1
|
Shen C, Mackeigan DT, Shoara AA, Bhoria P, Zhu G, Karakas D, Ma W, Chen ZY, Xu R, Slavkovic S, Zhang D, Prifti V, Liu Z, Cerenzia EG, Chen P, Neves MAD, Li H, Xue F, Yang R, Liu J, Lai R, Li R, Ni H. Novel GPIb-independent platelet aggregation induced by botrocetin: implications for diagnosis and antithrombotic therapy. J Thromb Haemost 2024; 22:3249-3265. [PMID: 39147240 DOI: 10.1016/j.jtha.2024.06.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/25/2024] [Accepted: 06/07/2024] [Indexed: 08/17/2024]
Abstract
BACKGROUND Snake venom botrocetin facilitates von Willebrand factor (VWF) binding to platelet GPIbα and has been widely used for the diagnosis of von Willebrand disease and GPIb-related disorders. Botrocetin is also commonly employed for the development/characterization of antithrombotics targeting the GPIb-VWF axis. OBJECTIVES To explore the alternative receptor(s)/mechanisms that participate in botrocetin-induced platelet aggregation. METHODS The effects of botrocetin on platelet aggregation were examined using platelets from wild-type, VWF- and fibrinogen-deficient, GPIbα-deficient, IL4Rα/GPIbα-transgenic, ITGA2B and ITGB3-deficient mice, and Bernard-Soulier syndrome and healthy human samples. Platelet-fibrinogen and platelet-VWF interaction were measured using flow cytometry. GPIbα-VWF binding was evaluated utilizing enzyme-linked immunosorbent assay. Botrocetin-αIIbβ3 and botrocetin-GPIbα interactions were measured using enzyme-linked immunosorbent assay and fluorescence anisotropy assays. Heparinized whole blood from healthy donors was examined for thrombus formation and growth in a perfusion chamber. RESULTS Botrocetin could induce aggregation of platelets from a Bernard-Soulier syndrome patient and GPIbα-deficient mice as well as platelets lacking the N-terminal extracellular domain of GPIbα. Botrocetin could interact with αIIbβ3 and facilitated αIIbβ3-VWF interaction independent of GPIb. Botrocetin competitively bound to the ligand-binding domain of activated rather than resting αIIbβ3. Although botrocetin-induced platelet aggregation requires VWF, strikingly, in the absence of VWF, botrocetin blocked fibrinogen and other ligand binding to αIIbβ3 and inhibited platelet aggregation and thrombus formation. Consistently, recombinant botrocetin defective in VWF binding inhibited αIIbβ3- and GPIb-mediated platelet aggregation, spreading, and thrombus formation. CONCLUSION Our study provides insights into avoiding the misdiagnosis of GPIb-related disorders and developing botrocetin mutants as potential new antithrombotics that may simultaneously target both αIIbβ3 and GPIbα.
Collapse
Affiliation(s)
- Chuanbin Shen
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada; Department of Laboratory Medicine, Li Ka Shing Knowledge Institute (LKSKI)-Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Toronto, Ontario, Canada; CCOA Therapeutics Inc, Toronto, Ontario, Canada; School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong, China.
| | - Daniel T Mackeigan
- Department of Laboratory Medicine, Li Ka Shing Knowledge Institute (LKSKI)-Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Toronto, Ontario, Canada; Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Aron A Shoara
- Department of Laboratory Medicine, Li Ka Shing Knowledge Institute (LKSKI)-Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Toronto, Ontario, Canada; Department of Physiology, University of Toronto, Toronto, Ontario, Canada; Canadian Blood Services Centre for Innovation, Toronto, Ontario, Canada
| | - Preeti Bhoria
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada; Department of Laboratory Medicine, Li Ka Shing Knowledge Institute (LKSKI)-Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Toronto, Ontario, Canada; CCOA Therapeutics Inc, Toronto, Ontario, Canada
| | - Guangheng Zhu
- Department of Laboratory Medicine, Li Ka Shing Knowledge Institute (LKSKI)-Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Toronto, Ontario, Canada; CCOA Therapeutics Inc, Toronto, Ontario, Canada
| | - Danielle Karakas
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada; Department of Laboratory Medicine, Li Ka Shing Knowledge Institute (LKSKI)-Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Toronto, Ontario, Canada
| | - Wenjing Ma
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada; Department of Laboratory Medicine, Li Ka Shing Knowledge Institute (LKSKI)-Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Toronto, Ontario, Canada; CCOA Therapeutics Inc, Toronto, Ontario, Canada
| | - Zi Yan Chen
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada; Department of Laboratory Medicine, Li Ka Shing Knowledge Institute (LKSKI)-Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Toronto, Ontario, Canada
| | - Runjia Xu
- Department of Laboratory Medicine, Li Ka Shing Knowledge Institute (LKSKI)-Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Toronto, Ontario, Canada
| | - Sladjana Slavkovic
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada; Department of Laboratory Medicine, Li Ka Shing Knowledge Institute (LKSKI)-Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Toronto, Ontario, Canada; CCOA Therapeutics Inc, Toronto, Ontario, Canada; Canadian Blood Services Centre for Innovation, Toronto, Ontario, Canada
| | - Dachuan Zhang
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada; Department of Laboratory Medicine, Li Ka Shing Knowledge Institute (LKSKI)-Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Toronto, Ontario, Canada; CCOA Therapeutics Inc, Toronto, Ontario, Canada
| | - Viktor Prifti
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada; Department of Laboratory Medicine, Li Ka Shing Knowledge Institute (LKSKI)-Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Toronto, Ontario, Canada
| | - Zhenze Liu
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada; Department of Laboratory Medicine, Li Ka Shing Knowledge Institute (LKSKI)-Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Toronto, Ontario, Canada; CCOA Therapeutics Inc, Toronto, Ontario, Canada
| | - Eric G Cerenzia
- Department of Laboratory Medicine, Li Ka Shing Knowledge Institute (LKSKI)-Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Toronto, Ontario, Canada; Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Pingguo Chen
- Department of Laboratory Medicine, Li Ka Shing Knowledge Institute (LKSKI)-Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Toronto, Ontario, Canada; Canadian Blood Services Centre for Innovation, Toronto, Ontario, Canada
| | - Miguel A D Neves
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada; Department of Laboratory Medicine, Li Ka Shing Knowledge Institute (LKSKI)-Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Toronto, Ontario, Canada; CCOA Therapeutics Inc, Toronto, Ontario, Canada; Canadian Blood Services Centre for Innovation, Toronto, Ontario, Canada
| | - Huiyuan Li
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Key Laboratory of Gene Therapy for Blood Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Feng Xue
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Key Laboratory of Gene Therapy for Blood Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Renchi Yang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Key Laboratory of Gene Therapy for Blood Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Junling Liu
- Department of Biochemistry and Molecular Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ren Lai
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Renhao Li
- Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Children's Healthcare of Atlanta, Emory University School of Medicine Atlanta, Atlanta, Georgia, Georgia, USA
| | - Heyu Ni
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada; Department of Laboratory Medicine, Li Ka Shing Knowledge Institute (LKSKI)-Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Toronto, Ontario, Canada; CCOA Therapeutics Inc, Toronto, Ontario, Canada; Department of Physiology, University of Toronto, Toronto, Ontario, Canada; Canadian Blood Services Centre for Innovation, Toronto, Ontario, Canada; Department of Medicine, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
2
|
Liang Q, Parker ET, Dean G, Karpen MS, Wu Y, Wang X, Di Paola J, Maier CL, Meeks SL, Lollar P, Sidonio RF, Li R. Nanobody activator improves sensitivity of the von Willebrand factor activity assay to multimer size. J Thromb Haemost 2024; 22:2052-2058. [PMID: 38704122 PMCID: PMC11214880 DOI: 10.1016/j.jtha.2024.04.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 04/15/2024] [Accepted: 04/19/2024] [Indexed: 05/06/2024]
Abstract
BACKGROUND The activity of von Willebrand factor (VWF) in facilitating platelet adhesion and aggregation correlates with its multimer size. Traditional ristocetin-dependent functional assays lack sensitivity to multimer sizes. Recently, nanobodies targeting the autoinhibitory module and activating VWF were identified. OBJECTIVES To develop an assay that can differentiate the platelet-binding activity of VWF multimers. METHODS A novel enzyme-linked immunosorbent assay (nanobody-triggered glycoprotein Ib binding assay [VWF:GPIbNab]) utilizing a VWF-activating nanobody was developed. Recombinant VWF, plasma-derived VWF (pdVWF), and selected gel-filtrated fractions of pdVWF were evaluated for VWF antigen and activity levels. A linear regression model was developed to estimate the specific activity of VWF multimers. RESULTS Of the 3 activating nanobodies tested, 6C11 with the lowest activation effect exhibited the highest sensitivity for high-molecular-weight multimers (HMWMs) of VWF. VWF:GPIbNab utilizing 6C11 (VWF:GPIbNab6C11) produced significantly higher activity/antigen ratios for recombinant VWF (>2.0) and HMWM-enriched pdVWF fractions (>2.0) than for pdVWF (∼1.0) or fractions enriched with shorter multimers (<1.0). The differences were much larger than those produced by VWF:GPIbNab utilizing other nanobodies, VWF:GPIbM, VWF:GPIbR, or VWF:CB assays. Linear regression analysis of 5 pdVWF fractions of various multimer sizes produced an estimated specific activity of 2.7 for HMWMs. The analysis attributed >90% of the VWF activity measured by VWF:GPIbNab6C11 to that of HMWMs, which is significantly higher than all other activity assays tested. CONCLUSION The VWF:GPIbNab6C11 assay exhibits higher sensitivity to HMWMs than ristocetin-based and collagen-binding assays. Future studies examining the application of this assay in clinical settings and any associated therapeutic benefit of doing so are warranted.
Collapse
Affiliation(s)
- Qian Liang
- Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Ernest T Parker
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Gabrielle Dean
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Matthew S Karpen
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Yujia Wu
- Department of Statistics, School of Statistics and Center of Statistical Research, Southwestern University of Finance and Economics, Chengdu, China
| | - Xuefeng Wang
- Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jorge Di Paola
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Cheryl L Maier
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Shannon L Meeks
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Pete Lollar
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Robert F Sidonio
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Renhao Li
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA.
| |
Collapse
|
3
|
Oh D, Chae YJ, Teoh JY, Yim B, Yoo D, Park Y, Kim J. Detection of α-Thrombin with Platelet Glycoprotein Ibα (GP1bα) for the Development of a Coagulation Marker. ACS OMEGA 2024; 9:13418-13426. [PMID: 38524497 PMCID: PMC10956406 DOI: 10.1021/acsomega.4c00010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 02/05/2024] [Accepted: 02/21/2024] [Indexed: 03/26/2024]
Abstract
The detection of prothrombotic markers is crucial for understanding thromboembolism and assessing the effectiveness of anticoagulant drugs. α-Thrombin is a marker that plays a critical role in the coagulation cascade process. However, the detection of this enzymatic molecule was hindered by the absence of an efficient modality in the clinical environment. Previously, we reported that one α-thrombin interacts with two α-chains of glycoprotein Ib (GPIbα), i.e., multivalent protein binding (MPB), using bioresponsive hydrogel nanoparticles (nanogels) and optical microscopy. In this study, we demonstrated that GPIbα-mediated platforms led to the highly sensitive and quantitative detection of α-thrombin in various diagnostic systems. Initially, a bioresponsive nanogel-based surface plasmon resonance (nSPR) assay was developed that responds to the MPB of α-thrombin to GPIbα. The use of GPIbα for the detection of α-thrombin was further validated using the enzyme-linked immunosorbent assay, which is a gold-standard protein detection technique. Additionally, GPIbα-functionalized latex beads were developed to perform latex agglutination (LA) assays, which are widely used with hospital diagnostic instruments. Notably, the nSPR and LA assays exhibited a nearly 1000-fold improvement in sensitivity for α-thrombin detection compared to our previous optical microscopy method. The superiority of our GPIbα-mediated platforms lies in their stability for α-thrombin detection through protein-protein interactions. By contrast, assays relying on α-thrombin enzymatic activity using substrates face the challenge of a rapid decrease in postsample collection. These results suggested that the MPB of α-thrombin to GPIbα is an ideal mode for clinical α-thrombin detection, particularly in outpatient settings.
Collapse
Affiliation(s)
- Doogie Oh
- R&D
Center, Scholar Foxtrot Co. Ltd., Seoul 02796, Republic of Korea
- Department
of Biomedical Sciences, College of Medicine, Korea University, Seoul 02841, Republic
of Korea
| | - Yun Jin Chae
- R&D
Center, Scholar Foxtrot Co. Ltd., Seoul 02796, Republic of Korea
| | - Jie Ying Teoh
- Department
of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Bora Yim
- R&D
Center, Scholar Foxtrot Co. Ltd., Seoul 02796, Republic of Korea
| | - Dongwon Yoo
- Department
of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
- Center
for Nanoparticle Research, Institute for
Basic Science (IBS), Seoul 08826, Republic
of Korea
| | - Yongdoo Park
- Department
of Biomedical Sciences, College of Medicine, Korea University, Seoul 02841, Republic
of Korea
| | - Jongseong Kim
- R&D
Center, Scholar Foxtrot Co. Ltd., Seoul 02796, Republic of Korea
- Department
of Biomedical Sciences, College of Medicine, Korea University, Seoul 02841, Republic
of Korea
| |
Collapse
|
4
|
Enomoto Y, Onuma T, Hori T, Tanabe K, Ueda K, Mizutani D, Doi T, Matsushima-Nishiwaki R, Ogura S, Iida H, Iwama T, Kozawa O, Tokuda H. Synergy by Ristocetin and CXCL12 in Human Platelet Activation: Divergent Regulation by Rho/Rho-Kinase and Rac. Int J Mol Sci 2023; 24:ijms24119716. [PMID: 37298667 DOI: 10.3390/ijms24119716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 05/25/2023] [Accepted: 06/02/2023] [Indexed: 06/12/2023] Open
Abstract
CXCL12, belonging to the CXC chemokine family, is a weak agonist of platelet aggregation. We previously reported that the combination of CXCL12 and collagen at low doses synergistically activates platelets via not CXCR7 but CXCR4, a specific receptor for CXCL12 on the plasma membrane. Recently, we reported that not Rho/Rho kinase, but Rac is involved in the platelet aggregation induced by this combination. Ristocetin is an activator of the von Willebrand factor that interacts with glycoprotein (GP) Ib/IX/V, which generates thromboxane A2 via phospholipase A2 activation, resulting in the release of the soluble CD40 ligand (sCD40L) from human platelets. In the present study, we investigated the effects of a combination of ristocetin and CXCL12 at low doses on human platelet activation and its underlying mechanisms. Simultaneous stimulation with ristocetin and CXCL12 at subthreshold doses synergistically induce platelet aggregation. A monoclonal antibody against not CXCR7 but CXCR4 suppressed platelet aggregation induced by the combination of ristocetin and CXCL12 at low doses. This combination induces a transient increase in the levels of both GTP-binding Rho and Rac, followed by an increase in phosphorylated cofilin. The ristocetin and CXCL12-induced platelet aggregation as well as the sCD40L release were remarkably enhanced by Y27362, an inhibitor of Rho-kinase, but reduced by NSC23766, an inhibitor of the Rac-guanine nucleotide exchange factor interaction. These results strongly suggest that the combination of ristocetin and CXCL12 at low doses synergistically induces human platelet activation via Rac and that this activation is negatively regulated by the simultaneous activation of Rho/Rho-kinase.
