h, while not affecting the anti-inflammatory effect of the modifications relative to the base PEU. This finding was most prominent on PEU-PPO, suggesting that the interaction of the adsorbed Fn with blood cells was different for the two additives. Hence, the results highlighted differentiating effects of Fn adsorption on specific blood activating processes related to inflammatory and thrombotic responses.
Collapse
|
23
|
Shah B, Allen N, Harchandani B, Pillinger M, Katz S, Sedlis SP, Echagarruga C, Samuels SK, Morina P, Singh P, Karotkin L, Berger JS. Effect of Colchicine on Platelet-Platelet and Platelet-Leukocyte Interactions: a Pilot Study in Healthy Subjects. Inflammation 2016; 39:182-189. [PMID: 26318864 PMCID: PMC4753094 DOI: 10.1007/s10753-015-0237-7] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The cardioprotective mechanisms of colchicine in patients with stable ischemic heart disease remain uncertain. We tested varying concentrations of colchicine on platelet activity in vitro and a clinically relevant 1.8-mg oral loading dose administered over 1 h in 10 healthy subjects. Data are shown as median [interquartile range]. Colchicine addition in vitro decreased light transmission platelet aggregation only at supratherapeutic concentrations but decreased monocyte- (MPA) and neutrophil-platelet aggregation (NPA) at therapeutic concentrations. Administration of 1.8 mg colchicine to healthy subjects had no significant effect on light transmission platelet aggregation but decreased the extent of MPA (28 % [22-57] to 22 % [19-31], p = 0.05) and NPA (19 % [16-59] to 15 % [11-30], p = 0.01), platelet surface expression of PAC-1 (370 mean fluorescence intensity (MFI) [328-555] to 333 MFI [232-407], p = 0.02) and P-selectin (351 MFI [269-492] to 279 [226-364], p = 0.03), and platelet adhesion to collagen (10.2 % [2.5-32.6] to 2.0 % [0.2-9.5], p = 0.09) 2 h post-administration. Thus, in clinically relevant concentrations, colchicine decreases expression of surface markers of platelet activity and inhibits leukocyte-platelet aggregation but does not inhibit homotypic platelet aggregation.
Collapse
Affiliation(s)
- Binita Shah
- Department of Medicine, Division of Cardiology, New York University School of Medicine, 227 E 30th Street, Office 835, New York, NY, 10016, USA.
- Department of Medicine, Section of Cardiology, Veterans Affairs New York Harbor Health Care System, New York, NY, USA.
| | - Nicole Allen
- Department of Medicine, Division of Cardiology, New York University School of Medicine, 227 E 30th Street, Office 835, New York, NY, 10016, USA
| | - Bhisham Harchandani
- Department of Medicine, Division of Cardiology, New York University School of Medicine, 227 E 30th Street, Office 835, New York, NY, 10016, USA
| | - Michael Pillinger
- Department of Medicine, Division of Rheumatology, New York University School of Medicine, New York, NY, USA
| | - Stuart Katz
- Department of Medicine, Division of Cardiology, New York University School of Medicine, 227 E 30th Street, Office 835, New York, NY, 10016, USA
| | - Steven P Sedlis
- Department of Medicine, Division of Cardiology, New York University School of Medicine, 227 E 30th Street, Office 835, New York, NY, 10016, USA
- Department of Medicine, Section of Cardiology, Veterans Affairs New York Harbor Health Care System, New York, NY, USA
| | - Christina Echagarruga
- Department of Medicine, Division of Cardiology, New York University School of Medicine, 227 E 30th Street, Office 835, New York, NY, 10016, USA
| | | | - Pajazit Morina
- Department of Medicine, Division of Cardiology, New York University School of Medicine, 227 E 30th Street, Office 835, New York, NY, 10016, USA
| | - Prabhjot Singh
- Department of Medicine, Division of Cardiology, New York University School of Medicine, 227 E 30th Street, Office 835, New York, NY, 10016, USA
| | - Liza Karotkin
- Department of Medicine, Division of Cardiology, New York University School of Medicine, 227 E 30th Street, Office 835, New York, NY, 10016, USA
| | - Jeffrey S Berger
- Department of Medicine, Divisions of Cardiology and Hematology, New York University School of Medicine, New York, NY, USA
| |
Collapse
|
24
|
Lanza GA, Stazi A, Villano A, Torrini F, Milo M, Laurito M, Flego D, Aurigemma C, Liuzzo G, Crea F. Effect of Remote Ischemic Preconditioning on Platelet Activation Induced by Coronary Procedures. Am J Cardiol 2016; 117:359-65. [PMID: 26739396 DOI: 10.1016/j.amjcard.2015.10.056] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2015] [Revised: 10/30/2015] [Accepted: 10/30/2015] [Indexed: 10/22/2022]
Abstract
In this study, we aim to assess whether remote ischemic preconditioning (RIPC) reduces platelet activation during coronary angiography (CA) and/or percutaneous coronary interventions. We studied 30 patients who underwent CA because of a suspect of stable angina. Patients were randomized to RIPC (3 short episodes of forearm ischemia) or sham RIPC (controls) before the procedure. Blood samples were collected at baseline, at the end of the procedure, and 24 hours later. Monocyte-platelet aggregate (MPA) formation and platelet CD41 in the MPA gate and CD41 and CD62 expression in the platelet gate were assessed by flow cytometry, in the absence and in the presence of adenosine diphosphate (ADP) stimulation. A significant increase in platelet activation occurred during the invasive procedure in controls, which persisted at 24 hours. However, compared with controls, RIPC group showed no or a lower increase in platelet variables, including MPA formation (p <0.0001) and CD41 (p = 0.002) in the MPA gate and CD41 (p <0.0001) and CD62 (p = 0.002) in the platelet gate. ADP increased platelet activation at baseline, but did not further increase platelet reactivity during the invasive procedure in either groups. Percutaneous coronary interventions, performed in 10 patients (6 in the RIPC group and 4 in controls), did not have any further significant effect on platelet activation and reactivity compared with CA alone. In conclusion, RIPC reduces platelet activation occurring during CA. In contrast, no effects were observed on platelet response to ADP stimulation, probably related to the administration of an ADP antagonist in all patients.
Collapse
|
25
|
Totani L, Amore C, Di Santo A, Dell'Elba G, Piccoli A, Martelli N, Tenor H, Beume R, Evangelista V. Roflumilast inhibits leukocyte-platelet interactions and prevents the prothrombotic functions of polymorphonuclear leukocytes and monocytes. J Thromb Haemost 2016; 14:191-204. [PMID: 26484898 DOI: 10.1111/jth.13173] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Accepted: 10/04/2015] [Indexed: 12/12/2022]
Abstract
UNLABELLED ESSENTIALS: Thrombosis is a major comorbidity in patients with chronic obstructive pulmonary disease (COPD). Roflumilast is a selective phosphodiesterase type-4 (PDE4) inhibitor approved for treatment of severe COPD. PDE4 blockade by roflumilast inhibits prothrombotic functions of neutrophils and monocytes. PDE4 inhibitors may reduce thrombotic risk in COPD as well as in other vascular diseases. BACKGROUND Roflumilast, an oral selective phosphodiesterase type 4 inhibitor, is approved for the treatment of severe chronic obstructive pulmonary disease (COPD). A recent meta-analysis of trials on COPD revealed that treatment with roflumilast was associated with a significant reduction in the rate of major cardiovascular events. The mechanisms of this effect remain unknown. OBJECTIVES We tested the hypothesis that roflumilast N-oxide (RNO), the active metabolite of roflumilast, curbs the molecular mechanisms required for leukocyte-platelet (PLT) interactions and prevents the prothrombotic functions of polymorphonuclear leukocytes (PMNs) and monocytes (MNs). METHODS Using well-characterized in vitro models, we analysed the effects of RNO on: (i) PMN adhesiveness; (ii) the release of neutrophil extracellular traps (NETs); and (iii) tissue factor expression in MNs. Key biochemical events underlying the inhibitory effects of RNO were defined. RESULTS AND CONCLUSIONS In PMNs, RNO prevented phosphoinositide 3-kinase (PI3K)-dependent phosphorylation of Akt on Ser473, and Src family kinase (SFK)-mediated Pyk2 phosphorylation on Tyr579-580, while inducing protein kinase A-mediated phosphorylation of C-terminal Src kinase, the major negative regulator of SFKs. Modulation of these signaling pathways by RNO resulted in a significant impairment of PMN adhesion to activated PLTs or human umbilical vein endothelial cells, mainly mediated by inhibition of the adhesive function of Mac-1. Moreover RNO curbed SFK/PI3K-mediated NET release by PMNs adherent on fibrinogen-coated surfaces. In MNs interacting with activated PLTs, RNO curbed PI3K-mediated expression of tissue factor. The efficacy of RNO was significantly potentiated by formoterol, a long acting β-adrenergic receptor agonist. This study reveals novel antithrombotic activities by which roflumilast may exert protective effects against cardiovascular comorbodities in COPD.
