1
|
Liu W, Li G, Shi J, Gao Y, Fang P, Zhao Y, Zhong F, Guo X, Lyu Y, Da X, Li Z, Fa J, Hu L, Yuan A, Chen L, Liu J, Chen AF, Sheng B, Ji Y, Lu X, Pu J. NR4A1 Acts as a Novel Regulator of Platelet Activation and Thrombus Formation. Circ Res 2025; 136:809-826. [PMID: 40035146 PMCID: PMC11984555 DOI: 10.1161/circresaha.124.325645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 02/16/2025] [Accepted: 02/19/2025] [Indexed: 03/05/2025]
Abstract
BACKGROUND Mounting evidence indicates that nuclear receptors play a critical regulatory role in platelet pathophysiology and thrombotic disorders. Although NR4A (the nuclear receptor subfamily 4 group A) plays an important role in cardiovascular pathophysiology, the expression profile and biological function of NR4A member 1 (NR4A1) in platelets have never been reported. METHODS We evaluated the functions and the underlying mechanisms of NR4A1 in platelet activation and thrombus formation using platelet-specific NR4A1-deficient mice and NR4A1-specific agonists. Using a hyperlipidemic mouse model and platelets from patients with hypercholesterolemia, we explored the influence of hypercholesterolemia on NR4A1 expression and the effects of NR4A1-specific agonists on platelet hyperreactivity induced by hypercholesterolemia. RESULTS NR4A1 was expressed in both human and mouse platelets. Platelet-specific NR4A1 deletion accelerated FeCl3-induced carotid arterial occlusive thrombus formation, enhanced collagen/epinephrine-induced pulmonary thromboembolism, and exacerbated microvascular microthrombi obstruction and infarct expansion in an acute myocardial infarction model. NR4A1-deficient platelets exhibited enhanced agonist-induced aggregation responses, integrin αIIbβ3 activation, dense granule release, α-granule release, platelet spreading, and clot retraction. Consistently, pharmacological activation of NR4A1 by specific agonists decreased platelet activation in both mouse and human platelets. Mechanistically, CAP1 (adenylyl cyclase-associated protein 1) was identified as the direct downstream interacting protein of NR4A1. NR4A1 deletion decreased cAMP levels and phosphorylation of VASP (vasodilator-stimulated phosphoprotein), while NR4A1-specific agonists increased cAMP levels and phosphorylation of VASP in platelets. Importantly, NR4A1 expression in platelets was upregulated in the setting of hypercholesterolemia, which was derived from its upregulation in megakaryocytes in a reactive oxygen species-dependent manner. Platelets from hypercholesterolemic patients and mice exhibited hyperreactivity. However, NR4A1-specific agonists significantly inhibited the activation of hypercholesterolemic platelets to the levels of healthy control platelets. CONCLUSIONS We provide the first evidence that nuclear receptor NR4A1 negatively regulates platelet activation and thrombus formation. NR4A1 may serve as a novel therapeutic target for managing thrombosis-based cardiovascular diseases, especially with hypercholesterolemia.
Collapse
MESH Headings
- Animals
- Nuclear Receptor Subfamily 4, Group A, Member 1/genetics
- Nuclear Receptor Subfamily 4, Group A, Member 1/deficiency
- Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism
- Nuclear Receptor Subfamily 4, Group A, Member 1/agonists
- Nuclear Receptor Subfamily 4, Group A, Member 1/blood
- Platelet Activation/physiology
- Humans
- Thrombosis/metabolism
- Thrombosis/blood
- Thrombosis/genetics
- Blood Platelets/metabolism
- Mice
- Mice, Knockout
- Mice, Inbred C57BL
- Male
- Hypercholesterolemia/blood
- Hypercholesterolemia/genetics
- Female
- Disease Models, Animal
Collapse
Affiliation(s)
- Wenhua Liu
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute (W.L., G.L., J.S., Y.G., P.F., Y.Z., F.Z., X.G., Y.L., X.D., Z.L., J.F., L.H., A.Y., L.C., J.L., X.L., J.P.), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Gaoxiang Li
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute (W.L., G.L., J.S., Y.G., P.F., Y.Z., F.Z., X.G., Y.L., X.D., Z.L., J.F., L.H., A.Y., L.C., J.L., X.L., J.P.), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Jianfeng Shi
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute (W.L., G.L., J.S., Y.G., P.F., Y.Z., F.Z., X.G., Y.L., X.D., Z.L., J.F., L.H., A.Y., L.C., J.L., X.L., J.P.), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Yu Gao
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute (W.L., G.L., J.S., Y.G., P.F., Y.Z., F.Z., X.G., Y.L., X.D., Z.L., J.F., L.H., A.Y., L.C., J.L., X.L., J.P.), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Peiliang Fang
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute (W.L., G.L., J.S., Y.G., P.F., Y.Z., F.Z., X.G., Y.L., X.D., Z.L., J.F., L.H., A.Y., L.C., J.L., X.L., J.P.), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Yichao Zhao
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute (W.L., G.L., J.S., Y.G., P.F., Y.Z., F.Z., X.G., Y.L., X.D., Z.L., J.F., L.H., A.Y., L.C., J.L., X.L., J.P.), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Fangyuan Zhong
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute (W.L., G.L., J.S., Y.G., P.F., Y.Z., F.Z., X.G., Y.L., X.D., Z.L., J.F., L.H., A.Y., L.C., J.L., X.L., J.P.), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Xiao Guo
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute (W.L., G.L., J.S., Y.G., P.F., Y.Z., F.Z., X.G., Y.L., X.D., Z.L., J.F., L.H., A.Y., L.C., J.L., X.L., J.P.), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Yuyan Lyu
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute (W.L., G.L., J.S., Y.G., P.F., Y.Z., F.Z., X.G., Y.L., X.D., Z.L., J.F., L.H., A.Y., L.C., J.L., X.L., J.P.), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Xingwen Da
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute (W.L., G.L., J.S., Y.G., P.F., Y.Z., F.Z., X.G., Y.L., X.D., Z.L., J.F., L.H., A.Y., L.C., J.L., X.L., J.P.), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Zhaoyan Li
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute (W.L., G.L., J.S., Y.G., P.F., Y.Z., F.Z., X.G., Y.L., X.D., Z.L., J.F., L.H., A.Y., L.C., J.L., X.L., J.P.), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Jingjing Fa
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute (W.L., G.L., J.S., Y.G., P.F., Y.Z., F.Z., X.G., Y.L., X.D., Z.L., J.F., L.H., A.Y., L.C., J.L., X.L., J.P.), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
- Baoshan Branch (J.F.), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Liuhua Hu
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute (W.L., G.L., J.S., Y.G., P.F., Y.Z., F.Z., X.G., Y.L., X.D., Z.L., J.F., L.H., A.Y., L.C., J.L., X.L., J.P.), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Ancai Yuan
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute (W.L., G.L., J.S., Y.G., P.F., Y.Z., F.Z., X.G., Y.L., X.D., Z.L., J.F., L.H., A.Y., L.C., J.L., X.L., J.P.), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Lei Chen
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute (W.L., G.L., J.S., Y.G., P.F., Y.Z., F.Z., X.G., Y.L., X.D., Z.L., J.F., L.H., A.Y., L.C., J.L., X.L., J.P.), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Junling Liu
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute (W.L., G.L., J.S., Y.G., P.F., Y.Z., F.Z., X.G., Y.L., X.D., Z.L., J.F., L.H., A.Y., L.C., J.L., X.L., J.P.), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
- Department of Biochemistry and Molecular Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education (J.L.), Shanghai Jiao Tong University School of Medicine, China
| | - Alex F. Chen
- Department of Cardiology and Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital (A.F.C.), Shanghai Jiao Tong University School of Medicine, China
| | - Bin Sheng
- Department of Computer Science and Engineering, Shanghai Jiao Tong University, China (B.S.)
| | - Yong Ji
- Key Laboratory of Drug Targets and Translational Medicine for Cardio-cerebrovascular Diseases, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Jiangsu, China(Y.J.)