Collapse
Affiliation(s)
- Yukiko Enomoto
- Department of Neurosurgery, Gifu University Graduate School of Medicine, Gifu 501-1193, Aichi, Japan
| | - Takashi Onuma
- Department of Anesthesiology and Pain Medicine, Gifu University Graduate School of Medicine, Gifu 501-1193, Aichi, Japan
| | - Takamitsu Hori
- Department of Neurosurgery, Gifu University Graduate School of Medicine, Gifu 501-1193, Aichi, Japan
- Department of Pharmacology, Gifu University Graduate School of Medicine, Gifu 501-1193, Aichi, Japan
- Department of Metabolic Research, Research Institute, National Center for Geriatrics and Gerontology, Obu 474-8511, Aichi, Japan
| | - Kumiko Tanabe
- Department of Anesthesiology and Pain Medicine, Gifu University Graduate School of Medicine, Gifu 501-1193, Aichi, Japan
| | - Kyohei Ueda
- Department of Anesthesiology and Pain Medicine, Gifu University Graduate School of Medicine, Gifu 501-1193, Aichi, Japan
| | - Daisuke Mizutani
- Department of Neurosurgery, Gifu University Graduate School of Medicine, Gifu 501-1193, Aichi, Japan
| | - Tomoaki Doi
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, Gifu 501-1193, Aichi, Japan
| | - Rie Matsushima-Nishiwaki
- Department of Pharmacology, Gifu University Graduate School of Medicine, Gifu 501-1193, Aichi, Japan
| | - Shinji Ogura
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, Gifu 501-1193, Aichi, Japan
| | - Hiroki Iida
- Department of Anesthesiology and Pain Medicine, Gifu University Graduate School of Medicine, Gifu 501-1193, Aichi, Japan
| | - Toru Iwama
- Department of Neurosurgery, Gifu University Graduate School of Medicine, Gifu 501-1193, Aichi, Japan
| | - Osamu Kozawa
- Department of Pharmacology, Gifu University Graduate School of Medicine, Gifu 501-1193, Aichi, Japan
- Department of Metabolic Research, Research Institute, National Center for Geriatrics and Gerontology, Obu 474-8511, Aichi, Japan
| | - Haruhiko Tokuda
- Department of Pharmacology, Gifu University Graduate School of Medicine, Gifu 501-1193, Aichi, Japan
- Department of Metabolic Research, Research Institute, National Center for Geriatrics and Gerontology, Obu 474-8511, Aichi, Japan
- Department of Clinical Laboratory, National Center for Geriatrics and Gerontology, Obu 474-8511, Aichi, Japan
| |
Collapse
|
5
|
Rosado AM, Zhang Y, Choi HK, Chen Y, Ehrlich SM, Jin F, Grakoui A, Evavold BD, Zhu C. Memory in repetitive protein–protein interaction series. APL Bioeng 2023. [DOI: 10.1063/5.0130805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Interactions between proteins coordinate biological processes in an organism and may impact its responses to changing environments and diseases through feedback systems. Feedback systems function by using changes in the past to influence behaviors in the future, which we refer to here as memory. Here, we summarized several observations made, ideas conceptualized, and mathematical models developed for quantitatively analyzing memory effects in repetitive protein–protein interactions (PPIs). Specifically, we consider how proteins on the cell or in isolation retain information about prior interactions to impact current interactions. The micropipette, biomembrane force probe, and atomic force microscopic techniques were used to repeatedly assay PPIs. The resulting time series were analyzed by a previous and two new models to extract three memory indices of short (seconds), intermediate (minutes), and long (hours) timescales. We found that interactions of cell membrane, but not soluble, T cell receptor (TCR) with peptide-major histocompatibility complex (pMHC) exhibits short-term memory that impacts on-rate, but not off-rate of the binding kinetics. Peptide dissociation from MHC resulted in intermediate- and long-term memories in TCR–pMHC interactions. However, we observed no changes in kinetic parameters by repetitive measurements on living cells over intermediate timescales using stable pMHCs. Parameters quantifying memory effects in PPIs could provide additional information regarding biological mechanisms. The methods developed herein also provide tools for future research.
Collapse
Affiliation(s)
- Aaron M. Rosado
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
- Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
- Medical Scientist Training Program, Emory University School of Medicine, Atlanta, Georgia 30332, USA
| | - Yan Zhang
- Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
- Georgia W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
| | - Hyun-Kyu Choi
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
- Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
| | - Yunfeng Chen
- Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
- Georgia W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
| | - Samuel M. Ehrlich
- Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
- Georgia W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
| | - Fengzhi Jin
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia 30332, USA
| | - Arash Grakoui
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia 30332, USA
| | - Brian D. Evavold
- Department of Immunology and Microbiology, Emory University School of Medicine, Atlanta, Georgia 30332 USA
| | - Cheng Zhu
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
- Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
- Georgia W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
| |
Collapse
|
6
|
Shen C, Mackeigan DT, Shoara AA, Xu R, Bhoria P, Karakas D, Ma W, Cerenzia E, Chen Z, Hoard B, Lin L, Lei X, Zhu G, Chen P, Johnson PE, Ni H. Dual roles of fucoidan-GPIbα interaction in thrombosis and hemostasis: implications for drug development targeting GPIbα. JOURNAL OF THROMBOSIS AND HAEMOSTASIS : JTH 2023; 21:1274-1288. [PMID: 36732162 DOI: 10.1016/j.jtha.2022.12.030] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 12/14/2022] [Accepted: 12/27/2022] [Indexed: 01/11/2023]
Abstract
BACKGROUND Platelet GPIbα-von Willebrand factor (VWF) interaction initiates platelet adhesion, activation, and thrombus growth, especially under high shear conditions. Therefore, the GPIb-VWF axis has been suggested as a promising target against arterial thrombosis. The polysaccharide fucoidan has been reported to have opposing prothrombotic and antithrombotic effects; however, its binding mechanism with platelets has not been adequately studied. OBJECTIVE The objective of this study was to explore the mechanism of fucoidan and its hydrolyzed products in thrombosis and hemostasis. METHODS Natural fucoidan was hydrolyzed by using hydrochloric acid and was characterized by using size-exclusion chromatography, UV-visible spectroscopy, and fluorometry techniques. The effects of natural and hydrolyzed fucoidan on platelet aggregation were examined by using platelets from wild-type, VWF and fibrinogen-deficient, GPIbα-deficient, and IL4Rα/GPIbα-transgenic and αIIb-deficient mice and from human beings. Platelet activation markers (P-selectin expression, PAC-1, and fibrinogen binding) and platelet-VWF A1 interaction were measured by using flow cytometry. GPIbα-VWF A1 interaction was evaluated by using enzyme-linked immunosorbent assay. GPIb-IX-induced signal transduction was detected by using western blot. Heparinized whole blood from healthy donors was used to test thrombus formation and growth in a perfusion chamber. RESULTS We found that GPIbα is critical for fucoidan-induced platelet activation. Fucoidan interacted with the extracellular domain of GPIbα and blocked its interaction with VWF but itself could lead to GPIbα-mediated signal transduction and, subsequently, αIIbβ3 activation and platelet aggregation. Conversely, low-molecular weight fucoidan inhibited GPIb-VWF-mediated platelet aggregation, spreading, and thrombus growth at high shear. CONCLUSION Fucoidan-GPIbα interaction may have unique therapeutic potential against bleeding disorders in its high-molecular weight state and protection against arterial thrombosis by blocking GPIb-VWF interaction after fucoidan is hydrolyzed.
Collapse
Affiliation(s)
- Chuanbin Shen
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, M5S 1A1, ON, Canada; Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Shandong, China; Department of Laboratory Medicine, LKSKI-Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Toronto, Canada
| | - Daniel T Mackeigan
- Department of Laboratory Medicine, LKSKI-Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Toronto, Canada; Department of Physiology, University of Toronto, Toronto, Canada
| | - Aron A Shoara
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, M5S 1A1, ON, Canada; Department of Laboratory Medicine, LKSKI-Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Toronto, Canada
| | - Runjia Xu
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, M5S 1A1, ON, Canada; Department of Laboratory Medicine, LKSKI-Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Toronto, Canada
| | - Preeti Bhoria
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, M5S 1A1, ON, Canada; Department of Laboratory Medicine, LKSKI-Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Toronto, Canada; CCOA Therapeutics Inc Toronto, Canada
| | - Danielle Karakas
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, M5S 1A1, ON, Canada; Department of Laboratory Medicine, LKSKI-Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Toronto, Canada
| | - Wenjing Ma
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, M5S 1A1, ON, Canada; Department of Laboratory Medicine, LKSKI-Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Toronto, Canada; CCOA Therapeutics Inc Toronto, Canada
| | - Eric Cerenzia
- Department of Laboratory Medicine, LKSKI-Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Toronto, Canada; Department of Physiology, University of Toronto, Toronto, Canada
| | - ZiYan Chen
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, M5S 1A1, ON, Canada; Department of Laboratory Medicine, LKSKI-Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Toronto, Canada
| | - Brock Hoard
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, M5S 1A1, ON, Canada; Department of Laboratory Medicine, LKSKI-Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Toronto, Canada
| | - Lisha Lin
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, M5S 1A1, ON, Canada; Department of Laboratory Medicine, LKSKI-Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Toronto, Canada; State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, China
| | - Xi Lei
- Department of Laboratory Medicine, LKSKI-Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Toronto, Canada; CCOA Therapeutics Inc Toronto, Canada
| | - Guangheng Zhu
- Department of Laboratory Medicine, LKSKI-Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Toronto, Canada; CCOA Therapeutics Inc Toronto, Canada
| | - Pingguo Chen
- Department of Laboratory Medicine, LKSKI-Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Toronto, Canada; Canadian Blood Services Centre for Innovation, Toronto, Canada
| | - Philip E Johnson
- Department of Chemistry and Centre for Research on Biomolecular Interactions, York University, Toronto, Canada
| | - Heyu Ni
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, M5S 1A1, ON, Canada; Department of Laboratory Medicine, LKSKI-Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Toronto, Canada; Department of Physiology, University of Toronto, Toronto, Canada; CCOA Therapeutics Inc Toronto, Canada; Canadian Blood Services Centre for Innovation, Toronto, Canada; Department of Medicine, University of Toronto, Toronto, Canada.
| |
Collapse
|
7
|
Nakayama M, Goto S, Goto S. Physical Characteristics of von Willebrand Factor Binding with Platelet Glycoprotein Ibɑ Mutants at Residue 233 Causing Various Biological Functions. TH OPEN : COMPANION JOURNAL TO THROMBOSIS AND HAEMOSTASIS 2022; 6:e421-e428. [PMID: 36632284 PMCID: PMC9729063 DOI: 10.1055/a-1937-9940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 08/05/2022] [Indexed: 01/14/2023]
Abstract
Glycoprotein (GP: HIS 1 -PRO 265 ) Ibɑ is a receptor protein expressed on the surface of the platelet. Its N-terminus domain binds with the A1 domain (ASP 1269 -PRO 1472 ) of its ligand protein von Willebrand factor (VWF) and plays a unique role in platelet adhesion under blood flow conditions. Single amino acid substitutions at residue 233 from glycine (G) to alanine (A), aspartic acid (D), or valine (V) are known to cause biochemically distinct functional alterations known as equal, loss, and gain of function, respectively. However, the underlying physical characteristics of VWF binding with GPIbɑ in wild-type and the three mutants exerting different biological functions are unclear. Here, we aimed to test the hypothesis: biological characteristics of macromolecules are influenced by small changes in physical parameters. The position coordinates and velocity vectors of all atoms and water molecules constructing the wild-type and the three mutants of GPIbɑ (G233A, G233D, and G233V) bound with VWF were calculated every 2 × 10 -15 seconds using the CHARMM (Chemistry at Harvard Macromolecular Mechanics) force field for 9 × 10 -10 seconds. Six salt bridges were detected for longer than 50% of the calculation period for the wild-type model generating noncovalent binding energy of -1096 ± 137.6 kcal/mol. In contrast, only four pairs of salt bridges were observed in G233D mutant with noncovalent binding energy of -865 ± 139 kcal/mol. For G233A and G233V, there were six and five pairs of salt bridges generating -929.8 ± 88.5 and -989.9 ± 94.0 kcal/mol of noncovalent binding energy, respectively. Our molecular dynamic simulation showing a lower probability of salt bridge formation with less noncovalent binding energy in VWF binding with the biologically loss of function G233D mutant of GPIbɑ as compared with wild-type, equal function, and gain of function mutant suggests that biological functions of macromolecules such as GPIbɑ are influenced by their small changes in physical characteristics.
Collapse
Affiliation(s)
- Masamitsu Nakayama
- Department of Medicine (Cardiology), Tokai University School of Medicine, Isehara, Japan
| | - Shinichi Goto
- Department of Medicine (Cardiology), Tokai University School of Medicine, Isehara, Japan
| | - Shinya Goto
- Department of Medicine (Cardiology), Tokai University School of Medicine, Isehara, Japan,Address for correspondence Shinya Goto, MD, PhD Department of Medicine (Cardiology), Tokai University School of Medicine143 Shimokasuya, IseharaJapan
| |
Collapse
|
8
|
Arce NA, Liu Y, Chen W, Zhang XF, Li R. Autoinhibitory module underlies species difference in shear activation of von Willebrand factor. J Thromb Haemost 2022; 20:2686-2696. [PMID: 36031939 PMCID: PMC9588639 DOI: 10.1111/jth.15837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 07/05/2022] [Accepted: 07/22/2022] [Indexed: 11/30/2022]
Abstract
BACKGROUND Von Willebrand factor (VWF) is a multimeric plasma protein that bridges the gap between vessel injury and platelet capture at high shear rates. Under high shear or tension, VWF can become activated upon the unfolding of its autoinhibitory module (AIM). AIM unfolding exposes the A1 domain, allowing for binding to platelet glycoprotein (GP)Ibα to initiate primary hemostasis. The characteristics of the AIM and its inhibitory properties within mouse VWF are unknown. OBJECTIVES To determine and characterize the autoinhibitory properties of mouse VWF. METHODS Recombinant mouse VWF A1 fragments containing or lacking the flanking regions around the A1 domain were generated. We tested the ability of these fragments to bind to human or mouse GPIbα and platelets. We compared the unfolding of mouse AIM-A1 to human AIM-A1 by single-molecule force spectroscopy. RESULTS Recombinant mouse AIM-A1 binds with higher affinity to GPIbα than its human counterpart. Recombinant mouse proteins lacking part of the AIM show increased binding to GPIbα. Activated A1 fragments lacking the AIM can effectively agglutinate platelets across the species barrier. Using single-molecule force spectroscopy, we determined that the mouse AIM unfolds under forces similar to the human AIM. Additionally, the human AIM paired with mouse A1 largely recapitulates the behavior of human AIM-A1. CONCLUSIONS Our results suggest that the regulation of VWF-GPIbα binding has been specifically tuned to work optimally in different rheological architectures. Differences in the AIM sequence may contribute to the difference in VWF shear response between human and mice.
Collapse
Affiliation(s)
- Nicholas A. Arce
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Yi Liu
- Department of Bioengineering, Lehigh University, Bethlehem, Pennsylvania, USA
| | - Wenchun Chen
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - X. Frank Zhang
- Department of Bioengineering, Lehigh University, Bethlehem, Pennsylvania, USA
| | - Renhao Li
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
9
|
Drabeck DH, Rucavado A, Hingst-Zaher E, Dean A, Jansa SA. Ancestrally Reconstructed von Willebrand Factor Reveals Evidence for Trench Warfare Coevolution between Opossums and Pit Vipers. Mol Biol Evol 2022; 39:msac140. [PMID: 35723968 PMCID: PMC9255381 DOI: 10.1093/molbev/msac140] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Opossums in the tribe Didelphini are resistant to pit viper venoms and are hypothesized to be coevolving with venomous snakes. Specifically, a protein involved in blood clotting (von Willebrand factor [vWF] which is targeted by snake venom C-type lectins [CTLs]) has been found to undergo rapid adaptive evolution in Didelphini. Several unique amino acid changes in vWF could explain their resistance; however, experimental evidence that these changes disrupt binding to venom CTLs was lacking. Furthermore, without explicit testing of ancestral phenotypes to reveal the mode of evolution, the assertion that this system represents an example of coevolution rather than noncoevolutionary adaptation remains unsupported. Using expressed vWF proteins and purified venom CTLs, we quantified binding affinity for vWF proteins from all resistant taxa, their venom-sensitive relatives, and their ancestors. We show that CTL-resistant vWF is present in opossums outside clade Didelphini and likely across a wider swath of opossums (family Didelphidae) than previously thought. Ancestral reconstruction and in vitro testing of vWF phenotypes in a clade of rapidly evolving opossums reveal a pattern consistent with trench warfare coevolution between opossums and their venomous snake prey.