Collapse
Affiliation(s)
- L Totani
- Laboratory of Vascular Biology and Pharmacology, Fondazione Mario Negri Sud, Santa Maria Imbaro, Italy
| | - C Amore
- Laboratory of Vascular Biology and Pharmacology, Fondazione Mario Negri Sud, Santa Maria Imbaro, Italy
| | - A Di Santo
- Laboratory of Vascular Biology and Pharmacology, Fondazione Mario Negri Sud, Santa Maria Imbaro, Italy
| | - G Dell'Elba
- Laboratory of Vascular Biology and Pharmacology, Fondazione Mario Negri Sud, Santa Maria Imbaro, Italy
| | - A Piccoli
- Laboratory of Vascular Biology and Pharmacology, Fondazione Mario Negri Sud, Santa Maria Imbaro, Italy
| | - N Martelli
- Laboratory of Vascular Biology and Pharmacology, Fondazione Mario Negri Sud, Santa Maria Imbaro, Italy
| | - H Tenor
- Takeda Pharmaceuticals International GmbH, Glattpark-Opfikon, Switzerland
| | - R Beume
- Takeda Pharmaceuticals International GmbH, Glattpark-Opfikon, Switzerland
| | - V Evangelista
- Laboratory of Vascular Biology and Pharmacology, Fondazione Mario Negri Sud, Santa Maria Imbaro, Italy
| |
Collapse
|
26
|
Glezeva N, Gilmer JF, Watson CJ, Ledwidge M. A Central Role for Monocyte-Platelet Interactions in Heart Failure. J Cardiovasc Pharmacol Ther 2015; 21:245-61. [PMID: 26519384 DOI: 10.1177/1074248415609436] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 08/04/2015] [Indexed: 01/08/2023]
Abstract
Heart failure (HF) is an increasingly prevalent and costly multifactorial syndrome with high morbidity and mortality rates. The exact pathophysiological mechanisms leading to the development of HF are not completely understood. Several emerging paradigms implicate cardiometabolic risk factors, inflammation, endothelial dysfunction, myocardial fibrosis, and myocyte dysfunction as key factors in the gradual progression from a healthy state to HF. Inflammation is now a recognized factor in disease progression in HF and a therapeutic target. Furthermore, the monocyte-platelet interaction has been highlighted as an important pathophysiological link between inflammation, thrombosis, endothelial activation, and myocardial malfunction. The contribution of monocytes and platelets to acute cardiovascular injury and acute HF is well established. However, their role and interaction in the pathogenesis of chronic HF are not well understood. In particular, the cross talk between monocytes and platelets in the peripheral circulation and in the vicinity of the vascular wall in the form of monocyte-platelet complexes (MPCs) may be a crucial element, which influences the pathophysiology and progression of chronic heart disease and HF. In this review, we discuss the role of monocytes and platelets as key mediators of cardiovascular inflammation in HF, the mechanisms of cell activation, and the importance of monocyte-platelet interaction and complexes in HF pathogenesis. Finally, we summarize recent information on pharmacological inhibition of inflammation and studies of antithrombotic strategies in the setting of HF that can inform opportunities for future work. We discuss recent data on monocyte-platelet interactions and the potential benefits of therapy directed at MPCs, particularly in the setting of HF with preserved ejection fraction.
Collapse
Affiliation(s)
- Nadezhda Glezeva
- School of Medicine & Medical Science, UCD Conway Institute, University College Dublin, Dublin, Belfield, Dublin, Ireland
| | - John F Gilmer
- School of Pharmacy & Pharmaceutical Sciences, TCD Centre for Health Sciences, Trinity College Dublin, College Green, Dublin, Ireland
| | - Chris J Watson
- School of Medicine & Medical Science, UCD Conway Institute, University College Dublin, Dublin, Belfield, Dublin, Ireland
| | - Mark Ledwidge
- Chronic Cardiovascular Disease Management Unit and Heart Failure Unit, St Vincent's Healthcare Group/St Michael's Hospital, County Dublin, Ireland
| |
Collapse
|
27
|
Hegedűs P, Korkmaz S, Radovits T, Schmidt H, Li S, Yoshikawa Y, Yasui H, Merkely B, Karck M, Szabó G. Bis (aspirinato) zinc (II) complex successfully inhibits carotid arterial neointima formation after balloon-injury in rats. Cardiovasc Drugs Ther 2015; 28:533-9. [PMID: 25129612 DOI: 10.1007/s10557-014-6549-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
PURPOSE Neointima formation following angioplasty is a serious consequence of endothelial damage in arteries. Inflammatory mediators and lack of endothelial regulatory mechanisms lead to migration and proliferation of smooth-muscle cells and thus to restenosis. This study examines the effect of the novel bis (aspirinato) zinc (II) complex on neointima formation in a rat model of carotid balloon-injury. METHODS Rats underwent balloon-injury of the right common carotid artery, then received PEG400 vehicle (untreated-group), acetylsalicylic-acid (ASA-group), zinc-chloride (Zn-group) and bis (aspirinato) zinc (II) complex (Zn(ASA) 2-group) orally for 18 consecutive days. From harvested carotid arteries, histology, immunohistochemistry and mRNA expression analysis were performed. RESULTS Compared to the untreated-group, Zn (ASA) 2-treatment significantly lowered stenosis ratio (54.0 ± 5.8% to 25.5 ± 3.9%) and reduced neointima/media ratio (1.5 ± 0.2 to 0.5 ± 0.1). Significantly higher alpha smooth muscle actin mRNA and protein expression were measured after Zn (ASA)2 and Zn-treatment in comparison with the untreated and ASA-groups while the expression of matrix-metalloproteinase-9 was significantly higher in these groups compared to Zn (ASA)2. The presence of collagen in media was significantly decreased in all treated groups. mRNA expressions of nuclear factor kappa-b, transforming growth-factor-β and proliferating cell nuclear antigen were significantly down-regulated, whereas a20 was up-regulated by Zn (ASA)2 treatment compared to the untreated and ASA-groups. CONCLUSION This study proves the effectivity of the novel bis (aspirinato) zinc complex in reducing neointima formation and restenosis after balloon-injury and supports the hypothesis that inhibition of smooth-muscle transformation/proliferation plays a key role in the prevention of restenosis.
Collapse
Affiliation(s)
- Péter Hegedűs
- Department of Cardiac Surgery, University of Heidelberg, INF 326, Heidelberg, 69120, Germany,
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Ed Rainger G, Chimen M, Harrison MJ, Yates CM, Harrison P, Watson SP, Lordkipanidzé M, Nash GB. The role of platelets in the recruitment of leukocytes during vascular disease. Platelets 2015. [PMID: 26196409 PMCID: PMC4673595 DOI: 10.3109/09537104.2015.1064881] [Citation(s) in RCA: 132] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Besides their role in the formation of thrombus during haemostasis, it is becoming clear that platelets contribute to a number of other processes within the vasculature. Indeed, the integrated function of the thrombotic and inflammatory systems, which results in platelet-mediated recruitment of leukocytes, is now considered to be of great importance in the propagation, progression and pathogenesis of atherosclerotic disease of the arteries. There are three scenarios by which platelets can interact with leukocytes: (1) during haemostasis, when platelets adhere to and are activated on sub-endothelial matrix proteins exposed by vascular damage and then recruit leukocytes to a growing thrombus. (2) Platelets adhere to and are activated on stimulated endothelial cells and then bridge blood borne leukocytes to the vessel wall and. (3) Adhesion between platelets and leukocytes occurs in the blood leading to formation of heterotypic aggregates prior to contact with endothelial cells. In the following review we will not discuss leukocyte recruitment during haemostasis, as this represents a physiological response to tissue trauma that can progress, at least in its early stages, in the absence of inflammation. Rather we will deal with scenarios 2 and 3, as these pathways of platelet–leukocyte interactions are important during inflammation and in chronic inflammatory diseases such as atherosclerosis. Indeed, these interactions mean that leukocytes possess means of adhesion to the vessel wall under conditions that may not normally be permissive of leukocyte–endothelial cell adhesion, meaning that the disease process may be able to bypass the regulatory pathways which would ordinarily moderate the inflammatory response.