- State Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Heilongjiang, China (Y.J.)
| | - Xiyuan Lu
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute (W.L., G.L., J.S., Y.G., P.F., Y.Z., F.Z., X.G., Y.L., X.D., Z.L., J.F., L.H., A.Y., L.C., J.L., X.L., J.P.), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Jun Pu
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute (W.L., G.L., J.S., Y.G., P.F., Y.Z., F.Z., X.G., Y.L., X.D., Z.L., J.F., L.H., A.Y., L.C., J.L., X.L., J.P.), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| |
Collapse
|
2
|
Jang J, Yu H, Oh EB, Park JW, Kim S, Kim T, Sohn J, Jin BR, Chang TS. Targeting NADPH Oxidase with APX-115: Suppression of Platelet Activation and Thrombotic Response. Antioxid Redox Signal 2025. [PMID: 40183134 DOI: 10.1089/ars.2024.0695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/05/2025]
Abstract
Aims: NADPH oxidase (NOX)-derived reactive oxygen species (ROS) are critical for platelet activation and thrombus formation. We hypothesized that inhibiting NOX-mediated ROS production with a pan-NOX inhibitor, APX-115, could effectively suppress platelet activation and thrombus formation, potentially serving as a novel antiplatelet therapeutic. This study aimed to explore the effects of APX-115 on human platelet functional responses and ROS-mediated signaling pathways. Results: APX-115 inhibited intracellular and extracellular ROS production in collagen-stimulated platelets, suppressing aggregation, P-selectin exposure, and ATP release. By preserving protein tyrosine phosphatase activity, APX-115 reduced tyrosine phosphorylation-dependent pathways inhibition, including spleen tyrosine kinase, LAT, Vav1, Bruton's tyrosine kinase, and phospholipase Cγ2, leading to decreased PKC activation and calcium mobilization. APX-115 also suppressed collagen-induced integrin αIIbβ3 activation, accompanied by elevated cGMP and vasodilator-stimulated phosphoprotein phosphorylation levels. In addition, APX-115 reduced p38 MAPK and ERK5 activation, leading to diminished phospholipase A2 phosphorylation, thromboxane production, and the exposure of procoagulant phosphatidylserine. These inhibitory effects extended to thrombus development caused by platelet adherence under shear and arterial thrombosis without prolonging bleeding time in murine models. Innovation: This study is the first to demonstrate that APX-115 inhibits NOX-mediated ROS production, platelet activation, and thrombus formation. By uncovering its effects on collagen receptor glycoprotein VI-mediated pathways, the work highlights the promise of APX-115 as an antiplatelet and antithrombotic agent. Conclusion: Our findings highlight the therapeutic potential of APX-115 in treating thrombotic and cardiovascular disorders by targeting NOX-mediated ROS production to mitigate platelet hyperreactivity and thrombus formation. Antioxid. Redox Signal. 00, 000-000. [Figure: see text].
Collapse
Affiliation(s)
- Joara Jang
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Hyunseong Yu
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Eun Bee Oh
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Ji Won Park
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Solee Kim
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Taeryeong Kim
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Jisue Sohn
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Bo-Ram Jin
- Division of Hematology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Tong-Shin Chang
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea
- Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
3
|
Gao X, Ma D, Mi L, Zhao J, An Q, Guo Z, Yang B, Zhang L, Xu K. Progress in the field of animal models of antiphospholipid syndrome. Autoimmunity 2024; 57:2391350. [PMID: 39155523 DOI: 10.1080/08916934.2024.2391350] [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: 01/29/2024] [Revised: 04/14/2024] [Accepted: 07/29/2024] [Indexed: 08/20/2024]
Abstract
Antiphospholipid syndrome (APS) is an autoimmune disease characterized by recurrent arteriovenous thrombosis and pathological pregnancy, accompanied by persistent antiphospholipid antibodies, (aPL). The incidence of APS is increasing year by year, clinicians lack of understanding of this type of disease, easy to misdiagnose and miss the diagnosis. Therefore, it is extremely important to establish a suitable animal model to reduce the process of disease development as much as possible and improve clinicians' understanding and understanding. This review will summarize the animal models of APS from the aspects of modeling methods, modeling mechanism, evaluation indicators and advantages and disadvantages of methods, providing a reference for finding an animal model highly similar to human APS, helping researchers to further clarify the pathogenesis of APS and find potential therapeutic targets, so as to achieve early diagnosis, early intervention, and ultimately improve the prognosis of patients.
Collapse
Affiliation(s)
- Xinnan Gao
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences,Tongji Shanxi Hospital, Taiyuan, China
| | - Dan Ma
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
| | - Liangyu Mi
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences,Tongji Shanxi Hospital, Taiyuan, China
| | - Jingwen Zhao
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences,Tongji Shanxi Hospital, Taiyuan, China
| | - Qi An
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences,Tongji Shanxi Hospital, Taiyuan, China
| | - Zhiying Guo
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences,Tongji Shanxi Hospital, Taiyuan, China
| | - Baoqi Yang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences,Tongji Shanxi Hospital, Taiyuan, China
| | - Liyun Zhang
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
| | - Ke Xu
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences,Tongji Shanxi Hospital, Taiyuan, China
| |
Collapse
|
4
|
Li C, Malloy M, Ture SK, Nieves-Lopez B, Thibord F, Johnson AD, Morrell CN. G protein-coupled receptor kinase 5 regulates thrombin signaling in platelets. Res Pract Thromb Haemost 2024; 8:102556. [PMID: 39309233 PMCID: PMC11415800 DOI: 10.1016/j.rpth.2024.102556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 06/28/2024] [Accepted: 08/16/2024] [Indexed: 09/25/2024] Open
Abstract
Background Our prior genome-wide association study of thrombin-induced platelet aggregation identified a G protein-coupled receptor kinase 5 (GRK5) noncoding variant (rs10886430-G) that is strongly associated with increased platelet reactivity to thrombin. This variant predisposes to increased risk of stroke, pulmonary embolism, and venous thromboembolism. Objectives To determine role of platelet specific GRK5 in platelet responses to agonists and injury. Methods Platelets from GRK5 mutant mice have been shown to have increased thrombin sensitivity, indicating that GRK5 may be a negative regulator of platelet activation. However, this has not been studied in a platelet-specific manner. We therefore used platelet-specific GRK5 mutant mice and models of thrombosis and pulmonary embolism. Results We now demonstrate that mice lacking GRK5 specifically in platelets had a mild increase in thrombin responses in vitro and a shortened time to arterial thrombosis in vivo. In addition, platelet GRK5 mutant mice had increased thrombin but not collagen-induced thrombus burden in a mouse model of pulmonary embolism. Conclusion These data indicate that platelet GRK5 has a significant role in limiting platelet responses to thrombin.