Collapse
Affiliation(s)
- Danielle H Drabeck
- Department of Ecology, Evolution, and Behavior, University of Minnesota, 1479 Gortner Ave., St Paul, MN 55108, USA
- Bell Museum, University of Minnesota, 1987 Upper Buford Circle, St. Paul, MN 55108, USA
| | - Alexandra Rucavado
- Instituto Clodomiro Picado, Facultad de Microbiología, Universidad de Costa Rica, San José, Costa Rica
| | - Erika Hingst-Zaher
- Museu Biológico, Instituto Butantan, CEP 05503-900 São Paulo, SP, Brazil
| | - Antony Dean
- Department of Ecology, Evolution, and Behavior, University of Minnesota, 1479 Gortner Ave., St Paul, MN 55108, USA
| | - Sharon A Jansa
- Department of Ecology, Evolution, and Behavior, University of Minnesota, 1479 Gortner Ave., St Paul, MN 55108, USA
- Bell Museum, University of Minnesota, 1987 Upper Buford Circle, St. Paul, MN 55108, USA
| |
Collapse
|
10
|
Yang M, Houck KL, Dong X, Hernandez M, Wang Y, Nathan SS, Wu X, Afshar-Kharghan V, Fu X, Cruz MA, Zhang J, Nascimbene A, Dong JF. Hyperadhesive von Willebrand Factor Promotes Extracellular Vesicle-Induced Angiogenesis: Implication for LVAD-Induced Bleeding. JACC Basic Transl Sci 2022; 7:247-261. [PMID: 35411318 PMCID: PMC8993768 DOI: 10.1016/j.jacbts.2021.12.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 12/13/2021] [Accepted: 12/15/2021] [Indexed: 11/22/2022]
Abstract
VWF in patients on LVAD supports was hyperadhesive, activated platelets, and generated platelet-derived extracellular vesicles. Extracellular vesicles from LVAD patients and those from shear-activated platelets promoted aberrant angiogenesis in a VWF-dependent manner. The activated VWF exposed the A1 domain through the synergistic actions of oxidative stress and HSS generated in LVAD-driven circulation.
Bleeding associated with left ventricular assist device (LVAD) implantation has been attributed to the loss of large von Willebrand factor (VWF) multimers to excessive cleavage by ADAMTS-13, but this mechanism is not fully supported by the current evidence. We analyzed VWF reactivity in longitudinal samples from LVAD patients and studied normal VWF and platelets exposed to high shear stress to show that VWF became hyperadhesive in LVAD patients to induce platelet microvesiculation. Platelet microvesicles activated endothelial cells, induced vascular permeability, and promoted angiogenesis in a VWF-dependent manner. Our findings suggest that LVAD-driven high shear stress primarily activates VWF, rather than inducing cleavage in the majority of patients.
Collapse
Key Words
- ADAMTS-13:Ag, ADAMTS-13 antigen
- AVS, aortic vascular segment
- EC, endothelial cell
- EV, extracellular vesicle
- EVFP, extracellular vesicle–free plasma
- GI, gastrointestinal
- GOF, gain of function
- GP, glycoprotein
- GPM, growth factor-poor medium
- GRM, growth factor-rich medium
- HSS, high shear stress
- LVAD, left ventricular assist device
- PS, phosphatidylserine
- SIPA, shear-induced platelet aggregation
- ULVWF, ultra-large von Willebrand factor
- VEGF, vascular endothelial growth factor
- VWF, von Willebrand factor
- VWF:Ag, von Willebrand factor antigen
- VWF:CB, von Willebrand factor binding to collagen
- VWF:pp, von Willebrand factor propeptide
- aVWS, acquired von Willebrand syndrome
- angiogenesis
- extracellular vesicles
- left ventricular assist devices
- pEV, extracellular vesicle from von Willebrand factor-activated platelets
- platelets
- shear stress
- von Willebrand factor
Collapse
Affiliation(s)
- Mengchen Yang
- Bloodworks Research Institute, Seattle, Washington, USA.,Department of Urology, Tianjin Medical University General Hospital, Tianjin, China
| | - Katie L Houck
- Bloodworks Research Institute, Seattle, Washington, USA
| | - Xinlong Dong
- Bloodworks Research Institute, Seattle, Washington, USA
| | - Maria Hernandez
- Center for Advanced Heart Failure, University of Texas at Houston, Houston, Texas, USA
| | - Yi Wang
- Bloodworks Research Institute, Seattle, Washington, USA
| | - Sriram S Nathan
- Center for Advanced Heart Failure, University of Texas at Houston, Houston, Texas, USA
| | - Xiaoping Wu
- Department of Pathology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Vahid Afshar-Kharghan
- Division of Internal Medicine, Department of Pulmonary Medicine, MD Anderson Cancer Center, University of Texas, Houston, Texas, USA
| | - Xiaoyun Fu
- Bloodworks Research Institute, Seattle, Washington, USA
| | - Miguel A Cruz
- Cardiovascular Research Section, Department of Medicine, Baylor College of Medicine.,Center for Translational Research on Inflammatory Diseases, Michael E. DeBakey VA Medical Center, Houston, Texas, USA
| | - Jianning Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Angelo Nascimbene
- Center for Advanced Heart Failure, University of Texas at Houston, Houston, Texas, USA
| | - Jing-Fei Dong
- Bloodworks Research Institute, Seattle, Washington, USA.,Division of Hematology, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
| |
Collapse
|
11
|
Hoefer T, Rana A, Niego B, Jagdale S, Albers HJ, Gardiner EE, Andrews RK, Van der Meer AD, Hagemeyer CE, Westein E. Targeting shear gradient activated von Willebrand factor by the novel single-chain antibody A1 reduces occlusive thrombus formation in vitro. Haematologica 2021; 106:2874-2884. [PMID: 33054112 PMCID: PMC8561297 DOI: 10.3324/haematol.2020.250761] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Indexed: 12/12/2022] Open
Abstract
Intraluminal thrombus formation precipitates conditions such as acute myocardial infarction and disturbs local blood flow resulting in areas of rapidly changing blood flow velocities and steep gradients of blood shear rate. Shear rate gradients are known to be pro-thrombotic with an important role for the shear-sensitive plasma protein von Willebrand factor (VWF). Here, we developed a single-chain antibody (scFv) that targets a shear gradient specific conformation of VWF to specifically inhibit platelet adhesion at sites of SRGs but not in areas of constant shear. Microfluidic flow channels with stenotic segments were used to create shear rate gradients during blood perfusion. VWF-GPIbα interactions were increased at sites of shear rate gradients compared to constant shear rate of matched magnitude. The scFv-A1 specifically reduced VWF-GPIbα binding and thrombus formation at sites of SRGs but did not block platelet deposition and aggregation under constant shear rate in upstream sections of the channels. Significantly, the scFv A1 attenuated platelet aggregation only in the later stages of thrombus formation. In the absence of shear, direct binding of scFv-A1 to VWF could not be detected and scFV-A1 did not inhibit ristocetin induced platelet agglutination. We have exploited the pro-aggregatory effects of SRGs on VWF dependent platelet aggregation and developed the shear-gradient sensitive scFv-A1 antibody that inhibits platelet aggregation exclusively at sites of shear rate gradients. The lack of VWF inhibition in non-stenosed vessel segments places scFV-A1 in an entirely new class of anti-platelet therapy for selective blockade of pathological thrombus formation while maintaining normal haemostasis.
Collapse
Affiliation(s)
- Thomas Hoefer
- Baker Heart and Diabetes Institute, Melbourne; TH and AR contributed equally to this work.
| | - Akshita Rana
- Australian Centre for Blood Diseases, Monash University, Melbourne; TH and AR contributed equally to this work.
| | - Be'eri Niego
- Australian Centre for Blood Diseases, Monash University, Melbourne.
| | - Shweta Jagdale
- Baker Heart and Diabetes Institute, Melbourne; Australian Centre for Blood Diseases, Monash University, Melbourne.
| | - Hugo J Albers
- Applied Stem Cell Technologies, University of Twente, Enschede; BIOS Lab-on-a-Chip, University of Twente, Enschede.
| | - Elizabeth E Gardiner
- ACRF Department of Cancer Biology and Therapeutics, John Curtin School of Medical Research, Australian National University, Canberra.
| | - Robert K Andrews
- Australian Centre for Blood Diseases, Monash University, Melbourne.
| | | | - Christoph E Hagemeyer
- Baker Heart and Diabetes Institute, Melbourne; Australian Centre for Blood Diseases, Monash University, Melbourne.
| | - Erik Westein
- Baker Heart and Diabetes Institute, Melbourne; Australian Centre for Blood Diseases, Monash University, Melbourne; CEH and EW contributed equally to this work.
| |
Collapse
|
12
|
Arce NA, Cao W, Brown AK, Legan ER, Wilson MS, Xu ER, Berndt MC, Emsley J, Zhang XF, Li R. Activation of von Willebrand factor via mechanical unfolding of its discontinuous autoinhibitory module. Nat Commun 2021; 12:2360. [PMID: 33883551 PMCID: PMC8060278 DOI: 10.1038/s41467-021-22634-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 03/16/2021] [Indexed: 01/05/2023] Open
Abstract
Von Willebrand factor (VWF) activates in response to shear flow to initiate hemostasis, while aberrant activation could lead to thrombosis. Above a critical shear force, the A1 domain of VWF becomes activated and captures platelets via the GPIb-IX complex. Here we show that the shear-responsive element controlling VWF activation resides in the discontinuous autoinhibitory module (AIM) flanking A1. Application of tensile force in a single-molecule setting induces cooperative unfolding of the AIM to expose A1. The AIM-unfolding force is lowered by truncating either N- or C-terminal AIM region, type 2B VWD mutations, or binding of a ristocetin-mimicking monoclonal antibody, all of which could activate A1. Furthermore, the AIM is mechanically stabilized by the nanobody that comprises caplacizumab, the only FDA-approved anti-thrombotic drug to-date that targets VWF. Thus, the AIM is a mechano-regulator of VWF activity. Its conformational dynamics may define the extent of VWF autoinhibition and subsequent activation under force.
Collapse
Affiliation(s)
- Nicholas A Arce
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Wenpeng Cao
- Department of Bioengineering, Department of Mechanical Engineering & Mechanics, Lehigh University, Bethlehem, PA, USA
| | - Alexander K Brown
- Biodiscovery Institute, School of Pharmacy, University of Nottingham, Nottingham, UK
| | - Emily R Legan
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Moriah S Wilson
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Emma-Ruoqi Xu
- Biodiscovery Institute, School of Pharmacy, University of Nottingham, Nottingham, UK
| | - Michael C Berndt
- Faculty of Health Sciences, Curtin University, Perth, WA, Australia
| | - Jonas Emsley
- Biodiscovery Institute, School of Pharmacy, University of Nottingham, Nottingham, UK
| | - X Frank Zhang
- Department of Bioengineering, Department of Mechanical Engineering & Mechanics, Lehigh University, Bethlehem, PA, USA.
| | - Renhao Li
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
13
|
Xu X, Wang C, Wu Y, Houck K, Hilton T, Zhou A, Wu X, Han C, Yang M, Yang W, Shi FD, Stolla M, Cruz MA, Li M, Zhang J, Dong JF. Conformation-dependent blockage of activated VWF improves outcomes of traumatic brain injury in mice. Blood 2021; 137:544-555. [PMID: 33507292 PMCID: PMC7845006 DOI: 10.1182/blood.2020007364] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 07/27/2020] [Indexed: 12/22/2022] Open
Abstract
Traumatic brain injury-induced coagulopathy (TBI-IC) causes life-threatening secondary intracranial bleeding. Its pathogenesis differs mechanistically from that of coagulopathy arising from extracranial injuries and hemorrhagic shock, but it remains poorly understood. We report results of a study designed to test the hypothesis that von Willebrand factor (VWF) released during acute TBI is intrinsically hyperadhesive because its platelet-binding A1-domain is exposed and contributes to TBI-induced vascular leakage and consumptive coagulopathy. This hyperadhesive VWF can be selectively blocked by a VWF A2-domain protein to prevent TBI-IC and to improve neurological function with a minimal risk of bleeding. We demonstrated that A2 given through intraperitoneal injection or IV infusion reduced TBI-induced death by >50% and significantly improved the neurological function of C57BL/6J male mice subjected to severe lateral fluid percussion injury. A2 protected the endothelium from extracellular vesicle-induced injury, reducing TBI-induced platelet activation and microvesiculation, and preventing a TBI-induced hypercoagulable state. A2 achieved this therapeutic efficacy by specifically blocking the A1 domain exposed on the hyperadhesive VWF released during acute TBI. These results suggest that VWF plays a causal role in the development of TBI-IC and is a therapeutic target for this life-threatening complication of TBI.
Collapse
Affiliation(s)
- Xin Xu
- Bloodworks Research Institute, Seattle, WA
- Departments of Neurosurgery, Neurology, and Obstetrics & Gynecology, Tianjin Medical University General Hospital, Tianjin, China
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Chenyu Wang
- Institute of Pathology, School of Medical Sciences, and the Gansu Provincial Key Laboratory of Preclinical Study for New Drug Development, Lanzhou University, Lanzhou, China
| | - Yingang Wu
- Department of Neurosurgery, the First Affiliated Hospital, University of Science and Technology, Hefei, China
| | | | | | | | | | - Cha Han
- Departments of Neurosurgery, Neurology, and Obstetrics & Gynecology, Tianjin Medical University General Hospital, Tianjin, China
| | - Mengchen Yang
- Departments of Neurosurgery, Neurology, and Obstetrics & Gynecology, Tianjin Medical University General Hospital, Tianjin, China
| | - Wei Yang
- Bloodworks Research Institute, Seattle, WA
- NanoString Technologies, Seattle, WA
| | - Fu-Dong Shi
- Departments of Neurosurgery, Neurology, and Obstetrics & Gynecology, Tianjin Medical University General Hospital, Tianjin, China
| | | | - Miguel A Cruz
- Cardiovascular Research Section, Department of Medicine, Baylor College of Medicine, Houston, TX
- Center for Translational Research on Inflammatory Diseases, Michael E. DeBakey Veterans Affairs (VA) Medical Center, Houston, TX; and
| | - Min Li
- Institute of Pathology, School of Medical Sciences, and the Gansu Provincial Key Laboratory of Preclinical Study for New Drug Development, Lanzhou University, Lanzhou, China
| | - Jianning Zhang
- Departments of Neurosurgery, Neurology, and Obstetrics & Gynecology, Tianjin Medical University General Hospital, Tianjin, China
| | - Jing-Fei Dong
- Bloodworks Research Institute, Seattle, WA
- Division of Hematology, Department of Medicine, University of Washington, School of Medicine, Seattle, WA
| |
Collapse
|
14
|
Drabeck DH, Rucavado A, Hingst-Zaher E, Cruz YP, Dean AM, Jansa SA. Resistance of South American opossums to vWF-binding venom C-type lectins. Toxicon 2020; 178:92-99. [PMID: 32135198 PMCID: PMC8522506 DOI: 10.1016/j.toxicon.2020.02.024] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 02/12/2020] [Accepted: 02/29/2020] [Indexed: 10/24/2022]
Abstract
Opossums in the clade Didelphini are well known to be resistant to snake venom due to endogenous circulating inhibitors which target metalloproteinases and phospholipases. However, the mechanisms through which these opossums cope with a variety of other damaging venom proteins are unknown. A protein involved in blood clotting (von Willebrand Factor) has been found to have undergone rapid adaptive evolution in venom-resistant opossums. This protein is a known target for a subset of snake venom C-type lectins (CTLs), which bind it and then induce it to bind platelets, causing hemostatic disruption. Several amino acid changes in vWF unique to these opossums could explain their resistance; however, experimental evidence that these changes disrupt venom CTL binding was lacking. We used platelet aggregation assays to quantify resistance to a venom-induced platelet response in two species of venom-resistant opossums (Didelphis virginiana, Didelphis aurita), and one venom-sensitive opossum (Monodelphis domestica). We found that all three species have lost nearly all their aggregation response to the venom CTLs tested. Using washed platelet assays we showed that this loss of aggregation response is not due to inhibitors in the plasma, but rather to the failure of either vWF or platelets (or both) to respond to venom CTLs. These results demonstrate the potential adaptive function of a trait previously shown to be evolving under positive selection. Surprisingly, these findings also expand the list of potentially venom tolerant species to include Monodelphis domestica and suggest that an ecological relationship between opossums and vipers may be a broader driver of adaptive evolution across South American marsupials than previously thought.
