1
|
Risman RA, Sen M, Tutwiler V, Hudson NE. Deconstructing fibrin(ogen) structure. J Thromb Haemost 2025; 23:368-380. [PMID: 39536819 PMCID: PMC11786978 DOI: 10.1016/j.jtha.2024.10.024] [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: 05/03/2024] [Revised: 09/18/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024]
Abstract
Fibrinogen and its insoluble degradation product fibrin are pivotal plasma proteins that play important roles in blood coagulation, wound healing, and immune responses. This review highlights research from the last 24 months connecting our progressing view of fibrin(ogen)'s structure, and in particular its conformational flexibility and posttranslational modifications, to its (patho)physiologic roles, molecular interactions, mechanical properties, use as a biomaterial, and potential as a therapeutic target. Recent work suggests that fibrinogen structure is highly dynamic, sampling multiple conformations, which may explain its myriad physiologic functions and the presence of cryptic binding sites. Investigations into fibrin clot structure elucidated the impact of posttranslational modifications, therapeutic interventions, and pathologic conditions on fibrin network morphology, offering insights into thrombus formation and embolization. Studies exploring the mechanical properties of fibrin reveal its response to blood flow and platelet-driven contraction, offering implications for clot stability and embolization risk. Moreover, advancements in tissue engineering leverage fibrin's biocompatibility and customizable properties for diverse applications, from wound healing to tissue regeneration and biomaterial interactions. These findings underscore the structural origins of fibrin(ogen)'s multifaceted roles and its potential as a target for therapeutic interventions.
Collapse
Affiliation(s)
- Rebecca A Risman
- Department of Biomedical Engineering, Rutgers University, New Brunswick, New Jersey, USA. https://twitter.com/rebecca_risman
| | - Mehmet Sen
- Department of Biology and Biochemistry, University of Houston, Houston, Texas, USA
| | - Valerie Tutwiler
- Department of Biomedical Engineering, Rutgers University, New Brunswick, New Jersey, USA. https://twitter.com/vatutwiler
| | - Nathan E Hudson
- Department of Physics, East Carolina University, Greenville, North Carolina, USA.
| |
Collapse
|
2
|
Kapustianenko L, Grinenko T, Rebriev A, Tykhomyrov A. The sequence 581Ser-610Val in the fibrinogen Aα chain is responsible for the formation of complexes between plasminogen and αC-regions of fibrin(ogen). Heliyon 2024; 10:e40852. [PMID: 39687172 PMCID: PMC11648866 DOI: 10.1016/j.heliyon.2024.e40852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 11/27/2024] [Accepted: 11/29/2024] [Indexed: 12/18/2024] Open
Abstract
Objective This study aimed to identify the binding sites for plasminogen (Pg) and its kringle-containing fragments within the αC-region of fibrin(ogen). This investigation is crucial while the conversion of fibrinogen into fibrin induces conformational changes that expose binding sites for Pg and tissue-type Pg activator (tPA), facilitating effective zymogen activation on the fibrin surface. Methods Two C-terminal fragments of the Aα chain ‒ 45 kDa (225Val-610Val) and 40 kDa (225Val-580Lys), were obtained through plasmin hydrolysis of human fibrinogen and subsequently characterized using MALDI TOF mass spectrometry. The interactions of Glu-Pg and Lys-Pg, as well as Pg kringle fragments (K1-3, K4, and K5), with the obtained αC truncated polypeptides were analyzed using ELISA and Western blot techniques with the use of specific antibodies. Results It was demonstrated that Pg and its fragments K1-3, K4, and K5 interact exclusively with the 45-kDa fragment (225Val-610Val) of the αC region of fibrinogen with high affinity in a concentration-dependent manner (Kd values for Glu-Pg = 7.10 × 10-9 M, Lys-Pg = 6.01 × 10-9 M, K1-3 = 1.08 × 10-7 M, K4 = 5.06 × 10-7 M, and K5 = 2.50 × 10-7 M). This fragment, unlike the 40-kDa polypeptide (225Val-580Lys), contains the α581Ser-610Val sequence. Conclusions It was shown that the sequence 581Ser-610Val of fibrinogen Aα-chain, which becomes exposed during the conversion of fibrinogen to fibrin, is essential for the formation of complexes between Pg and αC regions of fibrin(ogen), thereby contributing to the initiation and regulation of fibrinolysis.
Collapse
Affiliation(s)
| | - Tetiana Grinenko
- Palladin Institute of Biochemistry of NAS of Ukraine, Kyiv, Ukraine
| | - Andrew Rebriev
- Palladin Institute of Biochemistry of NAS of Ukraine, Kyiv, Ukraine
| | - Artem Tykhomyrov
- Palladin Institute of Biochemistry of NAS of Ukraine, Kyiv, Ukraine
| |
Collapse
|
3
|
Filatov NS, Khismatullin RR, Bilyalov AI, Khabirova AI, Salyakhutdinova SM, Ursan RV, Kasimova RN, Peshkova AD, Gazizov II, Shagimardanova EI, Woroncow MV, Kiyasov AP, Litvinov RI, Gusev OA. Distinct Hemostasis and Blood Composition in Spiny Mouse Acomys cahirinus. Int J Mol Sci 2024; 25:12867. [PMID: 39684578 DOI: 10.3390/ijms252312867] [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: 10/17/2024] [Revised: 11/24/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
The spiny mouse (Acomys species) is capable of scarless wound regeneration through largely yet unknown mechanisms. To investigate whether this capacity is related to peculiarities of the hemostatic system, we studied the blood of Acomys cahirinus in comparison to Mus musculus (Balb/c) to reveal differences in blood composition and clotting in both males and females. In response to surgical manipulations, blood clots formed in wounds of Acomys comprised a stronger hemostatic seal with reduced surgical bleeding in comparison with Balb/c. Acomys demonstrated notably shorter tail bleeding times and elevated clottable fibrinogen levels. Histological analysis revealed that clots from Acomys blood had densely packed fibrin-rich clots with pronounced fibrin segregation from erythrocytes. Acomys exhibited superior plasma clot stiffness as revealed with thromboelastography. The latter two characteristics are likely due to hyperfibrinogenemia. Light transmission platelet aggregometry demonstrated that ADP-induced platelet aggregates in Acomys males are stable, unlike the aggregates formed in the plasma of Balb/c undergoing progressive disaggregation over time. There were no apparent distinctions in platelet contractility and baseline expression of phosphatidylserine. Hematological profiling revealed a reduced erythrocytes count but increased mean corpuscular volume and hemoglobin content in Acomys. These results demonstrate the distinctive hemostatic potential of Acomys cahirinus, which may contribute to their remarkable regenerative capacity.
Collapse
Affiliation(s)
- Nikita S Filatov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | - Rafael R Khismatullin
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | - Airat I Bilyalov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
- Loginov Moscow Clinical Scientific Center, 111123 Moscow, Russia
| | - Alina I Khabirova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | | | - Roman V Ursan
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | | | - Alina D Peshkova
- Department of Pharmacology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Insaf I Gazizov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | - Elena I Shagimardanova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
- Loginov Moscow Clinical Scientific Center, 111123 Moscow, Russia
| | - Mary V Woroncow
- Institute for Regenerative Medicine, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Andrey P Kiyasov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | - Rustem I Litvinov
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Oleg A Gusev
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
- LIFT, Life Improvement by Future Technologies Institute, 121205 Moscow, Russia
- Intractable Disease Research Center, Graduate School of Medicine, Juntendo University, Tokyo 113-8421, Japan
| |
Collapse
|
4
|
Huang B, Tang P, Liu Y, Liu F, Zheng Y, Yang X, Zhang X, Xie H, Lin L, Lin B, Lin B. Xuefu Zhuyu decoction alleviates deep vein thrombosis through inhibiting the activation of platelets and neutrophils via sirtuin 1/nuclear factor kappa-B pathway. JOURNAL OF ETHNOPHARMACOLOGY 2024; 333:118485. [PMID: 38908490 DOI: 10.1016/j.jep.2024.118485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 06/19/2024] [Accepted: 06/20/2024] [Indexed: 06/24/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Xuefu Zhuyu Decoction (XZD), a renowned traditional Chinese medicine prescription, is widely employed for the management of conditions characterized by qi-stagnation and blood stasis. Although its anti-thrombotic effect on deep vein thrombosis (DVT) patients has been clinically observed, the underlying mechanism remains largely unexplored. AIM OF THE STUDY Our aim was to investigate the mechanisms by which XZD exerted its effect on DVT. MATERIALS AND METHODS The ultra performance liquid chromatography (UPLC) technique was employed to evaluate quality of XZD. To examine the effect of XZD on DVT, a DVT rat model with inferior vena cava (IVC) stenosis was established. The 4D-label-free proteomics approach was then utilized to uncover the possible mechanisms of XZD against DVT. Based on proteomics, citrullinated histone H3 (CitH3), along with serum levels of tumor necrosis factor-alpha (TNF-α) and interleukin-1 beta (IL-1β) were observed the inhibitory activity of XZD on neutrophil activation. Subsequently, the marker of platelet activation, specifically glycoprotein IIb (CD41) and glycoprotein IIIa (CD61), were assessed along with the secretion of von Willebrand factor (vWF) to investigate the inhibitory activity of XZD on platelet activation. Finally, we explored the impact of XZD on the sirtuin 1 (SIRT1)/nuclear factor kappa-B (NF-κB) pathway, which was associated with the activation of platelets and neutrophils. RESULTS Eight distinct components were identified for the quality control of XZD. XZD effectively reduced thrombus weight and length in DVT rats, without affecting the coagulation function or hematological parameters in the systemic circulation. Proteomics analysis revealed that XZD alleviated DVT by inhibiting the activation of platelets and neutrophils. The protein expression of CitH3, along with serum levels of TNF-α and IL-1β, were reduced in XZD-treated DVT rats. Similarly, protein expressions of CD41 and CD61, along with the release of vWF, were markedly down-regulated in XZD-treated DVT rats. Finally, treatment with XZD resulted in an up-regulation of SIRT1 protein expression and a down-regulation of both acetylated NF-κB/p65 and phosphorylated NF-κB/p65 protein expressions in endothelium. CONCLUSIONS XZD alleviates DVT by inhibiting the activation of platelets and neutrophils at the injured endothelium via the regulation of SIRT1/NF-κB pathway.
Collapse
Affiliation(s)
- Boning Huang
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; Guangdong Clinical Research Academy of Chinese Medicine, Guangdong, China
| | - Ping Tang
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; Guangdong Clinical Research Academy of Chinese Medicine, Guangdong, China
| | - Youchen Liu
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; Guangdong Clinical Research Academy of Chinese Medicine, Guangdong, China
| | - Fangle Liu
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; Guangdong Clinical Research Academy of Chinese Medicine, Guangdong, China
| | - Yuying Zheng
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; Guangdong Clinical Research Academy of Chinese Medicine, Guangdong, China
| | - Xinrong Yang
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; Guangdong Clinical Research Academy of Chinese Medicine, Guangdong, China
| | - Xiubing Zhang
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; Guangdong Clinical Research Academy of Chinese Medicine, Guangdong, China
| | - Huiyi Xie
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; Guangdong Clinical Research Academy of Chinese Medicine, Guangdong, China
| | - Liuqing Lin
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; Guangdong Clinical Research Academy of Chinese Medicine, Guangdong, China
| | - Bingqing Lin
- School of Mathematical Sciences, Shenzhen University, Shenzhen, Guangdong, China.
| | - Baoqin Lin
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; Guangdong Clinical Research Academy of Chinese Medicine, Guangdong, China.
| |
Collapse
|
5
|
Adair BD, Field CO, Alonso JL, Xiong JP, Deng SX, Ahn HS, Mashin E, Clish CB, van Agthoven J, Yeager M, Guo Y, Tess DA, Landry DW, Poncz M, Arnaout MA. Platelet integrin αIIbβ3 plays a key role in a venous thrombogenesis mouse model. Nat Commun 2024; 15:8612. [PMID: 39366965 PMCID: PMC11452527 DOI: 10.1038/s41467-024-52869-3] [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: 12/19/2023] [Accepted: 09/21/2024] [Indexed: 10/06/2024] Open
Abstract
Venous thrombosis (VT) is a common vascular disease associated with reduced survival and a high recurrence rate. VT is initiated by the accumulation of platelets and neutrophils at sites of endothelial cell activation. A role for platelet αIIbβ3 in VT is not established, a task complicated by the increased bleeding risk caused by partial agonists such as tirofiban. Here, we show that m-tirofiban, a modified version of tirofiban, does not agonize αIIbβ3 based on lack of neoepitope expression and the cryo-EM structure of m-tirofiban/full-length αIIbβ3 complex. m-tirofiban abolishes agonist-induced platelet aggregation while preserving clot retraction ex vivo and, unlike tirofiban, it suppresses venous thrombogenesis in a mouse model without increasing bleeding. These findings establish a key role for αIIbβ3 in VT initiation and suggest that m-tirofiban and compounds with a similar structurally-defined mechanism of action merit consideration as potential thromboprophylaxis agents in patients at high risk for VT and hemorrhage.
Collapse
Affiliation(s)
- Brian D Adair
- Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Leukocyte Biology and Inflammation Laboratory, Massachusetts General Hospital, Boston, MA, USA
- Structural Biology Program, Massachusetts General Hospital, Boston, MA, USA
| | - Conroy O Field
- Division of Hematology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - José L Alonso
- Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Leukocyte Biology and Inflammation Laboratory, Massachusetts General Hospital, Boston, MA, USA
| | - Jian-Ping Xiong
- Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Leukocyte Biology and Inflammation Laboratory, Massachusetts General Hospital, Boston, MA, USA
- Structural Biology Program, Massachusetts General Hospital, Boston, MA, USA
| | - Shi-Xian Deng
- Department of Medicine, New York-Presbyterian Hospital-Columbia and Cornell, New York, NY, USA
| | - Hyun Sook Ahn
- Division of Hematology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | | | | | - Johannes van Agthoven
- Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Leukocyte Biology and Inflammation Laboratory, Massachusetts General Hospital, Boston, MA, USA
- Structural Biology Program, Massachusetts General Hospital, Boston, MA, USA
| | - Mark Yeager
- The Frost Institute for Chemistry and Molecular Science, University of Miami, Coral Gables, FL, USA
| | - Youzhong Guo
- Department of Medicinal Chemistry, VCU School of Pharmacy, Richmond, VA, USA
| | - David A Tess
- Department of Pharmacokinetics, Dynamics, and Metabolism, Pfizer Inc, Cambridge, MA, USA
| | - Donald W Landry
- Department of Medicine, New York-Presbyterian Hospital-Columbia and Cornell, New York, NY, USA
| | - Mortimer Poncz
- Division of Hematology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - M Amin Arnaout
- Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
- Leukocyte Biology and Inflammation Laboratory, Massachusetts General Hospital, Boston, MA, USA.
