1
|
Miranda-Mosqueda ML, Ruiz-Oropeza SY, González-Barrios JA, Jaimez R, Peña-Ortega F, Gómez-Acevedo C. Agmatine: An Emerging Approach for Neuroprotection in Recurrent Ischemic Stroke Events in a Murine Model. Drug Dev Res 2024; 85:e70012. [PMID: 39470139 DOI: 10.1002/ddr.70012] [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: 05/08/2024] [Revised: 10/07/2024] [Accepted: 10/11/2024] [Indexed: 10/30/2024]
Abstract
This study investigates the effect of agmatine on reducing mortality, neurobehavioral alterations, infarct size, and expression of pro-inflammatory cytokines in mice subjected to bilateral carotid thrombosis. Under pentobarbital anesthesia, the left common carotid artery was exposed to 6% FeCl3. Thirty-two days later, the same procedure was performed on the right common carotid artery. Subsequently, Agmatine (100 mg/kg) was administered 15 min after the second procedure, and in another experimental group, the dose of Agmatine was repeated at 72 h. Administration of agmatine extended survival in ischemic animals up to 72 h for the single-dose group and up to 96 h for the repeated-dose group, without significant increases in neurological deficits or infarct area size. This neurobehavioral effect was also observed in sham animals treated with agmatine. In ischemic animals, agmatine administration improved digging behavior and reduced recovery times, consistently shorter in those animals treated with repeated doses. RT-PCR analyses revealed a positive regulation of the cytokine IL-1β in agmatine-treated animals, which has been associated with recovery stages. The results suggest that the observed effect may be attributed to the multiple interactions of agmatine with ischemic cascade events, highlighting its anti-inflammatory role.
Collapse
Affiliation(s)
- M L Miranda-Mosqueda
- Laboratorio de Farmacología Conductual, Departamento de Farmacología, Facultad de Medicina, UNAM, Juriquilla, México
| | - S Y Ruiz-Oropeza
- Laboratorio de Farmacología Conductual, Departamento de Farmacología, Facultad de Medicina, UNAM, Juriquilla, México
| | - J A González-Barrios
- Laboratorio de Medicina Genómica, Hospital Regional 1° de Octubre, ISSSTE, México City, México
| | - R Jaimez
- Laboratorio de Estrógenos y Hemostasis, Departamento de Farmacología, Facultad de Medicina, UNAM, Juriquilla, México
| | - Fernando Peña-Ortega
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Juriquilla, México
| | - C Gómez-Acevedo
- Laboratorio de Farmacología Conductual, Departamento de Farmacología, Facultad de Medicina, UNAM, Juriquilla, México
| |
Collapse
|
2
|
Roytenberg R, Yue H, DeHart A, Kim E, Bai F, Kim Y, Denning K, Kwei A, Zhang Q, Liu J, Zheng XL, Li W. Thymidine phosphorylase mediates SARS-CoV-2 spike protein enhanced thrombosis in K18-hACE2 TG mice. Thromb Res 2024; 244:109195. [PMID: 39442286 DOI: 10.1016/j.thromres.2024.109195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 09/09/2024] [Accepted: 10/17/2024] [Indexed: 10/25/2024]
Abstract
INTRODUCTION Thymidine phosphorylase (TYMP), which facilitates platelet activation and thrombosis, is significantly increased in COVID-19 patients. We hypothesize that TYMP mediates SARS-CoV-2 spike protein (SP)-induced thrombosis. MATERIALS AND METHODS Plasmids encoding wildtype SP or empty vector (p3.1) were transfected into COS-7 cells, and cell lysates were prepared as a reservoir for SP or p3.1 (control), respectively. K18-hACE2TG and K18-hACE2TG/Tymp-/- mice were treated with a single dose of SP or p3.1 by intraperitoneal injection and then subjected to thrombosis studies three days later. The role of SP on inflammatory signaling activation was assessed in BEAS-2B cells. RESULTS SARS-CoV-2 SP increased the expression of TYMP, resulting in the activation of STAT3 and NF-κB in BEAS-2B cells. A siRNA-mediated knockdown of TYMP attenuated SP-enhanced activation of STAT3. SP significantly promoted arterial thrombosis in K18-hACE2TG mice. SP-accelerated thrombosis was attenuated by inhibition or genetic ablation of TYMP. SP treatment did not influence ADP- or collagen-induced platelet aggregation but significantly increased platelet adhesion to fibrinogen. SP treatment also significantly shortened activated partial thromboplastin time, which was reversed and even prolonged by TYMP deficiency. Additionally, SP binds to platelet factor 4 (PF4) and TYMP. TYMP does not bind PF4 but enhances the formation of the SP/PF4 complex, which may augment the procoagulant and prothrombotic effect of PF4. CONCLUSIONS We conclude that SP is prothrombotic and upregulates TYMP expression, and TYMP inhibition or knockout mitigates SP-enhanced thrombosis. These findings suggest that inhibition of TYMP may be a novel therapeutic strategy for COVID-19-associated thrombosis.