Collapse
Affiliation(s)
- Danielle H Drabeck
- Department of Ecology, Evolution, and Behavior, University of Minnesota, 1479 Gortner Ave., St Paul, MN, 55108, USA; Bell Museum of Natural History, University of Minnesota, 1987 Upper Buford Circle, St. Paul, MN, 55108, USA.
| | - Alexandra Rucavado
- Instituto Clodomiro Picado, Facultad de Microbiología, Universidad de Costa Rica, San José, Costa Rica
| | - Erika Hingst-Zaher
- Museu Biológico, Instituto Butantan, CEP 05503-900, São Paulo, SP, Brazil
| | - Yolanda P Cruz
- Department of Biology, Oberlin College, Oberlin, OH, 44074, USA
| | - Antony M Dean
- Department of Ecology, Evolution, and Behavior, University of Minnesota, 1479 Gortner Ave., St Paul, MN, 55108, USA
| | - Sharon A Jansa
- Department of Ecology, Evolution, and Behavior, University of Minnesota, 1479 Gortner Ave., St Paul, MN, 55108, USA; Bell Museum of Natural History, University of Minnesota, 1987 Upper Buford Circle, St. Paul, MN, 55108, USA
| |
Collapse
|
15
|
Goto S, Oka H, Ayabe K, Yabushita H, Nakayama M, Hasebe T, Yokota H, Takagi S, Sano M, Tomita A, Goto S. Prediction of binding characteristics between von Willebrand factor and platelet glycoprotein Ibα with various mutations by molecular dynamic simulation. Thromb Res 2019; 184:129-135. [PMID: 31739151 DOI: 10.1016/j.thromres.2019.10.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Revised: 10/12/2019] [Accepted: 10/21/2019] [Indexed: 11/29/2022]
Abstract
INTRODUCTION Binding of platelet glycoprotein (GP)Ibα with von-Willebrand factor (VWF) exclusively mediates the initial platelet adhesion to injured vessel wall. To understand the mechanism of biomedical functions, we calculated the dynamic fluctuating three-dimensional (3D) structures and dissociation energy for GPIbα with various single amino-acid substitution at G233, which location is known to cause significant changes in platelet adhesive characteristics. MATERIAL AND METHODS Molecular dynamics (MD) simulation was utilized to calculate 3D structures and Potential of Mean Force (PMF) for wild-type VWF bound with wild-type, G233A (equal function), G233V (gain of function), and G233D (loss of function) GPIbα. Simulation was done on water-soluble condition with time-step of 2 × 10-15 s using NAnoscale Molecular Dynamics (NAMD) with Chemistry at HARvard Molecular Mechanics (CHARMM) force field. Initial structure for each mutant was obtained by inducing single amino-acid substitution to the stable water-soluble binding structure of wild-type. RESULTS The most stable structures of wild-type VWF bound to GPIbα in wild-type or any mutant did not differ. However, bond dissociation energy defined as difference of PMF between most stable structure and the structure at 65 Å mass center distances in G233D was 4.32 kcal/mol (19.5%) lower than that of wild-type. Approximately, 2.07 kcal/mol energy was required to dissociate VWF from GPIbα with G233V at mass center distance from 48 to 52 Å, which may explain the apparent "gain of function" in G233V. CONCLUSION The mechanism of substantially different biochemical characteristics of GPIbα with mutations in G233 location was predicted from physical movement of atoms constructing these proteins.
Collapse
Affiliation(s)
- Shinichi Goto
- Department of Medicine (Cardiology), Tokai University School of Medicine, Metabolic Disease Research Center, Tokai University Graduate School of Medicine, Isehara, Japan; Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - Hideki Oka
- Department of Medicine (Cardiology), Tokai University School of Medicine, Metabolic Disease Research Center, Tokai University Graduate School of Medicine, Isehara, Japan
| | - Kengo Ayabe
- Department of Medicine (Cardiology), Tokai University School of Medicine, Metabolic Disease Research Center, Tokai University Graduate School of Medicine, Isehara, Japan
| | - Hiroto Yabushita
- Department of Medicine (Cardiology), Tokai University School of Medicine, Metabolic Disease Research Center, Tokai University Graduate School of Medicine, Isehara, Japan
| | - Masamitsu Nakayama
- Department of Medicine (Cardiology), Tokai University School of Medicine, Metabolic Disease Research Center, Tokai University Graduate School of Medicine, Isehara, Japan
| | - Terumitsu Hasebe
- Department of Radiology, Tokai University Hachioji Hospital, Tokai University School of Medicine, Hachioji, Tokyo, Japan
| | - Hideo Yokota
- Image Processing Research Team, Center for Advanced Photonics Extreme Photonics Research, RIKEN, Wako, Japan
| | - Shu Takagi
- Department of Mechanical Engineering, The University of Tokyo, Tokyo, Japan
| | - Motoaki Sano
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - Aiko Tomita
- Department of Clinical Pharmacology, Tokai University School of Medicine, Isehara, Japan
| | - Shinya Goto
- Department of Medicine (Cardiology), Tokai University School of Medicine, Metabolic Disease Research Center, Tokai University Graduate School of Medicine, Isehara, Japan.
| |
Collapse
|
16
|
Ngo T, Kim K, Bian Y, Nam G, Park HJ, Lee K, Cho GS, Ryu JM, Lim KM, Chung JH. Antithrombotic effect of SP-8008, a benzoic acid derivative, through the selective inhibition of shear stress-induced platelet aggregation. Br J Pharmacol 2019; 177:929-944. [PMID: 31648364 PMCID: PMC7024737 DOI: 10.1111/bph.14894] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Revised: 07/30/2019] [Accepted: 09/05/2019] [Indexed: 01/23/2023] Open
Abstract
Background and Purpose Bleeding is one of the most critical adverse effects of antithrombotic drugs, and many efforts have been made to discover novel antiplatelet agents without bleeding complications. Shear stress‐induced platelet aggregation (SIPA), where the interaction of von Willebrand factor (vWF) and platelet glycoprotein (GP) Ib constitutes the initial step, is a promising target to overcome bleeding problems, as SIPA occurs only in pathological conditions. Here, we describe SP‐8008, a novel modulator of vWF–GP Ib interactions and evaluated its antiplatelet/antithrombotic effects. Experimental Approach Newly synthesized compounds were screened for antiplatelet effects in vitro, using human platelets exposed to high shear stress. Aggregation, intracellular calcium level, granule secretion, and integrin activation were assessed. Molecular modelling using virtual docking and flow cytometry were used to evaluate effects on vWF–GP Ib interactions. Antithrombotic effects in vivo were determined in rats, using arterial thrombosis and shear stress‐specific thrombosis. Transection tail bleeding time was used to evaluate adverse effects. Key Results SP‐8008 was a potent inhibitor of SIPA, with IC50 of 1.44 ± 0.09 μM. SP‐8008 effectively and broadly blocked shear stress‐induced platelet activation events, without any significant toxicity. Importantly, SP‐8008 was highly selective against SIPA, effectively interfering with vWF–GP Ib engagement. Most importantly, SP‐8008 exerted significant antithrombotic effects in vivo in both shear stress‐specific and arterial thrombosis, without prolonging bleeding time. Conclusions and Implications Our results demonstrated that SP‐8008 can be a novel selective antiplatelet agent with improved safety profile.
Collapse
Affiliation(s)
- Thien Ngo
- College of Pharmacy, Seoul National University, Seoul, Korea
| | - Keunyoung Kim
- College of Pharmacy, Seoul National University, Seoul, Korea
| | - Yiying Bian
- College of Pharmacy, Seoul National University, Seoul, Korea
| | - Gibeom Nam
- School of Pharmacy, Sungkyunkwan University, Suwon, Korea
| | - Hyun-Ju Park
- School of Pharmacy, Sungkyunkwan University, Suwon, Korea
| | - Kiho Lee
- College of Pharmacy, Korea University, Sejong, Korea
| | - Geum-Sil Cho
- Research Headquarters, Shin Poong Pharm. Co. Ltd., Ansan, Korea
| | - Jei-Man Ryu
- Research Headquarters, Shin Poong Pharm. Co. Ltd., Ansan, Korea
| | - Kyung-Min Lim
- College of Pharmacy, Ewha Womans University, Seoul, Korea
| | - Jin-Ho Chung
- College of Pharmacy, Seoul National University, Seoul, Korea
| |
Collapse
|
17
|
Ngo T, Kim K, Bian Y, Noh H, Lim KM, Chung JH, Bae ON. Antithrombotic Effects of Paeoniflorin from Paeonia suffruticosa by Selective Inhibition on Shear Stress-Induced Platelet Aggregation. Int J Mol Sci 2019; 20:ijms20205040. [PMID: 31614534 PMCID: PMC6834133 DOI: 10.3390/ijms20205040] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 09/30/2019] [Accepted: 10/09/2019] [Indexed: 12/16/2022] Open
Abstract
Antiplatelet agents are important in the pharmacotherapeutic regime for many cardiovascular diseases, including thrombotic disorders. However, bleeding, the most serious adverse effect associated with current antiplatelet therapy, has led to many efforts to discover novel anti-platelet drugs without bleeding issues. Of note, shear stress-induced platelet aggregation (SIPA) is a promising target to overcome bleeding since SIPA happens only in pathological conditions. Accordingly, this study was carried out to discover antiplatelet agents selectively targeting SIPA. By screening various herbal extracts, Paeonia suffruticosa and its major bioactive constituent, paeoniflorin, were identified to have significant inhibitory effects against shear-induced aggregation in human platelets. The effects of paeoniflorin on intraplatelet calcium levels, platelet degranulation, and integrin activation in high shear stress conditions were evaluated by a range of in vitro experiments using human platelets. The inhibitory effect of paeoniflorin was determined to be highly selective against SIPA, through modulating von Willebrand Factor (vWF)-platelet glycoprotein Ib (GP Ib) interaction. The effects of paeoniflorin on platelet functions under high shear stress were confirmed in the ex vivo SIPA models in rats, showing the good accordance with the anti-SIPA effects on human platelets. Treatment with paeoniflorin significantly prevented arterial thrombosis in vivo from the dose of 10 mg/kg without prolonging bleeding time or blood clotting time in rats. Collectively, our results demonstrated that paeoniflorin can be a novel anti-platelet agent selectively targeting SIPA with an improved safety profile.
Collapse
Affiliation(s)
- Thien Ngo
- College of Pharmacy, Seoul National University, Seoul 08826, Korea.
- Faculty of Pharmacy, Thai Binh University of Medicine and Pharmacy, Thai Binh city 410000, Vietnam.
| | - Keunyoung Kim
- College of Pharmacy, Seoul National University, Seoul 08826, Korea.
| | - Yiying Bian
- College of Pharmacy, Seoul National University, Seoul 08826, Korea.
- School of Public Health, China Medical University, Shenyang 110122, China.
| | - Hakjun Noh
- College of Pharmacy, Seoul National University, Seoul 08826, Korea.
| | - Kyung-Min Lim
- College of Pharmacy, Ewha Womans University, Seoul 03760, Korea.
| | - Jin-Ho Chung
- College of Pharmacy, Seoul National University, Seoul 08826, Korea.
| | - Ok-Nam Bae
- College of Pharmacy, Hanyang University, Ansan 15588, Korea.
| |
Collapse
|
18
|
Ngo T, Kim K, Bian Y, An GJ, Bae ON, Lim KM, Chung JH. Cyclocurcumin from Curcuma longa selectively inhibits shear stress-induced platelet aggregation. J Funct Foods 2019. [DOI: 10.1016/j.jff.2019.103462] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
19
|
Rana A, Westein E, Niego B, Hagemeyer CE. Shear-Dependent Platelet Aggregation: Mechanisms and Therapeutic Opportunities. Front Cardiovasc Med 2019; 6:141. [PMID: 31620451 PMCID: PMC6763557 DOI: 10.3389/fcvm.2019.00141] [Citation(s) in RCA: 109] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 09/03/2019] [Indexed: 01/04/2023] Open
Abstract
Cardiovascular diseases (CVD) are the number one cause of morbidity and death worldwide. As estimated by the WHO, the global death rate from CVD is 31% wherein, a staggering 85% results from stroke and myocardial infarction. Platelets, one of the key components of thrombi, have been well-investigated over decades for their pivotal role in thrombus development in healthy as well as diseased blood vessels. In hemostasis, when a vascular injury occurs, circulating platelets are arrested at the site of damage, where they are activated and aggregate to form hemostatic thrombi, thus preventing further bleeding. However, in thrombosis, pathological activation of platelets occurs, leading to uncontrolled growth of a thrombus, which in turn can occlude the blood vessel or embolize, causing downstream ischemic events. The molecular processes causing pathological thrombus development are in large similar to the processes controlling physiological thrombus formation. The biggest challenge of anti-thrombotics and anti-platelet therapeutics has been to decouple the pathological platelet response from the physiological one. Currently, marketed anti-platelet drugs are associated with major bleeding complications for this exact reason; they are not effective in targeting pathological thrombi without interfering with normal hemostasis. Recent studies have emphasized the importance of shear forces generated from blood flow, that primarily drive platelet activation and aggregation in thrombosis. Local shear stresses in obstructed blood vessels can be higher by up to two orders of magnitude as compared to healthy vessels. Leveraging abnormal shear forces in the thrombus microenvironment may allow to differentiate between thrombosis and hemostasis and develop shear-selective anti-platelet therapies. In this review, we discuss the influence of shear forces on thrombosis and the underlying mechanisms of shear-induced platelet activation. Later, we summarize the therapeutic approaches to target shear-sensitive platelet activation and pathological thrombus growth, with a particular focus on the shear-sensitive protein von Willebrand Factor (VWF). Inhibition of shear-specific platelet aggregation and targeted drug delivery may prove to be much safer and efficacious approaches over current state-of-the-art antithrombotic drugs in the treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Akshita Rana
- Nanobiotechnology Laboratory, Australian Centre for Blood Diseases, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Erik Westein
- Nanobiotechnology Laboratory, Australian Centre for Blood Diseases, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Be'eri Niego
- Nanobiotechnology Laboratory, Australian Centre for Blood Diseases, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Christoph E Hagemeyer
- Nanobiotechnology Laboratory, Australian Centre for Blood Diseases, Central Clinical School, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
20
|
Riswari SF, Tunjungputri RN, Kullaya V, Garishah FM, Utari GSR, Farhanah N, Overheul GJ, Alisjahbana B, Gasem MH, Urbanus RT, de Groot PG, Lefeber DJ, van Rij RP, van der Ven A, de Mast Q. Desialylation of platelets induced by Von Willebrand Factor is a novel mechanism of platelet clearance in dengue. PLoS Pathog 2019; 15:e1007500. [PMID: 30849118 PMCID: PMC6426266 DOI: 10.1371/journal.ppat.1007500] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 03/20/2019] [Accepted: 12/03/2018] [Indexed: 11/19/2022] Open
Abstract
Thrombocytopenia and platelet dysfunction are commonly observed in patients with dengue virus (DENV) infection and may contribute to complications such as bleeding and plasma leakage. The etiology of dengue-associated thrombocytopenia is multifactorial and includes increased platelet clearance. The binding of the coagulation protein von Willebrand factor (VWF) to the platelet membrane and removal of sialic acid (desialylation) are two well-known mechanisms of platelet clearance, but whether these conditions also contribute to thrombocytopenia in dengue infection is unknown. In two observational cohort studies in Bandung and Jepara, Indonesia, we show that adult patients with dengue not only had higher plasma concentrations of plasma VWF antigen and active VWF, but that circulating platelets had also bound more VWF to their membrane. The amount of platelet-VWF binding correlated well with platelet count. Furthermore, sialic acid levels in dengue patients were significantly reduced as assessed by the binding of Sambucus nigra lectin (SNA) and Maackia amurensis lectin II (MAL-II) to platelets. Sialic acid on the platelet membrane is neuraminidase-labile, but dengue virus has no known neuraminidase activity. Indeed, no detectable activity of neuraminidase was present in plasma of dengue patients and no desialylation was found of plasma transferrin. Platelet sialylation was also not altered by in vitro exposure of platelets to DENV nonstructural protein 1 or cultured DENV. In contrast, induction of binding of VWF to glycoprotein 1b on platelets using the VWF-activating protein ristocetin resulted in the removal of platelet sialic acid by translocation of platelet neuraminidase to the platelet surface. The neuraminidase inhibitor oseltamivir reduced VWF-induced platelet desialylation. Our data demonstrate that excessive binding of VWF to platelets in dengue results in neuraminidase-mediated platelet desialylation and platelet clearance. Oseltamivir might be a novel treatment option for severe thrombocytopenia in dengue infection. Dengue is the most common arbovirus infection in the world. A decrease in the number of blood platelets is an almost universal finding in severe dengue. Binding of the coagulation protein von Willebrand factor (VWF) and loss of sialic acid residues from the platelet membrane are two main mechanisms of clearance of senescent platelets under non-pathological conditions. Here, we show that platelets from patients with acute dengue have bound more VWF and have lost sialic acid from their membrane. Sialic acid can be cleaved by the enzyme neuraminidase. We show that neuraminidase activity in the plasma is not increased and that neither dengue virus itself nor nonstructural protein 1, a protein secreted by dengue virus, cleave sialic acid from the platelet membrane. In contrast, binding of VWF to platelets results in translocation of neuraminidase to the platelet membrane and subsequent cleavage of sialic acid. This process could be inhibited by the neuraminidase inhibitor oseltamivir, a commonly used anti-influenza drug. Altogether, our results indicate that VWF binding to platelets is increased in dengue infection, leading to the removal of sialic acid and platelet clearance. Oseltamivir may prevent this process and thus represent a novel treatment option for low platelet numbers in dengue infection.