Collapse
Affiliation(s)
- Chen Li
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - Michael Malloy
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - Sara K. Ture
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - Benjamin Nieves-Lopez
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
- University of Puerto Rico, Medical Sciences Campus, San Juan, Puerto Rico
| | - Florian Thibord
- Population Sciences Branch, National Heart, Lung and Blood Institute, Framingham, Massachusetts, USA
| | - Andrew D. Johnson
- Population Sciences Branch, National Heart, Lung and Blood Institute, Framingham, Massachusetts, USA
| | - Craig N. Morrell
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
- Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
- Department of Pathology and Laboratory Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| |
Collapse
|
5
|
Oliveira SNMCG, Bezerra FF, Piquet AA, Sales RA, Valle GCT, Capillé NV, Tovar AMF, Mourão PAS. A Unique Enoxaparin Derived from Bovine Intestinal Heparin: A Single Purification Step of the Starting Material Assures a Bovine Enoxaparin Like the Standard from Porcine Origin. ACS OMEGA 2024; 9:23111-23120. [PMID: 38826523 PMCID: PMC11137703 DOI: 10.1021/acsomega.4c02128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 04/05/2024] [Accepted: 04/09/2024] [Indexed: 06/04/2024]
Abstract
Low-molecular-weight heparin represent a significant advancement in anticoagulant therapy with enoxaparin being a prominent example obtained exclusively through the fragmentation of porcine intestinal heparin. However, escalating demand and limited resources have raised concerns about enoxaparin supplementation. The current challenge involves exploring alternative heparin sources for large-scale enoxaparin production with bovine intestinal heparin emerging as a promising option. Our study demonstrates that enoxaparin derived from the available bovine heparin preparation differs significantly from the reference compound. Yet, the implementation of a straightforward purification step yields a preparation termed "high-anticoagulant bovine heparin". Fragmentation of this purified product through β-elimination produces enoxaparin akin to the standard from a porcine origin. To ensure physicochemical similarity, we employed various spectroscopic, enzymatic, and chromatographic tests to compare the new bovine-derived enoxaparin with the original porcine compound. Biological activity was confirmed through in vitro coagulation assays and assessments using an animal model of venous thrombosis. Our study affirms that the β-elimination reaction cleaves the bovine heparin chain without preferential breaks in regions with different sulfation patterns. Additionally, we scrutinized decasaccharides purified from enoxaparin preparations, providing a comprehensive demonstration of the similarity between products obtained from porcine and bovine heparin. In summary, our findings indicate that an enoxaparin equivalent to the original porcine-derived product can be derived from bovine heparin, given that the starting material undergoes a simple purification step.
Collapse
Affiliation(s)
| | | | - Adriana A. Piquet
- Laboratório de Tecido
Conjuntivo, Hospital Universitário Clementino Fraga Filho and
Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-913, Brazil
| | - Rodrigo A. Sales
- Laboratório de Tecido
Conjuntivo, Hospital Universitário Clementino Fraga Filho and
Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-913, Brazil
| | - Gabrielly C. T. Valle
- Laboratório de Tecido
Conjuntivo, Hospital Universitário Clementino Fraga Filho and
Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-913, Brazil
| | - Nina V. Capillé
- Laboratório de Tecido
Conjuntivo, Hospital Universitário Clementino Fraga Filho and
Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-913, Brazil
| | - Ana M. F. Tovar
- Laboratório de Tecido
Conjuntivo, Hospital Universitário Clementino Fraga Filho and
Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-913, Brazil
| | - Paulo A. S. Mourão
- Laboratório de Tecido
Conjuntivo, Hospital Universitário Clementino Fraga Filho and
Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-913, Brazil
| |
Collapse
|
6
|
Ye Y, Yang L, Leng M, Wang Q, Wu J, Wan W, Wang H, Li L, Peng Y, Chai S, Meng Z. Luteolin inhibits GPVI-mediated platelet activation, oxidative stress, and thrombosis. Front Pharmacol 2023; 14:1255069. [PMID: 38026984 PMCID: PMC10644720 DOI: 10.3389/fphar.2023.1255069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Accepted: 10/02/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction: Luteolin inhibits platelet activation and thrombus formation, but the mechanisms are unclear. This study investigated the effects of luteolin on GPVI-mediated platelet activation in vitro and explored the effect of luteolin on thrombosis, coagulation, and platelet production in vivo. Methods: Washed human platelets were used for aggregation, membrane protein expression, ATP, Ca2+, and LDH release, platelet adhesion/spreading, and clot retraction experiments. Washed human platelets were used to detect collagen and convulxin-induced reactive oxygen species production and endogenous antioxidant effects. C57BL/6 male mice were used for ferric chloride-induced mesenteric thrombosis, collagen-epinephrine induced acute pulmonary embolism, tail bleeding, coagulation function, and luteolin toxicity experiments. The interaction between luteolin and GPVI was analyzed using solid phase binding assay and surface plasmon resonance (SPR). Results: Luteolin inhibited collagen- and convulxin-mediated platelet aggregation, adhesion, and release. Luteolin inhibited collagen- and convulxin-induced platelet ROS production and increased platelet endogenous antioxidant capacity. Luteolin reduced convulxin-induced activation of ITAM and MAPK signaling molecules. Molecular docking simulation showed that luteolin forms hydrogen bonds with GPVI. The solid phase binding assay showed that luteolin inhibited the interaction between collagen and GPVI. Surface plasmon resonance showed that luteolin bonded GPVI. Luteolin inhibited integrin αIIbβ3-mediated platelet activation. Luteolin inhibited mesenteric artery thrombosis and collagen- adrenergic-induced pulmonary thrombosis in mice. Luteolin decreased oxidative stress in vivo. Luteolin did not affect coagulation, hemostasis, or platelet production in mice. Discussion: Luteolin may be an effective and safe antiplatelet agent target for GPVI. A new mechanism (decreased oxidative stress) for the anti-platelet activity of luteolin has been identified.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Zhaohui Meng
- Laboratory of Molecular Cardiology, Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
7
|
Zhang S, Li J, Ren J, Xue Z, Qi X, Si Q. Cyclic RGD functionalized PLGA nanoparticles loaded with noncovalent complex of indocyanine green with urokinase for synergistic thrombolysis. Front Bioeng Biotechnol 2022; 10:945531. [PMID: 36032719 PMCID: PMC9399888 DOI: 10.3389/fbioe.2022.945531] [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: 05/18/2022] [Accepted: 07/04/2022] [Indexed: 11/16/2022] Open
Abstract
Thrombotic diseases have the characteristics of long latency period, rapid onset, and high mortality rate, which seriously threaten people's life and health. The aim of this research is to fabricate a novel indocyanine green complex of urokinase (ICG@uPA) and employ the amphiphilic PEG-PLGA polymer to deliver the complex as an enzyme-phototherapeutic synergistic thrombolysis platform. The noncovalent indocyanine green (ICG) complex of urokinase (ICG@uPA) was prepared via supramolecular self-assembly and then encapsulated into cRGD decorated polymeric nanoparticles (cRGD-ICG-uPA NPs) by double-emulsion solvent evaporation method. Then the nanoparticles (NPs) were characterized in terms of particle size, optical properties, in vitro release, etc. The targeting and thrombolytic effect of the nanoparticles were studied both in vitro and in vivo. ICG@uPA and cRGD-ICG-uPA NPs displayed significantly higher photostability and laser energy conversion efficiency than free ICG. Concomitantly, the NPs exhibited selective binding affinity to the activated platelets and specific accumulation in the mouse mesenteric vessel thrombus. Significant thrombolysis was achieved in vivo by photo-assisted synergistic therapy with reduced dose and systemic bleeding risk of uPA. Our results prove that the functional PLGA nanoparticle loaded with the ICG@uPA offers a novel option for effective and safe thrombolytic treatment.