- Structural Biology Program, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
6
|
Vidallon MLP, Moon MJ, Liu H, Song Y, Crawford S, Teo BM, McFadyen JD, Bishop AI, Tabor RF, Peter K, Wang X. Engineering Hyperechogenic Colloids with Clot-Targeting Capabilities from Platelet-Derived Membranes. ACS APPLIED MATERIALS & INTERFACES 2024; 16. [PMID: 39360874 PMCID: PMC11492166 DOI: 10.1021/acsami.4c12024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 09/25/2024] [Accepted: 09/25/2024] [Indexed: 10/25/2024]
Abstract
Thrombosis-related cardiovascular diseases remain the leading global cause of mortality and morbidity. In this study, we present a pioneering approach in the field of nanobiotechnology, with a focus on clinical translation, aimed at advancing early diagnosis and enhancing treatment options for thrombotic disorders. We introduce the fabrication of Platelet Membrane-Derived Bubbles (PMBs), which exhibit distinctive characteristics compared to conventional nanoparticles. These PMBs possess an average diameter of 700 nm and a negative ζ-potential, mirroring the attributes of parent platelet membranes. Utilizing diagnostic ultrasound imaging, we demonstrated the ability to visualize PMBs as hyperechogenic entities in agarose phantoms in vitro and in live mice in vivo. Furthermore, through confocal laser microscopy, we verified the retention of crucial transmembrane proteins, such as CD41 (GPIIb) and CD42 (GPIb), pivotal in conferring platelet-specific targeting functions. Importantly, our platelet aggregation studies confirmed that PMBs do not induce platelet aggregation but instead adhere to preformed platelet-rich in vitro thrombi. Overall, our work showcases the safe and precise utilization of PMBs to directly target acute thrombosis induced by laser injury in murine mesenteric veins in vivo, as visualized through intravital microscopy. In conclusion, we have successfully developed a rapid method for generating PMBs with unique ultrasound-directed and thrombus-targeting properties. These exceptional attributes of PMBs hold significant promise for advancing the field of ultrasound diagnostic thrombus imaging and clot-targeted therapy in the clinical context.
Collapse
Affiliation(s)
- Mark Louis P. Vidallon
- Molecular
Imaging and Theranostics Laboratory, Baker
Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
- Baker
Department of Cardiometabolic Health, University
of Melbourne, Parkville, VIC 3010, Australia
- School
of Chemistry, Monash University, Clayton, VIC 3800, Australia
- Baker
Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Bundoora, VIC 3086, Australia
| | - Mitchell J. Moon
- Baker
Department of Cardiometabolic Health, University
of Melbourne, Parkville, VIC 3010, Australia
- Atherothrombosis
and Vascular Biology Laboratory, Baker Heart
and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
| | - Haikun Liu
- Molecular
Imaging and Theranostics Laboratory, Baker
Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
- Baker
Department of Cardiometabolic Health, University
of Melbourne, Parkville, VIC 3010, Australia
| | - Yuyang Song
- Molecular
Imaging and Theranostics Laboratory, Baker
Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
- Baker
Department of Cardiometabolic Health, University
of Melbourne, Parkville, VIC 3010, Australia
| | - Simon Crawford
- Ramaciotti
Centre for Cryo-electron Microscopy, Monash
University, Clayton, VIC 3800, Australia
| | - Boon Mian Teo
- School
of Chemistry, Monash University, Clayton, VIC 3800, Australia
| | - James D. McFadyen
- Baker
Department of Cardiometabolic Health, University
of Melbourne, Parkville, VIC 3010, Australia
- Atherothrombosis
and Vascular Biology Laboratory, Baker Heart
and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
- Department
of Clinical Hematology, The Alfred Hospital, Melbourne, VIC 3004, Australia
- School
of Translational Medicine, Monash University, Melbourne, VIC 3004, Australia
| | - Alexis I. Bishop
- School
of Physics and Astronomy, Monash University, Clayton, VIC 3800, Australia
| | - Rico F. Tabor
- School
of Chemistry, Monash University, Clayton, VIC 3800, Australia
| | - Karlheinz Peter
- Baker
Department of Cardiometabolic Health, University
of Melbourne, Parkville, VIC 3010, Australia
- Baker
Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Bundoora, VIC 3086, Australia
- Atherothrombosis
and Vascular Biology Laboratory, Baker Heart
and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
- School
of Translational Medicine, Monash University, Melbourne, VIC 3004, Australia
| | - Xiaowei Wang
- Molecular
Imaging and Theranostics Laboratory, Baker
Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
- Baker
Department of Cardiometabolic Health, University
of Melbourne, Parkville, VIC 3010, Australia
- Baker
Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Bundoora, VIC 3086, Australia
- School
of Translational Medicine, Monash University, Melbourne, VIC 3004, Australia
| |
Collapse
|
7
|
Wang R, Tian Z, Zhu M, Zhang B, Li Y, Zheng Y, Mao Y, Zhao Y, Yang Y. SARS-CoV-2 spike protein potentiates platelet aggregation via upregulating integrin αIIbβ3 outside-in signaling pathway. J Thromb Thrombolysis 2024; 57:1225-1232. [PMID: 38981976 DOI: 10.1007/s11239-024-03008-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/25/2024] [Indexed: 07/11/2024]
Abstract
Platelet hyperreactivity is one of the crucial causes of coagulative disorders in patients with COVID-19. Few studies have indicated that integrin αIIbβ3 may be a potential target for spike protein binding to platelets. This study aims to investigate whether spike protein interacts with platelet integrin αIIbβ3 and upregulates outside-in signaling to potentiate platelet aggregation. In this study, we found that spike protein significantly potentiated platelet aggregation induced by different agonists and platelet spreading in vitro. Mechanism studies revealed that spike protein upregulated the outside-in signaling, such as increased thrombin-induced phosphorylation of β3, c-Src. Moreover, using tirofiban to inhibit spike protein binding to αIIbβ3 or using PP2 to block outside-in signaling, we found that the potentiating effect of spike protein on platelet aggregation was abolished. These results demonstrate that SARS-CoV-2 spike protein directly enhances platelet aggregation via integrin αIIbβ3 outside-in signaling, and suggest a potential target for platelet hyperreactivity in patients with COVID-19. HIGHLIGHTS: • Spike protein potentiates platelet aggregation and upregulates αIIbβ3 outside-in signaling. • Spike protein interacts with integrin αIIbβ3 to potentiate platelet aggregation. • Blocking outside-in signaling abolishes the effect of spike protein on platelets.
Collapse
Affiliation(s)
- Ruijie Wang
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Guangdong Province, Shenzhen, China
- Guangdong Engineering Technology Center of Nutrition Transformation, Sun Yat-sen University, Guangdong Province, Shenzhen, China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Sun Yat-sen University, Guangdong Province, Guangzhou, China
| | - Zezhong Tian
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Guangdong Province, Shenzhen, China
- Guangdong Engineering Technology Center of Nutrition Transformation, Sun Yat-sen University, Guangdong Province, Shenzhen, China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Sun Yat-sen University, Guangdong Province, Guangzhou, China
| | - Meiyan Zhu
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Guangdong Province, Shenzhen, China
| | - Bingying Zhang
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Guangdong Province, Shenzhen, China
| | - Yanzhang Li
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Guangdong Province, Shenzhen, China
| | - Yiqi Zheng
- Zhongshan School of Medicine, Sun Yat-sen University, Guangdong Province, Guangzhou, China
| | - Yuheng Mao
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Guangdong Province, Shenzhen, China
- Guangdong Engineering Technology Center of Nutrition Transformation, Sun Yat-sen University, Guangdong Province, Shenzhen, China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Sun Yat-sen University, Guangdong Province, Guangzhou, China
| | - Yimin Zhao
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Guangdong Province, Shenzhen, China
- Guangdong Engineering Technology Center of Nutrition Transformation, Sun Yat-sen University, Guangdong Province, Shenzhen, China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Sun Yat-sen University, Guangdong Province, Guangzhou, China
| | - Yan Yang
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Guangdong Province, Shenzhen, China.
- Guangdong Engineering Technology Center of Nutrition Transformation, Sun Yat-sen University, Guangdong Province, Shenzhen, China.
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Sun Yat-sen University, Guangdong Province, Guangzhou, China.
| |
Collapse
|
8
|
Hao Y, Tersteeg C, Hoekstra AG, Závodszky G. The effect of flow-derived mechanical cues on the growth and morphology of platelet aggregates under low, medium, and high shear rates. Comput Biol Med 2024; 180:109010. [PMID: 39159545 DOI: 10.1016/j.compbiomed.2024.109010] [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: 06/21/2024] [Revised: 08/04/2024] [Accepted: 08/07/2024] [Indexed: 08/21/2024]
Abstract
Platelet aggregation is a dynamic process that can obstruct blood flow, leading to cardiovascular diseases. While many studies have demonstrated clear connections between shear rate and platelet aggregation, the impact of flow-derived mechanical signals on this process is not fully understood. The objective of this work is to investigate the role of flow conditions on platelet aggregation dynamics, including effects on growth, shape, density composition, and their potential correlation with binding processes that are characterised by longer (e.g., via αIIbβ3 integrin) and shorter (e.g., via VWF) initial binding times. In vitro blood perfusion experiments were conducted at wall shear rates of 800, 1600 and 4000 s-1. Detailed analysis of two modalities of experimental images was performed to offer insights into the morphology of platelet aggregates. A consistent structural pattern was observed across all samples: a high-density core enveloped by a low-density outer shell. An image-based 3D computational blood flow model was subsequently employed to study the local flow conditions, including binding availability time and flow-derived mechanical signals via shear rate and rate of elongation. The results show substantial dependence of the aggregation dynamics on these flow parameters. We found that the different binding mechanisms that prefer different flow regimes do not have a monotonic cross-over in efficiency as the flow increases. There is a significant dip in the cumulative aggregation potential in-between the preferred regimes. The results suggest that treatments targeting the biomechanical pathways could benefit from creating conditions that exploit these low-efficiency zones of aggregation.
Collapse
Affiliation(s)
- Yue Hao
- Computational Science Lab, Informatics Institute, University of Amsterdam, Amsterdam, The Netherlands
| | - Claudia Tersteeg
- Laboratory for Thrombosis Research, IRF Life Sciences, KU Leuven Campus Kulak Kortrijk, Kortrijk, Belgium
| | - Alfons G Hoekstra
- Computational Science Lab, Informatics Institute, University of Amsterdam, Amsterdam, The Netherlands
| | - Gábor Závodszky
- Computational Science Lab, Informatics Institute, University of Amsterdam, Amsterdam, The Netherlands; Department of Hydrodynamic Systems, Budapest University of Technology and Economics, Budapest, Hungary.
| |
Collapse
|
9
|
Adair BD, Field CO, Alonso JL, Xiong JP, Deng SX, Ahn HS, Mashin E, Clish CB, van Agthoven J, Yeager M, Guo Y, Tess DA, Landry DW, Poncz M, Arnaout MA. Platelet integrin αIIbβ3 plays a key role in venous thrombogenesis in a mouse model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.11.602533. [PMID: 39026880 PMCID: PMC11257514 DOI: 10.1101/2024.07.11.602533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Venous thrombosis (VT) is a common vascular disease associated with reduced survival and a high recurrence rate. Previous studies have shown that the accumulation of platelets and neutrophils at sites of endothelial cell activation is a primary event in VT, but a role for platelet αIIbβ3 in the initiation of venous thrombosis has not been established. This task has been complicated by the increased bleeding linked to partial agonism of current αIIbβ3 inhibitory drugs such as tirofiban (Aggrastat ® ). Here, we show that m-tirofiban, an engineered version of tirofiban, is not a partial agonist of αIIbβ3. This is based on its cryo-EM structure in complex with human full-length αIIbβ3 and its inability to increase expression of an activation-sensitive epitope on platelet αIIbβ3. m-tirofiban abolished agonist-induced platelet aggregation ex vivo at concentrations that preserved clot retraction and markedly suppressed the accumulation of platelets, neutrophils, and fibrin on thrombin-activated endothelium in real-time using intravital microscopy in a mouse model of venous thrombogenesis. Unlike tirofiban, however, m-tirofiban did not increase bleeding at the thrombosis-inhibitory dose. These findings establish a key role for αIIbβ3 in the initiation of VT, provide a guiding principle for designing potentially safer inhibitors for other integrins, and suggest that pure antagonists of αIIbβ3 like m-tirofiban merit further consideration as potential thromboprophylaxis agents in patients at high-risk for VT and hemorrhage.
Collapse
|
10
|
Coffman RE, Bidone TC. Application of Funnel Metadynamics to the Platelet Integrin αIIbβ3 in Complex with an RGD Peptide. Int J Mol Sci 2024; 25:6580. [PMID: 38928286 PMCID: PMC11203998 DOI: 10.3390/ijms25126580] [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: 03/25/2024] [Revised: 06/11/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
Integrin αIIbβ3 mediates platelet aggregation by binding the Arginyl-Glycyl-Aspartic acid (RGD) sequence of fibrinogen. RGD binding occurs at a site topographically proximal to the αIIb and β3 subunits, promoting the conformational activation of the receptor from bent to extended states. While several experimental approaches have characterized RGD binding to αIIbβ3 integrin, applying computational methods has been significantly more challenging due to limited sampling and the need for a priori information regarding the interactions between the RGD peptide and integrin. In this study, we employed all-atom simulations using funnel metadynamics (FM) to evaluate the interactions of an RGD peptide with the αIIb and β3 subunits of integrin. FM incorporates an external history-dependent potential on selected degrees of freedom while applying a funnel-shaped restraint potential to limit RGD exploration of the unbound state. Furthermore, it does not require a priori information about the interactions, enhancing the sampling at a low computational cost. Our FM simulations reveal significant molecular changes in the β3 subunit of integrin upon RGD binding and provide a free-energy landscape with a low-energy binding mode surrounded by higher-energy prebinding states. The strong agreement between previous experimental and computational data and our results highlights the reliability of FM as a method for studying dynamic interactions of complex systems such as integrin.