Collapse
Affiliation(s)
- G Ed Rainger
- Centre for Cardiovascular Sciences, Institute for Biomedical Research, The Medical School, The University of Birmingham , Birmingham , UK and
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Men M, Zhang L, Li T, Mi B, Wang T, Fan Y, Chen Y, Shen G, Liang L, Ma A. Prognostic Value of the Percentage of Neutrophils on Admission in Patients with ST-elevated Myocardial Infarction Undergoing Primary Percutaneous Coronary Intervention. Arch Med Res 2015; 46:274-9. [PMID: 25989351 DOI: 10.1016/j.arcmed.2015.05.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 05/12/2015] [Indexed: 11/29/2022]
Abstract
BACKGROUND AND AIMS An elevated neutrophil count or neutrophil/lymphocyte ratio on admission has been reported to be an independent predictor of adverse cardiac events in patients with acute coronary syndrome (ACS). The relationship between the percentage of neutrophils (N%) at the time of admission and the long-term outcomes in patients with ST-segment elevated myocardial infarction (STEMI) who have undergone primary percutaneous coronary intervention (PCI) remains unclear. The aim of this study was to investigate the usefulness of the admission N% in predicting long-term mortality in patients with STEMI who were undergoing primary PCI. METHODS We evaluated 701 consecutive patients admitted to nine medical institutions in northwest China within 24 h after symptom onset from January 1, 2009-December 31, 2011. Using a receiver-operating characteristic analysis, N% ≥82.1% was the best predictor of long-term mortality. Patients were divided into two groups according to this criterion. Mean follow-up time was 39.03 months. RESULTS The long-term all-cause mortality rate was significantly higher in patients with a high N% level (7.17 vs. 3.11%, p = 0.015) as was the rate of cardiac mortality (6.48 vs. 2.59%, p = 0.013). In a multivariate logistic analysis, a high N% level was an independent predictor of long-term all-cause mortality (odds ratio 2.59, 95% confidence interval 1.21-5.53, p = 0.002) and long-term cardiac mortality (odds ratio 2.79, 95% confidence interval 1.24-6.28, p = 0.013). CONCLUSIONS A high N% level on admission is an independent predictor of long-term mortality in STEMI patients undergoing primary PCI.
Collapse
Affiliation(s)
- Min Men
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China; Department of Cardiovascular Medicine, Xi'an Central Hospital, Xi'an, China
| | - Li Zhang
- Department of Geriatrics, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Tao Li
- Department of Cardiovascular Medicine, Xi'an Central Hospital, Xi'an, China
| | - Baibing Mi
- Department of Statistics and Epidemiology, Xi'an Jiaotong University, Health Science Center, Xi'an, China
| | - Tingzhong Wang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China; Key Laboratory of Molecular Cardiology, Shaanxi Province, China; Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Xi'an, China
| | - Yan Fan
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yuewu Chen
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Guidong Shen
- Department of Cardiovascular Medicine, Ankang Central Hospital, Ankang, China
| | - Lei Liang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Aiqun Ma
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China; Key Laboratory of Molecular Cardiology, Shaanxi Province, China; Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
30
|
Araújo PV, Ribeiro MS, Dalio MB, Rocha LA, Viaro F, Joviliano RD, Piccinato CE, Évora PRB, Joviliano EE. Interleukins and Inflammatory Markers in In-Stent Restenosis after Femoral Percutaneous Transluminal Angioplasty. Ann Vasc Surg 2015; 29:731-7. [DOI: 10.1016/j.avsg.2014.12.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Revised: 11/06/2014] [Accepted: 12/12/2014] [Indexed: 10/23/2022]
|
31
|
Rong MY, Wang CH, Wu ZB, Zeng W, Zheng ZH, Han Q, Jia JF, Li XY, Zhu P. Platelets induce a proinflammatory phenotype in monocytes via the CD147 pathway in rheumatoid arthritis. Arthritis Res Ther 2014; 16:478. [PMID: 25404518 PMCID: PMC4298113 DOI: 10.1186/s13075-014-0478-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Accepted: 10/23/2014] [Indexed: 12/29/2022] Open
Abstract
Introduction Activated platelets exert a proinflammatory action that can be largely ascribed to their ability to interact with monocytes. However, the mechanisms that promote dynamic changes in monocyte subsets in rheumatoid arthritis (RA) have not been clearly identified. The aim of this study was to determine whether platelet activation and the consequent formation of monocyte-platelet aggregates (MPA) might induce a proinflammatory phenotype in circulating monocytes in RA. Methods The surface phenotype of platelets and the frequencies of monocyte subpopulations in the peripheral blood of RA patients were determined using flow cytometry. Platelets were sorted and co-cultured with monocytes. In addition, monocyte activation was assessed by measuring the nuclear factor kappa B (NF-κB) pathway. The disease activity was evaluated using the 28-joint disease activity score. Results Platelet activation, circulating intermediate monocytes (Mon2) and MPA formation were significantly elevated in RA, especially in those with active disease status. Furthermore, Mon2 monocytes showed higher CD147 expression and responded to direct cell contact with activated platelets with higher cytokine production and matrix metallopeptidase 9 (MMP-9) secretion, which increased the expression of CD147. After the addition of specific antibodies for CD147, those effects were abolished. Furthermore, the NF-κB-driven inflammatory pathway may be involved in this process. Conclusions These findings indicate an important role of platelet activation and the consequent formation of MPA in the generation of the proinflammatory cytokine milieu and for the promotion and maintenance of the pathogenically relevant Mon2 monocyte compartment in RA, which is likely to play an important role in the pathogenesis of autoimmunity.
Collapse
|
32
|
Lee CH, Yu CY, Chang SH, Hung KC, Liu SJ, Wang CJ, Hsu MY, Hsieh IC, Chen WJ, Ko YS, Wen MS. Promoting endothelial recovery and reducing neointimal hyperplasia using sequential-like release of acetylsalicylic acid and paclitaxel-loaded biodegradable stents. Int J Nanomedicine 2014; 9:4117-33. [PMID: 25206303 PMCID: PMC4157626 DOI: 10.2147/ijn.s67721] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
INTRODUCTION This work reports on the development of a biodegradable dual-drug-eluting stent with sequential-like and sustainable drug-release of anti-platelet acetylsalicylic acid and anti-smooth muscle cell (SMC) proliferative paclitaxel. METHODS To fabricate the biodegradable stents, poly-L-lactide strips are first cut from a solvent-casted film. They are rolled onto the surface of a metal pin to form spiral stents. The stents are then consecutively covered by acetylsalicylic acid and paclitaxel-loaded polylactide-polyglycolide nanofibers via electrospinning. RESULTS Biodegradable stents exhibit mechanical properties that are superior to those of metallic stents. Biodegradable stents sequentially release high concentrations of acetylsalicylic acid and paclitaxel for more than 30 and 60 days, respectively. In vitro, the eluted drugs promote endothelial cell numbers on days 3 and 7, and reduce the proliferation of SMCs in weeks 2, 4, and 8. The stents markedly inhibit the adhesion of platelets on days 3, 7, and 14 relative to a non-drug-eluting stent. In vivo, the implanted stent is intact, and no stent thrombosis is observed in the stent-implanted vessels without the administration of daily oral acetylsalicylic acid. Promotion of endothelial recovery and inhibition of neointimal hyperplasia are also observed on the stented vessels. CONCLUSION The work demonstrates the efficiency and safety of the biodegradable dual-drug-eluting stents with sequential and sustainable drug release to diseased arteries.
Collapse
Affiliation(s)
- Cheng-Hung Lee
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital-Linkou, Tao-Yuan, Taiwan
- Department of Mechanical Engineering, Chang Gung University, Tao-Yuan, Taiwan
| | - Chia-Ying Yu
- Department of Mechanical Engineering, Chang Gung University, Tao-Yuan, Taiwan
| | - Shang-Hung Chang
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital-Linkou, Tao-Yuan, Taiwan
| | - Kuo-Chun Hung
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital-Linkou, Tao-Yuan, Taiwan
| | - Shih-Jung Liu
- Department of Mechanical Engineering, Chang Gung University, Tao-Yuan, Taiwan
| | - Chao-Jan Wang
- Department of Medical Imaging and Intervention, Chang Gung Memorial Hospital, Linkou, Tao-Yuan, Taiwan
| | - Ming-Yi Hsu
- Department of Medical Imaging and Intervention, Chang Gung Memorial Hospital, Linkou, Tao-Yuan, Taiwan
| | - I-Chang Hsieh
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital-Linkou, Tao-Yuan, Taiwan
| | - Wei-Jan Chen
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital-Linkou, Tao-Yuan, Taiwan
| | - Yu-Shien Ko
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital-Linkou, Tao-Yuan, Taiwan
| | - Ming-Shien Wen
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital-Linkou, Tao-Yuan, Taiwan
| |
Collapse
|
33
|
Totani L, Piccoli A, Dell'Elba G, Concetta A, Di Santo A, Martelli N, Federico L, Pamuklar Z, Smyth SS, Evangelista V. Phosphodiesterase type 4 blockade prevents platelet-mediated neutrophil recruitment at the site of vascular injury. Arterioscler Thromb Vasc Biol 2014; 34:1689-96. [PMID: 24925970 DOI: 10.1161/atvbaha.114.303939] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
OBJECTIVE Platelet-neutrophil interactions play a key role in cardiovascular disease and inflammatory processes. Src family kinases mediate P-selectin glycoprotein ligand-1-Mac-1 cross talk necessary for firm platelet-neutrophil adhesion. Because Src family kinase activity can be regulated by cAMP-dependent pathways, in this work, we evaluated the role of phosphodiesterases in the signaling events that are required to sustain platelet-neutrophil interactions and neutrophil recruitment at the site of vascular injury. APPROACH AND RESULTS In neutrophils exposed to P-selectin, selective phosphodiesterase 4 (PDE4) inhibition prevented Src family kinase-mediated phosphorylation of the proline-rich tyrosine kinase 2 on Tyr579/Tyr580. The effects of PDE4 inhibition required protein kinase A, likely through protein kinase A-mediated activation of COOH-terminal Src kinase, a major negative regulator of Src family kinases. PDE4, but not other phosphodiesterase inhibitors, reduced platelet-neutrophil conjugates as well as neutrophil firm adhesion on spread platelets under flow conditions. The effect of PDE4 inhibition on neutrophil adhesion was primarily mediated by downregulation of P-selectin-induced activation of Mac-1. In a murine model of endovascular injury, selective inhibition of PDE4 significantly reduced neutrophil recruitment at the site of vascular damage. CONCLUSIONS This study identifies PDE4 as a central node in the signaling network that mediates platelet-neutrophil adhesion and suggests that pharmacological inhibition of PDE4 may represent a novel therapeutic avenue for the treatment of cardiovascular disease.
