51
|
Haworth JA, Mears RG, Jenkinson HF, Kerrigan SW, Nobbs AH. Oral hygiene as a risk factor in infective endocarditis. ACTA ACUST UNITED AC 2017. [DOI: 10.12968/denu.2017.44.9.877] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Jennifer A Haworth
- Academic Clinical Lecturer, Bristol Dental School, University of Bristol, Lower Maudlin Street, BS1 2LY, Bristol, UK
| | - Richard G Mears
- General Dental Practitioner, Combe Road Dental Practice, 6 Combe Road, Portishead, BS20 6BJ and Clinical Teaching Fellow, Restorative Dentistry, Bristol Dental School, University of Bristol, Lower Maudlin Street, BS1 2LY, Bristol, UK
| | - Howard F Jenkinson
- Professor of Oral Microbiology, Bristol Dental School, University of Bristol, Lower Maudlin Street, BS1 2LY, Bristol, UK
| | - Steve W Kerrigan
- Senior Lecturer in Pharmacology, Royal College of Surgeons in Ireland, 123 St Stephen's Green, Dublin 2, Ireland
| | - Angela H Nobbs
- Senior Lecturer in Oral Microbiology, Bristol Dental School, University of Bristol, Lower Maudlin Street, BS1 2LY, Bristol, UK
| |
Collapse
|
52
|
Force-activatable biosensor enables single platelet force mapping directly by fluorescence imaging. Biosens Bioelectron 2017; 100:192-200. [PMID: 28915383 DOI: 10.1016/j.bios.2017.09.007] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 08/25/2017] [Accepted: 09/05/2017] [Indexed: 11/23/2022]
Abstract
Integrin-transmitted cellular forces are critical for platelet adhesion, activation, aggregation and contraction during hemostasis and thrombosis. Measuring and mapping single platelet forces are desired in both research and clinical applications. Conventional force-to-strain based cell traction force microscopies have low resolution which is not ideal for cellular force mapping in small platelets. To enable platelet force mapping with submicron resolution, we developed a force-activatable biosensor named integrative tension sensor (ITS) which directly converts molecular tensions to fluorescent signals, therefore enabling cellular force mapping directly by fluorescence imaging. With ITS, we mapped cellular forces in single platelets at 0.4µm resolution. We found that platelet force distribution has strong polarization which is sensitive to treatment with the anti-platelet drug tirofiban, suggesting that the ITS force map can report anti-platelet drug efficacy. The ITS also calibrated integrin molecular tensions in platelets and revealed two distinct tension levels: 12-54 piconewton (nominal values) tensions generated during platelet adhesion and tensions above 54 piconewton generated during platelet contraction. Overall, the ITS is a powerful biosensor for the study of platelet mechanobiology, and holds great potential in antithrombotic drug development and assessing platelet activity in health and disease.
Collapse
|
53
|
Dissecting intrinsic and ligand-induced structural communication in the β3 headpiece of integrins. Biochim Biophys Acta Gen Subj 2017; 1861:2367-2381. [DOI: 10.1016/j.bbagen.2017.05.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 05/20/2017] [Accepted: 05/22/2017] [Indexed: 12/15/2022]
|
54
|
Abstract
Integrin αIIbβ3 is a highly abundant heterodimeric platelet receptor that can transmit information bidirectionally across the plasma membrane, and plays a critical role in hemostasis and thrombosis. Upon platelet activation, inside-out signaling pathways increase the affinity of αIIbβ3 for fibrinogen and other ligands. Ligand binding and integrin clustering subsequently stimulate outside-in signaling, which initiates and amplifies a range of cellular events driving essential platelet processes such as spreading, thrombus consolidation, and clot retraction. Integrin αIIbβ3 has served as an excellent model for the study of integrin biology, and it has become clear that integrin outside-in signaling is highly complex and involves a vast array of enzymes, signaling adaptors, and cytoskeletal components. In this review, we provide a concise but comprehensive overview of αIIbβ3 outside-in signaling, focusing on the key players involved, and how they cooperate to orchestrate this critical aspect of platelet biology. We also discuss gaps in the current understanding of αIIbβ3 outside-in signaling and highlight avenues for future investigation.
Collapse
|
55
|
Grönman M, Tarkia M, Kiviniemi T, Halonen P, Kuivanen A, Savunen T, Tolvanen T, Teuho J, Käkelä M, Metsälä O, Pietilä M, Saukko P, Ylä-Herttuala S, Knuuti J, Roivainen A, Saraste A. Imaging of α vβ 3 integrin expression in experimental myocardial ischemia with [ 68Ga]NODAGA-RGD positron emission tomography. J Transl Med 2017. [PMID: 28629432 PMCID: PMC5477135 DOI: 10.1186/s12967-017-1245-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Background Radiolabeled RGD peptides detect αvβ3 integrin expression associated with angiogenesis and extracellular matrix remodeling after myocardial infarction. We studied whether cardiac positron emission tomography (PET) with [68Ga]NODAGA-RGD detects increased αvβ3 integrin expression after induction of flow-limiting coronary stenosis in pigs, and whether αvβ3 integrin is expressed in viable ischemic or injured myocardium. Methods We studied 8 Finnish landrace pigs 13 ± 4 days after percutaneous implantation of a bottleneck stent in the proximal left anterior descending coronary artery. Antithrombotic therapy was used to prevent stent occlusion. Myocardial uptake of [68Ga]NODAGA-RGD (290 ± 31 MBq) was evaluated by a 62 min dynamic PET scan. The ischemic area was defined as the regional perfusion abnormality during adenosine-induced stress by [15O]water PET. Guided by triphenyltetrazolium chloride staining, tissue samples from viable and injured myocardial areas were obtained for autoradiography and histology. Results Stent implantation resulted in a partly reversible myocardial perfusion abnormality. Compared with remote myocardium, [68Ga]NODAGA-RGD PET showed increased tracer uptake in the ischemic area (ischemic-to-remote ratio 1.3 ± 0.20, p = 0.0034). Tissue samples from the injured areas, but not from the viable ischemic areas, showed higher [68Ga]NODAGA-RGD uptake than the remote non-ischemic myocardium. Uptake of [68Ga]NODAGA-RGD correlated with immunohistochemical detection of αvβ3 integrin that was expressed in the injured myocardial areas. Conclusions Cardiac [68Ga]NODAGA-RGD PET demonstrates increased myocardial αvβ3 integrin expression after induction of flow-limiting coronary stenosis in pigs. Localization of [68Ga]NODAGA-RGD uptake indicates that it reflects αvβ3 integrin expression associated with repair of recent myocardial injury.
Collapse
Affiliation(s)
- Maria Grönman
- Turku PET Centre, University of Turku, 20521, Turku, Finland
| | - Miikka Tarkia
- Turku PET Centre, University of Turku, 20521, Turku, Finland
| | | | - Paavo Halonen
- A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Joensuu, Finland
| | - Antti Kuivanen
- A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Joensuu, Finland
| | - Timo Savunen
- Heart Center, Turku University Hospital, Turku, Finland.,Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
| | - Tuula Tolvanen
- Turku PET Centre, Turku University Hospital, Turku, Finland.,Department of Medical Physics, Turku University Hospital and University of Turku, Turku, Finland
| | - Jarmo Teuho
- Turku PET Centre, Turku University Hospital, Turku, Finland.,Department of Medical Physics, Turku University Hospital and University of Turku, Turku, Finland
| | - Meeri Käkelä
- Turku PET Centre, University of Turku, 20521, Turku, Finland
| | - Olli Metsälä
- Turku PET Centre, University of Turku, 20521, Turku, Finland
| | - Mikko Pietilä
- Heart Center, Turku University Hospital, Turku, Finland
| | - Pekka Saukko
- Department of Forensic Medicine, University of Turku, Turku, Finland
| | - Seppo Ylä-Herttuala
- A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Joensuu, Finland
| | - Juhani Knuuti
- Turku PET Centre, University of Turku, 20521, Turku, Finland.,Turku PET Centre, Turku University Hospital, Turku, Finland
| | - Anne Roivainen
- Turku PET Centre, University of Turku, 20521, Turku, Finland.,Turku PET Centre, Turku University Hospital, Turku, Finland.,Turku Center for Disease Modeling, University of Turku, Turku, Finland
| | - Antti Saraste
- Turku PET Centre, University of Turku, 20521, Turku, Finland. .,Heart Center, Turku University Hospital, Turku, Finland. .,Turku PET Centre, Turku University Hospital, Turku, Finland. .,Institute of Clinical Medicine, University of Turku, Turku, Finland.
| |
Collapse
|
56
|
Abstract
Virus–platelet interplay is complex. Diverse virus types have been shown to associate with numerous distinct platelet receptors. This association can benefit the virus or the host, and thus the platelet is somewhat of a renegade. Evidence is accumulating to suggest that viruses are capable of entering platelets. For at least one type of RNA virus (dengue virus), the platelet has the necessary post-translational and packaging machinery required for production of replicative viral progeny. As a facilitator of immunity, the platelet also participates in eradicating the virus by direct and indirect mechanisms involving presentation of the pathogen to the innate and adaptive immune systems, thus enhancing inflammation by release of cytokines and other agonists. Virus-induced thrombocytopenia is caused by tangential imbalance of thrombopoeisis, autoimmunity, and loss of platelet function and integrity.
Collapse
|
57
|
Meikle CKS, Kelly CA, Garg P, Wuescher LM, Ali RA, Worth RG. Cancer and Thrombosis: The Platelet Perspective. Front Cell Dev Biol 2017; 4:147. [PMID: 28105409 PMCID: PMC5214375 DOI: 10.3389/fcell.2016.00147] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 12/12/2016] [Indexed: 01/03/2023] Open
Abstract
Platelets are critical to hemostatic and immunological function, and are key players in cancer progression, metastasis, and cancer-related thrombosis. Platelets interact with immune cells to stimulate anti-tumor responses and can be activated by immune cells and tumor cells. Platelet activation can lead to complex interactions between platelets and tumor cells. Platelets facilitate cancer progression and metastasis by: (1) forming aggregates with tumor cells; (2) inducing tumor growth, epithelial-mesenchymal transition, and invasion; (3) shielding circulating tumor cells from immune surveillance and killing; (4) facilitating tethering and arrest of circulating tumor cells; and (5) promoting angiogenesis and tumor cell establishment at distant sites. Tumor cell-activated platelets also predispose cancer patients to thrombotic events. Tumor cells and tumor-derived microparticles lead to thrombosis by secreting procoagulant factors, resulting in platelet activation and clotting. Platelets play a critical role in cancer progression and thrombosis, and markers of platelet-tumor cell interaction are candidates as biomarkers for cancer progression and thrombosis risk.
Collapse
Affiliation(s)
- Claire K S Meikle
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences Toledo, OH, USA
| | - Clare A Kelly
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences Toledo, OH, USA
| | - Priyanka Garg
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences Toledo, OH, USA
| | - Leah M Wuescher
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences Toledo, OH, USA
| | - Ramadan A Ali
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences Toledo, OH, USA
| | - Randall G Worth
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences Toledo, OH, USA
| |
Collapse
|
58
|
Taylor KA, Pugh N. The contribution of zinc to platelet behaviour during haemostasis and thrombosis. Metallomics 2016; 8:144-55. [PMID: 26727074 DOI: 10.1039/c5mt00251f] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Platelets are the primary cellular determinants of haemostasis and pathological thrombus formation leading to myocardial infarction and stroke. Following vascular injury or atherosclerotic plaque rupture, platelets are recruited to sites of damage and undergo activation induced by a variety of soluble and/or insoluble agonists. Platelet activation is a multi-step process culminating in the formation of thrombi, which contribute to the haemostatic process. Zinc (Zn(2+)) is acknowledged as an important signalling molecule in a diverse range of cellular systems, however there is limited understanding of the influence of Zn(2+) on platelet behaviour during thrombus formation. This review evaluates the contributions of exogenous and intracellular Zn(2+) to platelet function and assesses the potential pathophysiological implications of Zn(2+) signalling. We also provide a speculative assessment of the mechanisms by which platelets could respond to changes in extracellular and intracellular Zn(2+) concentration.
Collapse
Affiliation(s)
- K A Taylor
- Department of Biomedical and Forensic Sciences, Faculty of Science and Technology, Anglia Ruskin University, Cambridge, CB1 1PT, UK.
| | - N Pugh
- Department of Biomedical and Forensic Sciences, Faculty of Science and Technology, Anglia Ruskin University, Cambridge, CB1 1PT, UK.
| |
Collapse
|
59
|
Fan Z, Ley K. Leukocyte arrest: Biomechanics and molecular mechanisms of β2 integrin activation. Biorheology 2016; 52:353-77. [PMID: 26684674 DOI: 10.3233/bir-15085] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Integrins are a group of heterodimeric transmembrane receptors that play essential roles in cell-cell and cell-matrix interaction. Integrins are important in many physiological processes and diseases. Integrins acquire affinity to their ligand by undergoing molecular conformational changes called activation. Here we review the molecular biomechanics during conformational changes of integrins, integrin functions in leukocyte biorheology (adhesive functions during rolling and arrest) and molecules involved in integrin activation.