Collapse
Affiliation(s)
- Robert E. Coffman
- Scientific Computing and Imaging Institute, University of Utah, Salt Lake City, UT 84112, USA;
| | - Tamara C. Bidone
- Scientific Computing and Imaging Institute, University of Utah, Salt Lake City, UT 84112, USA;
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA
- Department of Biochemistry, University of Utah, Salt Lake City, UT 84112, USA
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
11
|
Fu X, Lei T, Song S, Xia L, Xiao Y, Xiao G. Preparation and Hemocompatibility of Novel Antioxidant-Modified Polyethersulfone Membranes as Red Blood Cell Thrombosis Inhibitors. Macromol Biosci 2024; 24:e2300496. [PMID: 38359399 DOI: 10.1002/mabi.202300496] [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: 10/31/2023] [Revised: 01/11/2024] [Indexed: 02/17/2024]
Abstract
The contact between the dialysis membrane and blood can induce oxidative stress and thrombosis, causing oxidative organ damage and impaired toxin clearance. To date, the selection of anticoagulants has focused on mechanisms inhibiting white, but not red (erythrocytes) thrombus formation. In the present study, polyethersulfone (PES) membranes are modified with the antioxidant drug tiopronin; the physicochemical properties and dialysis performance of the Tio-PES membranes are evaluated. The effects on erythrocyte thrombosis are evaluated in terms of erythrocyte morphology, prothrombotic properties (adhesion, aggregation, viscosity, sedimentation, and hemolysis), and fibrinogen (FIB)-erythrocyte interactions. The regular anticoagulant and antiplatelet properties are also assessed. Superoxide dismutase, malondialdehyde, plasma protein, and complement C3a are further determined. Finally, the biosafety of the Tio-PES membranes is evaluated both in vitro and in vivo. The Tio-PES membranes exhibit excellent physicochemical properties and improved dialysis performance. It is found that the Tio-PES membranes stabilize erythrocyte morphology, reduce erythrocyte prothrombotic properties, decrease FIB adsorption, and prevent red thrombus formation. In addition, the Tio-PES membranes exhibit excellent antioxidant properties and show biosafety in primary toxicity studies. Thus, Tio-PES membranes hold promise as novel, safe, and effective dialysis materials for potential clinical application.
Collapse
Affiliation(s)
- Xiao Fu
- Department of Hematology, Xiangya Hemophilia Diagnosis and Treatment Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ting Lei
- Powder Metallurgy Institute of Central South University, Changsha, Hunan, China
| | - Shitao Song
- Department of Laboratory Medicine, The 3rd Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Luling Xia
- Department of Pulmonary and Critical Care Medicine, The 3rd Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yuan Xiao
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Gong Xiao
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Nephropathy, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
12
|
Wang R, Tian Z, Wang C, Zhang B, Zhu M, Yang Y. 1,25-Dihydroxyvitamin D3 attenuates platelet aggregation potentiated by SARS-CoV-2 spike protein via inhibiting integrin αIIbβ3 outside-in signaling. Cell Biochem Funct 2024; 42:e4039. [PMID: 38751189 DOI: 10.1002/cbf.4039] [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: 01/14/2024] [Revised: 04/30/2024] [Accepted: 05/05/2024] [Indexed: 05/26/2024]
Abstract
Platelet hyperreactivity contributes to the pathogenesis of COVID-19, which is associated with a hypercoagulability state and thrombosis disorder. It has been demonstrated that Vitamin D deficiency is associated with the severity of COVID-19 infection. Vitamin D supplement is widely used as a dietary supplement due to its safety and health benefits. In this study, we investigated the direct effects and underlying mechanisms of 1,25(OH)2D3 on platelet hyperreactivity induced by SRAS-CoV-2 spike protein via Western blot and platelet functional studies in vitro. Firstly, we found that 1,25(OH)2D3 attenuated platelet aggregation and Src-mediated signaling. We further observed that 1,25(OH)2D3 attenuated spike protein-potentiated platelet aggregation in vitro. Mechanistically, 1,25(OH)2D3 attenuated spike protein upregulated-integrin αIIbβ3 outside-in signaling such as platelet spreading and the phosphorylation of β3, c-Src and Syk. Moreover, using PP2, the Src family kinase inhibitor to abolish spike protein-stimulated platelet aggregation and integrin αIIbβ3 outside-in signaling, the combination of PP2 and 1,25(OH)2D3 did not show additive inhibitory effects on spike protein-potentiated platelet aggregation and the phosphorylation of β3, c-Src and Syk. Thus, our data suggest that 1,25(OH)2D3 attenuates platelet aggregation potentiated by spike protein via downregulating integrin αIIbβ3 outside-in signaling.
Collapse
Affiliation(s)
- Ruijie Wang
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong Province, China
- Guangdong Engineering Technology Center of Nutrition Transformation, Sun Yat-sen University, Shenzhen, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Zezhong Tian
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong Province, China
- Guangdong Engineering Technology Center of Nutrition Transformation, Sun Yat-sen University, Shenzhen, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Caixia Wang
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong Province, China
- Guangdong Engineering Technology Center of Nutrition Transformation, Sun Yat-sen University, Shenzhen, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Bingying Zhang
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong Province, China
| | - Meiyan Zhu
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong Province, China
| | - Yan Yang
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong Province, China
- Guangdong Engineering Technology Center of Nutrition Transformation, Sun Yat-sen University, Shenzhen, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| |
Collapse
|
13
|
Güven B, Can M. Fibrinogen: Structure, abnormalities and laboratory assays. Adv Clin Chem 2024; 120:117-143. [PMID: 38762239 DOI: 10.1016/bs.acc.2024.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/20/2024]
Abstract
Fibrinogen is the primary precursor protein for the fibrin clot, which is the final target of blood clotting. It is also an acute phase reactant that can vary under physiologic and inflammatory conditions. Disorders in fibrinogen concentration and/or function have been variably linked to the risk of bleeding and/or thrombosis. Fibrinogen assays are commonly used in the management of bleeding as well as the treatment of thrombosis. This chapter examines the structure of fibrinogen, its role in hemostasis as well as in bleeding abnormalities and measurement thereof with respect to clinical management.
Collapse
Affiliation(s)
- Berrak Güven
- Department of Clinical Biochemistry, Zonguldak Bülent Ecevit University, Zonguldak, Turkey.
| | - Murat Can
- Department of Clinical Biochemistry, Zonguldak Bülent Ecevit University, Zonguldak, Turkey
| |
Collapse
|
14
|
Nurden AT. Molecular basis of clot retraction and its role in wound healing. Thromb Res 2023; 231:159-169. [PMID: 36008192 DOI: 10.1016/j.thromres.2022.08.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 08/08/2022] [Accepted: 08/10/2022] [Indexed: 11/30/2022]
Abstract
Clot retraction is important for the prevention of bleeding, in the manifestations of thrombosis and for tissue repair. The molecular mechanisms behind clot formation are complex. Platelet involvement begins with adhesion at sites of vessel injury followed by platelet aggregation, thrombin generation and fibrin production. Other blood cells incorporate into a fibrin mesh that is consolidated by FXIIIa-mediated crosslinking and platelet contractile activity. The latter results in the asymmetric redistribution of erythrocytes into a tighter central mass providing the clot with stability and resistance to fibrinolysis. Integrin αIIbβ3 on platelets is the key player in these events, bridging fibrin and the platelet cytoskeleton. Glycoprotein VI participates in thrombus formation but not in the retraction. Rheological and environmental factors influence clot construction with retraction driven by the platelet cytoskeleton with actomyosin acting as the motor. Activated platelets provide procoagulant activity stimulating thrombin generation together with the release of a plethora of biologically active proteins and substances from storage pools; many form chemotactic gradients within the fibrin or the underlying matrix. Also released are newly synthesized metabolites and lipid-rich vesicles that circulate within the vasculature and mimic platelet functions. Platelets and their released elements play key roles in wound healing. This includes promoting stem cell and mesenchymal stromal cell recruitment, fibroblast and endothelial cell migration, angiogenesis and matrix formation. These properties have led to the use of autologous clots in therapies designed to accelerate tissue repair while offering the potential for genetic manipulation in both inherited and acquired diseases.
Collapse
Affiliation(s)
- Alan T Nurden
- Institut Hospitalo-Universitaire LIRYC, Pessac, France.
| |
Collapse
|
15
|
Chang Y, Hsia CW, Huang WC, Jayakumar T, Hsia CH, Yen TL, Sheu JR, Hou SM. Myricetin as a promising inhibitor of platelet fibrinogen receptor in humans. Heliyon 2023; 9:e20286. [PMID: 37767484 PMCID: PMC10520825 DOI: 10.1016/j.heliyon.2023.e20286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 09/12/2023] [Accepted: 09/18/2023] [Indexed: 09/29/2023] Open
Abstract
Platelets play a vital role in the formation of dangerous arterial thrombosis. Platelets are activated by adhesive proteins or soluble agonists through their specific receptors. The receptor-mediated signaling pathways lead to common signaling events, which result in shape changes and inside-out signaling, leading fibrinogen binding to glycoprotein IIb/IIIa complex (integrin αIIbβ3). This interaction initiates integrin αIIbβ3-mediated outside-in signaling, subsequently culminating in granule secretion and aggregation. Myricetin is a flavonoid that occurs in a variety of plants. Although myricetin has been demonstrated to have several bioactive properties, its role in platelet activation has not been extensively studied. The present study demonstrated the ability of myricetin to inhibit platelet aggregation stimulated by collagen, thrombin, and U46619. Myricetin reduced the ATP-release, cytosolic Ca2+ mobilization, and P-selectin expression and the activation of PLCγ2/PKC, PI3K/Akt/GSK3β, and MAPK. Myricetin exerted a direct inhibitory effect on the activation of integrin αIIbβ3 by disrupting the binding between FITC-PAC-1 and the integrin. Moreover, myricetin suppressed integrin αIIbβ3-mediated outside-in signaling, such as integrin β3, Src, and Syk phosphorylation on immobilized fibrinogen. In animal studies, myricetin significantly prolonged the occlusion time of thrombotic platelet plug formation in mesenteric microvessels without extending bleeding time. This study concludes that myricetin is a natural integrin αIIbβ3 inhibitor and a novel antithrombotic agent.
Collapse
Affiliation(s)
- Yi Chang
- Department of Anesthesiology, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, 111, Taiwan
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei City, 242, Taiwan
| | - Chih-Wei Hsia
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan
| | - Wei-Chieh Huang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan
| | - Thanasekaran Jayakumar
- Department of Ecology and Environmental Sciences, Pondicherry University, Puducherry, 605014, India
| | - Chih-Hsuan Hsia
- Translational Medicine Center, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, 111, Taiwan
| | - Ting-Lin Yen
- Department of Medical Research, Cathay General Hospital, Taipei, 106, Taiwan
| | - Joen-Rong Sheu
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan
| | - Shaw-Min Hou
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei City, 242, Taiwan
- Department of Cardiovascular Center, Cathay General Hospital, Taipei, 106, Taiwan
| |
Collapse
|
16
|
Lambrichts I, Wolfs E, Bronckaers A, Gervois P, Vangansewinkel T. The Effect of Leukocyte- and Platelet-Rich Fibrin on Central and Peripheral Nervous System Neurons-Implications for Biomaterial Applicability. Int J Mol Sci 2023; 24:14314. [PMID: 37762617 PMCID: PMC10532231 DOI: 10.3390/ijms241814314] [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: 08/31/2023] [Revised: 09/12/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
Leukocyte- and Platelet-Rich Fibrin (L-PRF) is a second-generation platelet concentrate that is prepared directly from the patient's own blood. It is widely used in the field of regenerative medicine, and to better understand its clinical applicability we aimed to further explore the biological properties and effects of L-PRF on cells from the central and peripheral nervous system. To this end, L-PRF was prepared from healthy human donors, and confocal, transmission, and scanning electron microscopy as well as secretome analysis were performed on these clots. In addition, functional assays were completed to determine the effect of L-PRF on neural stem cells (NSCs), primary cortical neurons (pCNs), and peripheral dorsal root ganglion (DRG) neurons. We observed that L-PRF consists of a dense but porous fibrin network, containing leukocytes and aggregates of activated platelets that are distributed throughout the clot. Antibody array and ELISA confirmed that it is a reservoir for a plethora of growth factors. Key molecules that are known to have an effect on neuronal cell functions such as brain-derived neurotrophic factor (BDNF), nerve growth factor (NGF), vascular endothelial growth factor (VEGF), and platelet-derived growth factor (PDGF) were slowly released over time from the clots. Next, we found that the L-PRF secretome had no significant effect on the proliferative and metabolic activity of NSCs, but it did act as a chemoattractant and improved the migration of these CNS-derived stem cells. More importantly, L-PRF growth factors had a detrimental effect on the survival of pCNs, and consequently, also interfered with their neurite outgrowth. In contrast, we found a positive effect on peripheral DRG neurons, and L-PRF growth factors improved their survival and significantly stimulated the outgrowth and branching of their neurites. Taken together, our study demonstrates the positive effects of the L-PRF secretome on peripheral neurons and supports its use in regenerative medicine but care should be taken when using it for CNS applications.