Collapse
Affiliation(s)
- Sha Zhang
- Department of Geriatric Cardiology, Second Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Geriatric Diseases, Beijing, China
- Medical School of Chinese PLA, Beijing, China
| | - Jinjie Li
- Centre of Sport Nutrition and Health, Zhengzhou University, Zhengzhou, China
| | - Jiefeng Ren
- Department of Geriatric Cardiology, Second Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Geriatric Diseases, Beijing, China
- Medical School of Chinese PLA, Beijing, China
| | - Zaiyao Xue
- Department of Geriatric Cardiology, Second Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Geriatric Diseases, Beijing, China
- Medical School of Chinese PLA, Beijing, China
| | - Xinlian Qi
- Department of Geriatric Cardiology, Second Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Geriatric Diseases, Beijing, China
- Medical School of Chinese PLA, Beijing, China
| | - Quanjin Si
- Department of the Third Health Care, Second Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Geriatric Diseases, Beijing, China
| |
Collapse
|
8
|
Nappi F, Bellomo F, Avtaar Singh SS. Insights into the Role of Neutrophils and Neutrophil Extracellular Traps in Causing Cardiovascular Complications in Patients with COVID-19: A Systematic Review. Biomedicines 2022; 11:2460. [PMID: 35566589 PMCID: PMC9855935 DOI: 10.3390/biomedicines11010113] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 04/24/2022] [Accepted: 04/25/2022] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND The coronavirus disease 2019 (COVID-19) pandemic caused by the SARS-CoV-2 virus has resulted in significant mortality and burdening of healthcare resources. While initially noted as a pulmonary pathology, subsequent studies later identified cardiovascular involvement with high mortalities reported in specific cohorts of patients. While cardiovascular comorbidities were identified early on, the exact manifestation and etiopathology of the infection remained elusive. This systematic review aims to investigate the role of inflammatory pathways, highlighting several culprits including neutrophil extracellular traps (NETs) which have since been extensively investigated. METHOD A search was conducted using three databases (MEDLINE; MEDLINE In-Process & Other Non-Indexed Citations and EMBASE). Data from randomized controlled trials (RCT), prospective series, meta-analyses, and unmatched observational studies were considered for the processing of the algorithm and treatment of inflammatory response during SARS-CoV-2 infection. Studies without the SARS-CoV-2 Infection period and case reports were excluded. RESULTS A total of 47 studies were included in this study. The role of the acute inflammatory response in the propagation of the systemic inflammatory sequelae of the disease plays a major part in determining outcomes. Some of the mechanisms of activation of these pathways have been highlighted in previous studies and are highlighted. CONCLUSION NETs play a pivotal role in the pathogenesis of the inflammatory response. Despite moving into the endemic phase of the disease in most countries, COVID-19 remains an entity that has not been fully understood with long-term effects remaining uncertain and requiring ongoing monitoring and research.
Collapse
Affiliation(s)
- Francesco Nappi
- Department of Cardiac Surgery, Centre Cardiologique du Nord of Saint-Denis, 93200 Saint-Denis, France
| | - Francesca Bellomo
- Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy
| | | |
Collapse
|
9
|
Lin X, Zhao P, Lin Z, Chen J, Bingwa LA, Siaw-Debrah F, Zhang P, Jin K, Yang S, Zhuge Q. Establishment of a Modified and Standardized Ferric Chloride-Induced Rat Carotid Artery Thrombosis Model. ACS OMEGA 2022; 7:8919-8927. [PMID: 35309441 PMCID: PMC8928333 DOI: 10.1021/acsomega.1c07316] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 02/21/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND Ferric chloride is widely utilized in inducing thrombosis in small vessels of experimental animals. However, the lack of its application in large blood vessels of experimental animals and inconsistent concentration has limited its application. Therefore, we systematically tested the most suitable concentration and reliable induction time in the experiment of using ferric chloride to induce rat carotid artery thrombosis. METHODS In this study, we selected the common carotid artery of 59 Sprague-Dawley rats as the target vessel. The exploration process was divided into three stages. First, to determine the optimum induction concentration, we compared the effects of 30-60% ferric chloride on thrombus formation within 24 h. Second, to confirm the handling time, we tested different induction times from 3 min to 10 min. Lastly, we used the thrombolytic drug rt-PA to detect whether the formed thrombus can be lysed. Doppler blood flow imaging and H-E staining were employed to estimate the blood flow and thrombus. The ATP levels in the brain were measured using a bioluminescence ATP assay kit. RESULTS We found that the application of 50% ferric chloride for 10 min was enough to successfully induce thrombosis in the rat carotid artery and without spontaneous thrombolysis after 24 h. It is better than other concentrations and will lead to the decline of the ATP content in the ischemic hemisphere. CONCLUSIONS Our results indicate that the rat carotid artery thrombosis model induced by 50% ferric chloride for 10 min is stable and reliable.
Collapse
Affiliation(s)
- Xiao Lin
- Department
of Neurosurgery, The First Affiliated Hospital
of Wenzhou Medical University, Wenzhou 325000 China
- Zhejiang
Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000 China
| | - Peiqi Zhao
- Department
of Neurosurgery, The First Affiliated Hospital
of Wenzhou Medical University, Wenzhou 325000 China
- Zhejiang
Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000 China
| | - Zhongxiao Lin
- Department
of Neurosurgery, The First Affiliated Hospital
of Wenzhou Medical University, Wenzhou 325000 China
- Zhejiang
Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000 China
| | - Jiayu Chen
- Department
of Neurosurgery, The First Affiliated Hospital
of Wenzhou Medical University, Wenzhou 325000 China
- Zhejiang
Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000 China
| | - Lebohang Anesu Bingwa
- Department
of Neurosurgery, The First Affiliated Hospital
of Wenzhou Medical University, Wenzhou 325000 China
| | - Felix Siaw-Debrah
- Department
of Neurosurgery, Korlebu Teaching Hospital, Korlebu, Ghana 00233, West Africa
| | - Peng Zhang
- Department
of Neurosurgery, The First Affiliated Hospital
of Wenzhou Medical University, Wenzhou 325000 China
- Zhejiang
Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000 China
| | - Kunlin Jin
- Department
of Pharmacology and Neuroscience, University
of North Texas Health Science Center, Fort Worth, Texas 76107, United States
| | - Su Yang
- Department
of Neurosurgery, The First Affiliated Hospital
of Wenzhou Medical University, Wenzhou 325000 China
- Zhejiang
Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000 China
| | - Qichuan Zhuge
- Department
of Neurosurgery, The First Affiliated Hospital
of Wenzhou Medical University, Wenzhou 325000 China
- Zhejiang
Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000 China
| |
Collapse
|
10
|
Shim Y, Kwon I, Park Y, Lee HW, Kim J, Kim YD, Nam HS, Park S, Heo JH. Characterization of Ferric Chloride-Induced Arterial Thrombosis Model of Mice and the Role of Red Blood Cells in Thrombosis Acceleration. Yonsei Med J 2021; 62:1032-1041. [PMID: 34672137 PMCID: PMC8542466 DOI: 10.3349/ymj.2021.62.11.1032] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 08/06/2021] [Accepted: 08/19/2021] [Indexed: 11/27/2022] Open
Abstract
PURPOSE The ferric chloride (FeCl3)-induced thrombosis model is widely used for thrombosis research. However, it lacks standardization with uncertainty in the exact mechanism of thrombosis. This study aimed to characterize thrombus formation in a mouse model. MATERIALS AND METHODS We investigated thrombus formation and stability using various FeCl3 concentrations (10%, 20%, 30%, 40%, and 50%, w/v) in carotid arteries of the Institute of Cancer Research (ICR) and C57BL/6N mice using the FeCl3-induced thrombosis model. We also investigated thrombus histopathology using immunohistochemistry and electron microscopy. RESULTS Higher FeCl3 concentrations induced dose-dependent, faster, larger, and more stable thrombus formation in both strains of mice. However, the ICR mice showed better dose-responses in thrombus formation and stability compared to the C57BL/6N mice. Thrombi were fibrin- and platelet-rich without significant changes across FeCl3 concentrations. However, the content of red blood cells (RBCs) increased with increasing FeCl3 concentrations (p for trend <0.001) and inversely correlated with time to occlusion (r=-0.65, p<0.001). While platelets and fibrin were evenly distributed over the thrombus, RBCs were predominantly located near the FeCl3 treatment area. Transmission electron microscopy showed that RBCs attached to and were surrounded by aggregates of degranulated platelets, suggesting their potential role in platelet activation. CONCLUSION Faster and larger thrombus formation is induced in a dose-dependent manner by a wide range of FeCl3 concentrations, but the stable thrombus formation requires higher FeCl3 concentrations. Mouse strain affects thrombus formation and stability. RBCs and their interaction with platelets play a key role in the acceleration of FeCl3-induced thrombosis.