Collapse
Affiliation(s)
- Silvita Fitri Riswari
- Clinical Infectious Disease Research Center, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
- Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Rahajeng N. Tunjungputri
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
- Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
- Center for Tropical and Infectious Disease (CENTRID), Faculty of Medicine Diponegoro University, Dr Kariadi Hospital, Semarang, Indonesia
| | - Vesla Kullaya
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
- Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
- Kilimanjaro Christian Medical Center, Kilimanjaro Clinical Research Institute, Moshi, Tanzania
| | - Fadel M. Garishah
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
- Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
- Center for Tropical and Infectious Disease (CENTRID), Faculty of Medicine Diponegoro University, Dr Kariadi Hospital, Semarang, Indonesia
| | - Gloria S. R. Utari
- Center for Tropical and Infectious Disease (CENTRID), Faculty of Medicine Diponegoro University, Dr Kariadi Hospital, Semarang, Indonesia
| | - Nur Farhanah
- Center for Tropical and Infectious Disease (CENTRID), Faculty of Medicine Diponegoro University, Dr Kariadi Hospital, Semarang, Indonesia
| | - Gijs J. Overheul
- Center for Tropical and Infectious Disease (CENTRID), Faculty of Medicine Diponegoro University, Dr Kariadi Hospital, Semarang, Indonesia
- Department of Medical Microbiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Bachti Alisjahbana
- Clinical Infectious Disease Research Center, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| | - M. Hussein Gasem
- Center for Tropical and Infectious Disease (CENTRID), Faculty of Medicine Diponegoro University, Dr Kariadi Hospital, Semarang, Indonesia
| | - Rolf T. Urbanus
- Department of Clinical Chemistry and Haematology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Philip. G. de Groot
- Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Dirk J. Lefeber
- Department of Neurology, Translational Metabolic Laboratory, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Ronald P. van Rij
- Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Medical Microbiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Andre van der Ven
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
- Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Quirijn de Mast
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
- Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
- * E-mail:
| |
Collapse
|
21
|
Structure of von Willebrand factor A1 on polystyrene determined from experimental and calculated sum frequency generation spectra. Biointerphases 2018; 13:06E411. [PMID: 30551688 DOI: 10.1116/1.5056219] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The blood-clotting protein von Willebrand factor (vWF) can be activated by small molecules, high shear stress, and interactions with interfaces. It subsequently binds platelet receptor glycoprotein Ibα (GPIbα) at the surface of platelets, thereby playing a crucial role in blood clotting due to platelet activation, which is an important process to consider in the design of cardiovascular implants and biomaterials used in blood-contacting applications. The influence of surfaces on the activation and the molecular-level structure of surface-bound vWF is largely unknown. Recent studies have indicated that when bound to hydrophobic polystyrene (PS), the A1 domain of vWF remains accessible for GPIbα binding. However, the detailed secondary structure and exact orientation of vWF A1 at the PS surface is still unresolved. Here, the authors resolve these features by studying the system with sum-frequency generation (SFG) spectroscopy. The data are consistent with a scenario where vWF A1 maintains a native secondary structure when bound to PS. Comparison of experimental and calculated SFG spectra combined with previously reported time-of-flight secondary ion mass spectrometry data suggests that A1 assumes an orientation with the GPIbα binding domain oriented away from the solid surface and exposed to the solution phase. This structural information will benefit future in vitro experiments with surface-adsorbed A1 domain and may have relevance for the design of novel blood-contacting biomaterials and wound-healing applications.
Collapse
|
22
|
Deng W, Voos KM, Colucci JK, Legan ER, Ortlund EA, Lollar P, Li R. Delimiting the autoinhibitory module of von Willebrand factor. J Thromb Haemost 2018; 16:2097-2105. [PMID: 30053340 PMCID: PMC6173635 DOI: 10.1111/jth.14251] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2018] [Indexed: 12/12/2022]
Abstract
Essentials The self-inhibitory mechanism of von Willebrand factor (VWF) remains unclear. Residues flanking the A1 domain of VWF form a discontinuous autoinhibitory module (AIM). rVWF1238-1493 exhibited greater thermostability and inactivity than its shorter counterparts. The cooperative coupling between the N- and C- AIM regions are required for inhibiting A1. SUMMARY Background The hierarchical hemostasis response involves a self-inhibitory feature of von Willebrand factor (VWF) that has not been fully characterized. The residues flanking the A1 domain of VWF are important in this self-inhibition by forming an autoinhibitory module (AIM) that masks the A1 domain. Objectives To delimit the AIM sequence and to evaluate the cooperative interplay between the discontinuous AIM regions. Methods ELISA, flow cytometry, a thermal stability assay and hydrogen-deuterium exchange (HDX) mass spectrometry were used to characterize recombinant VWF A1 fragments varying in length. Results The longest A1 fragment (rVWF1238-1493 ) showed higher inactivity in binding the platelet receptor glycoprotein (GP) Ibα and greater thermostability than its shorter counterparts. The HDX results showed that most of the N-terminal residues and residues 1459-1478 at the C-terminus of rVWF1238-1493 have slower deuterium uptake than the residues in its denatured counterpart, implying that these residues may interact with the A1 domain. In contrast, residues 1479-1493 showed less difference from the denatured form, indicating that these residues are unlikely to be involved in binding the A1 domain. The A1 fragment that lacks either the entire C-terminal flanking region of the AIM (C-AIM), i.e. rVWF1238-1461 , or the entire N-terminal flanking region of the AIM (N-AIM), i.e. rVWF1271-1493 , showed high GPIbα-binding affinity and low thermostability, suggesting that removal of either N-terminal or C-terminal residues resulted in loss of AIM inhibition of the A1 domain. Conclusion The AIM is probably composed of residues 1238-1271 (N-AIM) and 1459-1478 (C-AIM). Neither the N-AIM nor the C-AIM alone could fully inhibit binding of the A1 domain to GPIbα.
Collapse
Affiliation(s)
- Wei Deng
- Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA
| | - Kayleigh M. Voos
- Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA
| | - Jennifer K. Colucci
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA
| | - Emily R. Legan
- Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA
| | - Eric A. Ortlund
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA
| | - Pete Lollar
- Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA
| | - Renhao Li
- Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA
| |
Collapse
|
23
|
Tokuda H, Kuroyanagi G, Onuma T, Enomoto Y, Doi T, Iida H, Otsuka T, Ogura S, Iwama T, Kojima K, Kozawa O. Ristocetin induces phosphorylated-HSP27 (HSPB1) release from the platelets of type 2 DM patients: Anti-platelet agent-effect on the release. Biomed Rep 2018. [PMID: 29541457 DOI: 10.3892/br.2018.1058] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
It has been previously reported that HSP27 is released from platelets in type 2 diabetes mellitus (DM) patients according to phosphorylation. In the present study, we investigated the effect of ristocetin, a glycoprotein (GP)Ib/IX/V activator, on the release of HSP27 and the effect of anti-platelet agents, such as acetylsalicylic acid (ASA), on this release. Forty-six patients with type 2 DM were recruited, and classified into two groups depending on administration of anti-platelet agents, resulting in 31 patients without these agents (control group) and 15 patients with them (anti-platelet group). Ristocetin potently induced the aggregation of platelets in the two groups. Ristocetin-induced changes of the area under the curve of light transmittance and the ratio of the size of platelet aggregates in the anti-platelet group were slightly different from those in the control group. On the other hand, the levels of phosphorylated-HSP27 induced by ristocetin in the platelets from a patient of the anti-platelet group taking ASA were significantly lower than those from a patient of the control group. The levels of HSP27 release from the ristocetin-stimulated platelets were significantly correlated with the levels of phosphorylated-HSP27 in the platelets from patients in the two groups. The levels of HSP27 release and those of platelet-derived growth factor-AB (PDGF-AB) secretion stimulated by ristocetin in the anti-platelet group were lower than those in the control group. In addition, the levels of HSP27 release and those of PDGF-AB secretion stimulated by ADP in the anti-platelet group were lower than those in the control group. These results strongly suggest that anti-platelet agents inhibit the HSP27 release from platelets stimulated by ristocetin but not the aggregation in type 2 DM patients.
Collapse
Affiliation(s)
- Haruhiko Tokuda
- Department of Clinical Laboratory/Biobank of Medical Genome Center, National Center for Geriatrics and Gerontology, Obu, Aichi 474-8511, Japan.,Department of Pharmacology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | - Gen Kuroyanagi
- Department of Pharmacology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan.,Department of Orthopedic Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya 467-8601, Japan
| | - Takashi Onuma
- Department of Pharmacology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan.,Department of Anesthesiology and Pain Medicine, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | - Yukiko Enomoto
- Department of Neurosurgery, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | - Tomoaki Doi
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | - Hiroki Iida
- Department of Anesthesiology and Pain Medicine, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | - Takanobu Otsuka
- Department of Orthopedic Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya 467-8601, Japan
| | - Shinji Ogura
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | - Toru Iwama
- Department of Neurosurgery, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | - Kumi Kojima
- Department of Clinical Laboratory/Biobank of Medical Genome Center, National Center for Geriatrics and Gerontology, Obu, Aichi 474-8511, Japan
| | - Osamu Kozawa
- Department of Pharmacology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| |
Collapse
|
24
|
Loon JEV, Jaegere PPTD, Vliet HHDMV, Maat MPMD, de Groot PG, Simoons ML, Leebeek FWG. The in vitro effect of the new antithrombotic drug candidate ALX-0081 on blood samples of patients undergoing percutaneous coronary intervention. Thromb Haemost 2017; 106:165-71. [DOI: 10.1160/th10-12-0804] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2010] [Accepted: 04/05/2011] [Indexed: 11/05/2022]
Abstract
SummaryCompound ALX-0081 is a bivalent humanised Nanobody® that binds the A1-domain of von Willebrand factor (VWF) with high affinity. Consequently, it can block the interaction between VWF and its platelet-receptor- glycoprotein Ib, which leads inevitably to formation of arterial thrombi. It was the objective of this study to assess the in vitro effects of ALX-0081 on platelet adhesion and aggregation in coronary artery disease (CAD) patients to determine the optimal concentration of ALX-0081 and the effect of co-medication. We included nine CAD patients, who were scheduled for elective percutaneous coronary intervention (PCI), and 11 healthy volunteers. At admission all patients received aspirin, clopidogrel and heparin. Blood was drawn 24 hours (h) before and 1 h after start of the PCI procedure and was subsequently spiked with different concentrations of ALX-0081 or buffer. The efficacy of ALX-0081 was assessed by in vitro experiments: flow chamber experiments, ristocetin-induced platelet aggregation (RIPA), and the platelet function analyser (PFA-100TM). VWF levels in CAD patients were significantly higher than in healthy controls. During PCI VWF levels did not rise. In all in vitro experiments, ALX-0081 led to complete inhibition of platelet adhesion and aggregation. However, the required effective concentration was higher in patients than in controls and was related to plasma VWF levels. In conclusion, ALX-0081 is able to completely inhibit in vitro platelet adhesion and aggregation in CAD patients scheduled for elective PCI. The efficacy of ALX-0081 is not influenced by PCI or co-medication. However, due to higher VWF levels in CAD patients a higher effective concentration of ALX-0081 was required than in healthy individuals.
Collapse
|
25
|
Deng W, Wang Y, Druzak SA, Healey JF, Syed AK, Lollar P, Li R. A discontinuous autoinhibitory module masks the A1 domain of von Willebrand factor. J Thromb Haemost 2017; 15:1867-1877. [PMID: 28692141 PMCID: PMC5585049 DOI: 10.1111/jth.13775] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Indexed: 12/18/2022]
Abstract
Essentials The mechanism for the auto-inhibition of von Willebrand factor (VWF) remains unclear. Hydrogen exchange of two VWF A1 fragments with disparate activities was measured and compared. Discontinuous residues flanking A1 form a structural module that blocks A1 binding to the platelet. Our results suggest a potentially unified model of VWF activation. Click to hear an ISTH Academy presentation on the domain architecture of VWF and activation by elongational flow by Dr Springer SUMMARY: Background How von Willebrand factor (VWF) senses and responds to shear flow remains unclear. In the absence of shear flow, VWF or its fragments can be induced to bind spontaneously to platelet GPIbα. Objectives To elucidate the auto-inhibition mechanism of VWF. Methods Hydrogen-deuterium exchange (HDX) of two recombinant VWF fragments expressed from baby hamster kidney cells were measured and compared. Results The shortA1 protein contains VWF residues 1261-1472 and binds GPIbα with a significantly higher affinity than the longA1 protein that contains VWF residues 1238-1472. Both proteins contain the VWF A1 domain (residues 1272-1458). Many residues in longA1, particularly those in the N- and C-terminal sequences flanking the A1 domain, and in helix α1, loops α1β2 and β3α2, demonstrated markedly reduced HDX compared with their counterparts in shortA1. The HDX-protected region in longA1 overlaps with the GPIbα-binding interface and is clustered with type 2B von Willebrand disease (VWD) mutations. Additional comparison with the HDX of denatured longA1 and ristocetin-bound longA1 indicates the N- and C-terminal sequences flanking the A1 domain form cooperatively an integrated autoinhibitory module (AIM) that interacts with the HDX-protected region. Binding of ristocetin to the C-terminal part of the AIM desorbs the AIM from A1 and enables longA1 binding to GPIbα. Conclusion The discontinuous AIM binds the A1 domain and prevents it from binding to GPIbα, which has significant implications for the pathogenesis of type 2B VWD and the shear-induced activation of VWF activity.