Collapse
Affiliation(s)
- Ivo Lambrichts
- Cardio and Organ Systems, Biomedical Research Institute, UHasselt—Hasselt University, 3590 Diepenbeek, Belgium; (E.W.); (A.B.); (P.G.)
| | - Esther Wolfs
- Cardio and Organ Systems, Biomedical Research Institute, UHasselt—Hasselt University, 3590 Diepenbeek, Belgium; (E.W.); (A.B.); (P.G.)
| | - Annelies Bronckaers
- Cardio and Organ Systems, Biomedical Research Institute, UHasselt—Hasselt University, 3590 Diepenbeek, Belgium; (E.W.); (A.B.); (P.G.)
| | - Pascal Gervois
- Cardio and Organ Systems, Biomedical Research Institute, UHasselt—Hasselt University, 3590 Diepenbeek, Belgium; (E.W.); (A.B.); (P.G.)
| | - Tim Vangansewinkel
- Cardio and Organ Systems, Biomedical Research Institute, UHasselt—Hasselt University, 3590 Diepenbeek, Belgium; (E.W.); (A.B.); (P.G.)
- VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, 3000 Leuven, Belgium
| |
Collapse
|
17
|
Gao D, Sun CW, Woodley AB, Dong JF. Clot Retraction and Its Correlation with the Function of Platelet Integrin α IIbβ 3. Biomedicines 2023; 11:2345. [PMID: 37760786 PMCID: PMC10525596 DOI: 10.3390/biomedicines11092345] [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: 06/19/2023] [Revised: 08/11/2023] [Accepted: 08/12/2023] [Indexed: 09/29/2023] Open
Abstract
Clot retraction results from retractions of platelet filopodia and fibrin fibers and requires the functional platelet αIIbβ3 integrin. This assay is widely used to test the functions of platelets and fibrinogen as well as the efficacy of fibrinolysis. Changes in clot retraction have been found in a variety of hemostatic abnormalities and, more recently, in arterial thrombosis. Despite its broad clinical use and low cost, many aspects of clot retraction are poorly understood. In the present study, we performed two clinical standard clot retraction assays using whole-blood and platelet-rich plasma (PRP) samples to determine how clot retraction correlates with platelet counts and mean volume, the density of αIIbβ3 integrin and PLA genotypes, and plasma fibrinogen levels. We found that clot retraction was affected by platelet counts, but not mean platelet volume. It correlated with the surface density of the integrin αIibβ3, but not PLA genotypes. These results indicate that clot retraction measures a unique aspect of platelet function and can serve as an additional means to detect functional changes in platelets.
Collapse
Affiliation(s)
- Daniel Gao
- Bloodworks Research Institute, 1551 Southlake Ave. E., Seattle, WA 98102, USA;
- Department of Chemistry, Pomona College, Claremont, CA 91711, USA
| | - Caroline W. Sun
- Section of Cardiovascular Sciences, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Angela B. Woodley
- Section of Cardiovascular Sciences, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jing-fei Dong
- Bloodworks Research Institute, 1551 Southlake Ave. E., Seattle, WA 98102, USA;
- Division of Hematology, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
| |
Collapse
|
18
|
Pinelo JEE, Manandhar P, Popovic G, Ray K, Tasdelen MF, Nguyen Q, Iavarone AT, Offenbacher AR, Hudson NE, Sen M. Systematic mapping of the conformational landscape and dynamism of soluble fibrinogen. J Thromb Haemost 2023; 21:1529-1543. [PMID: 36746319 PMCID: PMC10407912 DOI: 10.1016/j.jtha.2023.01.034] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 01/23/2023] [Accepted: 01/26/2023] [Indexed: 02/05/2023]
Abstract
BACKGROUND Fibrinogen is a soluble, multisubunit, and multidomain dimeric protein, which, upon its proteolytic cleavage by thrombin, is converted to insoluble fibrin, initiating polymerization that substantially contributes to clot growth. Fibrinogen contains numerous, transiently accessible "cryptic" epitopes for hemostatic and immunologic proteins, suggesting that fibrinogen exhibits conformational flexibility, which may play functional roles in its temporal and spatial interactions. Hitherto, there have been limited integrative approaches characterizing the solution structure and internal flexibility of fibrinogen. METHODS Here, utilizing a multipronged, biophysical approach involving 2 solution-based techniques, temperature-dependent hydrogen-deuterium exchange mass spectrometry and small angle X-ray scattering, corroborated by negative stain electron microscopy, we present a holistic, conformationally dynamic model of human fibrinogen in solution. RESULTS Our data reveal 4 major and distinct conformations of fibrinogen accommodated by a high degree of internal protein flexibility along its central scaffold. We propose that the fibrinogen structure in the solution consists of a complex, conformational landscape with multiple local minima. This is further supported by the location of numerous point mutations that are linked to dysfibrinogenemia and posttranslational modifications, residing near the identified fibrinogen flexions. CONCLUSION This work provides a molecular basis for the structural "dynamism" of fibrinogen that is expected to influence the broad swath of its functionally diverse macromolecular interactions and fine-tune the structural and mechanical properties of blood clots.
Collapse
Affiliation(s)
- Jose E E Pinelo
- Department of Biology and Biochemistry, University of Houston, Houston, Texas, USA
| | - Pragya Manandhar
- Department of Biology and Biochemistry, University of Houston, Houston, Texas, USA
| | - Grega Popovic
- Department of Chemistry, East Carolina University, Greenville, North Carolina, USA
| | - Katherine Ray
- Department of Chemistry, East Carolina University, Greenville, North Carolina, USA
| | - Mehmet F Tasdelen
- Department of Computer Science, University of Houston, Houston, Texas, USA
| | - Quoc Nguyen
- Department of Mathematics, University of Houston, Houston, Texas, USA
| | - Anthony T Iavarone
- QB3/Chemistry/Mass Spectrometry Facility, University of California, Berkeley, California, USA
| | - Adam R Offenbacher
- Department of Chemistry, East Carolina University, Greenville, North Carolina, USA
| | - Nathan E Hudson
- Department of Physics, East Carolina University, Greenville, North Carolina, USA
| | - Mehmet Sen
- Department of Biology and Biochemistry, University of Houston, Houston, Texas, USA.
| |
Collapse
|
19
|
Buerck JP, Foster KM, Larson PR, O'Rear EA. Shear stimulated red blood cell microparticles: Effect on clot structure, flow and fibrinolysis. Biorheology 2023; 59:43-59. [PMID: 36970891 DOI: 10.3233/bir-220012] [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] [Indexed: 06/12/2023]
Abstract
BACKGROUND Microparticles (MPs) have activity in thrombus promotion and generation. Erythrocyte microparticles (ErMPs) have been reported to accelerate fibrinolysis in the absence of permeation. We hypothesized that shear induced ErMPs would affect fibrin structure of clots and change flow with implications for fibrinolysis. OBJECTIVE To determine the effect of ErMPs on clot structure and fibrinolysis. METHODS Plasma with elevated ErMPs was isolated from whole blood or from washed red blood cells (RBCs) resuspended in platelet free plasma (PFP) after high shear. Dynamic light scattering (DLS) provided size distribution of ErMPs from sheared samples and unsheared PFP controls. Clots were formed by recalcification for flow/lysis experiments and examined by confocal microscopy and SEM. Flow rates through clots and time-to-lysis were recorded. A cellular automata model showed the effect of ErMPs on fibrin polymerization and clot structure. RESULTS Coverage of fibrin increased by 41% in clots formed from plasma of sheared RBCs in PFP over controls. Flow rate decreased by 46.7% under a pressure gradient of 10 mmHg/cm with reduction in time to lysis from 5.7 ± 0.7 min to 12.2 ± 1.1 min (p < 0.01). Particle size of ErMPs from sheared samples (200 nm) was comparable to endogenous microparticles. CONCLUSIONS ErMPs alter the fibrin network in a thrombus and affect hydraulic permeability resulting in decelerated delivery of fibrinolytic drugs.
Collapse
Affiliation(s)
- James P Buerck
- School of Chemical, Biological and Materials Engineering, University of Oklahoma, Norman, OK, USA
| | - Kylie M Foster
- School of Chemical, Biological and Materials Engineering, University of Oklahoma, Norman, OK, USA
| | - Preston R Larson
- Samuel Roberts Noble Electron Microscopy Laboratory, University of Oklahoma, Norman, OK, USA
| | - Edgar A O'Rear
- School of Chemical, Biological and Materials Engineering, University of Oklahoma, Norman, OK, USA
- Institute for Biomedical Engineering, Science and Technology, University of Oklahoma, Norman, OK, USA
| |
Collapse
|
20
|
Litvinov RI, Weisel JW. Blood clot contraction: Mechanisms, pathophysiology, and disease. Res Pract Thromb Haemost 2023; 7:100023. [PMID: 36760777 PMCID: PMC9903854 DOI: 10.1016/j.rpth.2022.100023] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 11/09/2022] [Accepted: 11/17/2022] [Indexed: 01/18/2023] Open
Abstract
A State of the Art lecture titled "Blood Clot Contraction: Mechanisms, Pathophysiology, and Disease" was presented at the International Society on Thrombosis and Haemostasis (ISTH) Congress in 2022. This was a systematic description of blood clot contraction or retraction, driven by activated platelets and causing compaction of the fibrin network along with compression of the embedded erythrocytes. The consequences of clot contraction include redistribution of the fibrin-platelet meshwork toward the periphery of the clot and condensation of erythrocytes in the core, followed by their deformation from the biconcave shape into polyhedral cells (polyhedrocytes). These structural signatures of contraction have been found in ex vivo thrombi derived from various locations, which indicated that clots undergo intravital contraction within the blood vessels. In hemostatic clots, tightly packed polyhedrocytes make a nearly impermeable seal that stems bleeding and is impaired in hemorrhagic disorders. In thrombosis, contraction facilitates the local blood flow by decreasing thrombus obstructiveness, reducing permeability, and changing susceptibility to fibrinolytic enzymes. However, in (pro)thrombotic conditions, continuous background platelet activation is followed by platelet exhaustion, refractoriness, and impaired intravital clot contraction, which is associated with weaker thrombi predisposed to embolization. Therefore, assays that detect imperfect in vitro clot contraction have potential diagnostic and prognostic values for imminent or ongoing thrombosis and thrombotic embolism. Collectively, the contraction of blood clots and thrombi is an underappreciated and understudied process that has a pathogenic and clinical significance in bleeding and thrombosis of various etiologies. Finally, we have summarized relevant new data on this topic presented during the 2022 ISTH Congress.
Collapse
Affiliation(s)
- Rustem I Litvinov
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - John W Weisel
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
21
|
Medved L, Weisel JW. The Story of the Fibrin(ogen) αC-Domains: Evolution of Our View on Their Structure and Interactions. Thromb Haemost 2022; 122:1265-1278. [PMID: 34902868 PMCID: PMC10658776 DOI: 10.1055/a-1719-5584] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Although much has been established concerning the overall structure and function of fibrinogen, much less has been known about its two αC regions, each consisting of an αC-connector and an αC-domain, but new information has been accumulating. This review summarizes the state of our current knowledge of the structure and interactions of fibrinogen's αC regions. A series of studies with isolated αC regions and their fragments demonstrated that the αC-domain forms compact ordered structures consisting of N- and C-terminal subdomains including β sheets and suggested that the αC-connector has a poly(L-proline) type II structure. Functionally, the αC-domains interact intramolecularly with each other and with the central region of the molecule, first demonstrated by electron microscopy and then quantified by optical trap force spectroscopy. Upon conversion of fibrinogen into fibrin, the αC-domains switch from intra- to intermolecular interactions to form ordered αC polymers. The formation of αC polymers occurs mainly through the homophilic interaction between the N-terminal subdomains; interaction between the C-terminal subdomains and the αC-connectors also contributes to this process. Considerable evidence supports the idea that the αC-regions accelerate fibrin polymerization and affect the final structure of fibrin clots. The interactions between αC-regions are important for the mechanical properties of clots, increasing their stiffness and extensibility. Conversion of fibrinogen into fibrin results in exposure of multiple binding sites in its αC regions, providing interaction of fibrin with different proteins and cell types during hemostasis and wound healing. This heretofore mysterious part of the fibrinogen molecule is finally giving up its secrets.
Collapse
Affiliation(s)
- Leonid Medved
- Center for Vascular and Inflammatory Diseases and the Department of Biochemistry, University of Maryland School of Medicine, Baltimore, Maryland 21201, United States
| | - John W. Weisel
- Department of Cell and Developmental Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| |
Collapse
|
22
|
Engineered Molecular Therapeutics Targeting Fibrin and the Coagulation System: a Biophysical Perspective. Biophys Rev 2022; 14:427-461. [PMID: 35399372 PMCID: PMC8984085 DOI: 10.1007/s12551-022-00950-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 03/25/2022] [Indexed: 02/07/2023] Open
Abstract
The coagulation cascade represents a sophisticated and highly choreographed series of molecular events taking place in the blood with important clinical implications. One key player in coagulation is fibrinogen, a highly abundant soluble blood protein that is processed by thrombin proteases at wound sites, triggering self-assembly of an insoluble protein hydrogel known as a fibrin clot. By forming the key protein component of blood clots, fibrin acts as a structural biomaterial with biophysical properties well suited to its role inhibiting fluid flow and maintaining hemostasis. Based on its clinical importance, fibrin is being investigated as a potentially valuable molecular target in the development of coagulation therapies. In this topical review, we summarize our current understanding of the coagulation cascade from a molecular, structural and biophysical perspective. We highlight single-molecule studies on proteins involved in blood coagulation and report on the current state of the art in directed evolution and molecular engineering of fibrin-targeted proteins and polymers for modulating coagulation. This biophysical overview will help acclimatize newcomers to the field and catalyze interdisciplinary work in biomolecular engineering toward the development of new therapies targeting fibrin and the coagulation system.