Collapse
Affiliation(s)
- Zhichao Fan
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA
| | - Klaus Ley
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA.,Department of Bioengineering, University of California San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
60
|
Zeller Meidell K, Robinson R, Vieira-de-Abreu A, Gormley AJ, Ghandehari H, W Grainger D, A Campbell R. RGDfK-functionalized gold nanorods bind only to activated platelets. J Biomed Mater Res A 2016; 105:209-217. [PMID: 27648522 DOI: 10.1002/jbm.a.35902] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 08/15/2016] [Accepted: 09/14/2016] [Indexed: 12/12/2022]
Abstract
Integrin-targeting peptide RGDfK-labeled gold nanorods (GNR) seek to improve hyperthermia targeted to solid tumors by exploiting the known up-regulation of integrin αvβ3 cell membrane proteins on solid tumor vasculature surfaces. Tumor binding specificity might be expected since surrounding tissues and endothelial cells have limited numbers of these receptors. However, RGD peptide binding to many proteins is promiscuous, with known affinity to several families of cell integrin receptors, and also possible binding to platelets after intravenous infusion via a different integrin receptor, αIIbβ3, on platelets. Binding of RGDfK-targeted GNR could considerably impact platelet function, ultimately leading to increased risk of bleeding or thrombosis depending on the degree of interaction. We sought to determine if RGDfK-labeled GNR could interact with platelets and alter platelet function. Targeted and untargeted nanorods exhibited little interaction with resting platelets in platelet rich plasma (PRP) preparations. However, upon platelet activation, peptide-targeted nanorods bound actively to platelets. Addition of RGDfK-GNR to unactivated platelets had little effect on markers of platelet activation, indicating that RGDfK-nanorods were incapable of inducing platelet activation. We next tested whether activated platelet function was altered in the presence of peptide-targeted nanorods. Platelet aggregation in whole blood and PRP in the presence of targeted nanorods had no significant effect on platelet aggregation. These data suggest that RGDfK-GNR alone have little impact on platelet function in plasma. However, nonspecific nanorod binding may occur in vascular beds where activated platelets are normally cleared, such as the spleen and liver, producing a possible toxicity risk for these nanomaterials. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 105A: 209-217, 2017.
Collapse
Affiliation(s)
- Krystin Zeller Meidell
- Department of Pharmaceutics and Pharmaceutical Chemistry, Health Sciences, University of Utah, Salt Lake City, Utah, 84112
| | - Ryan Robinson
- Department of Bioengineering, University of Utah, Salt Lake City, Utah, 84112.,Center for Nanomedicine, Nano Institute of Utah, University of Utah, Salt Lake City, Utah, 84112
| | - Adriana Vieira-de-Abreu
- Program in Molecular Medicine, School of Medicine, University of Utah, Salt Lake City, Utah, 84112
| | - Adam J Gormley
- Department of Bioengineering, University of Utah, Salt Lake City, Utah, 84112.,Center for Nanomedicine, Nano Institute of Utah, University of Utah, Salt Lake City, Utah, 84112
| | - Hamidreza Ghandehari
- Department of Pharmaceutics and Pharmaceutical Chemistry, Health Sciences, University of Utah, Salt Lake City, Utah, 84112.,Department of Bioengineering, University of Utah, Salt Lake City, Utah, 84112.,Center for Nanomedicine, Nano Institute of Utah, University of Utah, Salt Lake City, Utah, 84112
| | - David W Grainger
- Department of Pharmaceutics and Pharmaceutical Chemistry, Health Sciences, University of Utah, Salt Lake City, Utah, 84112.,Department of Bioengineering, University of Utah, Salt Lake City, Utah, 84112.,Center for Nanomedicine, Nano Institute of Utah, University of Utah, Salt Lake City, Utah, 84112
| | - Robert A Campbell
- Program in Molecular Medicine, School of Medicine, University of Utah, Salt Lake City, Utah, 84112
| |
Collapse
|
61
|
Benz PM, Laban H, Zink J, Günther L, Walter U, Gambaryan S, Dib K. Vasodilator-Stimulated Phosphoprotein (VASP)-dependent and -independent pathways regulate thrombin-induced activation of Rap1b in platelets. Cell Commun Signal 2016; 14:21. [PMID: 27620165 PMCID: PMC5020514 DOI: 10.1186/s12964-016-0144-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 09/06/2016] [Indexed: 11/13/2022] Open
Abstract
Background Vasodilator-Stimulated Phosphoprotein (VASP) is involved in the inhibition of agonist-induced platelet aggregation by cyclic nucleotides and the adhesion of platelets to the vascular wall. αIIbβ3 is the main integrin responsible for platelet activation and Rap1b plays a key role in integrin signalling. We investigated whether VASP is involved in the regulation of Rap1b in platelets since VASP-null platelets exhibit augmented adhesion to endothelial cells in vivo. Methods Washed platelets from wild type and VASP-deficient mice were stimulated with thrombin, the purinergic receptors agonist ADP, or the thromboxane A2 receptor agonist U46619 and Rap1b activation was measured using the GST-RalGDS-RBD binding assay. Interaction of VASP and Crkl was investigated by co-immunoprecipitation, confocal microscopy, and pull-down assays using Crkl domains expressed as GST-fusion proteins. Results Surprisingly, we found that activation of Rap1b in response to thrombin, ADP, or U46619 was significantly reduced in platelets from VASP-null mice compared to platelets from wild type mice. However, inhibition of thrombin-induced activation of Rap1b by nitric oxide (NO) was similar in platelets from wild type and VASP-null mice indicating that the NO/cGMP/PKG pathway controls inhibition of Rap1b independently from VASP. To understand how VASP regulated Rap1b, we investigated association between VASP and the Crk-like protein (Crkl), an adapter protein which activates the Rap1b guanine nucleotide exchange factor C3G. We demonstrated the formation of a Crkl/VASP complex by showing that: 1) Crkl co-immunoprecipitated VASP from platelet lysates; 2) Crkl and VASP dynamically co-localized at actin-rich protrusions reminiscent of focal adhesions, filopodia, and lamellipodia upon platelet spreading on fibronectin; 3) recombinant VASP bound directly to the N-terminal SH3 domain of Crkl; 4) Protein Kinase A (PKA) -mediated VASP phosphorylation on Ser157 abrogated the binding of Crkl. Conclusions We identified Crkl as a novel protein interacting with VASP in platelets. We propose that the C3G/Crkl/VASP complex plays a role in the regulation of Rap1b and this explains, at least in part, the reduced agonist-induced activation of Rap1b in VASP-null platelets. In addition, the fact that PKA-dependent VASP phosphorylation abrogated its interaction with Crkl may provide, at least in part, a rationale for the PKA-dependent inhibition of Rap1b and platelet aggregation.
Collapse
Affiliation(s)
- Peter M Benz
- Institute for Vascular Signalling, Centre for Molecular Medicine, Johann Wolfgang Goethe University and DZHK (German Centre for Cardiovascular Research) partner site Rhine-Main, 60590, Frankfurt, Germany
| | - Hebatullah Laban
- Institute for Vascular Signalling, Centre for Molecular Medicine, Johann Wolfgang Goethe University and DZHK (German Centre for Cardiovascular Research) partner site Rhine-Main, 60590, Frankfurt, Germany
| | - Joana Zink
- Institute for Vascular Signalling, Centre for Molecular Medicine, Johann Wolfgang Goethe University and DZHK (German Centre for Cardiovascular Research) partner site Rhine-Main, 60590, Frankfurt, Germany
| | - Lea Günther
- Institute for Vascular Signalling, Centre for Molecular Medicine, Johann Wolfgang Goethe University and DZHK (German Centre for Cardiovascular Research) partner site Rhine-Main, 60590, Frankfurt, Germany
| | - Ulrich Walter
- Centre for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Mainz, Germany
| | - Stepan Gambaryan
- Department of Cytology and Histology, St. Petersburg State University, St. Petersburg, Russia.,Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St. Petersburg, Russia
| | - Karim Dib
- Centre for Experimental Medicine, Medical Biology Center (MBC) building, Queen's University of Belfast, Third floor, 97 Lisburn Road, BT9 7BL, Belfast, Northern Ireland, UK.
| |
Collapse
|
62
|
Plaza JQ, Garzón LB, Gimenez BB, Moraleda BFM, Collía F, Rodríguez-Lorenzo LM. Application of calcium phosphates and fibronectin as complementary treatment for osteoporotic bone fractures. Injury 2016; 47 Suppl 3:S15-S21. [PMID: 27692101 DOI: 10.1016/s0020-1383(16)30601-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
INTRODUCTION The gradual aging of the population results in increased incidence of osteoporotic bone fractures. In a good quality bone, the fixation with the usual methods is adequate, but not in osteoporotic bone, in which consolidation delays and other complications are common, with failure rates for screws up to 25%. OBJECTIVE To test fibronectin loaded hydroxyapatite as a complementary treatment for osteoporotic fractures. MATERIAL AND METHODS This study was performed in a vivo model; 42 female osteoporotic adult rabbits 4-5kg (White New Zealand) were used. Two groups (hydroxyapatite and fibronectin loaded hydroxyapatite) and a control group were tested. 3 time points 24h, 48h and 5days were studied. Defects were created in both femurs, in one of them, a cannulated screw (4mm) and a biocompatible material were placed; in the other femur a screw was inserted without supplemented material forming the control group. Osteoporosis was induced from models already known throughout administration of steroids. Samples were analyzed histologically and through imaging (micro Ct). RESULTS Basal levels of BMD are observed below to normal when compared to other studies (0.25/0.3 instead of 0.4). Global and dependent of time analysis of samples, show no significant differences for samples analyzed. However, an important trend was noted for variables that define the trabecular bone microarchitecture. Indices that define trabecular microarchitecture in the comparative analysis found to have statistical differences (p<0.01). DISCUSSION Osteosynthesis in an osteoporotic bone is a challenge for the surgeon, due to a reduced bone mineral density and different bone architecture. The main finding was the verification of the hypothesis that the trabecular bone parameters increases with our augmentation material in weak rabbit bone quality. Also, the histological analyses of samples show an increase of non inflammatory cells in protein samples (OHAp-Fn) from the first 24hours. CONCLUSION An early response of rabbit osteroporotic bone to a complementary treatment with fibronectin loaded hydroxyapatite has been observed. This response is reflected in greater values for indices that define the trabecular bone microarchitecture, thickness and separation, a greater non-inflammatory cellularity after only 24hours and an increased amount of connective tissue observed at 48hours.
Collapse
Affiliation(s)
| | | | | | | | - Francisco Collía
- Facultad de Medicina, Universidad de Salamanca, Salamanca, Spain
| | - Luis M Rodríguez-Lorenzo
- Biomaterials group, ICTP-CSIC, Madrid, Spain; Polymeric Biomaterials Group, CIBER-BBN, Madrid, Spain
| |
Collapse
|
63
|
Yala S, Boustta M, Gallet O, Hindié M, Carreiras F, Benachour H, Sidane D, Khireddine H. New synthesis method of HA/P(D,L)LA composites: study of fibronectin adsorption and their effects in osteoblastic behavior for bone tissue engineering. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2016; 27:140. [PMID: 27534400 DOI: 10.1007/s10856-016-5756-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 07/09/2016] [Indexed: 06/06/2023]
Abstract
A novel synthetic method to synthesize hydroxyapatite/poly (D,L) lactic acid biocomposite is presented in this study by mixing only the precursors hydroxyapatite and (D,L) LA monomer without adding neither solvent nor catalyst. Three compositions were successfully synthesized with the weight ratios of 1/1, 1/3, and 3/5 (hydroxyapatite/(D,L) lactic acid), and the grafting efficiency of poly (D,L) lactic acid on hydroxyapatite surface reaches up to 84 %. Scanning electron microscopy and Fourier transform infrared spectroscopy showed that the hydroxyapatite particles were successfully incorporated into the poly (D,L) lactic acid polymer and X ray diffraction analysis showed that hydroxyapatite preserved its crystallinity after poly (D,L) lactic acid grafting. Differential scanning calorimetry shows that Tg of hydroxyapatite/poly (D,L) lactic acid composite is less than Tg of pure poly (D,L) lactic acid, which facilitates the shaping of the composite obtained. The addition of poly (D,L) lactic acid improves the adsorption properties of hydroxyapatite for fibronectin extracellular matrix protein. Furthermore, the presence of poly (D,L) lactic acid on hydroxyapatite surface coated with fibronectin enhanced pre-osteoblast STRO-1 adhesion and cell spreading. These results show the promising potential of hydroxyapatite/poly (D,L) lactic acid composite as a bone substitute material for orthopedic applications and bone tissue engineering.