Collapse
Affiliation(s)
- Renat Roytenberg
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine at Marshall University, Huntington, WV 25755, USA
| | - Hong Yue
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine at Marshall University, Huntington, WV 25755, USA
| | - Autumn DeHart
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine at Marshall University, Huntington, WV 25755, USA
| | - Eugene Kim
- Department of Chemistry, College of Sciences, Marshall University, Huntington, WV 25755, USA
| | - Fang Bai
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine at Marshall University, Huntington, WV 25755, USA
| | - Yongick Kim
- Department of Chemistry, College of Sciences, Marshall University, Huntington, WV 25755, USA
| | - Krista Denning
- Department of Pathology, Joan C. Edwards School of Medicine at Marshall University, Huntington, WV 25755, USA
| | - Alec Kwei
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine at Marshall University, Huntington, WV 25755, USA
| | - Quan Zhang
- Department of Pathology and Laboratory Medicine, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Jiang Liu
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine at Marshall University, Huntington, WV 25755, USA
| | - X Long Zheng
- Department of Pathology and Laboratory Medicine, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Wei Li
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine at Marshall University, Huntington, WV 25755, USA.
| |
Collapse
|
3
|
Han X, Knauss EA, Fuente MDL, Li W, Conlon RA, LePage DF, Jiang W, Renna SA, McKenzie SE, Nieman MT. A mouse model of the protease-activated receptor 4 Pro310Leu variant has reduced platelet reactivity. J Thromb Haemost 2024; 22:1715-1726. [PMID: 38508397 DOI: 10.1016/j.jtha.2024.03.004] [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: 12/01/2023] [Revised: 02/26/2024] [Accepted: 03/11/2024] [Indexed: 03/22/2024]
Abstract
BACKGROUND Protease-activated receptor 4 (PAR4) mediates thrombin signaling on platelets and other cells. Our recent structural studies demonstrated that a single nucleotide polymorphism in extracellular loop 3 and PAR4-P310L (rs2227376) leads to a hyporeactive receptor. OBJECTIVES The goal of this study was to determine how the hyporeactive PAR4 variant in extracellular loop 3 impacts platelet function in vivo using a novel knock-in mouse model (PAR4-322L). METHODS A point mutation was introduced into the PAR4 gene F2rl3 via CRISPR/Cas9 to create PAR4-P322L, the mouse homolog to human PAR4-P310L. Platelet response to PAR4 activation peptide (AYPGKF), thrombin, ADP, and convulxin was monitored by αIIbβ3 integrin activation and P-selectin translocation using flow cytometry or platelet aggregation. In vivo responses were determined by the tail bleeding assay and the ferric chloride-induced carotid artery injury model. RESULTS PAR4-P/L and PAR4-L/L platelets had a reduced response to AYPGKF and thrombin measured by P-selectin translocation or αIIbβ3 activation. The response to ADP and convulxin was unchanged among genotypes. In addition, both PAR4-P/L and PAR4-L/L platelets showed a reduced response to thrombin in aggregation studies. There was an increase in the tail bleeding time for PAR4-L/L mice. The PAR4-P/L and PAR4-L/L mice both showed an extended time to arterial thrombosis. CONCLUSION PAR4-322L significantly reduced platelet responsiveness to AYPGKF and thrombin, which is in agreement with our previous structural and cell signaling studies. In addition, PAR4-322L had prolonged arterial thrombosis time. Our mouse model provides a foundation to further evaluate the role of PAR4 in other pathophysiological contexts.
Collapse
Affiliation(s)
- Xu Han
- Case Western Reserve University School of Medicine, Department of Pharmacology, Cleveland, Ohio, USA
| | - Elizabeth A Knauss
- Case Western Reserve University School of Medicine, Department of Pharmacology, Cleveland, Ohio, USA
| | - Maria de la Fuente
- Case Western Reserve University School of Medicine, Department of Pharmacology, Cleveland, Ohio, USA
| | - Wei Li
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine at Marshall University, Huntington, West Virginia, USA
| | - Ronald A Conlon
- Case Transgenic and Targeting Facility, Case Western Reserve University, Cleveland, Ohio, USA
| | - David F LePage
- Case Transgenic and Targeting Facility, Case Western Reserve University, Cleveland, Ohio, USA
| | - Weihong Jiang
- Case Transgenic and Targeting Facility, Case Western Reserve University, Cleveland, Ohio, USA
| | - Stephanie A Renna
- Department of Medicine, The Cardeza Foundation for Hematologic Research, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Steven E McKenzie
- Department of Medicine, The Cardeza Foundation for Hematologic Research, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Marvin T Nieman
- Case Western Reserve University School of Medicine, Department of Pharmacology, Cleveland, Ohio, USA.