Collapse
Affiliation(s)
- Yeseul Shim
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea
- Integrative Research Institute for Cerebrovascular and Cardiovascular Diseases, Yonsei University College of Medicine, Seoul, Korea
- Department of Neurology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea
| | - Il Kwon
- Integrative Research Institute for Cerebrovascular and Cardiovascular Diseases, Yonsei University College of Medicine, Seoul, Korea
| | - Youngseon Park
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea
- Integrative Research Institute for Cerebrovascular and Cardiovascular Diseases, Yonsei University College of Medicine, Seoul, Korea
- Department of Neurology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea
| | - Heow Won Lee
- Integrative Research Institute for Cerebrovascular and Cardiovascular Diseases, Yonsei University College of Medicine, Seoul, Korea
| | - Jayoung Kim
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea
| | - Young Dae Kim
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea
- Integrative Research Institute for Cerebrovascular and Cardiovascular Diseases, Yonsei University College of Medicine, Seoul, Korea
| | - Hyo Suk Nam
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea
- Integrative Research Institute for Cerebrovascular and Cardiovascular Diseases, Yonsei University College of Medicine, Seoul, Korea
| | - Sungha Park
- Integrative Research Institute for Cerebrovascular and Cardiovascular Diseases, Yonsei University College of Medicine, Seoul, Korea
- Division of Cardiology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Ji Hoe Heo
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea
- Integrative Research Institute for Cerebrovascular and Cardiovascular Diseases, Yonsei University College of Medicine, Seoul, Korea
- Department of Neurology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
11
|
Jia Q, Li Z, Bai M, Yan H, Zhang R, Ji Y, Feng Y, Yang Z, Wang Z, Li J. Estimating dynamic vascular perfusion based on Er-based lanthanide nanoprobes with enhanced down-conversion emission beyond 1500 nm. Theranostics 2021; 11:9859-9872. [PMID: 34815791 PMCID: PMC8581431 DOI: 10.7150/thno.65771] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 09/22/2021] [Indexed: 12/24/2022] Open
Abstract
Peripheral artery disease (PAD) is a common, yet serious, circulatory condition that can increase the risk of amputation, heart attack or stroke. Accurate identification of PAD and dynamic monitoring of the treatment efficacy of PAD in real time are crucial for optimizing therapeutic outcomes. However, current imaging techniques do not enable these requirements. Methods: A lanthanide-based nanoprobe with emission in the second near-infrared window b (NIR-IIb, 1500-1700 nm), Er-DCNPs, was utilized for continuous imaging of dynamic vascular structures and hemodynamic alterations in real time using PAD-related mouse models. The NIR-IIb imaging capability, stability, and biocompatibility of Er-DCNPs were evaluated in vitro and in vivo. Results: Owing to their high temporal-spatial resolution in the NIR-IIb imaging window, Er-DCNPs not only exhibited superior capability in visualizing anatomical and pathophysiological features of the vasculature of mice but also provided dynamic information on blood perfusion for quantitative assessment of blood recovery, thereby achieving the synergistic integration of diagnostic and therapeutic imaging functions, which is very meaningful for the successful management of PAD. Conclusion: Our findings indicate that Er-DCNPs can serve as a promising system to facilitate the diagnosis and treatment of PAD as well as other vasculature-related diseases.
Collapse
Affiliation(s)
- Qian Jia
- Engineering Research Center of Molecular and Neuro-imaging of ministry of education, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710126 China
| | - Zheng Li
- Engineering Research Center of Molecular and Neuro-imaging of ministry of education, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710126 China
| | - Mingli Bai
- Engineering Research Center of Molecular and Neuro-imaging of ministry of education, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710126 China
| | - Haohao Yan
- Engineering Research Center of Molecular and Neuro-imaging of ministry of education, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710126 China
| | - Ruili Zhang
- Engineering Research Center of Molecular and Neuro-imaging of ministry of education, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710126 China
- Academy of Advanced Interdisciplinary Research, Xidian University, Xi'an, Shaanxi, 710071, China
| | - Yu Ji
- Engineering Research Center of Molecular and Neuro-imaging of ministry of education, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710126 China
| | - Yanbin Feng
- Engineering Research Center of Molecular and Neuro-imaging of ministry of education, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710126 China
| | - Zuo Yang
- Engineering Research Center of Molecular and Neuro-imaging of ministry of education, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710126 China
| | - Zhongliang Wang
- Engineering Research Center of Molecular and Neuro-imaging of ministry of education, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710126 China
- Academy of Advanced Interdisciplinary Research, Xidian University, Xi'an, Shaanxi, 710071, China
| | - Jianxiong Li
- Department of Radiotherapy, Chinese PLA General Hospital, Beijing, 100071, China
| |
Collapse
|
12
|
Effect of Yiqi Huoxue Granules on Platelet Activation Induced by Thrombin. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:6622848. [PMID: 34335832 PMCID: PMC8313338 DOI: 10.1155/2021/6622848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 07/10/2021] [Indexed: 11/18/2022]
Abstract
Objective To study the effects of Yiqi Huoxue (YQHX) granules on platelet activation and aggregation induced by thrombin. Methods The effect of YQHX on platelet aggregation rate was detected by platelet aggregation instrument; the effect of YQHX on thrombosis time was observed by the mouse mesentery thrombosis model. DAMI cells were induced to transform into platelet-like granules using PMA, and the effects of SCH (PAR-1 inhibitor) on thrombin-induced changes in platelet intracellular calcium concentration, PAR-1 protein expression, and phosphorylation of MAPK were examined. Results Compared with the control group, the platelet aggregation rate, PAR-1 protein expression, phosphorylation of ERK1/2, and p38 protein in the YQHX group decreased (P < 0.05), and there was no significant difference between the YQHX + SCH group and YQHX group (P > 0.05). Conclusion YQHX suppresses the platelet activation induced by thrombin by inhibiting PAR-1 expression.
Collapse
|
13
|
Understanding the Pathophysiology of Thrombotic APS through Animal Models. Int J Mol Sci 2021; 22:ijms22052588. [PMID: 33806694 PMCID: PMC7961365 DOI: 10.3390/ijms22052588] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 03/01/2021] [Accepted: 03/01/2021] [Indexed: 12/12/2022] Open
Abstract
Antiphospholipid syndrome (APS) is a leading acquired cause of thrombotic events, with a notable tendency to promote thrombosis in vascular beds of all sizes, including both arterial and venous circuits. While pathogenic antiphospholipid antibodies circulate at relatively stable levels in blood, thrombosis tends to manifest as discrete and acute events, suggesting the requirement for a “second hit.” While this two-hit model is generally accepted, much remains to be learned about exactly how antiphospholipid antibodies predispose to thrombosis in vivo and exactly how this predisposition interacts with the second hit. To this end, investigators have turned to animal models. Numerous approaches for modeling APS in animals have been described to date, each with potential advantages and disadvantages. This review will attempt to describe the most common APS models employed so far while discussing some pros and cons of each. Mechanisms of thrombotic APS that have thus far been explored in animal models will also be briefly addressed.