Collapse
Affiliation(s)
- Sabeha Yala
- Laboratoire de Génie de l'Environnement, Faculté de Technologie, Université de Bejaia, Bejaia, 06000, Algeria.
| | - Mahfoud Boustta
- Institut des Biomolécules Max Mousseron, UMR CNRS 5247, CRBA, Faculté des Sciences, Université de Montpellier, 15 avenue Charles Flahault, BP 14491, Montpellier cedex 5, 34093, France
| | - Olivier Gallet
- ERRMECe, Université de Cergy-Pontoise, Site Saint-Martin, 2 Avenue Adolphe Chauvin, Cergy-Pontoise Cedex, 95302, France
| | - Mathilde Hindié
- ERRMECe, Université de Cergy-Pontoise, Site Saint-Martin, 2 Avenue Adolphe Chauvin, Cergy-Pontoise Cedex, 95302, France
| | - Franck Carreiras
- ERRMECe, Université de Cergy-Pontoise, Site Saint-Martin, 2 Avenue Adolphe Chauvin, Cergy-Pontoise Cedex, 95302, France
| | - Hamanou Benachour
- ERRMECe, Université de Cergy-Pontoise, Site Saint-Martin, 2 Avenue Adolphe Chauvin, Cergy-Pontoise Cedex, 95302, France
| | - Djahida Sidane
- Laboratoire de Génie de l'Environnement, Faculté de Technologie, Université de Bejaia, Bejaia, 06000, Algeria
| | - Hafit Khireddine
- Laboratoire de Génie de l'Environnement, Faculté de Technologie, Université de Bejaia, Bejaia, 06000, Algeria
| |
Collapse
|
64
|
Fernández-Montes Moraleda B, San Román J, Rodríguez-Lorenzo LM. Adsorption and conformational modification of fibronectin and fibrinogen adsorbed on hydroxyapatite. A QCM-D study. J Biomed Mater Res A 2016; 104:2585-94. [PMID: 27254464 DOI: 10.1002/jbm.a.35802] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Revised: 03/11/2016] [Accepted: 03/16/2016] [Indexed: 01/06/2023]
Abstract
Hydroxyapatite is a bioactive ceramic frequently used for bone engineering/replacement. One of the parameters that influence the biological response to implanted materials is the conformation of the first adsorbed protein layer. In this work, the adsorption and conformational changes of two fibroid serum proteins; fibronectin and fibrinogen adsorbed onto four different hydroxyapatite powders are studied with a Quartz Crystal Microbalance with Dissipation (QCM-D). Each of the calcined apatites adsorbs less protein than their corresponding synthesized samples. Adsorption on synthesized samples yields always an extended conformation whereas a reorganization of the layer is observed for the calcined samples. Fg acquires a "Side on" conformation in all the samples at the beginning of the experiment except for one of the synthesized samples where an "End-on" conformation is obtained during the whole experiment. The Extended conformation is the active conformation for Fn. This conformation is favored by apatites with large specific surface area (SSA) and on highly concentrated media. Apatite surface features should be considered in the selection or design of materials for bone regeneration, since it is possible to control the conformation mode of attachment of Fn and Fg by an appropriate selection of them. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 104A: 2585-2594, 2016.
Collapse
Affiliation(s)
- Belén Fernández-Montes Moraleda
- Biomaterials Group, ICTP-CSIC, Juan De La Cierva, 3, Madrid, 28006, Spain.,Networking Biomedical Research Centre in Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN, Zaragoza, Spain
| | - Julio San Román
- Biomaterials Group, ICTP-CSIC, Juan De La Cierva, 3, Madrid, 28006, Spain.,Networking Biomedical Research Centre in Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN, Zaragoza, Spain
| | - Luís M Rodríguez-Lorenzo
- Biomaterials Group, ICTP-CSIC, Juan De La Cierva, 3, Madrid, 28006, Spain.,Networking Biomedical Research Centre in Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN, Zaragoza, Spain
| |
Collapse
|
65
|
Santoso S, Wihadmadyatami H, Bakchoul T, Werth S, Al-Fakhri N, Bein G, Kiefel V, Zhu J, Newman PJ, Bayat B, Sachs UJ. Antiendothelial αvβ3 Antibodies Are a Major Cause of Intracranial Bleeding in Fetal/Neonatal Alloimmune Thrombocytopenia. Arterioscler Thromb Vasc Biol 2016; 36:1517-24. [PMID: 27283740 DOI: 10.1161/atvbaha.116.307281] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 06/01/2016] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Fetal/neonatal alloimmune thrombocytopenia is a severe bleeding disorder, which can result in intracranial hemorrhage (ICH), leading to death or neurological sequelae. In whites, maternal anti-human platelet antigen-1a (HPA-1a) antibodies are responsible for the majority of cases. No predictive factors for ICH are available to guide prophylactic treatment during pregnancy. In this study, we investigated antibodies from mothers with ICH-positive fetal/neonatal alloimmune thrombocytopenia and with ICH-negative fetal/neonatal alloimmune thrombocytopenia to identify serological and functional differences between the groups. APPROACH AND RESULTS In an antigen capture assay, we observed a stronger binding of +ICH antibodies to endothelial cell (EC)-derived αvβ3. By absorption experiments, we subsequently identified anti-HPA-1a antibodies of anti-αvβ3 specificity in the +ICH but not in the -ICH cohort. Only the anti-αvβ3 subtype, but not the anti-β3 subtype, induced EC apoptosis of HPA-1a-positive ECs by caspase-3/7 activation, and mediated by reactive oxygen species. In addition, only the anti-αvβ3 subtype, but not the anti-β3 subtype, interfered with EC adhesion to vitronectin and with EC tube formation. CONCLUSIONS We conclude that the composition of the anti-HPA-1a antibody subtype(s) of the mother may determine whether ICH occurs. Analysis of anti-HPA-1a antibodies of the anti-αvβ3 subtype in maternal serum has potential in the diagnostic prediction of ICH development and may allow for modification of prophylactic treatment in fetal/neonatal alloimmune thrombocytopenia.
Collapse
Affiliation(s)
- Sentot Santoso
- From the Institute for Clinical Immunology and Transfusion Medicine, Justus Liebig University, Giessen, Germany (S.S., H.W., S.W., G.B., B.B., U.J.S.); Department of Anatomy, Faculty of Veterinary Medicine, Universitas Gadjah Mada, Yogyakarta, Indonesia (H.W.); Institute for Immunology and Transfusion Medicine, Ernst-Moritz-Arndt University, Greifswald, Germany (T.B.); Center for Transfusion Medicine and Hemotherapy (N.A.-F., U.J.S.) and German Center for Fetomaternal Incompatibility (DZFI) (G.B., U.J.S.), University Hospital Giessen and Marburg, Giessen, Germany; Institute for Transfusion Medicine, University of Rostock, Rostock, Germany (V.K.); and Blood Research Institute, BloodCenter of Wisconsin, Milwaukee (J.Z., P.J.N.).
| | - Hevi Wihadmadyatami
- From the Institute for Clinical Immunology and Transfusion Medicine, Justus Liebig University, Giessen, Germany (S.S., H.W., S.W., G.B., B.B., U.J.S.); Department of Anatomy, Faculty of Veterinary Medicine, Universitas Gadjah Mada, Yogyakarta, Indonesia (H.W.); Institute for Immunology and Transfusion Medicine, Ernst-Moritz-Arndt University, Greifswald, Germany (T.B.); Center for Transfusion Medicine and Hemotherapy (N.A.-F., U.J.S.) and German Center for Fetomaternal Incompatibility (DZFI) (G.B., U.J.S.), University Hospital Giessen and Marburg, Giessen, Germany; Institute for Transfusion Medicine, University of Rostock, Rostock, Germany (V.K.); and Blood Research Institute, BloodCenter of Wisconsin, Milwaukee (J.Z., P.J.N.)
| | - Tamam Bakchoul
- From the Institute for Clinical Immunology and Transfusion Medicine, Justus Liebig University, Giessen, Germany (S.S., H.W., S.W., G.B., B.B., U.J.S.); Department of Anatomy, Faculty of Veterinary Medicine, Universitas Gadjah Mada, Yogyakarta, Indonesia (H.W.); Institute for Immunology and Transfusion Medicine, Ernst-Moritz-Arndt University, Greifswald, Germany (T.B.); Center for Transfusion Medicine and Hemotherapy (N.A.-F., U.J.S.) and German Center for Fetomaternal Incompatibility (DZFI) (G.B., U.J.S.), University Hospital Giessen and Marburg, Giessen, Germany; Institute for Transfusion Medicine, University of Rostock, Rostock, Germany (V.K.); and Blood Research Institute, BloodCenter of Wisconsin, Milwaukee (J.Z., P.J.N.)
| | - Silke Werth
- From the Institute for Clinical Immunology and Transfusion Medicine, Justus Liebig University, Giessen, Germany (S.S., H.W., S.W., G.B., B.B., U.J.S.); Department of Anatomy, Faculty of Veterinary Medicine, Universitas Gadjah Mada, Yogyakarta, Indonesia (H.W.); Institute for Immunology and Transfusion Medicine, Ernst-Moritz-Arndt University, Greifswald, Germany (T.B.); Center for Transfusion Medicine and Hemotherapy (N.A.-F., U.J.S.) and German Center for Fetomaternal Incompatibility (DZFI) (G.B., U.J.S.), University Hospital Giessen and Marburg, Giessen, Germany; Institute for Transfusion Medicine, University of Rostock, Rostock, Germany (V.K.); and Blood Research Institute, BloodCenter of Wisconsin, Milwaukee (J.Z., P.J.N.)
| | - Nadia Al-Fakhri
- From the Institute for Clinical Immunology and Transfusion Medicine, Justus Liebig University, Giessen, Germany (S.S., H.W., S.W., G.B., B.B., U.J.S.); Department of Anatomy, Faculty of Veterinary Medicine, Universitas Gadjah Mada, Yogyakarta, Indonesia (H.W.); Institute for Immunology and Transfusion Medicine, Ernst-Moritz-Arndt University, Greifswald, Germany (T.B.); Center for Transfusion Medicine and Hemotherapy (N.A.-F., U.J.S.) and German Center for Fetomaternal Incompatibility (DZFI) (G.B., U.J.S.), University Hospital Giessen and Marburg, Giessen, Germany; Institute for Transfusion Medicine, University of Rostock, Rostock, Germany (V.K.); and Blood Research Institute, BloodCenter of Wisconsin, Milwaukee (J.Z., P.J.N.)
| | - Gregor Bein
- From the Institute for Clinical Immunology and Transfusion Medicine, Justus Liebig University, Giessen, Germany (S.S., H.W., S.W., G.B., B.B., U.J.S.); Department of Anatomy, Faculty of Veterinary Medicine, Universitas Gadjah Mada, Yogyakarta, Indonesia (H.W.); Institute for Immunology and Transfusion Medicine, Ernst-Moritz-Arndt University, Greifswald, Germany (T.B.); Center for Transfusion Medicine and Hemotherapy (N.A.-F., U.J.S.) and German Center for Fetomaternal Incompatibility (DZFI) (G.B., U.J.S.), University Hospital Giessen and Marburg, Giessen, Germany; Institute for Transfusion Medicine, University of Rostock, Rostock, Germany (V.K.); and Blood Research Institute, BloodCenter of Wisconsin, Milwaukee (J.Z., P.J.N.)
| | - Volker Kiefel
- From the Institute for Clinical Immunology and Transfusion Medicine, Justus Liebig University, Giessen, Germany (S.S., H.W., S.W., G.B., B.B., U.J.S.); Department of Anatomy, Faculty of Veterinary Medicine, Universitas Gadjah Mada, Yogyakarta, Indonesia (H.W.); Institute for Immunology and Transfusion Medicine, Ernst-Moritz-Arndt University, Greifswald, Germany (T.B.); Center for Transfusion Medicine and Hemotherapy (N.A.-F., U.J.S.) and German Center for Fetomaternal Incompatibility (DZFI) (G.B., U.J.S.), University Hospital Giessen and Marburg, Giessen, Germany; Institute for Transfusion Medicine, University of Rostock, Rostock, Germany (V.K.); and Blood Research Institute, BloodCenter of Wisconsin, Milwaukee (J.Z., P.J.N.)