Collapse
|
23
|
Maly M, Riedel T, Stikarova J, Suttnar J, Kotlin R, Hajsl M, Tousek P, Kaufmanova J, Kucerka O, Weisel JW, Dyr JE. Incorporation of Fibrin, Platelets, and Red Blood Cells into a Coronary Thrombus in Time and Space. Thromb Haemost 2021; 122:434-444. [PMID: 34781375 DOI: 10.1055/s-0041-1739193] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
We describe the internal structure, spatial organization and dynamic formation of coronary artery thrombi from ST-segment elevation myocardial infarction patients. Scanning electron microscopy (SEM) revealed significant differences among four groups of patients (<2 hours; 2-6 hours; 6-12 hours, and >12 hours) related to the time of ischemia. Coronary artery thrombi from patients presenting less than 2 hours after the infarction were almost entirely composed of platelets, with small amounts of fibrin and red blood cells. In contrast, thrombi from late presenters (>12 hours) consisted of mainly platelets at the distal end, where clotting was initiated, with almost no platelets at the proximal end, while the red blood cell content went from low at the initiating end to more than 90% at the proximal end. Furthermore, fibrin was present mainly on the outside of the thrombi and older thrombi contained thicker fibers. The red blood cells in late thrombi were compressed to a close-packed, tessellated array of polyhedral structures, called polyhedrocytes. Moreover, there was redistribution from the originally homogeneous composition to fibrin and platelets to the outside, with polyhedrocytes on the interior. The presence of polyhedrocytes and the redistribution of components are signs of in vivo clot contraction (or retraction). These results suggest why later thrombi are resistant to fibrinolytic agents and other treatment modalities, since the close-packed polyhedrocytes form a nearly impermeable seal. Furthermore, it is of particular clinical significance that these findings suggest specific disparate therapies that will be most effective at different stages of thrombus development.
Collapse
Affiliation(s)
- Martin Maly
- First Faculty of Medicine, Department of Medicine, Charles University in Prague and Military University Hospital, Prague, Czech Republic
| | - Tomas Riedel
- Department of Biochemistry, Institute of Hematology and Blood Transfusion, Prague, Czech Republic.,Chemistry and Physics of Surfaces and Biointerfaces, Institute of Macromolecular Chemistry, Czech Academy of Sciences, Prague, Czech Republic
| | - Jana Stikarova
- Department of Biochemistry, Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - Jiri Suttnar
- Department of Biochemistry, Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - Roman Kotlin
- Department of Biochemistry, Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - Martin Hajsl
- First Faculty of Medicine, Department of Medicine, Charles University in Prague and Military University Hospital, Prague, Czech Republic
| | - Petr Tousek
- Cardiocenter, University Hospital Kralovske Vinohrady and Third Medical Faculty, Charles University, Prague, Czech Republic
| | - Jirina Kaufmanova
- Department of Biochemistry, Institute of Hematology and Blood Transfusion, Prague, Czech Republic.,Department of Biochemistry and Microbiology, University of Chemistry and Technology Prague, Prague, Czech Republic
| | - Ondrej Kucerka
- First Faculty of Medicine, Department of Medicine, Charles University in Prague and Military University Hospital, Prague, Czech Republic
| | - John W Weisel
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Jan E Dyr
- Department of Biochemistry, Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| |
Collapse
|
24
|
Platelet binding to polymerizing fibrin is avidity driven and requires activated αIIbβ3 but not fibrin cross-linking. Blood Adv 2021; 5:3986-4002. [PMID: 34647980 PMCID: PMC8945615 DOI: 10.1182/bloodadvances.2021005142] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 07/12/2021] [Indexed: 11/20/2022] Open
Abstract
Platelet interaction with polymerizing fibrin is avidity driven and requires activated αIIbβ3 but not fibrin cross-linking. The mechanism by which αIIbβ3 interacts with polymerizing fibrin differs subtly from the interaction of αIIbβ3 with fibrinogen.
The molecular basis of platelet-fibrin interactions remains poorly understood despite the predominance of fibrin in thrombi. We have studied the interaction of platelets with polymerizing fibrin by adding thrombin to washed platelets in the presence of the peptide RGDW, which inhibits the initial platelet aggregation mediated by fibrinogen binding to αIIbβ3 but leaves intact a delayed increase in light transmission (delayed wave; DW) as platelets interact with the polymerizing fibrin. The DW was absent in platelets from a patient with Glanzmann thrombasthenia, indicating a requirement for αIIbβ3. The DW required αIIbb3 activation and it was inhibited by the αIIbβ3 antagonists eptifibatide and the monoclonal antibody (mAb) 7E3, but only at much higher concentrations than needed to inhibit platelet aggregation initiated by a thrombin receptor activating peptide (T6). Surface plasmon resonance and scanning electron microscopy studies both supported fibrin having greater avidity for αIIbβ3 than fibrinogen rather than greater affinity, consistent with fibrin’s multivalency. mAb 10E5, a potent inhibitor of T6-induced platelet aggregation, did not inhibit the DW, suggesting that fibrin differs from fibrinogen in its mechanism of binding. Inhibition of factor XIII–mediated fibrin cross-linking by >95% reduced the DW by only 32%. Clot retraction showed a pattern of inhibition similar to that of the DW. We conclude that activated αIIbβ3 is the primary mediator of platelet-fibrin interactions leading to clot retraction, and that the interaction is avidity driven, does not require fibrin cross-linking, and is mediated by a mechanism that differs subtly from that of the interaction of αIIbβ3 with fibrinogen.
Collapse
|
25
|
Galanakis DK, Protopopova A, Zhang L, Li K, Marmorat C, Scheiner T, Koo J, Savitt AG, Rafailovich M, Weisel J. Fibers Generated by Plasma Des-AA Fibrin Monomers and Protofibril/Fibrinogen Clusters Bind Platelets: Clinical and Nonclinical Implications. TH OPEN 2021; 5:e273-e285. [PMID: 34240000 PMCID: PMC8260279 DOI: 10.1055/s-0041-1725976] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 01/26/2021] [Indexed: 12/31/2022] Open
Abstract
Objective Soluble fibrin (SF) is a substantial component of plasma fibrinogen (fg), but its composition, functions, and clinical relevance remain unclear. The study aimed to evaluate the molecular composition and procoagulant function(s) of SF. Materials and Methods Cryoprecipitable, SF-rich (FR) and cryosoluble, SF-depleted (FD) fg isolates were prepared and adsorbed on one hydrophilic and two hydrophobic surfaces and scanned by atomic force microscopy (AFM). Standard procedures were used for fibrin polymerization, crosslinking by factor XIII, electrophoresis, and platelet adhesion. Results Relative to FD fg, thrombin-induced polymerization of FR fg was accelerated and that induced by reptilase was markedly delayed, attributable to its decreased (fibrinopeptide A) FpA. FR fg adsorption to each surface yielded polymeric clusters and co-cryoprecipitable solitary monomers. Cluster components were crosslinked by factor XIII and comprised ≤21% of FR fg. In contrast to FD fg, FR fg adsorption on hydrophobic surfaces resulted in fiber generation enabled by both clusters and solitary monomers. This began with numerous short protofibrils, which following prolonged adsorption increased in number and length and culminated in surface-linked three-dimensional fiber networks that bound platelets. Conclusion The abundance of adsorbed protofibrils resulted from (1) protofibril/fg clusters whose fg was dissociated during adsorption, and (2) adsorbed des-AA monomers that attracted solution counterparts initiating protofibril assembly and elongation by their continued incorporation. The substantial presence of both components in transfused plasma and cryoprecipitate augments hemostasis by accelerating thrombin-induced fibrin polymerization and by tightly anchoring the resulting clot to the underlying wound or to other abnormal vascular surfaces.
Collapse
Affiliation(s)
- Dennis K Galanakis
- Department of Pathology, Stony Brook University School of Medicine, Stony Brook, New York
| | - Anna Protopopova
- Department of Cell and Developmental Biology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Liudi Zhang
- Department of Materials Science and Engineering, Stony Brook University, Stony Brook, New York
| | - Kao Li
- Department of Materials Science and Engineering, Stony Brook University, Stony Brook, New York
| | - Clement Marmorat
- Department of Materials Science and Engineering, Stony Brook University, Stony Brook, New York
| | - Tomas Scheiner
- Department of Cell and Developmental Biology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Jaseung Koo
- Department of Materials Science and Engineering, Stony Brook University, Stony Brook, New York
| | - Anne G Savitt
- Department of Microbiology and Immunology, Stony Brook University School of Medicine, Stony Brook, New York
| | - Miriam Rafailovich
- Department of Materials Science and Engineering, Stony Brook University, Stony Brook, New York
| | - John Weisel
- Department of Cell and Developmental Biology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| |
Collapse
|
26
|
Grant DM, Macedo A, Toms D, Klein C. Fibrinogen in equine pregnancy as a mediator of cell adhesion, an epigenetic and functional investigation. Biol Reprod 2021; 102:170-184. [PMID: 31403677 DOI: 10.1093/biolre/ioz157] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 07/09/2019] [Accepted: 07/31/2019] [Indexed: 11/12/2022] Open
Abstract
Preimplantation equine embryos synthesize and secrete fibrinogen, which is a peculiar finding as fibrinogen synthesis almost exclusively occurs in the liver. This study investigated the hypothesis that conceptus-derived fibrinogen mediates cell adhesion during fixation. On day 21 of pregnancy, five integrin subunits, including ITGA5, ITGB1, ITGAV, and ITGB1, displayed significantly higher transcript abundance than on day 16 of pregnancy. Endometrial epithelial cells adhered to fibrinogen in an integrin-dependent manner in an in vitro cell adhesion assay. Bilaminar trophoblast and allantochorion expressed fibrinogen transcript, indicating that fibrinogen expression persists past fixation. Preimplantation-phase endometrium, conceptuses, and microcotyledonary tissue expressed components of the clotting cascade regulating fibrin homeostasis, leaving open the possibility that fibrinogen is converted to fibrin. Fibrinogen is likely to have functions beyond mediating cell adhesion, such trapping growth factors and triggering signaling cascades, and has remarkable parallels to the expression of fibrinogen by some tumors. The deposition of fibrinogen within tumor stroma is characteristic of breast carcinoma, and tumor-derived fibrinogen has been implicated in the metastatic potential of circulating tumor cells. DNA methylation of the fibrinogen locus in equine conceptuses was examined in comparison to liver and endometrium, and across the full gene cluster, was significantly higher for endometrium than liver and conceptus. DNA methylation of regulatory regions did not differ between liver and conceptus, and was significantly lower than in endometrium. These results, therefore, support the hypothesis of DNA methylation being a regulator of fibrinogen expression in the conceptus.
Collapse
Affiliation(s)
- Danielle M Grant
- Department of Veterinary and Clinical Diagnostic Sciences, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Alysson Macedo
- Department of Veterinary and Clinical Diagnostic Sciences, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Derek Toms
- Department of Veterinary and Clinical Diagnostic Sciences, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Claudia Klein
- Department of Veterinary and Clinical Diagnostic Sciences, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
27
|
Gupta P, Zhang P, Sheriff J, Bluestein D, Deng Y. A multiscale model for multiple platelet aggregation in shear flow. Biomech Model Mechanobiol 2021; 20:1013-1030. [PMID: 33782796 PMCID: PMC8274306 DOI: 10.1007/s10237-021-01428-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Accepted: 01/22/2021] [Indexed: 10/21/2022]
Abstract
We developed a multiscale model for simulating aggregation of multiple, free-flowing platelets in low-intermediate shear viscous flow, in which aggregation is mediated by the interaction of αIIbβ3 receptors on the platelet membrane and fibrinogen (Fg). This multiscale model uses coarse grained molecular dynamics (CGMD) for platelets at the microscales and dissipative particle dynamics (DPD) for the shear flow at the macroscales, employing our hybrid aggregation force field for modeling molecular level receptor ligand bonds. We define an aggregation tensor and use it to quantify the molecular level contact characteristics between platelets in an aggregate. We perform numerical studies under different flow conditions for platelet doublets and triplets and evaluate the contact area, detaching force and minimum distance between different pairs of platelets in an aggregate. We also present the dynamics of applied stress and velocity magnitude distributions on the platelet membrane during aggregation and quantify the increase in stress in the contact region under different flow conditions. Integrating the knowledge from our previously validated models, together with new aggregation scenarios, our model can dynamically quantify aggregation characteristics and map stress and velocity distribution on the platelet membrane which are difficult to measure in vitro, thus providing an insight into mechanotransduction bond formation response of platelets to flow-induced shear stresses. This modeling framework, together with the tensor method for quantifying inter-platelet contact, can be extended to simulate and analyze larger aggregates and their adhesive properties.
Collapse
Affiliation(s)
- Prachi Gupta
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Peng Zhang
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, NY, 11794, USA
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Jawaad Sheriff
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Danny Bluestein
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Yuefan Deng
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, NY, 11794, USA.
| |
Collapse
|
28
|
Kirwan DE, Chong DLW, Friedland JS. Platelet Activation and the Immune Response to Tuberculosis. Front Immunol 2021; 12:631696. [PMID: 34093524 PMCID: PMC8170316 DOI: 10.3389/fimmu.2021.631696] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 05/04/2021] [Indexed: 12/24/2022] Open
Abstract
In 2019 10 million people developed symptomatic tuberculosis (TB) disease and 1.2 million died. In active TB the inflammatory response causes tissue destruction, which leads to both acute morbidity and mortality. Tissue destruction in TB is driven by host innate immunity and mediated via enzymes, chiefly matrix metalloproteinases (MMPs) which are secreted by leukocytes and stromal cells and degrade the extracellular matrix. Here we review the growing evidence implicating platelets in TB immunopathology. TB patients typically have high platelet counts, which correlate with disease severity, and a hypercoagulable profile. Platelets are present in human TB granulomas and platelet-associated gene transcripts are increased in TB patients versus healthy controls. Platelets most likely drive TB immunopathology through their effect on other immune cells, particularly monocytes, to lead to upregulation of activation markers, increased MMP secretion, and enhanced phagocytosis. Finally, we consider current evidence supporting use of targeted anti-platelet agents in the treatment of TB due to growing interest in developing host-directed therapies to limit tissue damage and improve treatment outcomes. In summary, platelets are implicated in TB disease and contribute to MMP-mediated tissue damage via their cellular interactions with other leukocytes, and are potential targets for novel host-directed therapies.