| |
Collapse
|
4
|
Yang W, Feng R, Peng G, Wang Z, Cen M, Jing Y, Feng W, Long T, Liu Y, Li Z, Huang K, Chang G. Glycoursodeoxycholic Acid Alleviates Arterial Thrombosis via Suppressing Diacylglycerol Kinases Activity in Platelet. Arterioscler Thromb Vasc Biol 2024; 44:1283-1301. [PMID: 38572646 DOI: 10.1161/atvbaha.124.320728] [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: 01/16/2024] [Accepted: 03/19/2024] [Indexed: 04/05/2024]
Abstract
BACKGROUND Glycoursodeoxycholic acid (GUDCA) has been acknowledged for its ability to regulate lipid homeostasis and provide benefits for various metabolic disorders. However, the impact of GUDCA on arterial thrombotic events remains unexplored. The objective of this study is to examine the effects of GUDCA on thrombogenesis and elucidate its underlying mechanisms. METHODS Plasma samples from patients with arterial thrombotic events and diet-induced obese mice were collected to determine the GUDCA concentrations using mass spectrometry. Multiple in vivo murine thrombosis models and in vitro platelet functional assays were conducted to comprehensively evaluate the antithrombotic effects of GUDCA. Moreover, lipidomic analysis was performed to identify the alterations of intraplatelet lipid components following GUDCA treatment. RESULTS Plasma GUDCA level was significantly decreased in patients with arterial thrombotic events and negatively correlated with thrombotic propensity in diet-induced obese mice. GUDCA exhibited prominent suppressing effects on platelet reactivity as evidenced by the attenuation of platelet activation, secretion, aggregation, spreading, and retraction (P<0.05). In vivo, GUDCA administration robustly alleviated thrombogenesis (P<0.05) without affecting hemostasis. Mechanistically, GUDCA inhibited DGK (diacylglycerol kinase) activity, leading to the downregulation of the phosphatidic acid-mediated signaling pathway. Conversely, phosphatidic acid supplementation was sufficient to abolish the antithrombotic effects of GUDCA. More importantly, long-term oral administration of GUDCA normalized the enhanced DGK activity, thereby remarkably alleviating the platelet hyperreactivity as well as the heightened thrombotic tendency in diet-induced obese mice (P<0.05). CONCLUSIONS Our study implicated that GUDCA reduces platelet hyperreactivity and improves thrombotic propensity by inhibiting DGKs activity, which is a potentially effective prophylactic approach and promising therapeutic agent for arterial thrombotic events.
Collapse
Affiliation(s)
- Wenchao Yang
- Division of Vascular Surgery, National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China (W.Y., R.F., G.P., Z.W., Y.J., W.F., T.L., Y.L., Z.L, K.H., G.C.)
| | - Ruijia Feng
- Division of Vascular Surgery, National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China (W.Y., R.F., G.P., Z.W., Y.J., W.F., T.L., Y.L., Z.L, K.H., G.C.)
| | - Guiyan Peng
- Division of Vascular Surgery, National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China (W.Y., R.F., G.P., Z.W., Y.J., W.F., T.L., Y.L., Z.L, K.H., G.C.)
| | - Zhecun Wang
- Division of Vascular Surgery, National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China (W.Y., R.F., G.P., Z.W., Y.J., W.F., T.L., Y.L., Z.L, K.H., G.C.)
| | - Meifeng Cen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, China (M.C.)
| | - Yexiang Jing
- Division of Vascular Surgery, National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China (W.Y., R.F., G.P., Z.W., Y.J., W.F., T.L., Y.L., Z.L, K.H., G.C.)
| | - Weiqi Feng
- Division of Vascular Surgery, National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China (W.Y., R.F., G.P., Z.W., Y.J., W.F., T.L., Y.L., Z.L, K.H., G.C.)
| | - Ting Long
- Division of Vascular Surgery, National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China (W.Y., R.F., G.P., Z.W., Y.J., W.F., T.L., Y.L., Z.L, K.H., G.C.)