| | - Jieqing Zhu
- From the Institute for Clinical Immunology and Transfusion Medicine, Justus Liebig University, Giessen, Germany (S.S., H.W., S.W., G.B., B.B., U.J.S.); Department of Anatomy, Faculty of Veterinary Medicine, Universitas Gadjah Mada, Yogyakarta, Indonesia (H.W.); Institute for Immunology and Transfusion Medicine, Ernst-Moritz-Arndt University, Greifswald, Germany (T.B.); Center for Transfusion Medicine and Hemotherapy (N.A.-F., U.J.S.) and German Center for Fetomaternal Incompatibility (DZFI) (G.B., U.J.S.), University Hospital Giessen and Marburg, Giessen, Germany; Institute for Transfusion Medicine, University of Rostock, Rostock, Germany (V.K.); and Blood Research Institute, BloodCenter of Wisconsin, Milwaukee (J.Z., P.J.N.)
| | - Peter J Newman
- From the Institute for Clinical Immunology and Transfusion Medicine, Justus Liebig University, Giessen, Germany (S.S., H.W., S.W., G.B., B.B., U.J.S.); Department of Anatomy, Faculty of Veterinary Medicine, Universitas Gadjah Mada, Yogyakarta, Indonesia (H.W.); Institute for Immunology and Transfusion Medicine, Ernst-Moritz-Arndt University, Greifswald, Germany (T.B.); Center for Transfusion Medicine and Hemotherapy (N.A.-F., U.J.S.) and German Center for Fetomaternal Incompatibility (DZFI) (G.B., U.J.S.), University Hospital Giessen and Marburg, Giessen, Germany; Institute for Transfusion Medicine, University of Rostock, Rostock, Germany (V.K.); and Blood Research Institute, BloodCenter of Wisconsin, Milwaukee (J.Z., P.J.N.)
| | - Behnaz Bayat
- From the Institute for Clinical Immunology and Transfusion Medicine, Justus Liebig University, Giessen, Germany (S.S., H.W., S.W., G.B., B.B., U.J.S.); Department of Anatomy, Faculty of Veterinary Medicine, Universitas Gadjah Mada, Yogyakarta, Indonesia (H.W.); Institute for Immunology and Transfusion Medicine, Ernst-Moritz-Arndt University, Greifswald, Germany (T.B.); Center for Transfusion Medicine and Hemotherapy (N.A.-F., U.J.S.) and German Center for Fetomaternal Incompatibility (DZFI) (G.B., U.J.S.), University Hospital Giessen and Marburg, Giessen, Germany; Institute for Transfusion Medicine, University of Rostock, Rostock, Germany (V.K.); and Blood Research Institute, BloodCenter of Wisconsin, Milwaukee (J.Z., P.J.N.)
| | - Ulrich J Sachs
- From the Institute for Clinical Immunology and Transfusion Medicine, Justus Liebig University, Giessen, Germany (S.S., H.W., S.W., G.B., B.B., U.J.S.); Department of Anatomy, Faculty of Veterinary Medicine, Universitas Gadjah Mada, Yogyakarta, Indonesia (H.W.); Institute for Immunology and Transfusion Medicine, Ernst-Moritz-Arndt University, Greifswald, Germany (T.B.); Center for Transfusion Medicine and Hemotherapy (N.A.-F., U.J.S.) and German Center for Fetomaternal Incompatibility (DZFI) (G.B., U.J.S.), University Hospital Giessen and Marburg, Giessen, Germany; Institute for Transfusion Medicine, University of Rostock, Rostock, Germany (V.K.); and Blood Research Institute, BloodCenter of Wisconsin, Milwaukee (J.Z., P.J.N.).
| |
Collapse
|
66
|
Gaul V, Lopez SG, Lentz BR, Moran N, Forster RJ, Keyes TE. The lateral diffusion and fibrinogen induced clustering of platelet integrin αIIbβ3 reconstituted into physiologically mimetic GUVs. Integr Biol (Camb) 2015; 7:402-11. [PMID: 25720532 DOI: 10.1039/c5ib00003c] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Platelet integrin αIIbβ3 is a key mediator of platelet activation and thrombosis. Upon activation αIIbβ3 undergoes significant conformational rearrangement, inducing complex bidirectional signalling and protein recruitment leading to platelet activation. Reconstituted lipid models of the integrin can enhance our understanding of the structural and mechanistic details of αIIbβ3 behaviour away from the complexity of the platelet machinery. Here, a novel method of αIIbβ3 insertion into Giant Unilamellar Vesicles (GUVs) is described that allows for effective integrin reconstitution unrestricted by lipid composition. αIIbβ3 was inserted into two GUV lipid compositions that seek to better mimic the platelet membrane. First, "nature's own", comprising 32% DOPC, 25% DOPE, 20% CH, 15% SM and 8% DOPS, intended to mimic the platelet cell membrane. Fluorescence Lifetime Correlation Spectroscopy (FLCS) reveals that exposure of the integrin to the activators Mn(2+) or DTT does not influence the diffusion coefficient of αIIbβ3. Similarly, exposure to αIIbβ3's primary ligand fibrinogen (Fg) alone does not affect αIIbβ3's diffusion coefficient. However, addition of Fg with either activator reduces the integrin diffusion coefficient from 2.52 ± 0.29 to μm(2) s(-1) to 1.56 ± 0.26 (Mn(2+)) or 1.49 ± 0.41 μm(2) s(-1) (DTT) which is consistent with aggregation of activated αIIbβ3 induced by fibrinogen binding. The Multichannel Scaler (MCS) trace shows that the integrin-Fg complex diffuses through the confocal volume in clusters. Using the Saffman-Delbrück model as a first approximation, the diffusion coefficient of the complex suggests at least a 20-fold increase in the radius of membrane bound protein, consistent with integrin clustering. Second, αIIbβ3 was also reconstituted into a "raft forming" GUV with well defined liquid disordered (Ld) and liquid ordered (Lo) phases. Using confocal microscopy and lipid partitioning dyes, αIIbβ3 showed an affinity for the DOPC rich Ld phase of the raft forming GUVs, and was effectively excluded from the cholesterol and sphingomyelin rich Lo phase. Activation and Fg binding of the integrin did not alter the distribution of αIIbβ3 between the lipid phases. This observation suggests partitioning of the activated fibrinogen bound αIIbβ3 into cholesterol rich domains is not responsible for the integrin clustering observed.
Collapse
Affiliation(s)
- Vinnie Gaul
- School of Chemical Sciences and National Biophotonics and Imaging Platform, Dublin City University, Dublin 9, Ireland.
| | | | | | | | | | | |
Collapse
|
67
|
Platelets and physics: How platelets “feel” and respond to their mechanical microenvironment. Blood Rev 2015; 29:377-86. [DOI: 10.1016/j.blre.2015.05.002] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Revised: 05/04/2015] [Accepted: 05/04/2015] [Indexed: 01/08/2023]
|
68
|
Developments in cryo-electron tomography for in situ structural analysis. Arch Biochem Biophys 2015; 581:78-85. [DOI: 10.1016/j.abb.2015.04.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Revised: 04/14/2015] [Accepted: 04/19/2015] [Indexed: 12/31/2022]
|
69
|
Hyaluronan based hydrogels provide an improved model to study megakaryocyte-matrix interactions. Exp Cell Res 2015; 346:1-8. [PMID: 26027944 DOI: 10.1016/j.yexcr.2015.05.014] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Revised: 04/30/2015] [Accepted: 05/16/2015] [Indexed: 01/09/2023]
Abstract
Hyaluronan (HA) is a glycosamminoglican involved in cell biology as well as a relevant polymer for tissue engineering and regenerative medicine. Megakaryocytes (Mks) are immersed in a mesh of extracellular matrix (ECM) components that regulate their maturation in the bone marrow (BM) and the release of platelets into the bloodstream. While fibrous ECMs such as collagens and fibronectin have been demonstrated to differently regulate Mk function and platelet release, the role of HA, that fills the majority of the BM extracellular interstitial space, has not been investigated so far. Here we demonstrated that, although human Mks express HA receptors, they are not affected by HA in terms of in vitro differentiation, maturation and platelet formation. Importantly, chemical properties of HA were exploited to generate hydrogels with entrapped ECMs that represent a useful model to more closely mimic the tridimensional characteristics of the BM environment for studying Mk function. In conclusion, in this work we demonstrated that HA is an ideal candidate for a 3D ex vivo model of human BM ECM component environment.
Collapse
|
70
|
Buitrago L, Rendon A, Liang Y, Simeoni I, Negri A, Filizola M, Ouwehand WH, Coller BS. αIIbβ3 variants defined by next-generation sequencing: predicting variants likely to cause Glanzmann thrombasthenia. Proc Natl Acad Sci U S A 2015; 112:E1898-907. [PMID: 25827233 PMCID: PMC4403182 DOI: 10.1073/pnas.1422238112] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Next-generation sequencing is transforming our understanding of human genetic variation but assessing the functional impact of novel variants presents challenges. We analyzed missense variants in the integrin αIIbβ3 receptor subunit genes ITGA2B and ITGB3 identified by whole-exome or -genome sequencing in the ThromboGenomics project, comprising ∼32,000 alleles from 16,108 individuals. We analyzed the results in comparison with 111 missense variants in these genes previously reported as being associated with Glanzmann thrombasthenia (GT), 20 associated with alloimmune thrombocytopenia, and 5 associated with aniso/macrothrombocytopenia. We identified 114 novel missense variants in ITGA2B (affecting ∼11% of the amino acids) and 68 novel missense variants in ITGB3 (affecting ∼9% of the amino acids). Of the variants, 96% had minor allele frequencies (MAF) < 0.1%, indicating their rarity. Based on sequence conservation, MAF, and location on a complete model of αIIbβ3, we selected three novel variants that affect amino acids previously associated with GT for expression in HEK293 cells. αIIb P176H and β3 C547G severely reduced αIIbβ3 expression, whereas αIIb P943A partially reduced αIIbβ3 expression and had no effect on fibrinogen binding. We used receiver operating characteristic curves of combined annotation-dependent depletion, Polyphen 2-HDIV, and sorting intolerant from tolerant to estimate the percentage of novel variants likely to be deleterious. At optimal cut-off values, which had 69-98% sensitivity in detecting GT mutations, between 27% and 71% of the novel αIIb or β3 missense variants were predicted to be deleterious. Our data have implications for understanding the evolutionary pressure on αIIbβ3 and highlight the challenges in predicting the clinical significance of novel missense variants.
Collapse
Affiliation(s)
- Lorena Buitrago
- Allen and Frances Adler Laboratory of Blood and Vascular Biology and
| | - Augusto Rendon
- Department of Haematology, University of Cambridge, Cambridge CB2 0PT, United Kingdom; Medical Research Council Biostatistics Unit, Cambridge Biomedical Campus, Cambridge Institute of Public Health, Cambridge, United Kingdom; National Health Service Blood & Transplant, Cambridge, United Kingdom
| | - Yupu Liang
- Research Bioinformatics, The Rockefeller University, New York, NY 10065
| | - Ilenia Simeoni
- Department of Haematology, University of Cambridge, Cambridge CB2 0PT, United Kingdom; National Health Service Blood & Transplant, Cambridge, United Kingdom
| | - Ana Negri
- Department of Structural and Chemical Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029; and
| | - Marta Filizola
- Department of Structural and Chemical Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029; and
| | - Willem H Ouwehand
- Department of Haematology, University of Cambridge, Cambridge CB2 0PT, United Kingdom; National Health Service Blood & Transplant, Cambridge, United Kingdom; Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Barry S Coller
- Allen and Frances Adler Laboratory of Blood and Vascular Biology and
| |
Collapse
|
71
|
Abstract
While there are currently many well-established topical hemostatic agents for field administration, there are still limited tools to staunch bleeding at less accessible injury sites. Current clinical methods to restore hemostasis after large volume blood loss include platelet and clotting factor transfusion, which have respective drawbacks of short shelf life and risk of viral transmission. Therefore, synthetic hemostatic agents that can be delivered intravenously and encourage stable clot formation after localizing to sites of vascular injury are particularly appealing. In the past three decades, platelet substitutes have been prepared using drug delivery vehicles such as liposomes and PLGA nanoparticles that have been modified to mimic platelet properties. Additionally, structural considerations such as particle size, shape, and flexibility have been addressed in a number of reports. Since platelets are the first responders after vascular injury, platelet substitutes represent an important class of intravenous hemostats under development. More recently, materials affecting fibrin formation have been introduced to induce faster or more stable blood clot formation through fibrin cross-linking. Fibrin represents a major structural component in the final blood clot, and a fibrin-based hemostatic mechanism acting downstream of initial platelet plug formation may be a safer alternative to platelets to avoid undesired thrombotic activity. This Review explores intravenous hemostats under development and strategies to optimize their clotting activity.