Collapse
Affiliation(s)
- Licia Totani
- From the Department of Translational Pharmacology, Laboratory of Vascular Biology and Pharmacology, Fondazione Mario Negri Sud, Santa Maria Imbaro, Chieti, Italy (L.T., A.P., G.D., A.C., A.D.S., N.M., V.E.); Division of Cardiovascular Medicine, The Gill Heart Institute, Lexington, KY (L.F., Z.P., S.S.S.); and VA Medical Center, Lexington, KY (S.S.S.)
| | - Antonio Piccoli
- From the Department of Translational Pharmacology, Laboratory of Vascular Biology and Pharmacology, Fondazione Mario Negri Sud, Santa Maria Imbaro, Chieti, Italy (L.T., A.P., G.D., A.C., A.D.S., N.M., V.E.); Division of Cardiovascular Medicine, The Gill Heart Institute, Lexington, KY (L.F., Z.P., S.S.S.); and VA Medical Center, Lexington, KY (S.S.S.)
| | - Giuseppe Dell'Elba
- From the Department of Translational Pharmacology, Laboratory of Vascular Biology and Pharmacology, Fondazione Mario Negri Sud, Santa Maria Imbaro, Chieti, Italy (L.T., A.P., G.D., A.C., A.D.S., N.M., V.E.); Division of Cardiovascular Medicine, The Gill Heart Institute, Lexington, KY (L.F., Z.P., S.S.S.); and VA Medical Center, Lexington, KY (S.S.S.)
| | - Amore Concetta
- From the Department of Translational Pharmacology, Laboratory of Vascular Biology and Pharmacology, Fondazione Mario Negri Sud, Santa Maria Imbaro, Chieti, Italy (L.T., A.P., G.D., A.C., A.D.S., N.M., V.E.); Division of Cardiovascular Medicine, The Gill Heart Institute, Lexington, KY (L.F., Z.P., S.S.S.); and VA Medical Center, Lexington, KY (S.S.S.)
| | - Angelomaria Di Santo
- From the Department of Translational Pharmacology, Laboratory of Vascular Biology and Pharmacology, Fondazione Mario Negri Sud, Santa Maria Imbaro, Chieti, Italy (L.T., A.P., G.D., A.C., A.D.S., N.M., V.E.); Division of Cardiovascular Medicine, The Gill Heart Institute, Lexington, KY (L.F., Z.P., S.S.S.); and VA Medical Center, Lexington, KY (S.S.S.)
| | - Nicola Martelli
- From the Department of Translational Pharmacology, Laboratory of Vascular Biology and Pharmacology, Fondazione Mario Negri Sud, Santa Maria Imbaro, Chieti, Italy (L.T., A.P., G.D., A.C., A.D.S., N.M., V.E.); Division of Cardiovascular Medicine, The Gill Heart Institute, Lexington, KY (L.F., Z.P., S.S.S.); and VA Medical Center, Lexington, KY (S.S.S.)
| | - Lorenzo Federico
- From the Department of Translational Pharmacology, Laboratory of Vascular Biology and Pharmacology, Fondazione Mario Negri Sud, Santa Maria Imbaro, Chieti, Italy (L.T., A.P., G.D., A.C., A.D.S., N.M., V.E.); Division of Cardiovascular Medicine, The Gill Heart Institute, Lexington, KY (L.F., Z.P., S.S.S.); and VA Medical Center, Lexington, KY (S.S.S.)
| | - Zehra Pamuklar
- From the Department of Translational Pharmacology, Laboratory of Vascular Biology and Pharmacology, Fondazione Mario Negri Sud, Santa Maria Imbaro, Chieti, Italy (L.T., A.P., G.D., A.C., A.D.S., N.M., V.E.); Division of Cardiovascular Medicine, The Gill Heart Institute, Lexington, KY (L.F., Z.P., S.S.S.); and VA Medical Center, Lexington, KY (S.S.S.)
| | - Susan S Smyth
- From the Department of Translational Pharmacology, Laboratory of Vascular Biology and Pharmacology, Fondazione Mario Negri Sud, Santa Maria Imbaro, Chieti, Italy (L.T., A.P., G.D., A.C., A.D.S., N.M., V.E.); Division of Cardiovascular Medicine, The Gill Heart Institute, Lexington, KY (L.F., Z.P., S.S.S.); and VA Medical Center, Lexington, KY (S.S.S.)
| | - Virgilio Evangelista
- From the Department of Translational Pharmacology, Laboratory of Vascular Biology and Pharmacology, Fondazione Mario Negri Sud, Santa Maria Imbaro, Chieti, Italy (L.T., A.P., G.D., A.C., A.D.S., N.M., V.E.); Division of Cardiovascular Medicine, The Gill Heart Institute, Lexington, KY (L.F., Z.P., S.S.S.); and VA Medical Center, Lexington, KY (S.S.S.).
| |
Collapse
|
34
|
Shah B, Berger JS, Amoroso NS, Mai X, Lorin JD, Danoff A, Schwartzbard AZ, Lobach I, Guo Y, Feit F, Slater J, Attubato MJ, Sedlis SP. Periprocedural glycemic control in patients with diabetes mellitus undergoing coronary angiography with possible percutaneous coronary intervention. Am J Cardiol 2014; 113:1474-80. [PMID: 24630791 DOI: 10.1016/j.amjcard.2014.01.428] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Revised: 01/20/2014] [Accepted: 01/20/2014] [Indexed: 12/18/2022]
Abstract
Periprocedural hyperglycemia is an independent predictor of mortality in patients who underwent percutaneous coronary intervention (PCI). However, periprocedural management of blood glucose is not standardized. The effects of routinely continuing long-acting glucose-lowering medications before coronary angiography with possible PCI on periprocedural glycemic control have not been investigated. Patients with diabetes mellitus (DM; n = 172) were randomized to continue (Continue group; n = 86) or hold (Hold group; n = 86) their clinically prescribed long-acting glucose-lowering medications before the procedure. The primary end point was glucose level on procedural access. In a subset of patients (no DM group: n = 25; Continue group: n = 25; and Hold group: n = 25), selected measures of platelet activity that change acutely were assessed. Patients with DM randomized to the Continue group had lower blood glucose levels on procedural access compared with those randomized to the Hold group (117 [97 to 151] vs 134 [117 to 172] mg/dl, p = 0.002). There were two hypoglycemic events in the Continue group and none in the Hold group, and no adverse events in either group. Selected markers of platelet activity differed across the no DM, Continue, and Hold groups (leukocyte platelet aggregates: 8.1% [7.2 to 10.4], 8.7% [6.9 to 11.4], 10.9% [8.6 to 14.7], p = 0.007; monocyte platelet aggregates: 14.0% [10.3 to 16.3], 20.8% [16.2 to 27.0], 22.5% [15.2 to 35.4], p <0.001; soluble p-selectin: 51.9 ng/ml [39.7 to 74.0], 59.1 ng/ml [46.8 to 73.2], 72.2 ng/ml [58.4 to 77.4], p = 0.014). In conclusion, routinely continuing clinically prescribed long-acting glucose-lowering medications before coronary angiography with possible PCI help achieve periprocedural euglycemia, appear safe, and should be considered as a strategy for achieving periprocedural glycemic control.