Collapse
Affiliation(s)
- W Deng
- Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Y Wang
- Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - S A Druzak
- Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - J F Healey
- Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - A K Syed
- Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - P Lollar
- Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - R Li
- Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
26
|
Thrombosis in diabetes: a shear flow effect? Clin Sci (Lond) 2017; 131:1245-1260. [PMID: 28592700 DOI: 10.1042/cs20160391] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 02/14/2017] [Accepted: 02/27/2017] [Indexed: 12/16/2022]
Abstract
Cardiovascular events are the major cause of morbidity and mortality in Type 2 diabetes (T2D). This condition is associated with heightened platelet reactivity, contributing to increased atherothrombotic risk. Indeed, individuals with diabetes respond inadequately to standard antiplatelet therapy. Furthermore, they often experience recurrent events as well as side effects that include excess bleeding. This highlights the need for identification of novel regulators of diabetes-associated thrombosis to target for therapeutic intervention. It is well established that platelet aggregation, a process essential for thrombus formation, is tightly regulated by shear stress; however, the mechanisms underlying shear activation of platelets, particularly in the setting of diabetes, are still poorly understood. This review will address the limitations of current diagnostic systems to assess the importance of shear stress in the regulation of thrombus formation in T2D, and the inability to recapitulate the pro-thrombotic phenotype seen clinically in the setting of T2D. Moreover, we will discuss recent findings utilizing new technologies to define the importance of shear stress in thrombus formation and their potential application to the setting of diabetes. Finally, we will discuss the potential of targeting shear-dependent mechanisms of thrombus formation as a novel therapeutic approach in the setting of T2D.
Collapse
|
27
|
Cheng Z, Gao W, Fan X, Chen X, Mei H, Liu J, Luo X, Hu Y. Extracellular signal-regulated kinase 5 associates with casein kinase II to regulate GPIb-IX-mediated platelet activation via the PTEN/PI3K/Akt pathway. J Thromb Haemost 2017; 15:1679-1688. [PMID: 28603902 DOI: 10.1111/jth.13755] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Indexed: 12/19/2022]
Abstract
Essentials The mechanisms of extracellular signal-regulated kinase 5 (ERK5) in GPIb-IX signaling are unclear. Function of ERK5 in GPIb-IX was tested using aggregation, western blotting, and mass spectrometry. The protein interacting with ERK5 in human platelets was identified as casein kinase II (CKII). ERK5 associates with CKII to regulate the activation of the PI3K/Akt pathway in GPIb-IX signaling. SUMMARY Background The platelet glycoprotein (GP) Ib-IX complex plays essential roles in thrombosis and hemostasis. The mitogen-activated protein kinases (MAPKs) ERK1/2 and p38 have been shown to be important in the GPIb-IX-mediated signaling leading to integrin activation. However, the roles of the MAPK extracellular signal-regulated kinase 5 (ERK5) in GPIb-IX-mediated platelet activation are unknown. Objective To reveal the function and mechanisms of ERK5 in GPIb-IX-mediated platelet activation. Methods The functions of ERK5 in GPIb-IX-mediated human platelet activation were assessed using botrocetin/VWF, ristocetin/VWF, or platelet adhesion to von Willebrand factor (VWF) under shear stress in the presence of a specific inhibitor of ERK5. ERK5-associated proteins were pulled down from Chinese hamster ovary (CHO) cells transfected with HA-tagged-ERK5, identified by mass spectrometry, and confirmed in human platelets. Roles of ERK5-associated proteins in GPIb-IX-mediated platelet activation were clarified using specific inhibitors. Results The phosphorylation levels of ERK5 were significantly enhanced in human platelets stimulated with botrocetin/VWF or ristocetin/VWF. The ERK5 inhibitor XMD8-92 suppressed the second wave of human platelet aggregation induced by botrocetin/VWF or ristocetin/VWF and inhibited human platelet adhesion on immobilized VWF under shear stress. Casein kinase II (CKII) was identified as an ERK5-associated protein in human platelets. The CKII inhibitor TBB, similar to the ERK5 inhibitor XMD8-92, specifically restrained PTEN phosphorylation, therefore suppressing Akt phosphorylation in human platelets treated with botrocetin/VWF. Conclusion ERK5 associates with CKII to play essential roles in GPIb-IX-mediated platelet activation via the PTEN/PI3K/Akt pathway.
Collapse
Affiliation(s)
- Z Cheng
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - W Gao
- Department of Cardiology, Huashan Hospital, Fudan University, Shanghai, China
| | - X Fan
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - X Chen
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - H Mei
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Collaborative Innovation Center of Hematology, Huazhong University of Science and Technology, Wuhan, China
| | - J Liu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - X Luo
- Department of Cardiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Y Hu
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Collaborative Innovation Center of Hematology, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
28
|
Löf A, Müller JP, Brehm MA. A biophysical view on von Willebrand factor activation. J Cell Physiol 2017; 233:799-810. [PMID: 28256724 DOI: 10.1002/jcp.25887] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 03/01/2017] [Indexed: 01/01/2023]
Abstract
The process of hemostatic plug formation at sites of vascular injury crucially relies on the large multimeric plasma glycoprotein von Willebrand factor (VWF) and its ability to recruit platelets to the damaged vessel wall via interaction of its A1 domain with platelet GPIbα. Under normal blood flow conditions, VWF multimers exhibit a very low binding affinity for platelets. Only when subjected to increased hydrodynamic forces, which primarily occur in connection with vascular injury, VWF can efficiently bind to platelets. This force-regulation of VWF's hemostatic activity is not only highly intriguing from a biophysical perspective, but also of eminent physiological importance. On the one hand, it prevents undesired activity of VWF in intact vessels that could lead to thromboembolic complications and on the other hand, it enables efficient VWF-mediated platelet aggregation exactly where needed. Here, we review recent studies that mainly employed biophysical approaches in order to elucidate the molecular mechanisms underlying the complex mechano-regulation of the VWF-GPIbα interaction. Their results led to two main hypotheses: first, intramolecular shielding of the A1 domain is lifted upon force-induced elongation of VWF; second, force-induced conformational changes of A1 convert it from a low-affinity to a high-affinity state. We critically discuss these hypotheses and aim at bridging the gap between the large-scale behavior of VWF as a linear polymer in hydrodynamic flow and the detailed properties of the A1-GPIbα bond at the single-molecule level.
Collapse
Affiliation(s)
- Achim Löf
- Department of Physics and Center for NanoScience, LMU Munich, Munich, Germany
| | - Jochen P Müller
- Department of Physics and Center for NanoScience, LMU Munich, Munich, Germany
| | - Maria A Brehm
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
29
|
Petzold T, Thienel M, Konrad I, Schubert I, Regenauer R, Hoppe B, Lorenz M, Eckart A, Chandraratne S, Lennerz C, Kolb C, Braun D, Jamasbi J, Brandl R, Braun S, Siess W, Schulz C, Massberg S. Oral thrombin inhibitor aggravates platelet adhesion and aggregation during arterial thrombosis. Sci Transl Med 2016; 8:367ra168. [DOI: 10.1126/scitranslmed.aad6712] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 08/31/2016] [Indexed: 12/13/2022]
|
30
|
Machha VR, Tischer A, Moon-Tasson L, Auton M. The Von Willebrand Factor A1-Collagen III Interaction Is Independent of Conformation and Type 2 Von Willebrand Disease Phenotype. J Mol Biol 2016; 429:32-47. [PMID: 27889474 DOI: 10.1016/j.jmb.2016.11.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 11/11/2016] [Accepted: 11/14/2016] [Indexed: 10/20/2022]
Abstract
The blood von Willebrand factor (VWF) mediates platelet adhesion to injured vessels by sequestering platelets from blood flow and depositing them to collagen and other exposed subendothelial matrix proteins. This process of capturing platelets to facilitate formation of platelet plugs occurs through transient interactions with platelet glycoprotein Ibα via the VWF A1 domain which also binds collagen. Using a conformationally diverse collection of natively folded and mutation-induced misfolded von Willebrand disease (VWD) variants, we test a recently proposed affinity up-regulation hypothesis which states that collagen binding changes the conformation of the A1 domain to a high-affinity GPIbα binding competent state. With surface plasmon resonance (SPR), we present this diversified collection to collagen and quantify the kinetics of association and dissociation to ascertain the conformational selectivity of collagen. With analytical rheology, we quantify real-time platelet pause times and translocation velocities across a Cu2+ HisTag-chelated and collagen-bound A1 single domain and A1A2A3 tridomain fragment of VWF under shear stress in an ex vivo shear flow microfluidic chamber. In contrast to expected hypothetical outcomes, collagen has limited conformational selectivity for binding A1. A1-collagen binding is independent of gain- or loss-of-function phenotype and under shear stress, platelet translocation pause times on collagen-bound A1A2A3 are either normal or shorter depending on whether A1 is concertedly bound with the A3 domain to collagen. With respect to A1, collagen has an inhibitory role that provides an explanation for the lack of thrombosis in patients with gain-of-function VWD.
Collapse
Affiliation(s)
- Venkata R Machha
- Division of Hematology, Departments of Internal Medicine and Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, United States
| | - Alexander Tischer
- Division of Hematology, Departments of Internal Medicine and Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, United States
| | - Laurie Moon-Tasson
- Division of Hematology, Departments of Internal Medicine and Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, United States
| | - Matthew Auton
- Division of Hematology, Departments of Internal Medicine and Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, United States.
| |
Collapse
|
31
|
Platelet clearance via shear-induced unfolding of a membrane mechanoreceptor. Nat Commun 2016; 7:12863. [PMID: 27670775 PMCID: PMC5052631 DOI: 10.1038/ncomms12863] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Accepted: 08/10/2016] [Indexed: 12/26/2022] Open
Abstract
Mechanisms by which blood cells sense shear stress are poorly characterized. In platelets, glycoprotein (GP)Ib–IX receptor complex has been long suggested to be a shear sensor and receptor. Recently, a relatively unstable and mechanosensitive domain in the GPIbα subunit of GPIb–IX was identified. Here we show that binding of its ligand, von Willebrand factor, under physiological shear stress induces unfolding of this mechanosensory domain (MSD) on the platelet surface. The unfolded MSD, particularly the juxtamembrane ‘Trigger' sequence therein, leads to intracellular signalling and rapid platelet clearance. These results illustrate the initial molecular event underlying platelet shear sensing and provide a mechanism linking GPIb–IX to platelet clearance. Our results have implications on the mechanism of platelet activation, and on the pathophysiology of von Willebrand disease and related thrombocytopenic disorders. The mechanosensation via receptor unfolding may be applicable for many other cell adhesion receptors. The platelets detect and respond to shear stress generated by blood flow. Here the authors show that the binding of the soluble von Willebrand factor to its receptor GPIba under physiological shear stress induces receptor's domain unfolding on the platelet and signalling into the platelet, leading to platelets clearance.
Collapse
|
32
|
Ju L, Chen Y, Xue L, Du X, Zhu C. Cooperative unfolding of distinctive mechanoreceptor domains transduces force into signals. eLife 2016; 5. [PMID: 27434669 PMCID: PMC5021522 DOI: 10.7554/elife.15447] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 07/18/2016] [Indexed: 12/30/2022] Open
Abstract
How cells sense their mechanical environment and transduce forces into biochemical signals is a crucial yet unresolved question in mechanobiology. Platelets use receptor glycoprotein Ib (GPIb), specifically its α subunit (GPIbα), to signal as they tether and translocate on von Willebrand factor (VWF) of injured arterial surfaces against blood flow. Force elicits catch bonds to slow VWF-GPIbα dissociation and unfolds the GPIbα leucine-rich repeat domain (LRRD) and juxtamembrane mechanosensitive domain (MSD). How these mechanical processes trigger biochemical signals remains unknown. Here we analyze these extracellular events and the resulting intracellular Ca(2+) on a single platelet in real time, revealing that LRRD unfolding intensifies Ca(2+) signal whereas MSD unfolding affects the type of Ca(2+) signal. Therefore, LRRD and MSD are analog and digital force transducers, respectively. The >30 nm macroglycopeptide separating the two domains transmits force on the VWF-GPIbα bond (whose lifetime is prolonged by LRRD unfolding) to the MSD to enhance its unfolding, resulting in unfolding cooperativity at an optimal force. These elements may provide design principles for a generic mechanosensory protein machine.
Collapse
Affiliation(s)
- Lining Ju
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, United States.,Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, United States.,Heart Research Institute, Camperdown, Australia.,Charles Perkins Centre, The University of Sydney, Camperdown, Australia
| | - Yunfeng Chen
- Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, United States.,Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, United States
| | - Lingzhou Xue
- Department of Statistics, The Pennsylvania State University, University Park, United States
| | - Xiaoping Du
- Department of Pharmacology, College of Medicine, University of Illinois at Chicago, Chicago, United States
| | - Cheng Zhu
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, United States.,Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, United States.,Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, United States
| |
Collapse
|
33
|
Tronic EH, Yakovenko O, Weidner T, Baio JE, Penkala R, Castner DG, Thomas WE. Differential surface activation of the A1 domain of von Willebrand factor. Biointerphases 2016; 11:029803. [PMID: 26968213 PMCID: PMC4788635 DOI: 10.1116/1.4943618] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2016] [Revised: 02/25/2016] [Accepted: 02/29/2016] [Indexed: 12/19/2022] Open
Abstract
The clotting protein von Willebrand factor (VWF) binds to platelet receptor glycoprotein Ibα (GPIbα) when VWF is activated by chemicals, high shear stress, or immobilization onto surfaces. Activation of VWF by surface immobilization is an important problem in the failure of cardiovascular implants, but is poorly understood. Here, the authors investigate whether some or all surfaces can activate VWF at least in part by affecting the orientation or conformation of the immobilized GPIbα-binding A1 domain of VWF. Platelets binding to A1 adsorbed onto polystyrene surfaces translocated rapidly at moderate and high flow, but detached at low flow, while platelets binding to A1 adsorbed onto glass or tissue-culture treated polystyrene surfaces translocated slowly, and detached only at high flow. Both x-ray photoelectron spectroscopy and conformation independent antibodies reported comparable A1 amounts on all surfaces. Time-of-flight secondary ion mass spectrometry (ToF-SIMS) and near-edge x-ray absorption fine structure spectra suggested differences in orientation on the three surfaces, but none that could explain the biological data. Instead, ToF-SIMS data and binding of conformation-dependent antibodies were consistent with the stabilization of an alternative more activated conformation of A1 by tissue culture polystyrene and especially glass. These studies demonstrate that different material surfaces differentially affect the conformation of adsorbed A1 domain and its biological activity. This is important when interpreting or designing in vitro experiments with surface-adsorbed A1 domain, and is also of likely relevance for blood-contacting biomaterials.