Collapse
Affiliation(s)
- Leslie W Chan
- †Department of Bioengineering and Molecular Engineering and Sciences Institute, University of Washington, 3720 15th Avenue NE, Box 355061, Seattle, Washington 98195, United States
| | - Nathan J White
- ‡Department of Medicine, Division of Emergency Medicine, University of Washington, Seattle, Washington 98195, United States
| | - Suzie H Pun
- †Department of Bioengineering and Molecular Engineering and Sciences Institute, University of Washington, 3720 15th Avenue NE, Box 355061, Seattle, Washington 98195, United States
| |
Collapse
|
72
|
Connolly-Andersen AM, Sundberg E, Ahlm C, Hultdin J, Baudin M, Larsson J, Dunne E, Kenny D, Lindahl TL, Ramström S, Nilsson S. Increased Thrombopoiesis and Platelet Activation in Hantavirus-Infected Patients. J Infect Dis 2015; 212:1061-9. [PMID: 25762786 DOI: 10.1093/infdis/jiv161] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 03/06/2015] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Thrombocytopenia is a common finding during viral hemorrhagic fever, which includes hemorrhagic fever with renal syndrome (HFRS). The 2 main causes for thrombocytopenia are impaired thrombopoiesis and/or increased peripheral destruction of platelets. In addition, there is an increased intravascular coagulation risk during HFRS, which could be due to platelet activation. METHODS Thrombopoiesis was determined by quantification of platelet counts, thrombopoietin, immature platelet fraction, and mean platelet volume during HFRS. The in vivo platelet activation was determined by quantification of soluble P-selectin (sP-selectin) and glycoprotein VI (sGPVI). The function of circulating platelets was determined by ex vivo stimulation followed by flow cytometry analysis of platelet surface-bound fibrinogen and P-selectin exposure. Intravascular coagulation during disease was determined by scoring for disseminated intravascular coagulation (DIC) and recording thromboembolic complications. RESULTS The levels of thrombopoietin, immature platelet fraction, and mean platelet volume all indicate increased thrombopoiesis during HFRS. Circulating platelets had reduced ex vivo function during disease compared to follow-up. Most interestingly, we observed significantly increased in vivo platelet activation in HFRS patients with intravascular coagulation (DIC and thromboembolic complications) as shown by sP-selectin and sGPVI levels. CONCLUSIONS HFRS patients have increased thrombopoiesis and platelet activation, which contributes to intravascular coagulation.
Collapse
Affiliation(s)
| | - Erik Sundberg
- Infectious Diseases, Department of Clinical Microbiology Clinical Chemistry, Department of Medical Biosciences, Umeå University, Sweden
| | - Clas Ahlm
- Infectious Diseases, Department of Clinical Microbiology
| | - Johan Hultdin
- Clinical Chemistry, Department of Medical Biosciences, Umeå University, Sweden
| | - Maria Baudin
- Infectious Diseases, Department of Clinical Microbiology
| | - Johanna Larsson
- Clinical Chemistry, Department of Medical Biosciences, Umeå University, Sweden
| | - Eimear Dunne
- Clinical Research Centre, Royal College of Surgeons in Ireland, Dublin
| | - Dermot Kenny
- Clinical Research Centre, Royal College of Surgeons in Ireland, Dublin
| | - Tomas L Lindahl
- Department of Clinical and Experimental Medicine, Linköping University, Sweden
| | - Sofia Ramström
- Department of Clinical and Experimental Medicine, Linköping University, Sweden
| | - Sofie Nilsson
- Clinical Chemistry, Department of Medical Biosciences, Umeå University, Sweden
| |
Collapse
|
73
|
Hamzeh-Cognasse H, Damien P, Chabert A, Pozzetto B, Cognasse F, Garraud O. Platelets and infections - complex interactions with bacteria. Front Immunol 2015; 6:82. [PMID: 25767472 PMCID: PMC4341565 DOI: 10.3389/fimmu.2015.00082] [Citation(s) in RCA: 157] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 02/11/2015] [Indexed: 12/29/2022] Open
Abstract
Platelets can be considered sentinels of vascular system due to their high number in the circulation and to the range of functional immunoreceptors they express. Platelets express a wide range of potential bacterial receptors, including complement receptors, FcγRII, Toll-like receptors but also integrins conventionally described in the hemostatic response, such as GPIIb–IIIa or GPIb. Bacteria bind these receptors either directly, or indirectly via fibrinogen, fibronectin, the first complement C1q, the von Willebrand Factor, etc. The fate of platelet-bound bacteria is questioned. Several studies reported the ability of activated platelets to internalize bacteria such as Staphylococcus aureus or Porphyromonas gingivalis, though there is no clue on what happens thereafter. Are they sheltered from the immune system in the cytoplasm of platelets or are they lysed? Indeed, while the presence of phagolysosome has not been demonstrated in platelets, they contain antimicrobial peptides that were shown to be efficient on S. aureus. Besides, the fact that bacteria can bind to platelets via receptors involved in hemostasis suggests that they may induce aggregation; this has indeed been described for Streptococcus sanguinis, S. epidermidis, or C. pneumoniae. On the other hand, platelets are able to display an inflammatory response to an infectious triggering. We, and others, have shown that platelet release soluble immunomodulatory factors upon stimulation by bacterial components. Moreover, interactions between bacteria and platelets are not limited to only these two partners. Indeed, platelets are also essential for the formation of neutrophil extracellular traps by neutrophils, resulting in bacterial clearance by trapping bacteria and concentrating antibacterial factors but in enhancing thrombosis. In conclusion, the platelet–bacteria interplay is a complex game; its fine analysis is complicated by the fact that the inflammatory component adds to the aggregation response.
Collapse
Affiliation(s)
| | - Pauline Damien
- GIMAP-EA3064, Université de Lyon , Saint-Etienne , France
| | - Adrien Chabert
- GIMAP-EA3064, Université de Lyon , Saint-Etienne , France
| | - Bruno Pozzetto
- GIMAP-EA3064, Université de Lyon , Saint-Etienne , France
| | - Fabrice Cognasse
- GIMAP-EA3064, Université de Lyon , Saint-Etienne , France ; Etablissement Français du Sang Auvergne-Loire , Saint-Etienne , France
| | - Olivier Garraud
- GIMAP-EA3064, Université de Lyon , Saint-Etienne , France ; Institut National de la Transfusion Sanguine , Paris , France
| |
Collapse
|
74
|
Schmidt T, Suk JE, Ye F, Situ AJ, Mazumder P, Ginsberg MH, Ulmer TS. Annular anionic lipids stabilize the integrin αIIbβ3 transmembrane complex. J Biol Chem 2015; 290:8283-93. [PMID: 25632962 DOI: 10.1074/jbc.m114.623504] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Cationic membrane-proximal amino acids determine the topology of membrane proteins by interacting with anionic lipids that are restricted to the intracellular membrane leaflet. This mechanism implies that anionic lipids interfere with electrostatic interactions of membrane proteins. The integrin αIIbβ3 transmembrane (TM) complex is stabilized by a membrane-proximal αIIb(Arg(995))-β3(Asp(723)) interaction; here, we examine the influence of anionic lipids on this complex. Anionic lipids compete for αIIb(Arg(995)) contacts with β3(Asp(723)) but paradoxically do not diminish the contribution of αIIb(Arg(995))-β3(Asp(723)) to TM complex stability. Overall, anionic lipids in annular positions stabilize the αIIbβ3 TM complex by up to 0.50 ± 0.02 kcal/mol relative to zwitterionic lipids in a headgroup structure-dependent manner. Comparatively, integrin receptor activation requires TM complex destabilization of 1.5 ± 0.2 kcal/mol, revealing a sizeable influence of lipid composition on TM complex stability. We implicate changes in lipid headgroup accessibility to small molecules (physical membrane characteristics) and specific but dynamic protein-lipid contacts in this TM helix-helix stabilization. Thus, anionic lipids in ubiquitous annular positions can benefit the stability of membrane proteins while leaving membrane-proximal electrostatic interactions intact.
Collapse
Affiliation(s)
- Thomas Schmidt
- From the Department of Biochemistry & Molecular Biology and Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California 90033 and
| | - Jae-Eun Suk
- From the Department of Biochemistry & Molecular Biology and Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California 90033 and
| | - Feng Ye
- the Department of Medicine, University of California San Diego, La Jolla, California 92093
| | - Alan J Situ
- From the Department of Biochemistry & Molecular Biology and Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California 90033 and
| | - Parichita Mazumder
- From the Department of Biochemistry & Molecular Biology and Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California 90033 and
| | - Mark H Ginsberg
- the Department of Medicine, University of California San Diego, La Jolla, California 92093
| | - Tobias S Ulmer
- From the Department of Biochemistry & Molecular Biology and Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California 90033 and
| |
Collapse
|
75
|
Rafii S, Cao Z, Lis R, Siempos II, Chavez D, Shido K, Rabbany SY, Ding BS. Platelet-derived SDF-1 primes the pulmonary capillary vascular niche to drive lung alveolar regeneration. Nat Cell Biol 2015; 17:123-136. [PMID: 25621952 DOI: 10.1038/ncb3096] [Citation(s) in RCA: 110] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Accepted: 12/16/2014] [Indexed: 02/06/2023]
Abstract
The lung alveoli regenerate after surgical removal of the left lobe by pneumonectomy (PNX). How this alveolar regrowth/regeneration is initiated remains unknown. We found that platelets trigger lung regeneration by supplying stromal-cell-derived factor-1 (SDF-1, also known as CXCL12). After PNX, activated platelets stimulate SDF-1 receptors CXCR4 and CXCR7 on pulmonary capillary endothelial cells (PCECs) to deploy the angiocrine membrane-type metalloproteinase MMP14, stimulating alveolar epithelial cell (AEC) expansion and neo-alveolarization. In mice lacking platelets or platelet Sdf1, PNX-induced alveologenesis was diminished. Reciprocally, infusion of Sdf1(+/+) but not Sdf1-deficient platelets rescued lung regeneration in platelet-depleted mice. Endothelial-specific ablation of Cxcr4 and Cxcr7 in adult mice similarly impeded lung regeneration. Notably, mice with endothelial-specific Mmp14 deletion exhibited impaired expansion of AECs but not PCECs after PNX, which was not rescued by platelet infusion. Therefore, platelets prime PCECs to initiate lung regeneration, extending beyond their haemostatic contribution. Therapeutic targeting of this haemo-vascular niche could enable regenerative therapy for lung diseases.
Collapse
Affiliation(s)
- Shahin Rafii
- Ansary Stem Cell Institute, Weill Cornell Medical College, New York, NY 10065.,Department of Medicine, Weill Cornell Medical College, New York, NY 10065.,Department of Genetic Medicine, Weill Cornell Medical College, New York, NY 10065
| | - Zhongwei Cao
- Ansary Stem Cell Institute, Weill Cornell Medical College, New York, NY 10065.,Department of Medicine, Weill Cornell Medical College, New York, NY 10065
| | - Raphael Lis
- Ansary Stem Cell Institute, Weill Cornell Medical College, New York, NY 10065.,Department of Medicine, Weill Cornell Medical College, New York, NY 10065.,Department of Reproductive Medicine, Weill Cornell Medical College, New York, NY 10065
| | - Ilias I Siempos
- Department of Medicine, Weill Cornell Medical College, New York, NY 10065.,First Department of Critical Care Medicine and Pulmonary Services, Evangelismos Hospital, University of Athens Medical School, Athens 10675, Greece
| | - Deebly Chavez
- Ansary Stem Cell Institute, Weill Cornell Medical College, New York, NY 10065.,Department of Medicine, Weill Cornell Medical College, New York, NY 10065.,Department of Genetic Medicine, Weill Cornell Medical College, New York, NY 10065
| | - Koji Shido
- Ansary Stem Cell Institute, Weill Cornell Medical College, New York, NY 10065.,Department of Medicine, Weill Cornell Medical College, New York, NY 10065
| | - Sina Y Rabbany
- Ansary Stem Cell Institute, Weill Cornell Medical College, New York, NY 10065.,Department of Medicine, Weill Cornell Medical College, New York, NY 10065.,Bioengineering Program, Hofstra University, Hempstead, NY 11549
| | - Bi-Sen Ding
- Ansary Stem Cell Institute, Weill Cornell Medical College, New York, NY 10065.,Department of Genetic Medicine, Weill Cornell Medical College, New York, NY 10065
| |
Collapse
|
76
|
Roll, adhere, spread and contract: structural mechanics of platelet function. Eur J Cell Biol 2015; 94:129-38. [PMID: 25655000 DOI: 10.1016/j.ejcb.2015.01.001] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2014] [Revised: 12/26/2014] [Accepted: 01/07/2015] [Indexed: 12/31/2022] Open
Abstract
Platelets are involved in life-sustaining processes such as hemostasis, wound healing, atherothrombosis and angiogenesis. Mechanical trauma to blood vessels causes platelet activation resulting in their adherence and clot formation at the damaged site, culminating in clot retraction and tissue repair. Two of the major players underlying this process are the cytoskeleton, i.e., actin and microtubules, and the membrane integrin receptors. Rare congenital bleeding disorders such as Glanzmann thrombasthenia and Bernard-Soulier syndrome are associated with genetic alterations of platelet surface receptors, also affecting the platelet cytoskeletal structure. In this review, we summarize the current knowledge about platelet structure and adhesion, and delve into the mechanical aspects of platelet function. Platelets lack a nucleus, and can thus provide a minimal model of a biological cell. New biophysical tools may help to scrutinize platelets anew and to extend the existing knowledge on cell biology.