Collapse
Affiliation(s)
- Binita Shah
- Division of Cardiology, Department of Medicine, Veterans Affairs New York Harbor Health Care System and New York University School of Medicine, New York, New York.
| | - Jeffrey S Berger
- Division of Cardiology, Department of Medicine, New York University School of Medicine, New York, New York; Division of Hematology, Department of Medicine, New York University School of Medicine, New York, New York
| | - Nicholas S Amoroso
- Division of Cardiology, Department of Medicine, Veterans Affairs New York Harbor Health Care System and New York University School of Medicine, New York, New York
| | - Xingchen Mai
- Division of Cardiology, Department of Medicine, Veterans Affairs New York Harbor Health Care System and New York University School of Medicine, New York, New York
| | - Jeffrey D Lorin
- Division of Cardiology, Department of Medicine, Veterans Affairs New York Harbor Health Care System and New York University School of Medicine, New York, New York
| | - Ann Danoff
- Division of Endocrinology, Department of Medicine, Veterans Affairs New York Harbor Health Care System and New York University School of Medicine, New York, New York
| | - Arthur Z Schwartzbard
- Division of Cardiology, Department of Medicine, Veterans Affairs New York Harbor Health Care System and New York University School of Medicine, New York, New York
| | - Iryna Lobach
- Division of Biostatistics, Department of Population Health, New York University School of Medicine, New York, New York
| | - Yu Guo
- Division of Biostatistics, Department of Population Health, New York University School of Medicine, New York, New York
| | - Frederick Feit
- Division of Cardiology, Department of Medicine, Veterans Affairs New York Harbor Health Care System and New York University School of Medicine, New York, New York
| | - James Slater
- Division of Cardiology, Department of Medicine, Veterans Affairs New York Harbor Health Care System and New York University School of Medicine, New York, New York
| | - Michael J Attubato
- Division of Cardiology, Department of Medicine, Veterans Affairs New York Harbor Health Care System and New York University School of Medicine, New York, New York
| | - Steven P Sedlis
- Division of Cardiology, Department of Medicine, Veterans Affairs New York Harbor Health Care System and New York University School of Medicine, New York, New York
| |
Collapse
|
35
|
Li J, Kim K, Hahm E, Molokie R, Hay N, Gordeuk VR, Du X, Cho J. Neutrophil AKT2 regulates heterotypic cell-cell interactions during vascular inflammation. J Clin Invest 2014; 124:1483-96. [PMID: 24642468 PMCID: PMC3973084 DOI: 10.1172/jci72305] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Accepted: 12/17/2013] [Indexed: 12/22/2022] Open
Abstract
Interactions between platelets, leukocytes, and activated endothelial cells are important during microvascular occlusion; however, the regulatory mechanisms of these heterotypic cell-cell interactions remain unclear. Here, using intravital microscopy to evaluate mice lacking specific isoforms of the serine/threonine kinase AKT and bone marrow chimeras, we found that hematopoietic cell-associated AKT2 is important for neutrophil adhesion and crawling and neutrophil-platelet interactions on activated endothelial cells during TNF-α-induced venular inflammation. Studies with an AKT2-specific inhibitor and cells isolated from WT and Akt KO mice revealed that platelet- and neutrophil-associated AKT2 regulates heterotypic neutrophil-platelet aggregation under shear conditions. In particular, neutrophil AKT2 was critical for membrane translocation of αMβ2 integrin, β2-talin1 interaction, and intracellular Ca2+ mobilization. We found that the basal phosphorylation levels of AKT isoforms were markedly increased in neutrophils and platelets isolated from patients with sickle cell disease (SCD), an inherited hematological disorder associated with vascular inflammation and occlusion. AKT2 inhibition reduced heterotypic aggregation of neutrophils and platelets isolated from SCD patients and diminished neutrophil adhesion and neutrophil-platelet aggregation in SCD mice, thereby improving blood flow rates. Our results provide evidence that neutrophil AKT2 regulates αMβ2 integrin function and suggest that AKT2 is important for neutrophil recruitment and neutrophil-platelet interactions under thromboinflammatory conditions such as SCD.
Collapse
Affiliation(s)
- Jing Li
- Department of Pharmacology,
Section of Hematology/Oncology, and
Comprehensive Sickle Cell Center, University of Illinois College of Medicine, Chicago, Illinois, USA.
Jesse Brown VA Medical Center, Chicago, Illinois, USA.
Department of Biochemistry and Molecular Genetics and
Department of Anesthesiology, University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Kyungho Kim
- Department of Pharmacology,
Section of Hematology/Oncology, and
Comprehensive Sickle Cell Center, University of Illinois College of Medicine, Chicago, Illinois, USA.
Jesse Brown VA Medical Center, Chicago, Illinois, USA.
Department of Biochemistry and Molecular Genetics and
Department of Anesthesiology, University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Eunsil Hahm
- Department of Pharmacology,
Section of Hematology/Oncology, and
Comprehensive Sickle Cell Center, University of Illinois College of Medicine, Chicago, Illinois, USA.
Jesse Brown VA Medical Center, Chicago, Illinois, USA.
Department of Biochemistry and Molecular Genetics and
Department of Anesthesiology, University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Robert Molokie
- Department of Pharmacology,
Section of Hematology/Oncology, and
Comprehensive Sickle Cell Center, University of Illinois College of Medicine, Chicago, Illinois, USA.
Jesse Brown VA Medical Center, Chicago, Illinois, USA.
Department of Biochemistry and Molecular Genetics and
Department of Anesthesiology, University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Nissim Hay
- Department of Pharmacology,
Section of Hematology/Oncology, and
Comprehensive Sickle Cell Center, University of Illinois College of Medicine, Chicago, Illinois, USA.
Jesse Brown VA Medical Center, Chicago, Illinois, USA.
Department of Biochemistry and Molecular Genetics and
Department of Anesthesiology, University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Victor R. Gordeuk
- Department of Pharmacology,
Section of Hematology/Oncology, and
Comprehensive Sickle Cell Center, University of Illinois College of Medicine, Chicago, Illinois, USA.
Jesse Brown VA Medical Center, Chicago, Illinois, USA.
Department of Biochemistry and Molecular Genetics and
Department of Anesthesiology, University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Xiaoping Du
- Department of Pharmacology,
Section of Hematology/Oncology, and
Comprehensive Sickle Cell Center, University of Illinois College of Medicine, Chicago, Illinois, USA.
Jesse Brown VA Medical Center, Chicago, Illinois, USA.
Department of Biochemistry and Molecular Genetics and
Department of Anesthesiology, University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Jaehyung Cho
- Department of Pharmacology,
Section of Hematology/Oncology, and
Comprehensive Sickle Cell Center, University of Illinois College of Medicine, Chicago, Illinois, USA.
Jesse Brown VA Medical Center, Chicago, Illinois, USA.
Department of Biochemistry and Molecular Genetics and
Department of Anesthesiology, University of Illinois College of Medicine, Chicago, Illinois, USA
| |
Collapse
|
36
|
Lee CH, Chang SH, Lin YH, Liu SJ, Wang CJ, Hsu MY, Hung KC, Yeh YH, Chen WJ, Hsieh IC, Wen MS. Acceleration of re-endothelialization and inhibition of neointimal formation using hybrid biodegradable nanofibrous rosuvastatin-loaded stents. Biomaterials 2014; 35:4417-27. [PMID: 24582553 DOI: 10.1016/j.biomaterials.2014.02.017] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Accepted: 02/09/2014] [Indexed: 10/25/2022]
Abstract
Incomplete endothelialization and neointimal hyperplasia of injured arteries can cause acute and late stent thromboses. This work develops hybrid stent/biodegradable nanofibers for the local and sustained delivery of rosuvastatin to denuded artery walls. Biodegradable nanofibers were firstly prepared by dissolving poly(D,L)-lactide-co-glycolide and rosuvastatin in 1,1,1,3,3,3-hexafluoro-2-propanol. The solution was then electrospun into nanofibrous tubes, which were mounted onto commercially available bare-metal stents. The in vitro release rates of the pharmaceuticals from the nanofibers were determined using an elution method and a high-performance liquid chromatography assay. The experimental results thus obtained suggest that the biodegradable nanofibers released high concentrations of rosuvastatin for four weeks. The effectiveness of the local delivery of rosuvastatin in reducing platelets was studied. The tissue inflammatory reaction caused by the hybrid stents that were used to treat diseased arteries was also documented. The proposed hybrid stent/biodegradable rosuvastatin-loaded nanofibers contributed substantially to the local and sustainable delivery of a high concentration of drugs to promote re-endothelialization, improve endothelial function, reduce inflammatory reaction, and inhibit neointimal formation of the injured artery. The results of this work provide insight into how patients with a high risk of stent restenosis should be treated for accelerating re-endothelialization and inhibiting neointimal hyperplasia.