Collapse
|
14
|
Jimenez K, Leitner F, Leitner A, Scharbert G, Schwabl P, Kramer AM, Krnjic A, Friske J, Helbich T, Evstatiev R, Khare V, Gasche C. Iron deficiency-induced thrombocytosis increases thrombotic tendency in rats. Haematologica 2021; 106:782-794. [PMID: 32079699 PMCID: PMC7928018 DOI: 10.3324/haematol.2019.245092] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Indexed: 12/19/2022] Open
Abstract
Iron deficiency (ID) is globally prevalent, and apart from anemia is associated with thrombocytosis. While considered benign, studies linking thrombotic events with prior ID anemia suggest otherwise. In this study we used animal models to assess the influence of ID on thrombotic tendency. Sprague-Dawley rats were fed control or iron-deficient diets and ferric carboxymaltose was used to reverse ID. Thrombosis was induced by stenosis of the inferior vena cava or damage to the right carotid artery using ferric chloride. Thrombi were evaluated histologically and by high frequency ultrasound in the venous model. ID consistently induced thrombocytosis alongside anemia. The growth of venous thrombi and the final dimensions of both arterial and venous thrombi were greater in animals with ID. In both models, platelet numbers correlated with the final thrombus size, with thrombi in iron-deficient animals having the largest platelet areas. Platelet function was also evaluated in surgically-naïve rats. Coagulability, determined by thromboelasto - graphy, and hemostasis, evaluated by tail transection, were enhanced in the animals with ID. Platelet P-selectin expression and plasma P-selectin levels were both higher in animals with ID. Platelet adhesion and aggregation in ID was impaired under shear flow but was intact in static assays. Iron replacement therapy reversed all ID-related changes in hematologic parameters, thrombus dimensions, and platelet assays. In summary, ID alone increases thrombotic tendency. Iron replacement therapy reverses these changes, making it a viable strategy for the prevention of ID-related thrombotic disease. This may be of importance in patients with chronic illnesses who may already be at increased risk of thrombosis, such as those with inflammatory bowel disease, chronic kidney disease, or cancer.
Collapse
Affiliation(s)
- Kristine Jimenez
- Div. of Gastroenterology and Hepatology,Dept. of Internal Medicine III, Medical University of Vienna
| | - Florentina Leitner
- Div. of Gastroenterology and Hepatology,Dept. of Internal Medicine III, Medical University of Vienna
| | - Aran Leitner
- Div. of Gastroenterology and Hepatology,Dept. of Internal Medicine III, Medical University of Vienna
| | - Gisela Scharbert
- Department of Special Anaesthesiology and Pain Management, General Intensive Care and Pain Control
| | - Philipp Schwabl
- Div. of Gastroenterology and Hepatology,Dept. of Internal Medicine III, Medical University of Vienna
| | | | - Anita Krnjic
- Div. of Gastroenterology and Hepatology,Dept. of Internal Medicine III, Medical University of Vienna
| | - Joachim Friske
- Department of Biomedical Imaging and Image-guided Therapy, Division of Gender and Molecular Imaging
| | - Thomas Helbich
- Department of Biomedical Imaging and Image-guided Therapy, Division of Gender and Molecular Imaging
| | - Rayko Evstatiev
- Div. of Gastroenterology and Hepatology,Dept. of Internal Medicine III, Medical University of Vienna
| | - Vineeta Khare
- Div. of Gastroenterology and Hepatology,Dept. of Internal Medicine III, Medical University of Vienna
| | - Christoph Gasche
- Div. of Gastroenterology and Hepatology,Dept. of Internal Medicine III, Medical University of Vienna
| |
Collapse
|
15
|
Filling the gaps on stroke research: Focus on inflammation and immunity. Brain Behav Immun 2021; 91:649-667. [PMID: 33017613 PMCID: PMC7531595 DOI: 10.1016/j.bbi.2020.09.025] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 09/10/2020] [Accepted: 09/23/2020] [Indexed: 02/08/2023] Open
Abstract
For the last two decades, researchers have placed hopes in a new era in which a combination of reperfusion and neuroprotection would revolutionize the treatment of stroke. Nevertheless, despite the thousands of papers available in the literature showing positive results in preclinical stroke models, randomized clinical trials have failed to show efficacy. It seems clear now that the existing data obtained in preclinical research have depicted an incomplete picture of stroke pathophysiology. In order to ameliorate bench-to-bed translation, in this review we first describe the main actors on stroke inflammatory and immune responses based on the available preclinical data, highlighting the fact that the link between leukocyte infiltration, lesion volume and neurological outcome remains unclear. We then describe what is known on neuroinflammation and immune responses in stroke patients, and summarize the results of the clinical trials on immunomodulatory drugs. In order to understand the gap between clinical trials and preclinical results on stroke, we discuss in detail the experimental results that served as the basis for the summarized clinical trials on immunomodulatory drugs, focusing on (i) experimental stroke models, (ii) the timing and selection of outcome measuring, (iii) alternative entry routes for leukocytes into the ischemic region, and (iv) factors affecting stroke outcome such as gender differences, ageing, comorbidities like hypertension and diabetes, obesity, tobacco, alcohol consumption and previous infections like Covid-19. We can do better for stroke treatment, especially when targeting inflammation following stroke. We need to re-think the design of stroke experimental setups, notably by (i) using clinically relevant models of stroke, (ii) including both radiological and neurological outcomes, (iii) performing long-term follow-up studies, (iv) conducting large-scale preclinical stroke trials, and (v) including stroke comorbidities in preclinical research.
Collapse
|
16
|
Bray MA, Sartain SE, Gollamudi J, Rumbaut RE. Microvascular thrombosis: experimental and clinical implications. Transl Res 2020; 225:105-130. [PMID: 32454092 PMCID: PMC7245314 DOI: 10.1016/j.trsl.2020.05.006] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 05/12/2020] [Accepted: 05/17/2020] [Indexed: 02/07/2023]
Abstract
A significant amount of clinical and research interest in thrombosis is focused on large vessels (eg, stroke, myocardial infarction, deep venous thrombosis, etc.); however, thrombosis is often present in the microcirculation in a variety of significant human diseases, such as disseminated intravascular coagulation, thrombotic microangiopathy, sickle cell disease, and others. Further, microvascular thrombosis has recently been demonstrated in patients with COVID-19, and has been proposed to mediate the pathogenesis of organ injury in this disease. In many of these conditions, microvascular thrombosis is accompanied by inflammation, an association referred to as thromboinflammation. In this review, we discuss endogenous regulatory mechanisms that prevent thrombosis in the microcirculation, experimental approaches to induce microvascular thrombi, and clinical conditions associated with microvascular thrombosis. A greater understanding of the links between inflammation and thrombosis in the microcirculation is anticipated to provide optimal therapeutic targets for patients with diseases accompanied by microvascular thrombosis.
Collapse
Key Words
- adamts13, a disintegrin-like and metalloproteinase with thrombospondin type 1 motif 13
- ap, alternate pathway
- apc, activated protein c
- aps, antiphospholipid syndrome
- caps, catastrophic aps
- asfa, american society for apheresis
- atp, adenosine triphosphate
- cfh, complement factor h
- con a, concavalin a
- cox, cyclooxygenase
- damp, damage-associated molecular pattern
- dic, disseminated intravascular coagulation
- gbm, glomerular basement membrane
- hellp, hemolysis, elevated liver enzymes, low platelets
- hitt, heparin-induced thrombocytopenia and thrombosis
- hlh, hemophagocytic lymphohistiocytosis
- hus, hemolytic-uremic syndrome
- isth, international society for thrombosis and haemostasis
- ivig, intravenous immunoglobulin
- ldh, lactate nos, nitric oxide synthase
- net, neutrophil extracellular trap
- pai-1, plasminogen activator inhibitor 1
- pf4, platelet factor 4
- prr, pattern recognition receptor
- rbc, red blood cell
- scd, sickle cell disease
- sle, systemic lupus erythematosus
- tlr, toll-like receptor
- tf, tissue factor
- tfpi, tissue factor pathway inhibitor
- tma, thrombotic microangiopathy
- tnf-α, tumor necrosis factor-α
- tpe, therapeutic plasma exchange
- ulc, ultra large heparin-pf4 complexes
- ulvwf, ultra-large von willebrand factor
- vwf, von willebrand factor
Collapse
Affiliation(s)
- Monica A Bray
- Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey VA Medical Center, Houston, Texas; Baylor College of Medicine, Houston, Texas
| | - Sarah E Sartain
- Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey VA Medical Center, Houston, Texas; Baylor College of Medicine, Houston, Texas
| | - Jahnavi Gollamudi
- Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey VA Medical Center, Houston, Texas; Baylor College of Medicine, Houston, Texas
| | - Rolando E Rumbaut
- Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey VA Medical Center, Houston, Texas; Baylor College of Medicine, Houston, Texas.