Collapse
|
77
|
Joshi N, Kopec AK, O'Brien KM, Towery KL, Cline-Fedewa H, Williams KJ, Copple BL, Flick MJ, Luyendyk JP. Coagulation-driven platelet activation reduces cholestatic liver injury and fibrosis in mice. J Thromb Haemost 2015; 13:57-71. [PMID: 25353084 PMCID: PMC4487795 DOI: 10.1111/jth.12770] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Accepted: 10/17/2014] [Indexed: 01/14/2023]
Abstract
BACKGROUND The coagulation cascade has been shown to participate in chronic liver injury and fibrosis, but the contribution of various thrombin targets, such as protease activated receptors (PARs) and fibrin(ogen), has not been fully described. Emerging evidence suggests that in some experimental settings of chronic liver injury, platelets can promote liver repair and inhibit liver fibrosis. However, the precise mechanisms linking coagulation and platelet function to hepatic tissue changes following injury remain poorly defined. OBJECTIVES To determine the role of PAR-4, a key thrombin receptor on mouse platelets, and fibrin(ogen) engagement of the platelet αII b β3 integrin (αIIb β3 ) in a model of cholestatic liver injury and fibrosis. METHODS Biliary and hepatic injury was characterized following 4 week administration of the bile duct toxicant α-naphthylisothiocyanate (ANIT) (0.025%) in PAR-4-deficient mice, mice expressing a mutant form of fibrin(ogen) incapable of binding integrin αII b β3 (Fibγ(Δ5) ), and wild-type mice. RESULTS Elevated plasma thrombin-antithrombin and serotonin levels, hepatic fibrin deposition, and platelet accumulation in liver accompanied hepatocellular injury and fibrosis in ANIT-treated wild-type mice. PAR-4 deficiency reduced plasma serotonin levels, increased serum bile acid concentration, and exacerbated ANIT-induced hepatocellular injury and peribiliary fibrosis. Compared with PAR-4-deficient mice, ANIT-treated Fibγ(Δ5) mice displayed more widespread hepatocellular necrosis accompanied by marked inflammation, robust fibroblast activation, and extensive liver fibrosis. CONCLUSIONS Collectively, the results indicate that PAR-4 and fibrin-αII b β3 integrin engagement, pathways coupling coagulation to platelet activation, each exert hepatoprotective effects during chronic cholestasis.
Collapse
MESH Headings
- 1-Naphthylisothiocyanate
- Animals
- Antithrombin III
- Bile Acids and Salts/blood
- Blood Coagulation/genetics
- Blood Platelets/metabolism
- Chemical and Drug Induced Liver Injury/blood
- Chemical and Drug Induced Liver Injury/genetics
- Chemical and Drug Induced Liver Injury/pathology
- Chemical and Drug Induced Liver Injury/prevention & control
- Cholestasis/blood
- Cholestasis/chemically induced
- Cholestasis/genetics
- Cholestasis/pathology
- Cholestasis/prevention & control
- Fibrinogens, Abnormal/genetics
- Fibrinogens, Abnormal/metabolism
- Genotype
- Liver/metabolism
- Liver/pathology
- Liver Cirrhosis, Experimental/blood
- Liver Cirrhosis, Experimental/chemically induced
- Liver Cirrhosis, Experimental/genetics
- Liver Cirrhosis, Experimental/pathology
- Liver Cirrhosis, Experimental/prevention & control
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Mutation
- Necrosis
- Peptide Hydrolases/blood
- Phenotype
- Platelet Activation/genetics
- Platelet Glycoprotein GPIIb-IIIa Complex/metabolism
- Receptors, Thrombin/deficiency
- Receptors, Thrombin/genetics
- Serotonin/blood
- Signal Transduction
Collapse
Affiliation(s)
- N Joshi
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA; Center for Integrative Toxicology, Michigan State University, East Lansing, MI, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
78
|
Basit H, Gaul V, Maher S, Forster RJ, Keyes TE. Aqueous-filled polymer microcavity arrays: versatile & stable lipid bilayer platforms offering high lateral mobility to incorporated membrane proteins. Analyst 2015; 140:3012-8. [DOI: 10.1039/c4an02317j] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
A robust new supported cell membrane model is described comprising lipid bilayers supported on aqueous filled spherical cap pores in PDMS, both lipid and reconstituted membrane proteins diffuse unhindered by the underlying support.
Collapse
Affiliation(s)
- Hajra Basit
- School of Chemical Sciences
- National Centre for Sensor Research
- Dublin City University
- Dublin 9
- Ireland
| | - Vinnie Gaul
- School of Chemical Sciences
- National Centre for Sensor Research
- Dublin City University
- Dublin 9
- Ireland
| | - Sean Maher
- School of Chemical Sciences
- National Centre for Sensor Research
- Dublin City University
- Dublin 9
- Ireland
| | - Robert J. Forster
- School of Chemical Sciences
- National Centre for Sensor Research
- Dublin City University
- Dublin 9
- Ireland
| | - Tia E. Keyes
- School of Chemical Sciences
- National Centre for Sensor Research
- Dublin City University
- Dublin 9
- Ireland
| |
Collapse
|
79
|
Wang YQ, Wang B, Liang Y, Cao SH, Liu L, Xu XN. Role of platelet TLR4 expression in pathogensis of septic thrombocytopenia. World J Emerg Med 2014; 2:13-7. [PMID: 25214976 DOI: 10.5847/wjem.j.1920-8642.2011.01.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2010] [Accepted: 12/15/2010] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Infection-induced thrombocytopenia (TCP) is an independent risk factor for death of patients with sepsis, but its mechanism is unknown. This study aimed to explore the underlying mechanism of TCP based on the relationship between TLR4 expression and platelet activation in septic patients. METHODS A total of 64 patients with sepsis were prospectively studied. Platelet count (PC), mean platelet volume (MPV), platelet distribution width (PDW), platelet TLR4 expression, platelet PAC-1 expression, sCD40L and TNF-α concentrations were compared between the healthy control group (15 volunteers) and sepsis group (64 patients) at admission and on the 3, 5, and 9 days after admission. The changes of MPV and PDW in the TCP and non-TCP subgroups of sepsis before and after treatment were recorded. Prognostic index was analyzed. RESULTS PC was lower in the sepsis group (P=0.006), and MPV and PDW were higher in the sepsis group than those in the healthy control group (P=0.046, P=0.001). Platelet TLR4 and PAC-1 expressions, and sCD40L and TNF-α levels increased more significantly in the sepsis group (P<0.001). PAC-1 expression and TNF-α level were higher in the TCP group than in the non-TCP group before and after treatment (P=0.023, P=0.011). sCD40L concentration and platelet TLR4 expression were significantly higher in the treated TCP group than in the non-TCP group (P=0.047, P=0.001). Compared to the non-TCP group, the rate of bleeding was higher (P=0.024) and the length of ICU stay was longer (P=0.013). The APACHE II score and the 28-day mortality were higher in the TCP group (P<0.01, P=0.048). CONCLUSIONS The elevation of platelet TLR4 expression in sepsis along with platelet activation is closely related to the incidence of thrombocytopenia. The occurrence of TCP is a sign of poor prognosis in sepsis patients.
Collapse
Affiliation(s)
- Yong-Qiang Wang
- ICU, Tianjin First Center Hospitial, Tianjin 300192, China (WangYQ, Wang B, Cao SH, Xu XN); Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China (Liang Y); General Hospital of Tianjin Medical University, Tianjin 300052, China (Liu L)
| | - Bing Wang
- ICU, Tianjin First Center Hospitial, Tianjin 300192, China (WangYQ, Wang B, Cao SH, Xu XN); Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China (Liang Y); General Hospital of Tianjin Medical University, Tianjin 300052, China (Liu L)
| | - Yong Liang
- ICU, Tianjin First Center Hospitial, Tianjin 300192, China (WangYQ, Wang B, Cao SH, Xu XN); Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China (Liang Y); General Hospital of Tianjin Medical University, Tianjin 300052, China (Liu L)
| | - Shu-Hua Cao
- ICU, Tianjin First Center Hospitial, Tianjin 300192, China (WangYQ, Wang B, Cao SH, Xu XN); Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China (Liang Y); General Hospital of Tianjin Medical University, Tianjin 300052, China (Liu L)
| | - Li Liu
- ICU, Tianjin First Center Hospitial, Tianjin 300192, China (WangYQ, Wang B, Cao SH, Xu XN); Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China (Liang Y); General Hospital of Tianjin Medical University, Tianjin 300052, China (Liu L)
| | - Xin-Nv Xu
- ICU, Tianjin First Center Hospitial, Tianjin 300192, China (WangYQ, Wang B, Cao SH, Xu XN); Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China (Liang Y); General Hospital of Tianjin Medical University, Tianjin 300052, China (Liu L)
| |
Collapse
|
80
|
Shiu HT, Goss B, Lutton C, Crawford R, Xiao Y. Formation of blood clot on biomaterial implants influences bone healing. TISSUE ENGINEERING PART B-REVIEWS 2014; 20:697-712. [PMID: 24906469 DOI: 10.1089/ten.teb.2013.0709] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The first step in bone healing is forming a blood clot at injured bones. During bone implantation, biomaterials unavoidably come into direct contact with blood, leading to a blood clot formation on its surface prior to bone regeneration. Despite both situations being similar in forming a blood clot at the defect site, most research in bone tissue engineering virtually ignores the important role of a blood clot in supporting healing. Dental implantology has long demonstrated that the fibrin structure and cellular content of a peri-implant clot can greatly affect osteoconduction and de novo bone formation on implant surfaces. This article reviews the formation of a blood clot during bone healing in relation to the use of platelet-rich plasma (PRP) gels. It is implicated that PRP gels are dramatically altered from a normal clot in healing, resulting in conflicting effect on bone regeneration. These results indicate that the effect of clots on bone regeneration depends on how the clots are formed. Factors that influence blood clot structure and properties in relation to bone healing are also highlighted. Such knowledge is essential for developing strategies to optimally control blood clot formation, which ultimately alter the healing microenvironment of bone. Of particular interest are modification of surface chemistry of biomaterials, which displays functional groups at varied composition for the purpose of tailoring blood coagulation activation, resultant clot fibrin architecture, rigidity, susceptibility to lysis, and growth factor release. This opens new scope of in situ blood clot modification as a promising approach in accelerating and controlling bone regeneration.
Collapse
Affiliation(s)
- Hoi Ting Shiu
- 1 Science and Engineering Faculty, Institute of Health and Biomedical Innovation, Queensland University of Technology , Brisbane, Australia
| | | | | | | | | |
Collapse
|
81
|
Pietraforte D, Vona R, Marchesi A, de Jacobis IT, Villani A, Del Principe D, Straface E. Redox control of platelet functions in physiology and pathophysiology. Antioxid Redox Signal 2014; 21:177-93. [PMID: 24597688 DOI: 10.1089/ars.2013.5532] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
SIGNIFICANCE An imbalance between the production and the detoxification of reactive oxygen species and reactive nitrogen species (ROS/RNS) can be implicated in many pathological processes. Platelets are best known as primary mediators of hemostasis and can be either targets of ROS/RNS or generate radicals during cell activation. These conditions can dramatically affect platelet physiology, leading even, as an ultimate event, to the cell number modification. In this case, pathological conditions such as thrombocytosis (promoted by increased cell number) or thrombocytopenia and myelodysplasia (promoted by cell decrease mediated by accelerated apoptosis) can occur. RECENT ADVANCES Usually, in peripheral blood, ROS/RNS production is balanced by the rate of oxidant elimination. Under this condition, platelets are in a nonadherent "resting" state. During endothelial dysfunction or under pathological conditions, ROS/RNS production increases and the platelets respond with specific biochemical and morphologic changes. Mitochondria are at the center of these processes, being able to both generate ROS/RNS, that drive redox-sensitive events, and respond to ROS/RNS-mediated changes of the cellular redox state. Irregular function of platelets and enhanced interaction with leukocytes and endothelial cells can contribute to pathogenesis of atherosclerotic and thrombotic events. CRITICAL ISSUES The relationship between oxidative stress, platelet death, and the activation-dependent pathways that drive platelet pro-coagulant activity is unclear and deserves to be explored. FUTURE DIRECTIONS Expanding knowledge about how platelets can mediate hemostasis and modulate inflammation may lead to novel and effective therapeutic strategies for the long and growing list of pathological conditions that involve both thrombosis and inflammation.