Collapse
Affiliation(s)
- Elaine H Tronic
- Department of Bioengineering, University of Washington, Seattle, Washington 98195
| | - Olga Yakovenko
- Department of Bioengineering, University of Washington, Seattle, Washington 98195
| | - Tobias Weidner
- Department of Bioengineering, University of Washington, Seattle, Washington 98195
| | - Joe E Baio
- Department of Chemical Engineering, University of Washington, Seattle, Washington 98195
| | - Rebecca Penkala
- Department of Bioengineering, University of Washington, Seattle, Washington 98195
| | - David G Castner
- Departments of Bioengineering and Chemical Engineering, University of Washington, Seattle, Washington 98195
| | - Wendy E Thomas
- Department of Bioengineering, University of Washington, Seattle, Washington 98195
| |
Collapse
|
34
|
Parker DN, Tasneem S, Farndale RW, Bihan D, Sadler JE, Sebastian S, de Groot PG, Hayward CPM. The functions of the A1A2A3 domains in von Willebrand factor include multimerin 1 binding. Thromb Haemost 2016; 116:87-95. [PMID: 27052467 PMCID: PMC5175582 DOI: 10.1160/th15-09-0700] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 03/18/2016] [Indexed: 12/24/2022]
Abstract
Multimerin 1 (MMRN1) is a massive, homopolymeric protein that is stored in platelets and endothelial cells for activation-induced release. In vitro, MMRN1 binds to the outer surfaces of activated platelets and endothelial cells, the extracellular matrix (including collagen) and von Willebrand factor (VWF) to support platelet adhesive functions. VWF associates with MMRN1 at high shear, not static conditions, suggesting that shear exposes cryptic sites within VWF that support MMRN1 binding. Modified ELISA and surface plasmon resonance were used to study the structural features of VWF that support MMRN1 binding, and determine the affinities for VWF-MMRN1 binding. High shear microfluidic platelet adhesion assays determined the functional consequences for VWF-MMRN1 binding. VWF binding to MMRN1 was enhanced by shear exposure and ristocetin, and required VWF A1A2A3 region, specifically the A1 and A3 domains. VWF A1A2A3 bound to MMRN1 with a physiologically relevant binding affinity (KD: 2.0 ± 0.4 nM), whereas the individual VWF A1 (KD: 39.3 ± 7.7 nM) and A3 domains (KD: 229 ± 114 nM) bound to MMRN1 with lower affinities. VWF A1A2A3 was also sufficient to support the adhesion of resting platelets to MMRN1 at high shear, by a mechanism dependent on VWF-GPIbα binding. Our study provides new information on the molecular basis of MMRN1 binding to VWF, and its role in supporting platelet adhesion at high shear. We propose that at sites of vessel injury, MMRN1 that is released following activation of platelets and endothelial cells, binds to VWF A1A2A3 region to support platelet adhesion at arterial shear rates.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Catherine P M Hayward
- Catherine P. M. Hayward, McMaster University Medical Centre, HSC 2N29A, 1200 Main St. West, Hamilton, Ontario, Canada L8N 3Z5, Tel.: +1 905 521 2100 Ext. 76274, Fax: +1 905 521 2338, E-mail:
| |
Collapse
|
35
|
Distinct mechanisms account for acquired von Willebrand syndrome in plasma cell dyscrasias. Ann Hematol 2016; 95:945-57. [PMID: 27040683 DOI: 10.1007/s00277-016-2650-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Accepted: 03/18/2016] [Indexed: 02/01/2023]
Abstract
Acquired von Willebrand syndrome (AVWS) is a rare bleeding disorder that may cause life-threatening hemorrhages in patients with plasma cell dyscrasias (PCDs). Early diagnosis and treatment require a thorough understanding of its underlying pathophysiology. Two patients with IgG MGUS presented with dramatically decreased plasma von Willebrand factor (VWF) and a severe type-1 pattern on multimer analysis. A prompt response to intravenous immunoglobulins (IVIG), but not to VWF/FVIII, was consistent with accelerated immunologic clearance of plasma VWF. Another IgG MGUS patient showed a type-2 pattern and a less pronounced response to IVIG, suggesting that additional mechanism(s) contributed to AVWS evolution. In a patient with Waldenström's macroglobulinemia and severe depletion of plasma VWF, multimer analysis indicated association of the IgM paraprotein with VWF before, but not after plasmapheresis, resulting in destruction of the agarose gel and a characteristically distorted band structure of VWF multimers. A type-2 pattern with highly abnormal VWF triplets and laboratory evidence of excessive fibrinolytic activity suggested that plasmin-mediated VWF degradation contributed to AVWS in a patient with multiple myeloma (MM) and AL amyloidosis. Finally, in a patient with IgG MM, maximally prolonged PFA-100® closure times and a specific defect in ristocetin-induced platelet agglutination, both of which resolved after remission induction, indicated interference of the paraprotein with VWF binding to platelet GPIb. Importantly, in none of the six patients, circulating autoantibodies to VWF were detected by a specific in-house ELISA. In summary, when evaluating PCD patients with severe bleeding symptoms, AVWS due to various pathogenic mechanisms should be considered.
Collapse
|
36
|
Feys HB, Van Aelst B, Devloo R, Vandekerckhove P, Compernolle V. The contribution of von Willebrand factor-GPIbα interactions to persistent aggregate formation in apheresis platelet concentrates. Vox Sang 2015; 110:344-51. [PMID: 26646710 DOI: 10.1111/vox.12365] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Revised: 10/05/2015] [Accepted: 10/14/2015] [Indexed: 11/29/2022]
Abstract
BACKGROUND AND OBJECTIVES Apheresis platelet concentrates sometimes contain persistent aggregates (PA). Because apheresis involves extracorporeal circulation, we hypothesized that interactions between GPIbα and von Willebrand factor (VWF) underlie their origin. MATERIALS AND METHODS Platelets in donations with PA were compared to aggregate-free (AF) controls. Flow cytometry was used to determine platelet bound VWF. Degranulation was measured using P-selectin expression in flow cytometry and cytokine release using immunosorbent assays. Platelet adhesion to VWF was assessed in hydrodynamic flow and real-time video microscopy. RESULTS Platelets in PA concentrates had significantly more (P = 0·009, n ≥ 8) bound VWF compared to AF platelets, but differences in VWF concentration, VWF collagen binding, activated VWF or GPIbα expression were not found. Degranulation was higher (P = 0·030, n = 7) in PA than AF concentrates on day 1 of storage, but adhesion to immobilized VWF under hydrodynamic flow conditions was normal at that moment. On day 6, however, significantly less VWF adhesion (P = 0·009, n ≥ 6) was found for PA platelets compared to AF, indicating accelerated storage lesion in PA products. In a model that mimicks PA formation by chemically induced binding of VWF to platelets, we found that degranulation, phosphatidylserine expression and metabolism did not differ with paired controls at any time during subsequent storage. CONCLUSION Accelerated storage lesion is found in concentrates with PA, but this cannot be explained solely by increased platelet bound VWF following apheresis. Therefore, additional stressors are probably responsible for the increases observed in platelet degranulation and storage lesion in products with PA.
Collapse
Affiliation(s)
- H B Feys
- Transfusion Research Center, Belgian Red Cross-Flanders, Ghent, Belgium
| | - B Van Aelst
- Transfusion Research Center, Belgian Red Cross-Flanders, Ghent, Belgium
| | - R Devloo
- Transfusion Research Center, Belgian Red Cross-Flanders, Ghent, Belgium
| | - P Vandekerckhove
- Blood Service of the Belgian Red Cross-Flanders, Mechelen, Belgium.,Faculty of Medicine and Health Sciences, University of Ghent, Ghent, Belgium.,Department of Public Health and Primary Care, Catholic University of Leuven, Leuven, Belgium
| | - V Compernolle
- Transfusion Research Center, Belgian Red Cross-Flanders, Ghent, Belgium.,Blood Service of the Belgian Red Cross-Flanders, Mechelen, Belgium.,Department of Clinical Chemistry, Microbiology and Immunology, University of Ghent, Ghent, Belgium
| |
Collapse
|
37
|
Ju L, Chen Y, Zhou F, Lu H, Cruz MA, Zhu C. Von Willebrand factor-A1 domain binds platelet glycoprotein Ibα in multiple states with distinctive force-dependent dissociation kinetics. Thromb Res 2015; 136:606-12. [PMID: 26213126 PMCID: PMC4553094 DOI: 10.1016/j.thromres.2015.06.019] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2015] [Revised: 05/27/2015] [Accepted: 06/14/2015] [Indexed: 12/22/2022]
Abstract
Circulating von Willebrand factor (VWF) adopts a closed conformation that shields the platelet glycoprotein Ibα (GPIbα) binding site in the VWF-A1 domain. Immobilized at sites of vascular injury, VWF is activated by its interaction with collagen and the exertion of increased hemodynamic forces. Studies on native VWF strings and isolated A1 domains suggest the existence of multiple A1 binding states in different biophysical contexts. In this single-molecule study, we have used a biomembrane force probe (BFP) and a flow chamber to identify and characterize a collagen binding induced conformation with a higher affinity to platelet GPIbα. As force increases, our results show that collagen binding increases the stability of GPIbα bond with both VWF and isolated A1 domain. However, the collagen 2D binding affinity for VWF-A3 domain is 10 times of that for A1 domain, suggesting the initial VWF capture is mediated by A3-collagen interaction while A1-collagen regulates the subsequent VWF activation. Our results reveal the molecular mechanism of collagen-regulated, A1-mediated platelet adhesion enhancement. Characterization of different A1 states provides insights into binding heterogeneity of VWF in different scenarios of inflammation and thrombosis.
Collapse
Affiliation(s)
- Lining Ju
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA; Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA 30332, USA; Heart Research Institute, Newtown, NSW 2042, Australia; Charles Perkins Centre, Camperdown, NSW 2006, Australia
| | - Yunfeng Chen
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA; Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Fangyuan Zhou
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA; Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Hang Lu
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA; Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Miguel A Cruz
- Cardiovascular Research Section, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Cheng Zhu
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA; Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA; Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA 30332, USA.
| |
Collapse
|
38
|
Exploiting the kinetic interplay between GPIbα-VWF binding interfaces to regulate hemostasis and thrombosis. Blood 2014; 124:3799-807. [PMID: 25293780 DOI: 10.1182/blood-2014-04-569392] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Platelet-von Willebrand factor (VWF) interactions must be tightly regulated in order to promote effective hemostasis and prevent occlusive thrombus formation. However, it is unclear what role the inherent properties of the bond formed between the platelet receptor glycoprotein Ibα and the A1 domain of VWF play in these processes. Using VWF-A1 knock-in mice with mutations that enhance (I1309V) or disrupt (R1326H) platelet receptor glycoprotein Ibα binding, we now demonstrate that the kinetic interplay between two distinct contact surfaces influences the site and extent to which platelets bind VWF. Incorporation of R1326H mutation into the major site shortened bond lifetime, yielding defects in hemostasis and thrombosis comparable to VWF-deficient animals. Similarly, disrupting this region of contact with an allosteric inhibitor impaired human platelet accrual in damaged arterioles. In contrast, the I1309V mutation near the minor site prolonged bond lifetime, which was essential for the development of a type 2B-like VWD phenotype. However, combining the R1326H and I1309V mutations normalized both bond kinetics and the hemostatic and thrombotic properties of VWF. These findings broaden our understanding of mechanisms governing platelet-VWF interactions in health and disease, and underscore the importance of combined biophysical and genetic approaches in identifying potential therapeutic avenues for treating bleeding and thrombotic disorders.
Collapse
|
39
|
Iida Y, Doi T, Matsushima-Nishiwaki R, Tokuda H, Ogura S, Kozawa O, Iida H. (-)-Epigallocatechin gallate selectively inhibits adenosine diphosphate‑stimulated human platelet activation: suppression of heat shock protein 27 phosphorylation via p38 mitogen‑activated protein kinase. Mol Med Rep 2014; 10:1383-8. [PMID: 25059833 DOI: 10.3892/mmr.2014.2389] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Accepted: 04/25/2014] [Indexed: 11/05/2022] Open
Abstract
(‑)‑Epigallocatechin gallate (EGCG) is a major component of green tea. It has been demonstrated that EGCG has an antithrombotic effect by inhibiting platelet aggregation. However, the detailed mechanisms underlying the effects of EGCG remain to be elucidated. The present study examined the effects of EGCG on human platelet activation by various stimulators and the exact underlying mechanisms. EGCG suppressed adenosine diphosphate (ADP)‑stimulated platelet aggregation dose dependently between 30 and 70 µM. By contrast, EGCG failed to affect platelet aggregation stimulated by collagen, U46619 (a TP agonist) or ristocetin (an activator of GPIb/IX/V). EGCG attenuated the ADP‑induced phosphorylation of p38 mitogen‑activated protein (MAP) kinase and heat shock protein 27 (HSP27). The ADP‑stimulated release of platelet‑derived growth factor (PDGF)‑AB and the soluble CD40 (sCD40) ligand was inhibited by EGCG. These findings suggest that EGCG selectively inhibits ADP‑stimulated human platelet activation and that EGCG reduces the release of PDGF‑AB and the sCD40 ligand due to suppressing HSP27 phosphorylation via p38 MAP kinase.
Collapse
Affiliation(s)
- Yuko Iida
- Department of Anesthesiology, Gifu University Graduate School of Medicine, Gifu, Gifu 501-1194, Japan
| | - Tomoaki Doi
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, Gifu, Gifu 501-1194, Japan
| | - Rie Matsushima-Nishiwaki
- Department of Pharmacology, Gifu University Graduate School of Medicine, Gifu, Gifu 501-1194, Japan
| | - Haruhiko Tokuda
- Department of Pharmacology, Gifu University Graduate School of Medicine, Gifu, Gifu 501-1194, Japan
| | - Shinji Ogura
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, Gifu, Gifu 501-1194, Japan
| | - Osamu Kozawa
- Department of Pharmacology, Gifu University Graduate School of Medicine, Gifu, Gifu 501-1194, Japan
| | - Hiroki Iida
- Department of Anesthesiology, Gifu University Graduate School of Medicine, Gifu, Gifu 501-1194, Japan
| |
Collapse
|
40
|
Abstract
When blood vessels are cut, the forces in the bloodstream increase and change character. The dark side of these forces causes hemorrhage and death. However, von Willebrand factor (VWF), with help from our circulatory system and platelets, harnesses the same forces to form a hemostatic plug. Force and VWF function are so closely intertwined that, like members of the Jedi Order in the movie Star Wars who learn to use "the Force" to do good, VWF may be considered the Jedi knight of the bloodstream. The long length of VWF enables responsiveness to flow. The shape of VWF is predicted to alter from irregularly coiled to extended thread-like in the transition from shear to elongational flow at sites of hemostasis and thrombosis. Elongational force propagated through the length of VWF in its thread-like shape exposes its monomers for multimeric binding to platelets and subendothelium and likely also increases affinity of the A1 domain for platelets. Specialized domains concatenate and compact VWF during biosynthesis. A2 domain unfolding by hydrodynamic force enables postsecretion regulation of VWF length. Mutations in VWF in von Willebrand disease contribute to and are illuminated by VWF biology. I attempt to integrate classic studies on the physiology of hemostatic plug formation into modern molecular understanding, and point out what remains to be learned.
Collapse
|
41
|
Haji-Valizadeh H, Modery-Pawlowski CL, Sen Gupta A. A factor VIII-derived peptide enables von Willebrand factor (VWF)-binding of artificial platelet nanoconstructs without interfering with VWF-adhesion of natural platelets. NANOSCALE 2014; 6:4765-73. [PMID: 24658160 PMCID: PMC4300948 DOI: 10.1039/c3nr06400j] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
There is substantial clinical interest in synthetic platelet analogs for potential application in transfusion medicine. To this end, our research is focused on self-assembled peptide-lipid nanoconstructs that can undergo injury site-selective adhesion and subsequently promote site-directed active platelet aggregation, thus mimicking platelet's primary hemostatic actions. For injury site-selective adhesion, we have utilized a coagulation factor FVIII-derived VWF-binding peptide (VBP). FVIII binds to VWF's D'-D3 domain while natural platelet GPIbα binds to VWF's A1 domain. Therefore, we hypothesized that the VBP-decorated nanoconstructs will adhere to VWF without mutual competition with natural platelets. We further hypothesized that the adherent VBP-decorated constructs can enhance platelet aggregation when co-decorated with a fibrinogen-mimetic peptide (FMP). To test these hypotheses, we used glycocalicin to selectively block VWF's A1 domain and, using fluorescence microscopy, studied the binding of fluorescently labeled VBP-decorated nanoconstructs versus platelets to ristocetin-treated VWF. Subsequently, we co-decorated the nanoconstructs with VBP and FMP and incubated them with human platelets to study construct-mediated enhancement of platelet aggregation. Decoration with VBP resulted in substantial construct adhesion to ristocetin-treated VWF even if the A1-domain was blocked by glycocalicin. In comparison, such A1-blocking resulted in significant reduction of platelet adhesion. Without A1-blocking, the VBP-decorated constructs and natural platelets could adhere to VWF concomitantly. Furthermore, the constructs co-decorated with VBP and FMP enhanced active platelet aggregation. The results indicate significant promise in utilizing the FVIII-derived VBP in developing synthetic platelet analogs that do not interfere with VWF-binding of natural platelets but allow site-directed enhancement of platelet aggregation when combined with FMP.