| | - Yunchong Liu
- Division of Vascular Surgery, National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China (W.Y., R.F., G.P., Z.W., Y.J., W.F., T.L., Y.L., Z.L, K.H., G.C.)
| | - Zilun Li
- Division of Vascular Surgery, National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China (W.Y., R.F., G.P., Z.W., Y.J., W.F., T.L., Y.L., Z.L, K.H., G.C.)
| | - Kan Huang
- Division of Vascular Surgery, National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China (W.Y., R.F., G.P., Z.W., Y.J., W.F., T.L., Y.L., Z.L, K.H., G.C.)
| | - Guangqi Chang
- Division of Vascular Surgery, National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China (W.Y., R.F., G.P., Z.W., Y.J., W.F., T.L., Y.L., Z.L, K.H., G.C.)
| |
Collapse
|
5
|
Li OQ, Yue H, DeHart AR, Roytenberg R, Aguilar R, Olanipekun O, Bai F, Liu J, Fedorova O, Kennedy D, Thompson E, Pierre SV, Li W. Sodium/Potassium ATPase Alpha 1 Subunit Fine-tunes Platelet GPCR Signaling Function and is Essential for Thrombosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.13.593923. [PMID: 38798556 PMCID: PMC11118499 DOI: 10.1101/2024.05.13.593923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Background Thrombosis is a major cause of myocardial infarction and ischemic stroke. The sodium/potassium ATPase (NKA), comprising α and β subunits, is crucial in maintaining intracellular sodium and potassium gradients. However, the role of NKA in platelet function and thrombosis remains unclear. Methods We utilized wild-type (WT, α1+/+) and NKA α1 heterozygous (α1+/-) mice, aged 8 to 16 weeks, of both sexes. An intravital microscopy-based, FeCl3-induced carotid artery injury thrombosis model was employed for in vivo thrombosis assessment. Platelet transfusion assays were used to evaluate platelet NKA α1 function on thrombosis. Human platelets isolated from healthy donors and heart failure patients treated with/without digoxin were used for platelet function and signaling assay. Complementary molecular approaches were used for mechanistic studies. Results NKA α1 haplodeficiency significantly reduced its expression on platelets without affecting sodium homeostasis. It significantly inhibited 7.5% FeCl3-induced thrombosis in male but not female mice without disturbing hemostasis. Transfusion of α1+/-, but not α1+/+, platelets to thrombocytopenic WT mice substantially prolonged thrombosis. Treating WT mice with low-dose ouabain or marinobufagenin, both binding NKA α1 and inhibiting its ion-transporting function, markedly inhibited thrombosis in vivo. NKA α1 formed complexes with leucine-glycine-leucine (LGL)-containing platelet receptors, including P2Y12, PAR4, and thromboxane A2 receptor. This binding was significantly attenuated by LGL>SFT mutation or LGL peptide. Haplodeficiency of NKA α1 in mice or ouabain treatment of human platelets notably inhibited ADP-induced platelet aggregation. While not affecting 10% FeCl3-induced thrombosis, NKA α1 haplodeficiency significantly prolonged thrombosis time in mice treated with an ineffective dose of clopidogrel. Conclusion NKA α1 plays an essential role in enhancing platelet activation through binding to LGL-containing platelet GPCRs. NKA α1 haplodeficiency or inhibition with low-dose ouabain and marinobufagenin significantly inhibited thrombosis and sensitized clopidogrel's anti-thrombotic effect. Targeting NKA α1 emerges as a promising antiplatelet and antithrombotic therapeutic strategy.