Collapse
Affiliation(s)
- Donatella Pietraforte
- 1 Department of Cell Biology and Neurosciences, Section of Cell Aging and Gender Medicine, Istituto Superiore di Sanità , Rome, Italy
| | | | | | | | | | | | | |
Collapse
|
82
|
Adamson K, Dolan C, Moran N, Forster RJ, Keyes TE. RGD Labeled Ru(II) Polypyridyl Conjugates for Platelet Integrin αIIbβ3 Recognition and as Reporters of Integrin Conformation. Bioconjug Chem 2014; 25:928-44. [DOI: 10.1021/bc5000737] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Kellie Adamson
- School
of Chemical Sciences, Dublin City University, Dublin 9, Ireland
| | - Ciaran Dolan
- School
of Chemical Sciences, Dublin City University, Dublin 9, Ireland
| | - Niamh Moran
- The
Department of Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Robert J. Forster
- School
of Chemical Sciences, Dublin City University, Dublin 9, Ireland
| | - Tia E. Keyes
- School
of Chemical Sciences, Dublin City University, Dublin 9, Ireland
| |
Collapse
|
83
|
Immobilisation of a fibrillin-1 fragment enhances the biocompatibility of PTFE. Colloids Surf B Biointerfaces 2014; 116:544-52. [DOI: 10.1016/j.colsurfb.2014.01.042] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Revised: 01/24/2014] [Accepted: 01/24/2014] [Indexed: 11/23/2022]
|
84
|
|
85
|
Kalli AC, Campbell ID, Sansom MSP. Conformational changes in talin on binding to anionic phospholipid membranes facilitate signaling by integrin transmembrane helices. PLoS Comput Biol 2013; 9:e1003316. [PMID: 24204243 PMCID: PMC3814715 DOI: 10.1371/journal.pcbi.1003316] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2013] [Accepted: 09/19/2013] [Indexed: 01/01/2023] Open
Abstract
Integrins are heterodimeric (αβ) cell surface receptors that are activated to a high affinity state by the formation of a complex involving the α/β integrin transmembrane helix dimer, the head domain of talin (a cytoplasmic protein that links integrins to actin), and the membrane. The talin head domain contains four sub-domains (F0, F1, F2 and F3) with a long cationic loop inserted in the F1 domain. Here, we model the binding and interactions of the complete talin head domain with a phospholipid bilayer, using multiscale molecular dynamics simulations. The role of the inserted F1 loop, which is missing from the crystal structure of the talin head, PDB:3IVF, is explored. The results show that the talin head domain binds to the membrane predominantly via cationic regions on the F2 and F3 subdomains and the F1 loop. Upon binding, the intact talin head adopts a novel V-shaped conformation which optimizes its interactions with the membrane. Simulations of the complex of talin with the integrin α/β TM helix dimer in a membrane, show how this complex promotes a rearrangement, and eventual dissociation of, the integrin α and β transmembrane helices. A model for the talin-mediated integrin activation is proposed which describes how the mutual interplay of interactions between transmembrane helices, the cytoplasmic talin protein, and the lipid bilayer promotes integrin inside-out activation. Transmission of signals across the cell membrane is an essential process for all living organisms. Integrins are one example of cell surface receptors (αβ) which, uniquely, form a bidirectional signalling pathway across the membrane. Integrins are crucial for many cellular processes and play key roles in pathological defects such as cardiovascular diseases and cancer. They are activated to a high affinity state by the intracellular protein talin in a process known as ‘inside-out activation’. Despite their importance and the existence of functional and structural data, the mechanism by which talin activates integrin remains elusive. In this study we use a multi-scale computational approach, which combines coarse-grained and atomistic molecular dynamics simulations, to suggest how the formation of the complex between the talin head domain, the cell membrane and the integrin moves the integrin equilibrium towards an active state. Our results show that conformational changes within the talin head domains optimize its interactions with the cell membrane. Upon binding to the integrin, talin facilitates rearrangement of the integrin TM region thus promoting integrin activation. This study also provides a demonstration of the strengths of a computational multi-scale approach in studies of membrane interactions and receptor conformational changes and associated proteins that enable transmembrane signaling.
Collapse
Affiliation(s)
- Antreas C. Kalli
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
| | - Iain D. Campbell
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
| | - Mark S. P. Sansom
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
- * E-mail:
| |
Collapse
|
86
|
Peptide-grafted poly(ethylene glycol) hydrogels support dynamic adhesion of endothelial progenitor cells. Acta Biomater 2013; 9:8279-89. [PMID: 23770139 DOI: 10.1016/j.actbio.2013.05.023] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Revised: 05/16/2013] [Accepted: 05/21/2013] [Indexed: 12/20/2022]
Abstract
This study investigated the dynamic adhesion of endothelial progenitor cells (EPCs) to peptide-grafted poly(ethylene glycol) diacrylate (PEGDA) hydrogels and determined the relative ability of RGDS, REDV and YIGSRG peptides to reduce the velocity of EPC rolling. Circulating EPCs are key mediators of endothelium repair and have been shown to accelerate re-endothelialization, which is important in reducing the incidence of restenosis following stent placement and occlusion of small diameter vascular grafts. However, to exploit these capabilities for tissue engineering applications, more knowledge is needed about EPC binding to the vascular wall under shear and, in particular, whether the incorporation of peptide ligands into biomaterials can support the process of EPC rolling or maintain EPC adhesion. This study specifically examined one type of EPCs endothelial colony forming cells (ECFCs), based on their ability to be expanded in culture and differentiate into mature endothelial cells. The amount of grafted PEG-peptide was shown to be dependent on the concentration of PEG-peptide grafting solution photopolymerized onto the hydrogel surface. The ECFC strength of adhesion on PEG-RDGS grafted hydrogels exceeded 350 dyn cm(-2) for 85% of adherent cells. PEG-RGDS grafted hydrogels supported ECFC rolling, whereas ECFC velocity on the negative control PEG-RGES grafted hydrogels and on the "blank slate" PEGDA hydrogels was substantially higher than the cutoff velocity for cell rolling. The ECFC rolling velocity on PEG-RDGS grafted hydrogels depended on the shear rate; as shear rate was increased from 20 s(-1) to 120 s(-1), ECFC rolling velocity increased from 103±3 μm s(-1) to 741±28 μm s(-1). REDV and YIGSRG, which are known to preferentially support endothelial cell adhesion, also supported ECFC rolling. Interestingly, the rolling velocity of ECFCs on PEG-REDV grafted hydrogels was significantly lower than on PEG-YIGSRG or on PEG-RGDS grafted hydrogels. Understanding the dynamic adhesion of ECFCs to peptide-grafted hydrogels is the first step towards understanding the similarities and differences of EPCs from mature endothelial cells and improving the ability to sequester EPCs to biomaterial surfaces in order to promote intravascular re-endothelialization.
Collapse
|
87
|
|
88
|
Fernández-Montes Moraleda B, Román JS, Rodríguez-Lorenzo LM. Influence of surface features of hydroxyapatite on the adsorption of proteins relevant to bone regeneration. J Biomed Mater Res A 2013; 101:2332-9. [DOI: 10.1002/jbm.a.34528] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2012] [Accepted: 10/25/2012] [Indexed: 01/04/2023]
|
89
|
Diz-Küçükkaya R. Inherited platelet disorders including Glanzmann thrombasthenia and Bernard-Soulier syndrome. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2013; 2013:268-275. [PMID: 24319190 DOI: 10.1182/asheducation-2013.1.268] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Inherited platelet disorders (IPDs) are a heterogeneous group of diseases affecting platelet production, morphology, and function. The degree of thrombocytopenia and functional abnormality of platelets determines the clinical manifestations. Although severe deficiencies may cause excessive bleeding beginning in early childhood, most of IPDs have mild bleeding tendencies and therefore are not always easy to distinguish from acquired platelet disorders. The diagnosis of IPD may require extensive laboratory investigation, because current routine laboratory tests are not satisfactory for differential diagnosis in some cases, and most of the specific tests are not readily available in many countries. This review summarizes the classification and clinical and molecular characteristics of known IPDs, including Bernard-Soulier syndrome and Glanzmann thrombasthenia, with a focus on current challenges in the laboratory diagnosis and management of bleeding in these patients.
Collapse
Affiliation(s)
- Reyhan Diz-Küçükkaya
- 1Department of Internal Medicine, Division of Hematology, Faculty of Medicine, Istanbul Bilim University, Istanbul, Turkey
| |
Collapse
|
90
|
New insights into adhesion signaling in bone formation. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2013; 305:1-68. [PMID: 23890379 DOI: 10.1016/b978-0-12-407695-2.00001-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Mineralized tissues that are protective scaffolds in the most primitive species have evolved and acquired more specific functions in modern animals. These are as diverse as support in locomotion, ion homeostasis, and precise hormonal regulation. Bone formation is tightly controlled by a balance between anabolism, in which osteoblasts are the main players, and catabolism mediated by the osteoclasts. The bone matrix is deposited in a cyclic fashion during homeostasis and integrates several environmental cues. These include diffusible elements that would include estrogen or growth factors and physicochemical parameters such as bone matrix composition, stiffness, and mechanical stress. Therefore, the microenvironment is of paramount importance for controlling this delicate equilibrium. Here, we provide an overview of the most recent data highlighting the role of cell-adhesion molecules during bone formation. Due to the very large scope of the topic, we focus mainly on the role of the integrin receptor family during osteogenesis. Bone phenotypes of some deficient mice as well as diseases of human bones involving cell adhesion during this process are discussed in the context of bone physiology.
Collapse
|
91
|
Wise SG, Waterhouse A, Michael P, Ng MKC. Extracellular matrix molecules facilitating vascular biointegration. J Funct Biomater 2012; 3:569-87. [PMID: 24955633 PMCID: PMC4031001 DOI: 10.3390/jfb3030569] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2012] [Revised: 08/01/2012] [Accepted: 08/06/2012] [Indexed: 12/16/2022] Open
Abstract
All vascular implants, including stents, heart valves and graft materials exhibit suboptimal biocompatibility that significantly reduces their clinical efficacy. A range of biomolecules in the subendothelial space have been shown to play critical roles in local regulation of thrombosis, endothelial growth and smooth muscle cell proliferation, making these attractive candidates for modulation of vascular device biointegration. However, classically used biomaterial coatings, such as fibronectin and laminin, modulate only one of these components; enhancing endothelial cell attachment, but also activating platelets and triggering thrombosis. This review examines a subset of extracellular matrix molecules that have demonstrated multi-faceted vascular compatibility and accordingly are promising candidates to improve the biointegration of vascular biomaterials.