Collapse
Affiliation(s)
- Daniela E Kirwan
- Institute for Infection & Immunity, St. George's, University of London, London, United Kingdom
| | - Deborah L W Chong
- Institute for Infection & Immunity, St. George's, University of London, London, United Kingdom
| | - Jon S Friedland
- Institute for Infection & Immunity, St. George's, University of London, London, United Kingdom
| |
Collapse
|
29
|
Kaneva VN, Dunster JL, Volpert V, Ataullahanov F, Panteleev MA, Nechipurenko DY. Modeling Thrombus Shell: Linking Adhesion Receptor Properties and Macroscopic Dynamics. Biophys J 2021; 120:334-351. [PMID: 33472026 DOI: 10.1016/j.bpj.2020.10.049] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 09/10/2020] [Accepted: 10/15/2020] [Indexed: 02/06/2023] Open
Abstract
Damage to arterial vessel walls leads to the formation of platelet aggregate, which acts as a physical obstacle for bleeding. An arterial thrombus is heterogeneous; it has a dense inner part (core) and an unstable outer part (shell). The thrombus shell is very dynamic, being composed of loosely connected discoid platelets. The mechanisms underlying the observed mobility of the shell and its (patho)physiological implications are unclear. To investigate arterial thrombus mechanics, we developed a novel, to our knowledge, two-dimensional particle-based computational model of microvessel thrombosis. The model considers two types of interplatelet interactions: primary reversible (glycoprotein Ib (GPIb)-mediated) and stronger integrin-mediated interaction, which intensifies with platelet activation. At high shear rates, the former interaction leads to adhesion, and the latter is primarily responsible for stable platelet aggregation. Using a stochastic model of GPIb-mediated interaction, we initially reproduced experimental curves that characterize individual platelet interactions with a von Willebrand factor-coated surface. The addition of the second stabilizing interaction results in thrombus formation. The comparison of thrombus dynamics with experimental data allowed us to estimate the magnitude of critical interplatelet forces in the thrombus shell and the characteristic time of platelet activation. The model predicts moderate dependence of maximal thrombus height on the injury size in the absence of thrombin activity. We demonstrate that the developed stochastic model reproduces the observed highly dynamic behavior of the thrombus shell. The presence of primary stochastic interaction between platelets leads to the properties of thrombus consistent with in vivo findings; it does not grow upstream of the injury site and covers the whole injury from the first seconds of the formation. А simplified model, in which GPIb-mediated interaction is deterministic, does not reproduce these features. Thus, the stochasticity of platelet interactions is critical for thrombus plasticity, suggesting that interaction via a small number of bonds drives the dynamics of arterial thrombus shell.
Collapse
Affiliation(s)
- Valeriia N Kaneva
- Center for Theoretical Problems of Physico-chemical Pharmacology, Russian Academy of Sciences, Moscow, Russia
| | - Joanne L Dunster
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Whiteknights, Reading, United Kingdom
| | - Vitaly Volpert
- Institut Camille Jordan, UMR 5208 CNRS, University Lyon 1, Villeurbanne, France; INRIA Team Dracula, INRIA Lyon La Doua, Villeurbanne, France; Peoples Friendship University of Russia (RUDN University), Moscow, Russian Federation
| | - Fazoil Ataullahanov
- Center for Theoretical Problems of Physico-chemical Pharmacology, Russian Academy of Sciences, Moscow, Russia; Dmitry Rogachev National Medical Research Centre of Pediatric Hematology, Oncology and Immunology, Moscow, Russia; Faculty of Physics, Lomonosov Moscow State University, Moscow, Russia; Faculty of Biological and Medical Physics, Moscow Institute of Physics and Technology, Dolgoprudnyi, Russia
| | - Mikhail A Panteleev
- Center for Theoretical Problems of Physico-chemical Pharmacology, Russian Academy of Sciences, Moscow, Russia; Dmitry Rogachev National Medical Research Centre of Pediatric Hematology, Oncology and Immunology, Moscow, Russia; Faculty of Physics, Lomonosov Moscow State University, Moscow, Russia; Faculty of Biological and Medical Physics, Moscow Institute of Physics and Technology, Dolgoprudnyi, Russia
| | - Dmitry Yu Nechipurenko
- Center for Theoretical Problems of Physico-chemical Pharmacology, Russian Academy of Sciences, Moscow, Russia; Dmitry Rogachev National Medical Research Centre of Pediatric Hematology, Oncology and Immunology, Moscow, Russia; Faculty of Physics, Lomonosov Moscow State University, Moscow, Russia.
| |
Collapse
|
30
|
Branfield S, Washington AV. The enigmatic nature of the triggering receptor expressed in myeloid cells -1 (TLT- 1). Platelets 2021; 32:753-760. [PMID: 33560928 DOI: 10.1080/09537104.2021.1881948] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Receptors are important pharmacological targets on cells. The Triggering Receptor Expressed on Myeloid Cells (TREM) - Like Transcript - 1 is an abundant, yet little understood, platelet receptor. It is a single Ig domain containing receptor isolated in the α-granules of resting platelets and brought to the platelet surface upon activation. On platelets, the integrin αIIbβ3 is the major receptor having roughly 80,000 copies. αIIbβ3 is a heterodimeric multidomain structure that mediates platelet aggregation through its interaction with the plasma protein fibrinogen. Anti-platelet drugs have successfully targeted αIIbβ3 to control thrombosis. Like αIIbβ3, TLT-1 also binds fibrinogen, making its role in platelet function somewhat obscure. In this review, we highlight the known structural features of TLT-1 and present the challenges of understanding TLT-1 function. In our analysis of the dynamics of the platelet surface after activation we propose a model in which TLT-1 supports αIIbβ3 function as a mechanoreceptor that may direct platelets toward immune function.
Collapse
Affiliation(s)
- Siobhan Branfield
- , Department of Biology, University of Puerto Rico- Rio Piedras- Molecular Science Research Center, San Juan, Puerto Rico
| | - A Valance Washington
- , Department of Biology, University of Puerto Rico- Rio Piedras- Molecular Science Research Center, San Juan, Puerto Rico
| |
Collapse
|
31
|
Firkowska-Boden I, Helbing C, Dauben TJ, Pieper M, Jandt KD. How Nanotopography-Induced Conformational Changes of Fibrinogen Affect Platelet Adhesion and Activation. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2020; 36:11573-11580. [PMID: 32921061 DOI: 10.1021/acs.langmuir.0c02094] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
The conformational state of adsorbed human plasma fibrinogen (HPF) has been recognized as the determinant factor in platelet adhesion and thrombus formation on blood-contacting biomaterials. Studies have highlighted the ability to control the HPF conformation merely by tailoring surface nanotopographical features. However, a clear relationship between the conformational changes of adsorbed HPF and the degree of platelet adhesion and activation achieved with different surface nanotopographies is still unclear. Here, we examined HPF assembly characteristics on nanostructured polybutene-1 (PB-1) surfaces with nanosized lamellar crystals (LCs), needle-like crystals (NLCs), and a nanostructured high-density polyethylene (HDPE) surface with shish-kebab crystals (SKCs), at a biologically relevant HPF concentration. By exposing the nanostructured surfaces with preadsorbed HPF to human platelets, significant differences in platelet response on LCs/SKCs and NLCs were identified. The former presented a uniform monolayer in the advanced stage of activation, whereas the latter exhibited minimal adhesion and the early stage of activation. Distinct platelet response was related to the postadsorption conformational changes in HPF, which were confirmed by topography-dependent shifts of the amide I band in attenuated total reflection-Fourier transform infrared (ATR-FTIR) analysis. Supported by atomic force microscopy (AFM) characterization, we propose that the mechanism behind the nanotopography-induced HPF conformation is driven by the interplay between the aspect ratios of polymeric crystals and HPF. From the biomedical perspective, our work reveals that surface structuring in a nanoscale size regime can provide a fine-tuning mechanism to manipulate HPF conformation, which can be exploited for the design of thromboresistant biomaterials surfaces.
Collapse
Affiliation(s)
- Izabela Firkowska-Boden
- Chair of Materials Science, Otto Schott Institute of Materials Research, Faculty of Physics and Astronomy, Friedrich Schiller University Jena, Löbdergraben 32, 07743 Jena, Germany
| | - Christian Helbing
- Chair of Materials Science, Otto Schott Institute of Materials Research, Faculty of Physics and Astronomy, Friedrich Schiller University Jena, Löbdergraben 32, 07743 Jena, Germany
| | - Thomas J Dauben
- Chair of Materials Science, Otto Schott Institute of Materials Research, Faculty of Physics and Astronomy, Friedrich Schiller University Jena, Löbdergraben 32, 07743 Jena, Germany
| | - Maja Pieper
- Chair of Materials Science, Otto Schott Institute of Materials Research, Faculty of Physics and Astronomy, Friedrich Schiller University Jena, Löbdergraben 32, 07743 Jena, Germany
| | - Klaus D Jandt
- Chair of Materials Science, Otto Schott Institute of Materials Research, Faculty of Physics and Astronomy, Friedrich Schiller University Jena, Löbdergraben 32, 07743 Jena, Germany
- Jena Center for Soft Matter, Friedrich Schiller University Jena, Philosophenweg 7, 07743 Jena, Germany
- Jena School for Microbial Communication (JSMC), Neugasse 23, 07743 Jena, Germany
| |
Collapse
|
32
|
Chen X, Zhao X, Cooper M, Ma P. The Roles of GRKs in Hemostasis and Thrombosis. Int J Mol Sci 2020; 21:ijms21155345. [PMID: 32731360 PMCID: PMC7432802 DOI: 10.3390/ijms21155345] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 07/20/2020] [Accepted: 07/27/2020] [Indexed: 12/20/2022] Open
Abstract
Along with cancer, cardiovascular and cerebrovascular diseases remain by far the most common causes of death. Heart attacks and strokes are diseases in which platelets play a role, through activation on ruptured plaques and subsequent thrombus formation. Most platelet agonists activate platelets via G protein-coupled receptors (GPCRs), which make these receptors ideal targets for many antiplatelet drugs. However, little is known about the mechanisms that provide feedback regulation on GPCRs to limit platelet activation. Emerging evidence from our group and others strongly suggests that GPCR kinases (GRKs) are critical negative regulators during platelet activation and thrombus formation. In this review, we will summarize recent findings on the role of GRKs in platelet biology and how one specific GRK, GRK6, regulates the hemostatic response to vascular injury. Furthermore, we will discuss the potential role of GRKs in thrombotic disorders, such as thrombotic events in COVID-19 patients. Studies on the function of GRKs during platelet activation and thrombus formation have just recently begun, and a better understanding of the role of GRKs in hemostasis and thrombosis will provide a fruitful avenue for understanding the hemostatic response to injury. It may also lead to new therapeutic options for the treatment of thrombotic and cardiovascular disorders.
Collapse
Affiliation(s)
- Xi Chen
- Cardeza Foundation for Hematologic Research, Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA; (X.C.); (X.Z.); (M.C.)
| | - Xuefei Zhao
- Cardeza Foundation for Hematologic Research, Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA; (X.C.); (X.Z.); (M.C.)
- Cyrus Tang Hematology Center, Soochow University, Suzhou 215123, China
| | - Matthew Cooper
- Cardeza Foundation for Hematologic Research, Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA; (X.C.); (X.Z.); (M.C.)
| | - Peisong Ma
- Cardeza Foundation for Hematologic Research, Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA; (X.C.); (X.Z.); (M.C.)
- Correspondence: ; Tel.: +1-215-955-3966
| |
Collapse
|
33
|
Buitrago L, Zafar H, Zhang Y, Li J, Walz T, Coller BS. Dominant role of αIIbβ3 in platelet interactions with cross-linked fibrin fragment D-dimer. Blood Adv 2020; 4:2939-2949. [PMID: 32603423 PMCID: PMC7362365 DOI: 10.1182/bloodadvances.2020001545] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 05/23/2020] [Indexed: 12/14/2022] Open
Abstract
Although much is known about the interaction of fibrinogen with αIIbβ3, much less is known about the interaction of platelets with cross-linked fibrin. Fibrinogen residue Lys406 plays a vital role in the interaction of fibrinogen with αIIbβ3, but because it participates in fibrin cross-linking, it is not available for interacting with αIIbβ3. We studied the adhesion of platelets and HEK cells expressing normal and constitutively active αIIbβ3 to both immobilized fibrinogen and D-dimer, a proteolytic fragment of cross-linked fibrin, as well as platelet-mediated clot retraction. Nonactivated platelets and HEK cells expressing normal αIIbβ3 adhered to fibrinogen but not D-dimer, whereas activated platelets as well as HEK cells expressing activated αIIbβ3 both bound to D-dimer. Small-molecule antagonists of the αIIbβ3 RGD (Arg-Gly-Asp) binding pocket inhibited adhesion to D-dimer, and an Asp119Ala mutation that disrupts the β3 metal ion-dependent adhesion site inhibited αIIbβ3-mediated adhesion to D-dimer. D-dimer and a polyclonal antibody against D-dimer inhibited clot retraction. The monoclonal antibody (mAb) 10E5, directed at αIIb and a potent inhibitor of platelet interactions with fibrinogen, did not inhibit the interaction of activated platelets with D-dimer or clot retraction, whereas the mAb 7E3, directed at β3, inhibited both phenomena. We conclude that activated, but not nonactivated, αIIbβ3 mediates interactions between platelets and D-dimer, and by extrapolation, to cross-linked fibrin. Although the interaction of αIIbβ3 with D-dimer differs from that with fibrinogen, it probably involves contributions from regions on β3 that are close to, or that are affected by, changes in the RGD binding pocket.
Collapse
Affiliation(s)
| | - Hina Zafar
- Laboratory of Blood and Vascular Biology and
| | - Yixiao Zhang
- Laboratory of Molecular Electron Microscopy, Rockefeller University, New York, NY
| | - Jihong Li
- Laboratory of Blood and Vascular Biology and
| | - Thomas Walz
- Laboratory of Molecular Electron Microscopy, Rockefeller University, New York, NY
| | | |
Collapse
|
34
|
Chen Y, Ju LA. Biomechanical thrombosis: the dark side of force and dawn of mechano-medicine. Stroke Vasc Neurol 2020; 5:185-197. [PMID: 32606086 PMCID: PMC7337368 DOI: 10.1136/svn-2019-000302] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 11/14/2019] [Indexed: 12/19/2022] Open
Abstract
Arterial thrombosis is in part contributed by excessive platelet aggregation, which can lead to blood clotting and subsequent heart attack and stroke. Platelets are sensitive to the haemodynamic environment. Rapid haemodynamcis and disturbed blood flow, which occur in vessels with growing thrombi and atherosclerotic plaques or is caused by medical device implantation and intervention, promotes platelet aggregation and thrombus formation. In such situations, conventional antiplatelet drugs often have suboptimal efficacy and a serious side effect of excessive bleeding. Investigating the mechanisms of platelet biomechanical activation provides insights distinct from the classic views of agonist-stimulated platelet thrombus formation. In this work, we review the recent discoveries underlying haemodynamic force-reinforced platelet binding and mechanosensing primarily mediated by three platelet receptors: glycoprotein Ib (GPIb), glycoprotein IIb/IIIa (GPIIb/IIIa) and glycoprotein VI (GPVI), and their implications for development of antithrombotic 'mechano-medicine' .