Collapse
Affiliation(s)
- Hassan Haji-Valizadeh
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio 44106, USA.
| | | | | |
Collapse
|
42
|
Navdaev A, Subramanian H, Petunin A, Clemetson KJ, Gambaryan S, Walter U. Echicetin coated polystyrene beads: a novel tool to investigate GPIb-specific platelet activation and aggregation. PLoS One 2014; 9:e93569. [PMID: 24705415 PMCID: PMC3976279 DOI: 10.1371/journal.pone.0093569] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Accepted: 03/05/2014] [Indexed: 01/01/2023] Open
Abstract
von Willebrand factor/ristocetin (vWF/R) induces GPIb-dependent platelet agglutination and activation of αIIbβ3 integrin, which also binds vWF. These conditions make it difficult to investigate GPIb-specific signaling pathways in washed platelets. Here, we investigated the specific mechanisms of GPIb signaling using echicetin-coated polystyrene beads, which specifically activate GPIb. We compared platelet activation induced by echicetin beads to vWF/R. Human platelets were stimulated with polystyrene beads coated with increasing amounts of echicetin and platelet activation by echicetin beads was then investigated to reveal GPIb specific signaling. Echicetin beads induced αIIbβ3-dependent aggregation of washed platelets, while under the same conditions vWF/R treatment led only to αIIbβ3-independent platelet agglutination. The average distance between the echicetin molecules on the polystyrene beads must be less than 7 nm for full platelet activation, while the total amount of echicetin used for activation is not critical. Echicetin beads induced strong phosphorylation of several proteins including p38, ERK and PKB. Synergistic signaling via P2Y12 and thromboxane receptor through secreted ADP and TxA2, respectively, were important for echicetin bead triggered platelet activation. Activation of PKG by the NO/sGC/cGMP pathway inhibited echicetin bead-induced platelet aggregation. Echicetin-coated beads are powerful and reliable tools to study signaling in human platelets activated solely via GPIb and GPIb-triggered pathways.
Collapse
Affiliation(s)
- Alexey Navdaev
- Institute of Clinical Biochemistry and Pathobiochemistry, University of Wuerzburg, Wuerzburg, Germany
| | - Hariharan Subramanian
- Institute of Clinical Biochemistry and Pathobiochemistry, University of Wuerzburg, Wuerzburg, Germany
| | - Alexey Petunin
- Institute of Biophysics, Siberian Branch of the Russian Academy of Sciences, Krasnoyarsk, Russia
| | | | - Stepan Gambaryan
- Institute of Clinical Biochemistry and Pathobiochemistry, University of Wuerzburg, Wuerzburg, Germany
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St. Petersburg, Russia
| | - Ulrich Walter
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Mainz, Germany
| |
Collapse
|
43
|
Blenner MA, Dong X, Springer TA. Structural basis of regulation of von Willebrand factor binding to glycoprotein Ib. J Biol Chem 2014; 289:5565-79. [PMID: 24391089 DOI: 10.1074/jbc.m113.511220] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Activation by elongational flow of von Willebrand factor (VWF) is critical for primary hemostasis. Mutations causing type 2B von Willebrand disease (VWD), platelet-type VWD (PT-VWD), and tensile force each increase affinity of the VWF A1 domain and platelet glycoprotein Ibα (GPIbα) for one another; however, the structural basis for these observations remains elusive. Directed evolution was used to discover a further gain-of-function mutation in A1 that shifts the long range disulfide bond by one residue. We solved multiple crystal structures of this mutant A1 and A1 containing two VWD mutations complexed with GPIbα containing two PT-VWD mutations. We observed a gained interaction between A1 and the central leucine-rich repeats (LRRs) of GPIbα, previously shown to be important at high shear stress, and verified its importance mutationally. These findings suggest that structural changes, including central GPIbα LRR-A1 contact, contribute to VWF affinity regulation. Among the mutant complexes, variation in contacts and poor complementarity between the GPIbα β-finger and the region of A1 harboring VWD mutations lead us to hypothesize that the structures are on a pathway to, but have not yet reached, a force-induced super high affinity state.
Collapse
Affiliation(s)
- Mark A Blenner
- From the Program in Cellular and Molecular Medicine, Boston Children's Hospital and Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115
| | | | | |
Collapse
|
44
|
Gitz E, Koopman CD, Giannas A, Koekman CA, van den Heuvel DJ, Deckmyn H, Akkerman JWN, Gerritsen HC, Urbanus RT. Platelet interaction with von Willebrand factor is enhanced by shear-induced clustering of glycoprotein Ibα. Haematologica 2013; 98:1810-8. [PMID: 23753027 DOI: 10.3324/haematol.2013.087221] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Initial platelet arrest at the exposed arterial vessel wall is mediated through glycoprotein Ibα binding to the A1 domain of von Willebrand factor. This interaction occurs at sites of elevated shear force, and strengthens upon increasing hydrodynamic drag. The increased interaction requires shear-dependent exposure of the von Willebrand factor A1 domain, but the contribution of glycoprotein Ibα remains ill defined. We have previously found that glycoprotein Ibα forms clusters upon platelet cooling and hypothesized that such a property enhances the interaction with von Willebrand factor under physiological conditions. We analyzed the distribution of glycoprotein Ibα with Förster resonance energy transfer using time-gated fluorescence lifetime imaging microscopy. Perfusion at a shear rate of 1,600 s(-1) induced glycoprotein Ibα clusters on platelets adhered to von Willebrand factor, while clustering did not require von Willebrand factor contact at 10,000 s(-1). Shear-induced clustering was reversible, not accompanied by granule release or αIIbβ3 activation and improved glycoprotein Ibα-dependent platelet interaction with von Willebrand factor. Clustering required glycoprotein Ibα translocation to lipid rafts and critically depended on arachidonic acid-mediated binding of 14-3-3ζ to its cytoplasmic tail. This newly identified mechanism emphasizes the ability of platelets to respond to mechanical force and provides new insights into how changes in hemodynamics influence arterial thrombus formation.
Collapse
|
45
|
Abstract
The study of blood ex vivo can occur in closed or open systems, with or without flow. Microfluidic devices, which constrain fluids to a small (typically submillimeter) scale, facilitate analysis of platelet function, coagulation biology, cellular biorheology, adhesion dynamics, and pharmacology and, as a result, can be an invaluable tool for clinical diagnostics. An experimental session can accommodate hundreds to thousands of unique clotting, or thrombotic, events. Using microfluidics, thrombotic events can be studied on defined surfaces of biopolymers, matrix proteins, and tissue factor, under constant flow rate or constant pressure drop conditions. Distinct shear rates can be generated on a device using a single perfusion pump. Microfluidics facilitated both the determination of intraluminal thrombus permeability and the discovery that platelet contractility can be activated by a sudden decrease in flow. Microfluidic devices are ideal for multicolor imaging of platelets, fibrin, and phosphatidylserine and provide a human blood analog to mouse injury models. Overall, microfluidic advances offer many opportunities for research, drug testing under relevant hemodynamic conditions, and clinical diagnostics.
Collapse
Affiliation(s)
- Thomas V Colace
- Department of Chemical and Biomolecular Engineering, Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | | | |
Collapse
|
46
|
Bartunek J, Barbato E, Heyndrickx G, Vanderheyden M, Wijns W, Holz JB. Novel antiplatelet agents: ALX-0081, a Nanobody directed towards von Willebrand factor. J Cardiovasc Transl Res 2013; 6:355-63. [PMID: 23307200 DOI: 10.1007/s12265-012-9435-y] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2012] [Accepted: 12/05/2012] [Indexed: 10/27/2022]
Abstract
This manuscript reviews the studies performed with ALX-0081 (INN: caplacizumab), a Nanobody targeting von Willebrand factor, in the context of current antithrombotic therapy in coronary artery disease. ALX-0081 specifically inhibits platelet adhesion to the vessel wall, and may control platelet aggregation and subsequent clot formation without increasing bleeding risk. A substantial number of antithrombotics are aimed at this cascade; however, their generally indiscriminative mode of action can result in a narrow therapeutic window, defined by the risk for bleeding complications, and thrombotic events. Nonclinically, ALX-0081 compared favorably to several antithrombotics. In Phase I studies in healthy subjects and stable angina patients undergoing percutaneous coronary intervention (PCI), ALX-0081 was well tolerated, and effectively inhibited pharmacodynamic markers. Following these results, a phase II study was initiated in high-risk acute coronary syndrome patients undergoing PCI. Based on its mechanism of action, ALX-0081 is also being developed for acquired thrombotic thrombocytopenic purpura.
Collapse
Affiliation(s)
- Jozef Bartunek
- Cardiovascular Center Aalst, OLV Clinic, Moorselbaan 164, 9300 Aalst, Belgium
| | | | | | | | | | | |
Collapse
|
47
|
Colace TV, Diamond SL. Direct observation of von Willebrand factor elongation and fiber formation on collagen during acute whole blood exposure to pathological flow. Arterioscler Thromb Vasc Biol 2012; 33:105-13. [PMID: 23104847 DOI: 10.1161/atvbaha.112.300522] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE In severe stenosis, von Willebrand factor (vWF) experiences millisecond exposures to pathological wall shear rates (γ(w)). We sought to evaluate the deposition of vWF onto collagen surfaces under flow in these environments. METHODS AND RESULTS Distinct from viscometry experiments that last many seconds, we deployed microfluidic devices for single-pass perfusion of whole blood or platelet-free plasma over fibrillar type 1 collagen (<50 ms transit time) at pathological γ(w) or spatial wall shear rate gradients (grad γ(w)). Using fluorescent anti-vWF, long thick vWF fibers (>20 μm) bound to collagen were visualized at constant γ(w)>30000 s(-1) during perfusion of platelet-free plasma, a process enhanced by EDTA. Rapid acceleration or deceleration of EDTA platelet-free plasma at grad γ(w)=±1.1×10(5) to ±4.3×10(7) s(-1)/cm did not promote vWF deposition. At 19400 s(-1), EDTA blood perfusion resulted in rolling vWF-platelet nets, although blood perfusion (normal Ca(2+)) generated large vWF/platelet deposits that repeatedly embolized and were blocked by anti-glycoprotein Ib or the α(IIb)β(3) inhibitor GR144053 and did not require grad γ(w). Blood perfusion at venous shear rate (200 s(-1)) produced a stable platelet deposit that was a substrate for massive but unstable vWF-platelet aggregates when flow was increased to 7800 s(-1). CONCLUSIONS Triggered by collagen and enhanced by platelet glycoprotein Ib and α(IIb)β(3), vWF fiber formation occurred during acute exposures to pathological γ(w) and did not require gradients in wall shear rate.
Collapse
Affiliation(s)
- Thomas V Colace
- Department of Chemical and Biomolecular Engineering, Institute for Medicine and Engineering, 1024 Vagelos Research Laboratory, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | |
Collapse
|
48
|
Montgomery RK, Reddoch KM, Evani SJ, Cap AP, Ramasubramanian AK. Enhanced shear-induced platelet aggregation due to low-temperature storage. Transfusion 2012; 53:1520-30. [PMID: 23043289 DOI: 10.1111/j.1537-2995.2012.03917.x] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2012] [Revised: 08/08/2012] [Accepted: 08/22/2012] [Indexed: 12/01/2022]
Abstract
BACKGROUND Refrigeration of platelets (PLTs) offers an attractive alternative to the currently practiced storage at room temperature since it may mitigate problems associated with bacterial contamination and extend storage lifetime. Refrigeration causes a number of biophysical and biochemical changes in PLTs and decreases PLT circulation time in vivo. However, the effect of refrigeration on PLT hemostatic functions under physiologic and pathophysiologic shear conditions has not been adequately characterized. STUDY DESIGN AND METHODS Washed PLTs prepared from either fresh PLT-rich plasma (PRP) or PRP stored at 4°C for 2 days was mixed with exogenous von Willebrand factor (VWF) and fibrinogen and sheared in a cone-and-plate viscometer. PLT aggregation, activation, and VWF binding after shear and glycoprotein (GP) Ibα receptor expression and ristocetin-induced PLT agglutination were measured. RESULTS PLTs stored at 4°C for 2 days aggregated significantly more than fresh PLTs particularly at high shear rates (10,000/sec), and this increase was independent of PLT concentration or suspension viscosity. Further, refrigerated PLTs showed a greater increase in GP Ibα-dependent PLT activation under shear and also bound more VWF than fresh PLTs. However, the GP Ibα expression levels as measured by three different antibodies were significantly lower in refrigerated PLTs than in fresh PLTs, and refrigeration resulted in a modest decrease in ristocetin-induced PLT agglutination. CONCLUSION The combined results demonstrate that refrigeration increases PLT aggregation under high shear, but not static, conditions and also increases shear-induced VWF binding and PLT activation. Clinically, enhanced shear-induced PLT aggregation due to low temperature storage may be a beneficial strategy to prevent severe bleeding in trauma.
Collapse
Affiliation(s)
- Robbie K Montgomery
- Department of Biomedical Engineering and South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, Texas 78249, USA
| | | | | | | | | |
Collapse
|
49
|
Kim K, Bae ON, Lim KM, Noh JY, Kang S, Chung KY, Chung JH. Novel antiplatelet activity of protocatechuic acid through the inhibition of high shear stress-induced platelet aggregation. J Pharmacol Exp Ther 2012; 343:704-11. [PMID: 22984226 DOI: 10.1124/jpet.112.198242] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Bleeding is the most common and serious adverse effect of currently available antiplatelet drugs. Many efforts are being made to develop novel antithrombotic agents without bleeding risks. Shear stress-induced platelet aggregation (SIPA), which occurs under abnormally high shear stress, plays a crucial role in the development of arterial thrombotic diseases. Here, we demonstrate that protocatechuic acid (PCA), a bioactive phytochemical from Lonicera (honeysuckle) flowers, selectively and potently inhibits high shear (>10,000 s(-1))-induced platelet aggregation. In isolated human platelets, PCA decreased SIPA and attenuated accompanying platelet activation, including intracellular calcium mobilization, granule secretion, and adhesion receptor expression. The anti-SIPA effect of PCA was mediated through blockade of von Willebrand factor binding to activated glycoprotein Ib, a primary and initial event for the accomplishment of SIPA. Conspicuously, PCA did not inhibit platelet aggregation induced by other endogenous agonists like collagen, thrombin, or ADP that are important in both pathological thrombosis and normal hemostasis. Antithrombotic effects of PCA were confirmed in vivo in a rat arterial thrombosis model, where PCA significantly delayed the arterial occlusion induced by FeCl(3). Of particular note, PCA did not increase bleeding times in a rat tail transection model, whereas conventional antiplatelet drugs, aspirin, and clopidogrel substantially prolonged it. Collectively, these results suggest that PCA may be a novel antiplatelet agent that can prevent thrombosis without increasing bleeding risks.
Collapse
Affiliation(s)
- Keunyoung Kim
- College of Pharmacy, Seoul National University, Seoul, Korea
| | | | | | | | | | | | | |
Collapse
|
50
|
Castro-Núñez L, Dienava-Verdoold I, Herczenik E, Mertens K, Meijer AB. Shear stress is required for the endocytic uptake of the factor VIII-von Willebrand factor complex by macrophages. J Thromb Haemost 2012; 10:1929-37. [PMID: 22812646 DOI: 10.1111/j.1538-7836.2012.04860.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND Low-density lipoprotein (LDL) receptor family members contribute to the cellular uptake of factor VIII. How von Willebrand factor fits into this endocytic pathway has remained poorly understood. OBJECTIVES It has been suggested that macrophages contribute to the clearance of the factor VIII (FVIII)-von Willebrand factor (VWF) complex. We now assessed the mechanisms of uptake employing human monocyte-derived macrophages. METHODS A confocal microscopy study was employed to study the uptake by monocyte-derived macrophages of a functional green fluorescent FVIII-GFP derivative in the presence and absence of VWF. RESULTS The results revealed that FVIII-GFP is internalized by macrophages. We found that FVIII-GFP co-localizes with LDL receptor-related protein (LRP), and that the LRP antagonist Receptor Associated Protein (RAP) blocks the uptake of FVIII-GFP. However, FVIII-GFP was not detected in the macrophages in the presence of VWF, suggesting that the FVIII-VWF complex is not internalized by these cells at all. Apart from static conditions, we also investigated the effect of shear stress on the uptake of FVIII-GFP in presence of VWF. Immunofluorescence studies demonstrated that VWF does not block endocytosis of FVIII-GFP under flow conditions. Moreover, VWF itself was also internalized by the macrophages. Strikingly, in the presence of RAP, endocytosis of FVIII-GFP and VWF was inhibited. CONCLUSION The results show that shear stress is required for macrophages to internalize both constituents of the FVIII-VWF complex.
Collapse
Affiliation(s)
- L Castro-Núñez
- Department of Plasma Proteins, Sanquin Research, Amsterdam, The Netherlands
| | | | | | | | | |
Collapse
|