Collapse
Affiliation(s)
- Steven G Wise
- The Heart Research Institute, Eliza Street, Newtown, NSW 2042, Australia.
| | - Anna Waterhouse
- Wyss Institute for Biologically Inspired Engineering at Harvard, Boston, MA 02115, USA.
| | - Praveesuda Michael
- The Heart Research Institute, Eliza Street, Newtown, NSW 2042, Australia.
| | - Martin K C Ng
- The Heart Research Institute, Eliza Street, Newtown, NSW 2042, Australia.
| |
Collapse
|
92
|
Litvinov RI, Mekler A, Shuman H, Bennett JS, Barsegov V, Weisel JW. Resolving two-dimensional kinetics of the integrin αIIbβ3-fibrinogen interactions using binding-unbinding correlation spectroscopy. J Biol Chem 2012; 287:35275-35285. [PMID: 22893701 DOI: 10.1074/jbc.m112.404848] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Using a combined experimental and theoretical approach named binding-unbinding correlation spectroscopy (BUCS), we describe the two-dimensional kinetics of interactions between fibrinogen and the integrin αIIbβ3, the ligand-receptor pair essential for platelet function during hemostasis and thrombosis. The methodology uses the optical trap to probe force-free association of individual surface-attached fibrinogen and αIIbβ3 molecules and forced dissociation of an αIIbβ3-fibrinogen complex. This novel approach combines force clamp measurements of bond lifetimes with the binding mode to quantify the dependence of the binding probability on the interaction time. We found that fibrinogen-reactive αIIbβ3 pre-exists in at least two states that differ in their zero force on-rates (k(on1) = 1.4 × 10(-4) and k(on2) = 2.3 × 10(-4) μm(2)/s), off-rates (k(off1) = 2.42 and k(off2) = 0.60 s(-1)), and dissociation constants (K(d)(1) = 1.7 × 10(4) and K(d)(2) = 2.6 × 10(3) μm(-2)). The integrin activator Mn(2+) changed the on-rates and affinities (K(d)(1) = 5 × 10(4) and K(d)(2) = 0.3 × 10(3) μm(-2)) but did not affect the off-rates. The strength of αIIbβ3-fibrinogen interactions was time-dependent due to a progressive increase in the fraction of the high affinity state of the αIIbβ3-fibrinogen complex characterized by a faster on-rate. Upon Mn(2+)-induced integrin activation, the force-dependent off-rates decrease while the complex undergoes a conformational transition from a lower to higher affinity state. The results obtained provide quantitative estimates of the two-dimensional kinetic rates for the low and high affinity αIIbβ3 and fibrinogen interactions at the single molecule level and offer direct evidence for the time- and force-dependent changes in αIIbβ3 conformation and ligand binding activity, underlying the dynamics of fibrinogen-mediated platelet adhesion and aggregation.
Collapse
Affiliation(s)
- Rustem I Litvinov
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Andrey Mekler
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Henry Shuman
- Department of Physiology, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Joel S Bennett
- Department of Hematology-Oncology Division of the Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Valeri Barsegov
- Department of Chemistry, University of Massachusetts, Lowell, Massachusetts 01854.
| | - John W Weisel
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, Pennsylvania 19104.
| |
Collapse
|
93
|
Abstract
Platelet aggregation is not only an essential part of hemostasis, but also initiates acute coronary syndrome or ischemic stroke. The precise understanding of the activation mechanism of platelet aggregation is fundamental for the development of more effective agents against platelet aggregation. Adenosine diphosphate, thrombin, and thromboxane A(2) activate platelet integrin αIIbβ3 through G protein-coupled receptors. G protein-mediated signaling pathways, which are initiated by G(q), G(12)/G(13) or G(i), include phospholipase C with calcium signaling, Rho signaling, protein kinase C and phosphatidylinositol 3-kinase. Rap1b, Ca(2+) and diacylglycerol-regulated guanine nucleotide exchange factor I, Rap1-GTP-interacting adaptor molecule, and Akt are important proteins involved in G protein-mediated activation of integrin αIIbβ3. Binding of talin-1 and kindlin-3 to cytoplasmic domains of β3-integrin triggers a conformational change in the extracellular domains that increases its affinity for ligands, such as fibrinogen or von Willebrand factor. Fibrinogens act as bridges between adjacent platelets to generate a platelet aggregate.
Collapse
Affiliation(s)
- Seung-Jae Joo
- Cardiology Division, Department of Internal Medicine, Jeju National University Hospital, Jeju, Korea
| |
Collapse
|
94
|
Goggs R, Poole AW. Platelet signaling-a primer. J Vet Emerg Crit Care (San Antonio) 2012; 22:5-29. [PMID: 22316389 DOI: 10.1111/j.1476-4431.2011.00704.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2011] [Accepted: 11/25/2011] [Indexed: 12/28/2022]
Abstract
OBJECTIVE To review the receptors and signal transduction pathways involved in platelet plug formation and to highlight links between platelets, leukocytes, endothelium, and the coagulation system. DATA SOURCES Original studies, review articles, and book chapters in the human and veterinary medical fields. DATA SYNTHESIS Platelets express numerous surface receptors. Critical among these are glycoprotein VI, the glycoprotein Ib-IX-V complex, integrin α(IIb) β(3) , and the G-protein-coupled receptors for thrombin, ADP, and thromboxane. Activation of these receptors leads to various important functional events, in particular activation of the principal adhesion receptor α(IIb) β(3) . Integrin activation allows binding of ligands such as fibrinogen, mediating platelet-platelet interaction in the process of aggregation. Signals activated by these receptors also couple to 3 other important functional events, secretion of granule contents, change in cell shape through cytoskeletal rearrangement, and procoagulant membrane expression. These processes generate a stable thrombus to limit blood loss and promote restoration of endothelial integrity. CONCLUSIONS Improvements in our understanding of how platelets operate through their signaling networks are critical for diagnosis of unusual primary hemostatic disorders and for rational antithrombotic drug design.
Collapse
Affiliation(s)
- Robert Goggs
- School of Physiology and Pharmacology, Faculty of Medical and Veterinary Sciences, University of Bristol, UK.
| | | |
Collapse
|
95
|
Abstract
The adhesion and aggregation of platelets during hemostasis and thrombosis represents one of the best-understood examples of cell-matrix adhesion. Platelets are exposed to a wide variety of extracellular matrix (ECM) proteins once blood vessels are damaged and basement membranes and interstitial ECM are exposed. Platelet adhesion to these ECM proteins involves ECM receptors familiar in other contexts, such as integrins. The major platelet-specific integrin, αIIbβ3, is the best-understood ECM receptor and exhibits the most tightly regulated switch between inactive and active states. Once activated, αIIbβ3 binds many different ECM proteins, including fibrinogen, its major ligand. In addition to αIIbβ3, there are other integrins expressed at lower levels on platelets and responsible for adhesion to additional ECM proteins. There are also some important nonintegrin ECM receptors, GPIb-V-IX and GPVI, which are specific to platelets. These receptors play major roles in platelet adhesion and in the activation of the integrins and of other platelet responses, such as cytoskeletal organization and exocytosis of additional ECM ligands and autoactivators of the platelets.
Collapse
Affiliation(s)
- Wolfgang Bergmeier
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, North Carolina 27599-7035, USA
| | | |
Collapse
|
96
|
Singh A, Winterbottom E, Daar IO. Eph/ephrin signaling in cell-cell and cell-substrate adhesion. Front Biosci (Landmark Ed) 2012; 17:473-97. [PMID: 22201756 DOI: 10.2741/3939] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Cell-cell and cell-matrix adhesion are critical processes for the formation and maintenance of tissue patterns during development, as well as control of invasion and metastasis of cancer cells. Although great strides have been made regarding our understanding of the processes that play a role in cell adhesion and cell movement, the precise mechanisms by which diverse signaling events regulate cell and tissue architecture are poorly understood. One group of cell surface molecules, Eph receptor tyrosine kinases, and their membrane-bound ligands, ephrins, are key regulators in these processes. It is the ability of Eph/ephrin signaling pathways to regulate cell-cell adhesion and motility that establishes this family as a formidable system for regulating tissue separation and morphogenesis. Moreover, the de-regulation of this signaling system is linked to the promotion of more aggressive and metastatic tumors in humans.
Collapse
Affiliation(s)
- Arvinder Singh
- Laboratory of Cell and Developmental Signaling, National Cancer Institute-Frederick, Frederick, Maryland 21702, USA
| | | | | |
Collapse
|
97
|
Affinity of talin-1 for the β3-integrin cytosolic domain is modulated by its phospholipid bilayer environment. Proc Natl Acad Sci U S A 2011; 109:793-8. [PMID: 22210111 DOI: 10.1073/pnas.1117220108] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Binding of the talin-1 FERM (4.1/ezrin/radixin/moesin) domain to the β3 cytosolic tail causes activation of the integrin αIIbβ3. The FERM domain also binds to acidic phospholipids. Although much is known about the interaction of talin-1 with integrins and lipids, the relative contribution of each interaction to integrin regulation and possible synergy between them remain to be clarified. Here, we examined the thermodynamic interplay between FERM domain binding to phospholipid bilayers and to its binding sites in the β3 tail. We found that although both the F0F1 and F2F3 subdomains of the talin-1 FERM domain bind acidic bilayers, the full-length FERM domain binds with an affinity similar to F2F3, indicating that F0F1 contributes little to the overall interaction. When free in solution, the β3 tail has weak affinity for the FERM domain. However, appending the tail to acidic phospholipids increased its affinity for the FERM domain by three orders of magnitude. Nonetheless, the affinity of the FERM for the appended tail was similar to its affinity for binding to bilayers alone. Thus, talin-1 binding to the β3 tail is a ternary interaction dominated by a favorable surface interaction with phospholipid bilayers and set by lipid composition. Nonetheless, interactions between the FERM domain, the β3 tail, and lipid bilayers are not optimized for a high-affinity synergistic interaction, even at the membrane surface. Instead, the interactions appear to be tuned in such a way that the equilibrium between inactive and active integrin conformations can be readily regulated.
Collapse
|
98
|
Abstract
A close homologue to protein disulfide isomerase (PDI) called ERp57 forms disulfide bonds in glycoproteins in the endoplasmic reticulum and is expressed on the platelet surface. We generated 2 rabbit Abs to ERp57. One Ab strongly inhibited ERp57 in a functional assay and strongly inhibited platelet aggregation. There was minimal cross-reactivity of this Ab with PDI by Western blot or in the functional assay. This Ab substantially inhibited activation of the αIIbβ3 fibrinogen receptor and P-selectin expression. Furthermore, adding ERp57 to platelets potentiated aggregation. In contrast, adding a catalytically inactive ERp57 inhibited platelet aggregation. When infused into mice the inactive ERp57 prolonged the tail bleeding times. We generated 2 IgG2a mAbs that reacted with ERp57 by immunoblot. One of these Abs inhibited both ERp57 activity and platelet aggregation. The other Ab did not inhibit ERp57 activity or platelet aggregation. The inhibitory Ab inhibited activation of αIIbβ3 and P-selectin expression, prolonged tail bleeding times, and inhibited FeCl(3)-induced thrombosis in mice. Finally, we found that a commonly used mAb to PDI also inhibited ERp57 activity. We conclude that a glycoprotein-specific member of the PDI family, ERp57, is required for platelet aggregation, hemostasis, and thrombosis.
Collapse
|
99
|
Yagi M, Murray J, Strand K, Blystone S, Interlandi G, Suda Y, Sobel M. Heparin modulates the conformation and signaling of platelet integrin αIIbβ3. Thromb Res 2011; 129:743-9. [PMID: 22197178 DOI: 10.1016/j.thromres.2011.11.054] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2011] [Revised: 11/07/2011] [Accepted: 11/30/2011] [Indexed: 10/14/2022]
Abstract
INTRODUCTION The glycosaminoglycan heparin has been shown to bind to platelet integrin αIIbβ3 and induce platelet activation and aggregation, although the relationship between binding and activation is unclear. We analyzed the interaction of heparin and αIIbβ3 in detail, to obtain a better understanding of the mechanism by which heparin acts on platelets. METHODS We assessed conformational changes in αIIbβ3 by flow cytometry of platelets exposed to unfractionated heparin. In human platelets and K562 cells engineered to express αIIbβ3, we assayed the effect of heparin on key steps in integrin signaling: phosphorylation of the β3 chain cytoplasmic tail, and activation of src kinase. We measured the heparin binding affinity of purified αIIbβ3, and of recombinant fragments of αIIb and β3, by surface plasmon resonance. RESULTS AND CONCLUSIONS Heparin binding results in conformational changes in αIIbβ3, similar to those observed upon ligand binding. Heparin binding alone is not sufficient to induce tyrosine phosphorylation of the integrin β3 cytoplasmic domain, but the presence of heparin increased both β3 phosphorylation and src kinase activation in response to ligand binding. Specific recombinant fragments derived from αIIb bound heparin, while recombinant β3 did not bind. This pattern of heparin binding, compared to the crystal structure of αIIbβ3, suggests that heparin-binding sites are located in clusters of basic amino acids in the headpiece and/or leg domains of αIIb. Binding of heparin to these clusters may stabilize the transition of αIIbβ3 to an open conformation with enhanced affinity for ligand, facilitating outside-in signaling and platelet activation.
Collapse
Affiliation(s)
- Mayumi Yagi
- Research & Development, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA.
| | | | | | | | | | | | | |
Collapse
|
100
|
Modery CL, Ravikumar M, Wong TL, Dzuricky MJ, Durongkaveroj N, Sen Gupta A. Heteromultivalent liposomal nanoconstructs for enhanced targeting and shear-stable binding to active platelets for site-selective vascular drug delivery. Biomaterials 2011; 32:9504-14. [DOI: 10.1016/j.biomaterials.2011.08.067] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2011] [Accepted: 08/20/2011] [Indexed: 11/28/2022]
|