Collapse
Affiliation(s)
- Cheng-Hung Lee
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou, Chang Gung University College of Medicine, Tao-Yuan, Taiwan; Department of Mechanical Engineering, Chang Gung University, Tao-Yuan, Taiwan
| | - Shang-Hung Chang
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou, Chang Gung University College of Medicine, Tao-Yuan, Taiwan
| | - Yu-Huang Lin
- Graduate Institute of Medical Mechatronics, Chang Gung University, Tao-Yuan, Taiwan
| | - Shih-Jung Liu
- Department of Mechanical Engineering, Chang Gung University, Tao-Yuan, Taiwan.
| | - Chao-Jan Wang
- Department of Medical Imaging and Intervention, Chang Gung Memorial Hospital, Linkou, Tao-Yuan, Taiwan
| | - Ming-Yi Hsu
- Department of Medical Imaging and Intervention, Chang Gung Memorial Hospital, Linkou, Tao-Yuan, Taiwan
| | - Kuo-Chun Hung
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou, Chang Gung University College of Medicine, Tao-Yuan, Taiwan
| | - Yung-Hsin Yeh
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou, Chang Gung University College of Medicine, Tao-Yuan, Taiwan
| | - Wei-Jan Chen
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou, Chang Gung University College of Medicine, Tao-Yuan, Taiwan
| | - I-Chang Hsieh
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou, Chang Gung University College of Medicine, Tao-Yuan, Taiwan
| | - Ming-Shien Wen
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou, Chang Gung University College of Medicine, Tao-Yuan, Taiwan
| |
Collapse
|
37
|
Rodriguez-Menocal L, Faridi MH, Martinez L, Shehadeh LA, Duque JC, Wei Y, Mesa A, Pena A, Gupta V, Pham SM, Vazquez-Padron RI. Macrophage-derived IL-18 and increased fibrinogen deposition are age-related inflammatory signatures of vascular remodeling. Am J Physiol Heart Circ Physiol 2014; 306:H641-53. [PMID: 24414074 DOI: 10.1152/ajpheart.00641.2013] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Aging has been associated with pathological vascular remodeling and increased neointimal hyperplasia. The understanding of how aging exacerbates this process is fundamental to prevent cardiovascular complications in the elderly. This study proposes a mechanism by which aging sustains leukocyte adhesion, vascular inflammation, and increased neointimal thickness after injury. The effect of aging on vascular remodeling was assessed in the rat balloon injury model using microarray analysis, immunohistochemistry, and LINCOplex assays. The injured arteries in aging rats developed thicker neointimas than those in younger animals, and this significantly correlated with a higher number of tissue macrophages and increased vascular IL-18. Indeed, IL-18 was 23-fold more abundant in the injured vasculature of aged animals compared with young rats, while circulating levels were similar in both groups of animals. The depletion of macrophages in aged rats with clodronate liposomes ameliorated vascular accumulation of IL-18 and significantly decreased neointimal formation. IL-18 was found to inhibit apoptosis of vascular smooth muscle cells (VSMC) and macrophages, thus favoring both the formation and inflammation of the neointima. In addition, injured arteries of aged rats accumulated 18-fold more fibrinogen-γ than those of young animals. Incubation of rat peritoneal macrophages with immobilized IL-18 increased leukocyte adhesion to fibrinogen and suggested a proinflammatory positive feedback loop among macrophages, VSMC, and the deposition of fibrinogen during neointimal hyperplasia. In conclusion, our data reveal that concentration changes in vascular cytokine and fibrinogen following injury in aging rats contribute to local inflammation and postinjury neointima formation.
Collapse
Affiliation(s)
- Luis Rodriguez-Menocal
- Department of Surgery and Vascular Biology Institute, University of Miami Miller School of Medicine, Miami, Florida
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Lee CH, Lin YH, Chang SH, Tai CD, Liu SJ, Chu Y, Wang CJ, Hsu MY, Chang H, Chang GJ, Hung KC, Hsieh MJ, Lin FC, Hsieh IC, Wen MS, Huang Y. Local sustained delivery of acetylsalicylic acid via hybrid stent with biodegradable nanofibers reduces adhesion of blood cells and promotes reendothelialization of the denuded artery. Int J Nanomedicine 2014; 9:311-26. [PMID: 24421640 PMCID: PMC3888352 DOI: 10.2147/ijn.s51258] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Incomplete endothelialization, blood cell adhesion to vascular stents, and inflammation of arteries can result in acute stent thromboses. The systemic administration of acetylsalicylic acid decreases endothelial dysfunction, potentially reducing thrombus, enhancing vasodilatation, and inhibiting the progression of atherosclerosis; but, this is weakened by upper gastrointestinal bleeding. This study proposes a hybrid stent with biodegradable nanofibers, for the local, sustained delivery of acetylsalicylic acid to injured artery walls. Biodegradable nanofibers are prepared by first dissolving poly(D,L)-lactide-co-glycolide and acetylsalicylic acid in 1,1,1,3,3,3-hexafluoro-2-propanol. The solution is then electrospun into nanofibrous tubes, which are then mounted onto commercially available bare-metal stents. In vitro release rates of pharmaceuticals from nanofibers are characterized using an elution method, and a highperformance liquid chromatography assay. The experimental results suggest that biodegradable nanofibers release high concentrations of acetylsalicylic acid for three weeks. The in vivo efficacy of local delivery of acetylsalicylic acid in reducing platelet and monocyte adhesion, and the minimum tissue inflammatory reaction caused by the hybrid stents in treating denuded rabbit arteries, are documented. The proposed hybrid stent, with biodegradable acetylsalicylic acid-loaded nanofibers, substantially contributed to local, sustained delivery of drugs to promote re-endothelialization and reduce thrombogenicity in the injured artery. The stents may have potential applications in the local delivery of cardiovascular drugs. Furthermore, the use of hybrid stents with acetylsalicylic acid-loaded nanofibers that have high drug loadings may provide insight into the treatment of patients with high risk of acute stent thromboses.
Collapse
Affiliation(s)
- Cheng-Hung Lee
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Linkou, Taiwan ; Department of Mechanical Engineering, Taiwan
| | - Yu-Huang Lin
- Graduate Institute of Medical Mechatronics, Chang Gung University, Taiwan
| | - Shang-Hung Chang
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Linkou, Taiwan
| | - Chun-Der Tai
- Graduate Institute of Medical Mechatronics, Chang Gung University, Taiwan
| | | | - Yen Chu
- Laboratory of Cardiovascular Physiology, Division of Thoracic and Cardiovascular Surgery, Taiwan
| | - Chao-Jan Wang
- Department of Medical Imaging and Intervention, Taiwan
| | - Ming-Yi Hsu
- Department of Medical Imaging and Intervention, Taiwan
| | - Hung Chang
- Hematology-Oncology Division, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Gwo-Jyh Chang
- Graduate Institute of Clinical Medicinal Sciences, Chang Gung University College of Medicine, Linkou, Taiwan
| | - Kuo-Chun Hung
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Linkou, Taiwan
| | - Ming-Jer Hsieh
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Linkou, Taiwan
| | - Fen-Chiung Lin
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Linkou, Taiwan
| | - I-Chang Hsieh
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Linkou, Taiwan
| | - Ming-Shien Wen
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Linkou, Taiwan
| | - Yenlin Huang
- Department of Anatomical Pathology, Chang Gung Memorial Hospital, Linkou, Tao-Yuan, Taiwan
| |
Collapse
|
39
|
Relation between white blood cell count and final infarct size in patients with ST-segment elevation acute myocardial infarction undergoing primary percutaneous coronary intervention (from the INFUSE AMI trial). Am J Cardiol 2013; 112:1860-6. [PMID: 24063843 DOI: 10.1016/j.amjcard.2013.08.010] [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: 07/04/2013] [Revised: 08/14/2013] [Accepted: 08/14/2013] [Indexed: 11/23/2022]
Abstract
Although it has been shown that elevated white blood cell count (WBCc) on presentation is associated with an increased risk of cardiac mortality in patients with ST-segment elevation myocardial infarction (STEMI), the responsible mechanisms are unknown. We therefore sought to investigate whether elevated WBCc is associated with increased infarct size measured with cardiac magnetic resonance imaging 30 days after primary percutaneous coronary intervention in the Intracoronary Abciximab and Aspiration Thrombectomy in Patients With Large Anterior Myocardial Infarction trial. INFUSE AMI randomized patients with STEMI and proximal or mid-left anterior descending coronary artery occlusion to bolus intracoronary abciximab versus no abciximab and to manual aspiration versus no aspiration. WBCc at hospital admission was available in 407 of 452 randomized patients. Patients were stratified according to tertiles of WBCc. At 30 days, a significant stepwise increase in infarct size (percentage of total left ventricular mass) was apparent across tertiles of increasing WBCc (median [interquartile range] for tertiles I vs II vs III = 11.2% [3.8% to 19.6%] vs 17.5% [0.5% to 22.9%] vs 19.1% [13.7 to 26.0], respectively, p <0.0001). Absolute infarct mass in grams and abnormal wall motion score were also significantly increased across tertiles of WBC. By multivariate linear regression analysis, WBCc was an independent predictor of infarct size along with intracoronary abciximab randomization, age, time from symptom onset to first device, proximal left anterior descending location, and baseline TIMI flow of 0/1. In conclusion, in patients with anterior wall STEMI, an elevated admission WBCc is a powerful independent predictor of infarct size measured with cardiac magnetic resonance imaging 30 days after primary percutaneous coronary intervention.