Collapse
Affiliation(s)
- Yunfeng Chen
- Molecular Medicine, Scripps Research Institute, La Jolla, California, USA
| | - Lining Arnold Ju
- School of Biomedical Engineering, Heart Research Institute and Charles Perkins Centre, The University of Sydney, Camperdown, New South Wales, Australia
| |
Collapse
|
35
|
Horev MB, Zabary Y, Zarka R, Sorrentino S, Medalia O, Zaritsky A, Geiger B. Differential dynamics of early stages of platelet adhesion and spreading on collagen IV- and fibrinogen-coated surfaces. F1000Res 2020; 9. [PMID: 32566134 PMCID: PMC7281675 DOI: 10.12688/f1000research.23598.2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/25/2020] [Indexed: 12/29/2022] Open
Abstract
Background: Upon wound formation, platelets adhere to the neighboring extracellular matrix and spread on it, a process which is critical for physiological wound healing. Multiple external factors, such as the molecular composition of the environment and its mechanical properties, play a key role in this process and direct its speed and outcome. Methods: We combined live cell imaging, quantitative interference reflection microscopy and cryo-electron tomography to characterize, at a single platelet level, the differential spatiotemporal dynamics of the adhesion process to fibrinogen- and collagen IV-functionalized surfaces. Results: Initially, platelets sense both substrates by transient rapid extensions of filopodia. On collagen IV, a short-term phase of filopodial extension is followed by lamellipodia-based spreading. This transition is preceded by the extension of a single or couple of microtubules into the platelet's periphery and their apparent insertion into the core of the filopodia. On fibrinogen surfaces, the filopodia-to-lamellipodia transition was partial and microtubule extension was not observed leading to limited spreading, which could be restored by manganese or thrombin. Conclusions: Based on these results, we propose that interaction with collagen IV stimulate platelets to extend microtubules to peripheral filopodia, which in turn, enhances filopodial-to-lamellipodial transition and overall lamellipodia-based spreading. Fibrinogen, on the other hand, fails to induce these early microtubule extensions, leading to full lamellipodia spreading in only a fraction of the seeded platelets. We further suggest that activation of integrin αIIbβ3 is essential for filopodial-to-lamellipodial transition, based on the capacity of integrin activators to enhance lamellipodia spreading on fibrinogen.
Collapse
Affiliation(s)
- Melanie B Horev
- Department of Immunology, Weizmann Institute of Science, Rehovot, Rehovot, 76100, Israel
| | - Yishaia Zabary
- Department of Software and Information Systems Engineering, Ben-Gurion University of the Negev, Beer-Sheva, 84105, Israel
| | - Revital Zarka
- Department of Immunology, Weizmann Institute of Science, Rehovot, Rehovot, 76100, Israel
| | - Simona Sorrentino
- Department of Biochemistry, University of Zurich, Zurich, CH-8057, Switzerland
| | - Ohad Medalia
- Department of Biochemistry, University of Zurich, Zurich, CH-8057, Switzerland
| | - Assaf Zaritsky
- Department of Software and Information Systems Engineering, Ben-Gurion University of the Negev, Beer-Sheva, 84105, Israel
| | - Benjamin Geiger
- Department of Immunology, Weizmann Institute of Science, Rehovot, Rehovot, 76100, Israel
| |
Collapse
|
36
|
Structure-guided design of pure orthosteric inhibitors of αIIbβ3 that prevent thrombosis but preserve hemostasis. Nat Commun 2020; 11:398. [PMID: 31964886 PMCID: PMC6972956 DOI: 10.1038/s41467-019-13928-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 12/06/2019] [Indexed: 02/07/2023] Open
Abstract
A prevailing dogma is that inhibition of vascular thrombosis by antagonizing platelet integrin αIIbβ3 cannot be achieved without compromising hemostasis, thus causing serious bleeding and increased morbidity and mortality. It is speculated that these adverse outcomes result from drug-induced activating conformational changes in αIIbβ3 but direct proof is lacking. Here, we report the structure-guided design of peptide Hr10 and a modified form of the partial agonist drug tirofiban that act as "pure" antagonists of αIIbβ3, i.e., they no longer induce the conformational changes in αIIbβ3. Both agents inhibit human platelet aggregation but preserve clot retraction. Hr10 and modified tirofiban are as effective as partial agonist drugs in inhibiting vascular thrombosis in humanized mice, but neither causes serious bleeding, establishing a causal link between partial agonism and impaired hemostasis. Pure orthosteric inhibitors of αIIbβ3 may thus provide safer alternatives for human therapy, and valuable tools to probe structure-activity relationships in integrins.
Collapse
|
37
|
Abstract
Acute kidney injury (AKI), a major public health problem associated with high mortality and increased risk of progression towards end-stage renal disease, is characterized by the activation of intra-renal haemostatic and inflammatory processes. Platelets, which are present in high numbers in the circulation and can rapidly release a broad spectrum of bioactive mediators, are important acute modulators of inflammation and haemostasis, as they are the first cells to arrive at sites of acute injury, where they interact with endothelial cells and leukocytes. Diminished control of platelet reactivity by endothelial cells and/or an increased release of platelet-activating mediators can lead to uncontrolled platelet activation in AKI. As increased platelet sequestration and increased expression levels of the markers P-selectin, thromboxane A2, CC-chemokine ligand 5 and platelet factor 4 on platelets have been reported in kidneys following AKI, platelet activation likely plays a part in AKI pathology. Results from animal models and some clinical studies highlight the potential of antiplatelet therapies in the preservation of renal function in the context of AKI, but as current strategies also affect other cell types and non-platelet-derived mediators, additional studies are required to further elucidate the extent of platelet contribution to the pathology of AKI and to determine the best therapeutic approach by which to specifically target related pathogenic pathways.
Collapse
Affiliation(s)
- Marcel P B Jansen
- Department of Pathology, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Sandrine Florquin
- Department of Pathology, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Joris J T H Roelofs
- Department of Pathology, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands.
| |
Collapse
|
38
|
Abstract
The vasculature is a dynamic environment in which blood platelets constantly survey the endothelium for sites of vessel damage. The formation of a mechanically coherent hemostatic plug to prevent blood loss relies on a coordinated series of ligand-receptor interactions governing the recruitment, activation, and aggregation of platelets. The physical biology of each step is distinct in that the recruitment of platelets depends on the mechanosensing of the platelet receptor glycoprotein Ib for the adhesive protein von Willebrand factor, whereas platelet activation and aggregation are responsive to the mechanical forces sensed at adhesive junctions between platelets and at the platelet-matrix interface. Herein we take a biophysical perspective to discuss the current understanding of platelet mechanotransduction as well as the measurement techniques used to quantify the physical biology of platelets in the context of thrombus formation under flow.
Collapse
Affiliation(s)
- Caroline E Hansen
- Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Children's Healthcare of Atlanta/Emory University School of Medicine, Atlanta, Georgia 30332, USA; .,Wallace H. Coulter Department of Biomedical Engineering and Institute for Electronics and Nanotechnology, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30332, USA
| | - Yongzhi Qiu
- Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Children's Healthcare of Atlanta/Emory University School of Medicine, Atlanta, Georgia 30332, USA; .,Wallace H. Coulter Department of Biomedical Engineering and Institute for Electronics and Nanotechnology, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30332, USA
| | - Owen J T McCarty
- Department of Cell, Developmental, and Cancer Biology, Oregon Health & Science University, Portland, Oregon 97239, USA.,Division of Hematology and Medical Oncology and Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, Oregon 97239, USA
| | - Wilbur A Lam
- Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Children's Healthcare of Atlanta/Emory University School of Medicine, Atlanta, Georgia 30332, USA; .,Wallace H. Coulter Department of Biomedical Engineering and Institute for Electronics and Nanotechnology, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30332, USA
| |
Collapse
|
39
|
Gupta P, Zhang P, Sheriff J, Bluestein D, Deng Y. A Multiscale Model for Recruitment Aggregation of Platelets by Correlating with In Vitro Results. Cell Mol Bioeng 2019; 12:327-343. [PMID: 31662802 DOI: 10.1007/s12195-019-00583-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Introduction We developed a multiscale model to simulate the dynamics of platelet aggregation by recruitment of unactivated platelets flowing in viscous shear flows by an activated platelet deposited onto a blood vessel wall. This model uses coarse grained molecular dynamics (CGMD) for platelets at the microscale and dissipative particle dynamics (DPD) for the shear flow at the macroscale. Under conditions of relatively low shear, aggregation is mediated by fibrinogen via αIIbβ3 receptors. Methods The binding of αIIbβ3 and fibrinogen is modeled by a molecular-level hybrid force field consisting of Morse potential and Hooke law for the nonbonded and bonded interactions, respectively. The force field, parametrized in two different interaction scales, is calculated by correlating with the platelet contact area measured in vitro and the detaching force between αIIbβ3 and fibrinogen. Results Using our model, we derived, the relationship between recruitment force and distance between the centers of mass of two platelets, by integrating the molecular-scale inter-platelet interactions during recruitment aggregation in shear flows. Our model indicates that assuming a rigid-platelet model, underestimates the contact area by 89% and the detaching force by 93% as compared to a model that takes into account the platelet deformability leading to a prediction of a significantly lower attachment during recruitment. Conclusions The molecular-level predictive capability of our model sheds a light on differences observed between transient and permanent platelet aggregation patterns. The model and simulation framework can be further adapted to simulate initial thrombus formation involving multiple flowing platelets as well as deposition and adhesion onto blood vessels.
Collapse
Affiliation(s)
- Prachi Gupta
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, NY 11794-3600 USA
| | - Peng Zhang
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY 11794 USA
| | - Jawaad Sheriff
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY 11794 USA
| | - Danny Bluestein
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY 11794 USA
| | - Yuefan Deng
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, NY 11794-3600 USA
| |
Collapse
|
40
|
Unique transmembrane domain interactions differentially modulate integrin αvβ3 and αIIbβ3 function. Proc Natl Acad Sci U S A 2019; 116:12295-12300. [PMID: 31160446 DOI: 10.1073/pnas.1904867116] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Lateral transmembrane (TM) helix-helix interactions between single-span membrane proteins play an important role in the assembly and signaling of many cell-surface receptors. Often, these helices contain two highly conserved yet distinct interaction motifs, arranged such that the motifs cannot be engaged simultaneously. However, there is sparse experimental evidence that dual-engagement mechanisms play a role in biological signaling. Here, we investigate the function of the two conserved interaction motifs in the TM domain of the integrin β3-subunit. The first motif uses reciprocating "large-large-small" amino acid packing to mediate the interaction of the β3 and αIIb TM domains and maintain the inactive resting conformation of the platelet integrin αIIbβ3. The second motif, S-x3-A-x3-I, is a variant of the classical "G-x3-G" motif. Using site-directed mutagenesis, optical trap-based force spectroscopy, and molecular modeling, we show that S-x3-A-x3-I does not engage αIIb but rather mediates the interaction of the β3 TM domain with the TM domain of the αv-subunit of the integrin αvβ3. Like αIIbβ3, αvβ3 on circulating platelets is inactive, and in the absence of platelet stimulation is unable to interact with components of the subendothelial matrix. However, disrupting any residue in the β3 S-x3-A-x3-I motif by site-directed mutations is sufficient to induce αvβ3 binding to the αvβ3 ligand osteopontin and to the monoclonal antibody WOW-1. Thus, the β3-integrin TM domain is able to engage in two mutually exclusive interactions that produce alternate α-subunit pairing, creating two integrins with distinct biological functions.
Collapse
|
41
|
Lanza GM, Cui G, Schmieder AH, Zhang H, Allen JS, Scott MJ, Williams T, Yang X. An unmet clinical need: The history of thrombus imaging. J Nucl Cardiol 2019; 26:986-997. [PMID: 28608182 PMCID: PMC5741521 DOI: 10.1007/s12350-017-0942-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 05/24/2017] [Indexed: 11/24/2022]
Abstract
Robust thrombus imaging is an unresolved clinical unmet need dating back to the mid 1970s. While early molecular imaging approaches began with nuclear SPECT imaging, contrast agents for virtually all biomedical imaging modalities have been demonstrated in vivo with unique strengths and common weaknesses. Two primary molecular imaging targets have been pursued for thrombus imaging: platelets and fibrin. Some common issues noted over 40 years ago persist today. Acute thrombus is readily imaged with all probes and modalities, but aged thrombus remains a challenge. Similarly, anti-coagulation continues to interfere with and often negate thrombus imaging efficacy, but heparin is clinically required in patients suspected of pulmonary embolism, deep venous thrombosis or coronary ruptured plaque prior to confirmatory diagnostic studies have been executed and interpreted. These fundamental issues can be overcome, but an innovative departure from the prior approaches will be needed.