| |
Collapse
|
17
|
Huttinger AL, Wheeler DG, Gnyawali S, Dornbos D, Layzer JM, Venetos N, Talentino S, Musgrave NJ, Jones C, Bratton C, Joseph ME, Sen C, Sullenger BA, Nimjee SM. Ferric Chloride-induced Canine Carotid Artery Thrombosis: A Large Animal Model of Vascular Injury. J Vis Exp 2018. [PMID: 30247470 DOI: 10.3791/57981] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Occlusive arterial thrombosis leading to cerebral ischemic stroke and myocardial infarction contributes to ~13 million deaths every year globally. Here, we have translated a vascular injury model from a small animal into a large animal (canine), with slight modifications that can be used for pre-clinical screening of prophylactic and thrombolytic agents. In addition to the surgical methods, the modified protocol describes the step-by-step methods to assess carotid artery canalization by angiography, detailed instructions to process both the brain and carotid artery for histological analysis to verify carotid canalization and cerebral hemorrhage, and specific parameters to complete an assessment of downstream thromboembolic events by utilizing magnetic resonance imaging (MRI). In addition, specific procedural changes from the previously well-established small animal model necessary to translate into a large animal (canine) vascular injury are discussed.
Collapse
Affiliation(s)
| | | | | | - David Dornbos
- Department of Neurological Surgery, Ohio State University
| | | | | | | | | | - Cheyenne Jones
- Department of Neurological Surgery, Ohio State University
| | | | | | - Chandan Sen
- Department of Surgery, Ohio State University
| | | | | |
Collapse
|
18
|
Bonnard T, Jayapadman A, Putri JA, Cui J, Ju Y, Carmichael C, Angelovich TA, Cody SH, French S, Pascaud K, Pearce HA, Jagdale S, Caruso F, Hagemeyer CE. Low-Fouling and Biodegradable Protein-Based Particles for Thrombus Imaging. ACS NANO 2018; 12:6988-6996. [PMID: 29874911 DOI: 10.1021/acsnano.8b02588] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Nanomedicine holds great promise for vascular disease diagnosis and specific therapy, yet rapid sequestration by the mononuclear phagocytic system limits the efficacy of particle-based agents. The use of low-fouling polymers, such as poly(ethylene glycol), efficiently reduces this immune recognition, but these nondegradable polymers can accumulate in the human body and may cause adverse effects after prolonged use. Thus, new particle formulations combining stealth, low immunogenicity and biocompatible features are required to enable clinical use. Here, a low-fouling particle platform is described using exclusively protein material. A recombinant protein with superior hydrophilic characteristics provided by the amino acid repeat proline, alanine, and serine (PAS) is designed and cross-linked into particles with lysine (K) and polyglutamic acid (E) using mesoporous silica templating. The obtained PASKE particles have low-fouling behavior, have a prolonged circulation time compared to albumin-based particles, and are rapidly degraded in the cell's lysosomal compartment. When labeled with near-infrared fluorescent molecules and functionalized with an anti-glycoprotein IIb/IIIa single-chain antibody targeting activated platelets, the particles show potential as a noninvasive molecular imaging tool in a mouse model of carotid artery thrombosis. The PASKE particles constitute a promising biodegradable and versatile platform for molecular imaging of vascular diseases.
Collapse
Affiliation(s)
- Thomas Bonnard
- Nanobiotechnology Laboratory, Australian Centre for Blood Diseases, Central Clinical School , Monash University , Melbourne 3004 , Victoria , Australia
| | - Anand Jayapadman
- Nanobiotechnology Laboratory, Australian Centre for Blood Diseases, Central Clinical School , Monash University , Melbourne 3004 , Victoria , Australia
| | - Jasmine A Putri
- Nanobiotechnology Laboratory, Australian Centre for Blood Diseases, Central Clinical School , Monash University , Melbourne 3004 , Victoria , Australia
| | - Jiwei Cui
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, and the Department of Chemical Engineering , The University of Melbourne , Parkville 3010 , Victoria , Australia
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, and the School of Chemistry and Chemical Engineering , Shandong University , Jinan 250100 , China
| | - Yi Ju
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, and the Department of Chemical Engineering , The University of Melbourne , Parkville 3010 , Victoria , Australia
| | - Catherine Carmichael
- Mammalian Functional Genetics Laboratory, Australian Centre for Blood Diseases, Central Clinical School , Monash University , Melbourne 3004 , Victoria , Australia
| | - Thomas A Angelovich
- Chronic Infectious and Inflammatory Diseases Program , School of Health and Biomedical Sciences, RMIT University , Melbourne 3004 , Australia
- Life Sciences , Burnet Institute , Melbourne 3004 , Victoria , Australia
| | - Stephen H Cody
- Monash Micro Imaging , Monash University , Melbourne 3004 , Victoria , Australia
| | - Shauna French
- Platelets and Thrombosis Laboratory, Australian Centre for Blood Diseases, Central Clinical School , Monash University , Melbourne 3004 , Victoria , Australia
| | - Karline Pascaud
- Nanobiotechnology Laboratory, Australian Centre for Blood Diseases, Central Clinical School , Monash University , Melbourne 3004 , Victoria , Australia
| | - Hannah A Pearce
- Nanobiotechnology Laboratory, Australian Centre for Blood Diseases, Central Clinical School , Monash University , Melbourne 3004 , Victoria , Australia
| | - Shweta Jagdale
- Nanobiotechnology Laboratory, Australian Centre for Blood Diseases, Central Clinical School , Monash University , Melbourne 3004 , Victoria , Australia
| | - Frank Caruso
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, and the Department of Chemical Engineering , The University of Melbourne , Parkville 3010 , Victoria , Australia
| | - Christoph E Hagemeyer
- Nanobiotechnology Laboratory, Australian Centre for Blood Diseases, Central Clinical School , Monash University , Melbourne 3004 , Victoria , Australia
| |
Collapse
|
19
|
Kung PH, Hsieh PW, Lin YT, Lee JH, Chen IH, Wu CC. HPW-RX40 prevents human platelet activation by attenuating cell surface protein disulfide isomerases. Redox Biol 2017; 13:266-277. [PMID: 28600983 PMCID: PMC5466588 DOI: 10.1016/j.redox.2017.05.019] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 05/22/2017] [Accepted: 05/25/2017] [Indexed: 01/01/2023] Open
Abstract
Protein disulfide isomerase (PDI) present at platelet surfaces has been considered to play an important role in the conformational change and activation of the integrin glycoprotein IIb/IIIa (GPIIb/IIIa) and thus enhances platelet aggregation. Growing evidences indicated that platelet surface PDI may serve as a potential target for developing of a new class of antithrombotic agents. In the present study, we investigated the effects of HPW-RX40, a chemical derivative of β-nitrostyrene, on platelet activation and PDI activity. HPW-RX40 inhibited platelet aggregation, GPIIb/IIIa activation, and P-selectin expression in human platelets. Moreover, HPW-RX40 reduced thrombus formation in human whole blood under flow conditions, and protects mice from FeCl3-induced carotid artery occlusion. HPW-RX40 inhibited the activity of recombinant PDI family proteins (PDI, ERp57, and ERp5) as well as suppressed cell surface PDI activity of platelets in a reversible manner. Exogenous addition of PDI attenuated the inhibitory effect of HPW-RX40 on GPIIb/IIIa activation. Structure-based molecular docking simulations indicated that HPW-RX40 binds to the active site of PDI by forming hydrogen bonds. In addition, HPW-RX40 neither affected the cell viability nor induced endoplasmic reticulum stress in human cancer A549 and MDA-MB-231 cells. Taken together, our results suggest that HPW-RX40 is a reversible and non-cytotoxic PDI inhibitor with antiplatelet effects, and it may have a potential for development of novel antithrombotic agents.