Collapse
|
40
|
Cho HO, Nam CW, Lee HM, Shin HW, Cho YK, Yoon HJ, Park HS, Kim H, Chung IS, Hur SH, Kim YN, Kim KB. Fever after primary percutaneous coronary intervention in ST-segment elevation myocardial infarction is associated with adverse outcomes. Int J Cardiol 2013; 170:376-80. [PMID: 24290071 DOI: 10.1016/j.ijcard.2013.11.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Revised: 09/24/2013] [Accepted: 11/02/2013] [Indexed: 01/19/2023]
Abstract
BACKGROUND Fever is a common finding after primary percutaneous coronary intervention (PPCI) in patients with ST-segment elevation myocardial infarction (STEMI). However, its prognostic value is not validated yet. OBJECTIVES This study sought to evaluate the impact of fever after PPCI in STEMI on adverse clinical outcomes. METHODS Five hundred fourteen consecutive patients who underwent PPCI due to STEMI were enrolled. Body temperature (BT) was checked every 6 h for 5 days after PPCI. Patients were divided into two groups according to the highest quartile of peak BT; peak BT≤37.6 °C (control group) and peak BT>37.6 °C (fever group). Rates of 1-year major adverse cardiovascular events (MACE; death, myocardial infarction, any revascularization) were compared. RESULTS The prevalence of fever group (peak BT>37.6 °C) was 24.7% (127/514). White blood cell count, highly sensitive C-reactive protein and serum cardiac troponin I level were higher in fever group than control group (12,162±4199/μL vs. 10,614±3773/μL, p<0.001; 22.9±49.4 mg/L vs. 7.4±2.5 mg/L, p=0.001, 16.7±36.9 ng/dl vs. 8.70±26.2 ng/dl, p=0.027, respectively). The frequency of a history of previous myocardial infarction and left ventricular ejection fraction was lower in fever group (0.0% vs. 4.7%, p=0.010; 47±8 % vs. 49±9 %, p=0.002, respectively). There was no significant difference in angiographic characteristics between 2 groups. 1-year MACE rates were higher in fever group (11.0% vs. 4.7%, p=0.010). Multivariate analysis revealed fever (OR 2.358, 95% CI 1.113-4.998, p=0.025), diabetes mellitus as risk factor (2.227, 1.031-4.812, 0.042), and left anterior descending artery as infarct related artery (2.443, 1.114-5.361, 0.026) as independent predictors for 1-year MACE. CONCLUSIONS Fever after PPCI in patients with STEMI is frequently developed and it can predict adverse clinical outcome.
Collapse
Affiliation(s)
- Hyun-Ok Cho
- Keimyung University, Dongsan Medical Center, Daegu, Republic of Korea; Andong Medical Group, Andong, Republic of Korea
| | - Chang-Wook Nam
- Keimyung University, Dongsan Medical Center, Daegu, Republic of Korea.
| | - Ho-Myung Lee
- Keimyung University, Dongsan Medical Center, Daegu, Republic of Korea
| | - Hong-Won Shin
- Keimyung University, Dongsan Medical Center, Daegu, Republic of Korea
| | - Yun-Kyeong Cho
- Keimyung University, Dongsan Medical Center, Daegu, Republic of Korea
| | - Hyuck-Jun Yoon
- Keimyung University, Dongsan Medical Center, Daegu, Republic of Korea
| | - Hyoung-Seob Park
- Keimyung University, Dongsan Medical Center, Daegu, Republic of Korea
| | - Hyungseop Kim
- Keimyung University, Dongsan Medical Center, Daegu, Republic of Korea
| | - In-Sung Chung
- Keimyung University, Dongsan Medical Center, Daegu, Republic of Korea
| | - Seung-Ho Hur
- Keimyung University, Dongsan Medical Center, Daegu, Republic of Korea
| | - Yoon-Nyun Kim
- Keimyung University, Dongsan Medical Center, Daegu, Republic of Korea
| | - Kwon-Bae Kim
- Keimyung University, Dongsan Medical Center, Daegu, Republic of Korea
| |
Collapse
|
41
|
Palmerini T, Généreux P, Mehran R, Dangas G, Caixeta A, Riva DD, Mariani A, Xu K, Stone GW. Association among leukocyte count, mortality, and bleeding in patients with non-ST-segment elevation acute coronary syndromes (from the Acute Catheterization and Urgent Intervention Triage StrategY [ACUITY] trial). Am J Cardiol 2013; 111:1237-45. [PMID: 23411106 DOI: 10.1016/j.amjcard.2012.12.056] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Revised: 12/26/2012] [Accepted: 12/26/2012] [Indexed: 11/29/2022]
Abstract
Although inflammation is involved in the pathogenesis of acute coronary syndromes, the extent of inflammation is not routinely assessed, and its prognostic implications in patients with non-ST-segment elevation acute coronary syndrome have not been investigated in depth. We analyzed the prognostic implications of an elevated white blood cell count (WBCc) in patients with moderate and high-risk non-ST-segment elevation acute coronary syndrome undergoing an early invasive strategy in the large-scale Acute Catheterization and Urgent Intervention Triage StrategY trial. The WBCc at admission was available for 13,678 of 13,819 patients (98.9%). The patients in the upper tertile of the WBCc had an increased risk of 30-day major bleeding, 1-year mortality, and definite/probable stent thrombosis compared to those in the mid or lower tertiles. On multivariate analysis, the WBCc was an independent predictor of 30-day major bleeding and 1-year cardiac, noncardiac, and all-cause mortality. The association between the WBCc and cardiac mortality was present in multiple prespecified subgroups, with no significant interaction between the WBCc and age, gender, diabetes, smoking, renal dysfunction, elevated baseline biomarkers, antithrombotic therapy, revascularization, and Thrombolysis In Myocardial Infarction risk score. The WBCc remained an independent predictor of mortality after adjusting for bleeding, C-reactive protein level, and angiographic variables, including left ventricular ejection fraction, Thrombolysis In Myocardial Infarction flow, and number of diseased vessels. The WBCc significantly improved the prognostic accuracy of the Thrombolysis In Myocardial Infarction risk score, with a net reclassification improvement of 11% (p <0.0001). In conclusion, in patients with moderate- and high-risk non-ST-segment elevation acute coronary syndrome, an elevated admission WBCc was an independent predictor of 30-day major bleeding, and 1-year cardiac, noncardiac, and all-cause mortality.
Collapse
|
42
|
Johnson G, Curry B, Cahalan L, Prater R, Biggerstaff J, Hussain A, Gartner M, Cahalan P. Effects of surface-bound and intravenously administered heparin on cell-surface interactions: inflammation and coagulation. Perfusion 2013; 28:263-71. [DOI: 10.1177/0267659113475834] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Intravenous administration of heparin and heparin-bonded extracorporeal circuits are frequently used to mitigate the deleterious effects of blood contact with synthetic materials. The work described here utilized human blood in a micro-perfusion circuit to experimentally examine the effects of intravenous and surface-bound heparin on cellular activation. Activation markers of coagulation and of the inflammatory response were examined using flow cytometry; specifically, markers of platelet, monocyte, polymorphonuclear leukocyte (PMN), and lymphocyte activation were quantified. The results indicate that surface-bound heparin reduces the inflammatory response whereas systemically administered heparin does not. This finding has important implications for blood-contacting devices, particularly within the context of recently elucidated connections between inflammation pathways and coagulation disorders. Data presented indicate that surface-bound heparin and intravenously administered heparin play distinct, but vital roles in rendering biomaterial surfaces compatible with blood.
Collapse
Affiliation(s)
| | - B Curry
- University of Tennessee – Knoxville, Knoxville, TN, USA
| | | | - R Prater
- University of Tennessee – Knoxville, Knoxville, TN, USA
| | - J Biggerstaff
- University of Tennessee – Knoxville, Knoxville, TN, USA
| | | | | | | |
Collapse
|
43
|
Abstract
Flow cytometry is a powerful and versatile tool which can be used to provide substantial phenotypic data on platelets by yielding quantitative information of their physical and antigenic properties. This includes surface expression of functional receptors, bound ligands, expression of granule components, interaction of platelets with other platelets via aggregation, or interaction with other blood components, such as leukocytes or the plasma coagulation system. Quantitative assessment of these parameters may facilitate the diagnosis of inherited or acquired platelet disorders, assist in the diagnosis of diseases associated with platelet activation, or assist in the monitoring of safety and efficacy of antiplatelet therapy.
Collapse
Affiliation(s)
- Matthew D Linden
- Centre for Microscopy, Characterisation and Analysis, University of Western Australia, Crawley, WA, Australia
| |
Collapse
|
44
|
Abstract
Platelets are cell fragments which circulate in blood. They are of pivotal importance in blood clot formation, affecting thrombosis and haemostasis. By rapidly altering the activation and expression of surface receptors, platelets are able to quickly undergo structural and phenotypic changes in response to stimulation, such as collagen exposure on injured vascular endothelium. This response to stimulation allows platelets to become adhesive, aggregate to form a thrombus, and release a variety of mediators affecting coagulation, inflammation, and chemotaxis at the site of injury. Therefore, in addition to their critical role in thrombosis and haemostasis, platelets also play a role in immunity, inflammation, wound healing, haematologic malignancies, and metabolic disorders. The role of platelets in disease, particularly in atherothrombosis, is increasingly the focus of current research and antiplatelet therapy plays a significant role in the prevention and treatment of atherothrombotic and inflammatory diseases.