Collapse
Affiliation(s)
- Gregory M Lanza
- Department of Medicine, Division of Cardiology, Washington University Medical School, St. Louis, MO, 63108, USA.
| | - Grace Cui
- Department of Medicine, Division of Cardiology, Washington University Medical School, St. Louis, MO, 63108, USA
| | - Anne H Schmieder
- Department of Medicine, Division of Cardiology, Washington University Medical School, St. Louis, MO, 63108, USA
| | - Huiying Zhang
- Department of Medicine, Division of Cardiology, Washington University Medical School, St. Louis, MO, 63108, USA
| | - John S Allen
- Department of Medicine, Division of Cardiology, Washington University Medical School, St. Louis, MO, 63108, USA
| | - Michael J Scott
- Department of Medicine, Division of Cardiology, Washington University Medical School, St. Louis, MO, 63108, USA
| | - Todd Williams
- Department of Medicine, Division of Cardiology, Washington University Medical School, St. Louis, MO, 63108, USA
| | - Xiaoxia Yang
- Department of Medicine, Division of Cardiology, Washington University Medical School, St. Louis, MO, 63108, USA
| |
Collapse
|
42
|
Rahman SM, Hlady V. Downstream platelet adhesion and activation under highly elevated upstream shear forces. Acta Biomater 2019; 91:135-143. [PMID: 31004847 DOI: 10.1016/j.actbio.2019.04.028] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 04/04/2019] [Accepted: 04/11/2019] [Indexed: 12/17/2022]
Abstract
Elevated shear force caused by an anastomotic stenosis is a common complication at the blood vessel-vascular implant interface. Although elevated shear forces were found to cause platelet aggregation around a stenotic region, transient platelet exposure to elevated shear forces and subsequent downstream events occurring under lower shear force were not extensively studied. We hypothesize that effects of elevated shear forces on pre-activation of platelets for downstream adhesion and activation are relevant in understanding the increased thrombotic risk associated with blood-contacting devices. We designed a microfluidic flow system to mimic the hemodynamic environment of vasculature with an upstream anastomotic stenosis with five wall shear strain rates ranging from 1620 s-1 to 11560 s-1. Under shear flow conditions, transient exposure of whole blood to elevated shear forces resulted in higher downstream platelet adhesion onto three different immobilized platelet agonists: fibrinogen, collagen, or von Willebrand factor. Platelet expression of four activation markers (P-selectin, GPIIb/IIIa, lysosomal glycoprotein, and phosphatidylserine) significantly increased after transient exposure to higher upstream wall shear strain rates of 2975-11560 s-1. A significant lysis was observed when platelets were primed by upstream wall shear strain rate of 11560 s-1. These experimental results could be helpful to understand how altered hemodynamics around an anastomotic stenosis promotes thrombus formation downstream. STATEMENT OF SIGNIFICANCE: Studying the downstream response of platelets following transient exposure to an upstream agonist is important because of significant clinical implications to the implantation of vascular devices. Due to intimal fibrous hyperplasia, vascular biomaterials such as synthetic small-diameter vascular grafts sometimes become stenotic (narrow), leading to transient platelet exposure to elevated shear forces. In this study, a microfluidic flow system was developed to mimic a stenosed vascular graft and to investigate how highly elevated, transient upstream shear forces, typically found in severe stenosis, results in the pre-activation of platelets for downstream adhesion and activation. The findings of the present study have implications for optimizing the design of blood-contacting biomaterials in order to minimize thrombotic risk associated with transiently elevated shear forces. The findings also provide additional insights into the mechanisms of thrombus formation at the post-stenotic regions of vascular implants.
Collapse
|
43
|
Kaneva VN, Martyanov AA, Morozova DS, Panteleev MA, Sveshnikova AN. Platelet Integrin αIIbβ3: Mechanisms of Activation and Clustering; Involvement into the Formation of the Thrombus Heterogeneous Structure. BIOCHEMISTRY (MOSCOW), SUPPLEMENT SERIES A: MEMBRANE AND CELL BIOLOGY 2019. [DOI: 10.1134/s1990747819010033] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
44
|
Qiu J, Lingna W, Jinghong H, Yongqing Z. Oral administration of leeches (Shuizhi): A review of the mechanisms of action on antiplatelet aggregation. JOURNAL OF ETHNOPHARMACOLOGY 2019; 232:103-109. [PMID: 30543914 DOI: 10.1016/j.jep.2018.12.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 12/08/2018] [Accepted: 12/08/2018] [Indexed: 05/20/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The leeches (Shuizhi) comprise approximately 680 species distributed throughout the world. As recorded, they have been used as traditional Chinese medicines since the Eastern Han Dynasty, where they were claimed for promote blood circulation and eliminate blood stasis. And have been used to prevent CVDs by exerting multiple effects when orally administered, one of which is the significant inhibition of platelet aggregation. Its ability to exert this effect has been extensively investigated in vivo and in clinical practice. AIM OF STUDY The aim of this review is to summarize and analyse the antiplatelet aggregation mechanisms of leeches by oral administration, support their therapeutic potential and uncover opportunities for future research. MATERIALS AND METHODS Relevant studies from 1980 to 2018 on leeches and platelet aggregation were collected from ancient books, pharmacopoeia, reports and theses via library and internet databases (PubMed, CNKI, Google Scholar, Web of science, SciFinder, Springer and Elsevier). RESULTS Leeches is a unique animal medicine, they can prevent platelet aggregation by inhibiting ADP-induced platelet aggregation, increasing PGI2, decreasing TXA2 and Ca2+, and possibly recovering endothelial cell dysfunction. Leeches also exhibit a strong ability to activate eNOS, leading to an increase in platelet-derived NO. Additionally, the pteridine compounds obtained and identified from leeches have sulfur structure similar to those of other antiplatelet aggregation agents, such as ticlopidine, clopidogrel and ticagrelor. CONCLUSION The present review has focused on the related antiplatelet aggregation mechanisms, dipyridine compounds and toxicological information of leeches. According to the reported data, leeches have emerged as a good source of natural medicine for the treatment of antiplatelet aggregation agents and also make educated guesses for material basis of effects on antiplatelet aggregation. This review can help provide new insights for further studies in association with the development of effective antiplatelet aggregation drugs from natural medicines, especially leeches.
Collapse
Affiliation(s)
- Jiang Qiu
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China.
| | - Wang Lingna
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China.
| | - Hu Jinghong
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China.
| | - Zhang Yongqing
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China.
| |
Collapse
|
45
|
WEISEL JW, LITVINOV RI. Red blood cells: the forgotten player in hemostasis and thrombosis. J Thromb Haemost 2019; 17:271-282. [PMID: 30618125 PMCID: PMC6932746 DOI: 10.1111/jth.14360] [Citation(s) in RCA: 267] [Impact Index Per Article: 44.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Indexed: 12/14/2022]
Abstract
New evidence has stirred up a long-standing but undeservedly forgotten interest in the role of erythrocytes, or red blood cells (RBCs), in blood clotting and its disorders. This review summarizes the most recent research that describes the involvement of RBCs in hemostasis and thrombosis. There are both quantitative and qualitative changes in RBCs that affect bleeding and thrombosis, as well as interactions of RBCs with cellular and molecular components of the hemostatic system. The changes in RBCs that affect hemostasis and thrombosis include RBC counts or hematocrit (modulating blood rheology through viscosity) and qualitative changes, such as deformability, aggregation, expression of adhesive proteins and phosphatidylserine, release of extracellular microvesicles, and hemolysis. The pathogenic mechanisms implicated in thrombotic and hemorrhagic risk include variable adherence of RBCs to the vessel wall, which depends on the functional state of RBCs and/or endothelium, modulation of platelet reactivity and platelet margination, alterations of fibrin structure and reduced susceptibility to fibrinolysis, modulation of nitric oxide availability, and the levels of von Willebrand factor and factor VIII in blood related to the ABO blood group system. RBCs are involved in platelet-driven contraction of clots and thrombi that results in formation of a tightly packed array of polyhedral erythrocytes, or polyhedrocytes, which comprises a nearly impermeable barrier that is important for hemostasis and wound healing. The revisited notion of the importance of RBCs is largely based on clinical and experimental associations between RBCs and thrombosis or bleeding, implying that RBCs are a prospective therapeutic target in hemostatic and thrombotic disorders.
Collapse
Affiliation(s)
- J. W. WEISEL
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - R. I. LITVINOV
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russian Federation
| |
Collapse
|
46
|
Regulatory element in fibrin triggers tension-activated transition from catch to slip bonds. Proc Natl Acad Sci U S A 2018; 115:8575-8580. [PMID: 30087181 DOI: 10.1073/pnas.1802576115] [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] [Indexed: 12/12/2022] Open
Abstract
Fibrin formation and mechanical stability are essential in thrombosis and hemostasis. To reveal how mechanical load impacts fibrin, we carried out optical trap-based single-molecule forced unbinding experiments. The strength of noncovalent A:a knob-hole bond stabilizing fibrin polymers first increases with tensile force (catch bonds) and then decreases with force when the force exceeds a critical value (slip bonds). To provide the structural basis of catch-slip-bond behavior, we analyzed crystal structures and performed molecular modeling of A:a knob-hole complex. The movable flap (residues γ295 to γ305) containing the weak calcium-binding site γ2 serves as a tension sensor. Flap dissociation from the B domain in the γ-nodule and translocation to knob 'A' triggers hole 'a' closure, resulting in the increase of binding affinity and prolonged bond lifetimes. The discovery of biphasic kinetics of knob-hole bond rupture is quantitatively explained by using a theory, formulated in terms of structural transitions in the binding pocket between the low-affinity (slip) and high-affinity (catch) states. We provide a general framework to understand the mechanical response of protein pairs capable of tension-induced remodeling of their association interface. Strengthening of the A:a knob-hole bonds at 30- to 40-pN forces might favor formation of nascent fibrin clots subject to hydrodynamic shear in vivo.
Collapse
|
47
|
Nurden AT. Acquired Antibodies to αIIbβ3 in Glanzmann Thrombasthenia: From Transfusion and Pregnancy to Bone Marrow Transplants and Beyond. Transfus Med Rev 2018; 32:S0887-7963(18)30037-3. [PMID: 29884513 DOI: 10.1016/j.tmrv.2018.05.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Revised: 05/14/2018] [Accepted: 05/20/2018] [Indexed: 11/23/2022]
Abstract
Patients with the inherited bleeding disorder Glanzmann thrombasthenia (GT) possess platelets that lack αIIbβ3 integrin and fail to aggregate, and have moderate to severe mucocutaneous bleeding. Many become refractory to platelet transfusions due to the formation of isoantibodies to αIIbβ3 with the rapid elimination of donor platelets and/or a block of function. Epitope characterization has shown isoantibodies to be polyclonal and to recognize different epitopes on the integrin with β3 a major site and αvβ3 on endothelial and vascular cells a newly recognized target. Pregnancy in GT can also lead to isoantibody formation when fetal cells with β3 integrins pass into the circulation of a mother lacking them; a consequence is neonatal thrombocytopenia and a high risk of mortality. Antibody removal prior to donor transfusions can provide transient relief, but all evidence points to recombinant FVIIa as the first choice for GT patients either to stop bleeding or as prophylaxis. Promoting thrombin generation by rFVIIa favors GT platelet interaction with fibrin, and the risk of deep vein thrombosis also associated with prolonged immobilization and catheter use requires surveillance. Although having a high risk, allogeneic bone marrow transplantation associated with different stem cell sources and conditioning regimens has proved successful in many cases of severe GT with antibodies, and often, the associated conditioning and immunosuppressive therapy leads to loss of isoantibody production. Animal models of gene therapy for GT show promising results, but isoantibody production can be stimulated and CRISPR/Cas9 technology has yet to be applied. Up-to-date consensus protocols for dealing with isoantibodies in GT are urgently required, and networks providing patient care should be expanded.
Collapse
Affiliation(s)
- Alan T Nurden
- Institut de Rhythmologie et de Modélisation Cardiaque, Plateforme Technologique d'Innovation Biomédicale, Hôpital Xavier Arnozan, Pessac, France.
| |
Collapse
|
48
|
Platelet integrins exhibit anisotropic mechanosensing and harness piconewton forces to mediate platelet aggregation. Proc Natl Acad Sci U S A 2017; 115:325-330. [PMID: 29269394 DOI: 10.1073/pnas.1710828115] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Platelet aggregation at the site of vascular injury is essential in clotting. During this process, platelets are bridged by soluble fibrinogen that binds surface integrin receptors. One mystery in the mechanism of platelet aggregation pertains to how resting platelets ignore soluble fibrinogen, the third most abundant protein in the bloodstream, and yet avidly bind immobile fibrinogen on the surface of other platelets at the primary injury site. We speculate that platelet integrins are mechanosensors that test their ligands across the platelet-platelet synapse. To investigate this model, we interrogate human platelets using approaches that include the supported lipid bilayer platform as well as DNA tension sensor technologies. Experiments suggest that platelet integrins require lateral forces to mediate platelet-platelet interactions. Mechanically labile ligands dampen platelet activation, and the onset of piconewton integrin tension coincides with calcium flux. Activated platelets display immobilized fibrinogen on their surface, thus mediating further recruitment of resting platelets. The distribution of integrin tension was shown to be spatially regulated through two myosin-signaling pathways, myosin light chain kinase and Rho-associated kinase. Finally, we discovered that the termination of integrin tension is coupled with the exposure of phosphatidylserine. Our work reveals the highest spatial and temporal resolution maps of platelet integrin mechanics and its role in platelet aggregation, suggesting that platelets are physical substrates for one another that establish mechanical feedback loops of activation. The results are reminiscent of mechanical regulation of the T-cell receptor, E-cadherin, and Notch pathways, suggesting a common feature for signaling at cell junctions.
Collapse
|
49
|
Fibrin opens the "gate" for leukocytes in the endothelium. Thromb Res 2017; 162:101-103. [PMID: 29249275 DOI: 10.1016/j.thromres.2017.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 12/11/2017] [Indexed: 11/22/2022]
|
50
|
Procoagulant platelets: generation, function, and therapeutic targeting in thrombosis. Blood 2017; 130:2171-2179. [DOI: 10.1182/blood-2017-05-787259] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 09/12/2017] [Indexed: 11/20/2022] Open
Abstract
Abstract
Current understanding of how platelets localize coagulation to wound sites has come mainly from studies of a subpopulation of activated platelets. In this review, we summarize data from the last 4 decades that have described these platelets with a range of descriptive titles and attributes. We identify striking overlaps in the reported characteristics of these platelets, which imply a single subpopulation of versatile platelets and thus suggest that their commonality requires unification of their description. We therefore propose the term procoagulant platelet as the unifying terminology. We discuss the agonist requirements and molecular drivers for the dramatic morphological transformation platelets undergo when becoming procoagulant. Finally, we provide perspectives on the biomarker potential of procoagulant platelets for thrombotic events as well as on the possible clinical benefits of inhibitors of carbonic anhydrase enzymes and the water channel Aquaporin-1 for targeting this subpopulation of platelets as antiprocoagulant antithrombotics.
Collapse
|