Collapse
Affiliation(s)
- Po-Hsiung Kung
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Pei-Wen Hsieh
- Graduate Institute of Natural Products, School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Department of Anesthesiology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Ying-Ting Lin
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jia-Hau Lee
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - I-Hua Chen
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chin-Chung Wu
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung, Taiwan.
| |
Collapse
|
20
|
张 家, 陈 坤, 张 福, 李 少, 吴 源, 冯 靖, 王 武, 闫 玉. [Establishment of a rabbit model of small diameter vascular graft replacement]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2017; 37:687-692. [PMID: 28539296 PMCID: PMC6780461 DOI: 10.3969/j.issn.1673-4254.2017.05.21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Indexed: 06/07/2023]
Abstract
OBJECTIVE To establish an rabbit model that mimics the hemodynamics of the bypass graft after coronary artery bypass surgery. METHODS Sixteen New Zealand rabbits were randomly divided into two groups for abdominal aortic artery replacement using a 3-cm-long ePTFE graft with an inner diameter 4 mm through an incision at 1/3 from the middle to the lower part of the abdomen (group A) or in the lower abdomen (group B). The general conditions of the rabbits, operative time, number of collateral vessels that needed to be ligated, rate of massive intraoperative bleeding, fluctuation of vascular anastomosis after surgery, patency rate of the graft on day 7 after the operation were compared between the two groups. RESULTS The two groups of rabbits had similar body weight, diameter of the abdominal aortic artery, intraoperative bleeding rate and occlusion rate of the vascular graft at 7 days after the procedure. The operative time was longer in group A, but the difference was not statistically significant. In group A, the number of the vascular branches that needed to be ligated was smaller and the rate normal femoral artery pulsation was higher than those in group B. CONCLUSION It is feasible to establish models of small diameter vascular graft replacement in rabbits, and the patency rate of the graft can be monitored by observation of the general condition and ultrasound examination of the rabbits.
Collapse
Affiliation(s)
- 家庆 张
- 南方医科大学珠江医院胸心外科,广东 广州 510280Department of Cardiothoracic Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - 坤棠 陈
- 南方医科大学珠江医院胸心外科,广东 广州 510280Department of Cardiothoracic Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - 福伟 张
- 南方医科大学珠江医院胸心外科,广东 广州 510280Department of Cardiothoracic Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - 少彬 李
- 南方医科大学珠江医院胸心外科,广东 广州 510280Department of Cardiothoracic Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - 源周 吴
- 南方医科大学珠江医院胸心外科,广东 广州 510280Department of Cardiothoracic Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - 靖 冯
- 南方医科大学珠江医院胸心外科,广东 广州 510280Department of Cardiothoracic Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - 武军 王
- 南方医科大学南方医院胸心外科,广东 广州 510515Department of Cardiothoracic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - 玉生 闫
- 南方医科大学珠江医院胸心外科,广东 广州 510280Department of Cardiothoracic Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| |
Collapse
|
21
|
Bonnard T, Tennant Z, Niego B, Kanojia R, Alt K, Jagdale S, Law LS, Rigby S, Medcalf RL, Peter K, Hagemeyer CE. Novel Thrombolytic Drug Based on Thrombin Cleavable Microplasminogen Coupled to a Single-Chain Antibody Specific for Activated GPIIb/IIIa. J Am Heart Assoc 2017; 6:JAHA.116.004535. [PMID: 28159824 PMCID: PMC5523756 DOI: 10.1161/jaha.116.004535] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Background Thrombolytic therapy for acute thrombosis is limited by life‐threatening side effects such as major bleeding and neurotoxicity. New treatment options with enhanced fibrinolytic potential are therefore required. Here, we report the development of a new thrombolytic molecule that exploits key features of thrombosis. We designed a recombinant microplasminogen modified to be activated by the prothrombotic serine‐protease thrombin (HtPlg), fused to an activation‐specific anti–glycoprotein IIb/IIIa single‐chain antibody (SCE5), thereby hijacking the coagulation system to initiate thrombolysis. Methods and Results The resulting fusion protein named SCE5‐HtPlg shows in vitro targeting towards the highly abundant activated form of the fibrinogen receptor glycoprotein IIb/IIIa expressed on activated human platelets. Following thrombin formation, SCE5‐HtPlg is activated to contain active microplasmin. We evaluate the effectiveness of our targeted thrombolytic construct in two models of thromboembolic disease. Administration of SCE5‐HtPlg (4 μg/g body weight) resulted in effective thrombolysis 20 minutes after injection in a ferric chloride–induced model of mesenteric thrombosis (48±3% versus 92±5% for saline control, P<0.01) and also reduced emboli formation in a model of pulmonary embolism (P<0.01 versus saline). Furthermore, at these effective therapeutic doses, the SCE5‐HtPlg did not prolong bleeding time compared with saline (P=0.99). Conclusions Our novel fusion molecule is a potent and effective treatment for thrombosis that enables in vivo thrombolysis without bleeding time prolongation. The activation of this construct by thrombin generated within the clot itself rather than by a plasminogen activator, which needs to be delivered systemically, provides a novel targeted approach to improve thrombolysis.
Collapse
Affiliation(s)
- Thomas Bonnard
- NanoBiotechnology Laboratory, Australian Centre for Blood Diseases, Monash University, Melbourne, Australia.,Vascular Biotechnology Laboratory, Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| | - Zachary Tennant
- Vascular Biotechnology Laboratory, Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| | - Be'Eri Niego
- Molecular Neurotrauma and Haemostasis Laboratory, Australian Centre for Blood Diseases, Monash University, Melbourne, Australia
| | - Ruchi Kanojia
- Vascular Biotechnology Laboratory, Baker IDI Heart and Diabetes Institute, Melbourne, Australia.,Atherothrombosis and Vascular Biology Laboratory, Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| | - Karen Alt
- NanoBiotechnology Laboratory, Australian Centre for Blood Diseases, Monash University, Melbourne, Australia.,Vascular Biotechnology Laboratory, Baker IDI Heart and Diabetes Institute, Melbourne, Australia.,Atherothrombosis and Vascular Biology Laboratory, Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| | - Shweta Jagdale
- NanoBiotechnology Laboratory, Australian Centre for Blood Diseases, Monash University, Melbourne, Australia.,Vascular Biotechnology Laboratory, Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| | - Lok Soon Law
- Vascular Biotechnology Laboratory, Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| | - Sheena Rigby
- Vascular Biotechnology Laboratory, Baker IDI Heart and Diabetes Institute, Melbourne, Australia.,Atherothrombosis and Vascular Biology Laboratory, Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| | - Robert Lindsay Medcalf
- Molecular Neurotrauma and Haemostasis Laboratory, Australian Centre for Blood Diseases, Monash University, Melbourne, Australia
| | - Karlheinz Peter
- Atherothrombosis and Vascular Biology Laboratory, Baker IDI Heart and Diabetes Institute, Melbourne, Australia.,RMIT University, Melbourne, Australia
| | - Christoph Eugen Hagemeyer
- NanoBiotechnology Laboratory, Australian Centre for Blood Diseases, Monash University, Melbourne, Australia .,Vascular Biotechnology Laboratory, Baker IDI Heart and Diabetes Institute, Melbourne, Australia.,RMIT University, Melbourne, Australia
| |
Collapse
|