Collapse
Affiliation(s)
- Matthew D Linden
- Centre for Microscopy, Characterisation and Analysis, University of Western Australia, Crawley, WA, Australia
| |
Collapse
|
45
|
|
46
|
Montagnana M, Fava C, Arosio E, Degan M, Tommasoli RM, De Marchi S, Delva P, Spadaro R, Guidi GC, Lechi A, Santonastaso CL, Minuz P. Inflammation and platelet activation in peripheral arterial occlusive disease. Int J Angiol 2012; 16:84-8. [PMID: 22477298 DOI: 10.1055/s-0031-1278255] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
OBJECTIVES Epidemiological evidence indicates that inflammation accompanies the progression of atherosclerosis. The aim of the present cross-sectional study was to define relationships between platelet activation and inflammation in patients with mild to severe (stages II to IV) peripheral arterial occlusive disease (PAOD) and matched controls. The effect of chronic administration of low-dose acetylsalicylic acid was investigated. METHODS Subjects were studied on a single occasion. C-reactive protein (CRP) and two indexes of in vivo platelet activation were measured - the urinary excretion of 11-dehydrothromboxane (TX) B(2) by immunoassay and circulating platelet-monocyte aggregates (PMAs) by flow cytometry. RESULTS Plasma PMAs and urinary 11-dehydro-TXB(2) were significantly increased in PAOD patients compared with controls (P<0.01 for all). A positive correlation between 11-dehydro-TXB(2) and CRP was found in the study population (r(s)=0.63, P<0.001). Using logistic regression analysis, CRP was the only independent correlate of 11-dehydro-TXB(2) (β(CRP)=11.9, P<0.01), whereas only the presence of PAOD was an independent predictor of high PMA levels (β(PAOD)=13.7, P=0.001). Chronic administration of acetylsalicylic acid reduced 11-dehydro-TXB(2), but not PMA and CRP. CONCLUSIONS There is evidence that platelet activation in patients with PAOD is related to the vascular disease and is dependent on the severity of inflammation.
Collapse
|
47
|
Abstract
The invited special lecture at the 76(th) Annual Scientific Meeting of the Japanese Circulation Society focused on the central role of inflammation in vascular injury and repair. Early studies pioneered the concept that mechanical injury, such as balloon angioplasty and endovascular stent deployment, elicits an inflammatory response from the vessel wall. This hypothesis was developed and substantiated at a time when the prevailing dogma viewed restenosis following angioplasty as a primarily proliferative smooth muscle cell disease. Antibody targeting of Mac-1 reduced leukocyte accumulation and limited neointimal formation following balloon injury or stent implantation. Genetic absence of Mac-1 resulted in diminished leukocyte accumulation and neointimal thickening after carotid artery injury in mice. In the course of those studies, our laboratory made fundamental discoveries regarding the mechanism of leukocyte recruitment at sites of vascular injury and identified platelet glycoprotein (GP) Ibα, a component of the GPIb-IX-V complex, as the previously unknown platelet counter-receptor for Mac-1. Follow-on studies have focused extensively on the structure, function, and signaling of the leukocyte integrin Mac-1. The binding site for GPIbα in Mac-1 has been mapped and subsequently showed that leukocyte engagement of platelet GPIbα via Mac-1 is critical not only for the biological response to vascular injury, but also for thrombosis, vasculitis, glomerulonephritis, and multiple sclerosis, thereby advancing the hypothesis that virtually all inflammation is platelet-dependent. Furthermore, ligand engagement of Mac-1 initiates a novel gene program that promotes inflammation by activating NFκB and downregulating the expression of the forkhead transcription factor Foxp1 that controls monocyte differentiation. Small molecule inhibitors of Mac-1 function have been pursued, including targeting of Mac-1-GPIbα binding or the downstream tyrosine kinase spleen tyrosine kinase. Drs Teruo Inoue, Koichi Node, Tatsuya Fukotomi, Masashi Sakuma, Toshifumi Morooka, and Kohsuke Nakajima, valued Japanese collaborators and post-doctoral fellows, have contributed enormously to these discoveries.
Collapse
Affiliation(s)
- Daniel I Simon
- Division of Cardiovascular Medicine, Harrington Heart & Vascular Institute, University Hospitals Case Medical Center, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA.
| |
Collapse
|
48
|
Liposomal alendronate for the treatment of restenosis. J Control Release 2012; 161:619-27. [DOI: 10.1016/j.jconrel.2011.11.037] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2011] [Revised: 11/29/2011] [Accepted: 11/30/2011] [Indexed: 12/24/2022]
|
49
|
Totani L, Dell'Elba G, Martelli N, Di Santo A, Piccoli A, Amore C, Evangelista V. Prasugrel inhibits platelet-leukocyte interaction and reduces inflammatory markers in a model of endotoxic shock in the mouse. Thromb Haemost 2012; 107:1130-40. [PMID: 22436970 DOI: 10.1160/th11-12-0867] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2011] [Accepted: 02/27/2012] [Indexed: 01/20/2023]
Abstract
Prasugrel, through its active metabolite, reduces atherothrombosis and its clinical manifestations by inhibiting platelet activation and aggregation. Platelets also contribute to inflammation through interaction with different classes of leukocytes. We investigated whether the inhibitory effect of prasugrel on platelets also counteract inflammatory responses. The effect of prasugrel active metabolite, R-138727, was investigated on platelet P-selectin expression, platelet adhesion to polymorphonuclear leukocytes (PMN) and monocytes (MN) and Mac-1 expression in PMN and MN, in vitro, in human cells. The ex vivo effect of prasugrel administration on P-selectin, thromboxane (TXB)2 formation, platelet-PMN conjugates and Mac-1 expression in PMN triggered by PAR-4 agonist peptide was examined in whole blood from healthy mice as well as from mice in which an acute inflammatory reaction was induced by treatment with endotoxin. The effect of prasugrel on inflammatory markers in endotoxin-treated animals was also tested in vivo. R-138727 inhibited agonist-stimulated expression of platelet P-selectin, platelet-PMN and platelet-MN adhesion and platelet-dependent Mac-1 expression in leukocytes. Addition of aspirin did not modify the inhibitory effect elicited by R-138727. Treatment of mice with prasugrel resulted in a profound inhibition of platelet P-selectin expression, TXB2 production, platelet-PMN adhesion and Mac-1 expression in PMN induced by ex vivo stimulation with PAR-4 agonist peptide of whole blood from healthy or endotoxin-treated mice. Measurement of markers revealed that prasugrel reduced TXB2 and tumour necrosis factor-α synthesis and increased nitric oxide metabolites in endotoxin-treated mice in vivo. In conclusion, prasugrel reduces platelet interactions with PMN and MN. Through these effects prasugrel may curb platelet-mediated inflammatory responses.
Collapse
Affiliation(s)
- Licia Totani
- Laboratory of Vascular Biology and Pharmacology, Consorzio Mario Negri Sud, Via Nazionale 8/A, 66030 Santa Maria Imbaro, Italy
| | | | | | | | | | | | | |
Collapse
|
50
|
Passacquale G, Ferro A. Current concepts of platelet activation: possibilities for therapeutic modulation of heterotypic vs. homotypic aggregation. Br J Clin Pharmacol 2012; 72:604-18. [PMID: 21223359 DOI: 10.1111/j.1365-2125.2011.03906.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Thrombogenic and inflammatory activity are two distinct aspects of platelet biology, which are sustained by the ability of activated platelets to interact with each other (homotypic aggregation) and to adhere to circulating leucocytes (heterotypic aggregation). These two events are regulated by distinct biomolecular mechanisms that are selectively activated in different pathophysiological settings. They can occur simultaneously, for example, as part of a pro-thrombotic/pro-inflammatory response induced by vascular damage, or independently, as in certain clinical conditions in which abnormal heterotypic aggregation has been observed in the absence of intravascular thrombosis. Current antiplatelet drugs have been developed to target specific molecular signalling pathways mainly implicated in thrombus formation, and their ever increasing clinical use has resulted in clear benefits in the treatment and prevention of arterial thrombotic events. However, the efficacy of currently available antiplatelet drugs remains suboptimal, most likely because their therapeutic action is limited to only few of the signalling pathways involved in platelet homotypic aggregation. In this context, modulation of heterotypic aggregation, which is believed to contribute importantly to acute thrombotic events, as well to the pathophysiology of atherosclerosis itself, may offer benefits over and above the classical antiplatelet approach. This review will focus on the distinct biomolecular pathways that, following platelet activation, underlie homotypic and heterotypic aggregation, aiming potentially to identify novel therapeutic targets.
Collapse
Affiliation(s)
- Gabriella Passacquale
- Department of Clinical Pharmacology, Cardiovascular Division, King's College London, London, UK
| | | |
Collapse
|