Collapse
Affiliation(s)
- Oliver Q. Li
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine at Marshall University, Huntington, WV, USA
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV, USA
| | - Hong Yue
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine at Marshall University, Huntington, WV, USA
| | - Autumn R. DeHart
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine at Marshall University, Huntington, WV, USA
| | - Renat Roytenberg
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine at Marshall University, Huntington, WV, USA
| | - Rodrigo Aguilar
- Department of Medicine, Joan C. Edwards School of Medicine at Marshall University, Huntington, WV, USA
| | - Olalekan Olanipekun
- Department of Medicine, Joan C. Edwards School of Medicine at Marshall University, Huntington, WV, USA
| | - Fang Bai
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine at Marshall University, Huntington, WV, USA
| | - Jiang Liu
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine at Marshall University, Huntington, WV, USA
| | - Olga Fedorova
- National Institute on Aging, Laboratory of Cardiovascular Science of Biomedical Research Center Baltimore, MD, USA
| | - David Kennedy
- Department of Medicine, University of Toledo, Toledo, OH, USA
| | - Ellen Thompson
- Department of Medicine, Joan C. Edwards School of Medicine at Marshall University, Huntington, WV, USA
| | - Sandrine V. Pierre
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine at Marshall University, Huntington, WV, USA
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV, USA
| | - Wei Li
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine at Marshall University, Huntington, WV, USA
| |
Collapse
|
6
|
Han G, Zhang Y, Zhong L, Wang B, Qiu S, Song J, Lin C, Zou F, Wu J, Yu H, Liang C, Wen K, Seow Y, Yin H. Generalizable anchor aptamer strategy for loading nucleic acid therapeutics on exosomes. EMBO Mol Med 2024; 16:1027-1045. [PMID: 38448545 PMCID: PMC11018858 DOI: 10.1038/s44321-024-00049-7] [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: 11/30/2023] [Revised: 02/15/2024] [Accepted: 02/19/2024] [Indexed: 03/08/2024] Open
Abstract
Clinical deployment of oligonucleotides requires delivery technologies that improve stability, target tissue accumulation and cellular internalization. Exosomes show potential as ideal delivery vehicles. However, an affordable generalizable system for efficient loading of oligonucleotides on exosomes remain lacking. Here, we identified an Exosomal Anchor DNA Aptamer (EAA) via SELEX against exosomes immobilized with our proprietary CP05 peptides. EAA shows high binding affinity to different exosomes and enables efficient loading of nucleic acid drugs on exosomes. Serum stability of thrombin inhibitor NU172 was prolonged by exosome-loading, resulting in increased blood flow after injury in vivo. Importantly, Duchenne Muscular Dystrophy PMO can be readily loaded on exosomes via EAA (EXOEAA-PMO). EXOEAA-PMO elicited significantly greater muscle cell uptake, tissue accumulation and dystrophin expression than PMO in vitro and in vivo. Systemic administration of EXOEAA-PMO elicited therapeutic levels of dystrophin restoration and functional improvements in mdx mice. Altogether, our study demonstrates that EAA enables efficient loading of different nucleic acid drugs on exosomes, thus providing an easy and generalizable strategy for loading nucleic acid therapeutics on exosomes.
Collapse
Affiliation(s)
- Gang Han
- State Key Laboratory of Experimental Hematology & The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics & International Joint Laboratory of Ocular Diseases (Ministry of Education), School of Medical Technology & School of Basic Medical Sciences, Tianjin Medical University, Qixiangtai Road, Heping District, 300070, Tianjin, China
| | - Yao Zhang
- State Key Laboratory of Experimental Hematology & The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics & International Joint Laboratory of Ocular Diseases (Ministry of Education), School of Medical Technology & School of Basic Medical Sciences, Tianjin Medical University, Qixiangtai Road, Heping District, 300070, Tianjin, China
| | - Li Zhong
- State Key Laboratory of Experimental Hematology & The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics & International Joint Laboratory of Ocular Diseases (Ministry of Education), School of Medical Technology & School of Basic Medical Sciences, Tianjin Medical University, Qixiangtai Road, Heping District, 300070, Tianjin, China
| | - Biaobiao Wang
- State Key Laboratory of Experimental Hematology & The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics & International Joint Laboratory of Ocular Diseases (Ministry of Education), School of Medical Technology & School of Basic Medical Sciences, Tianjin Medical University, Qixiangtai Road, Heping District, 300070, Tianjin, China
| | - Shuai Qiu
- State Key Laboratory of Experimental Hematology & The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics & International Joint Laboratory of Ocular Diseases (Ministry of Education), School of Medical Technology & School of Basic Medical Sciences, Tianjin Medical University, Qixiangtai Road, Heping District, 300070, Tianjin, China
| | - Jun Song
- State Key Laboratory of Experimental Hematology & The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics & International Joint Laboratory of Ocular Diseases (Ministry of Education), School of Medical Technology & School of Basic Medical Sciences, Tianjin Medical University, Qixiangtai Road, Heping District, 300070, Tianjin, China
| | - Caorui Lin
- State Key Laboratory of Experimental Hematology & The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics & International Joint Laboratory of Ocular Diseases (Ministry of Education), School of Medical Technology & School of Basic Medical Sciences, Tianjin Medical University, Qixiangtai Road, Heping District, 300070, Tianjin, China
| | - Fangdi Zou
- Public Laboratory & Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center & Tianjin's Clinical Research Center for Cancer, 300060, Tianjin, China
| | - Jingqiao Wu
- State Key Laboratory of Experimental Hematology & The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics & International Joint Laboratory of Ocular Diseases (Ministry of Education), School of Medical Technology & School of Basic Medical Sciences, Tianjin Medical University, Qixiangtai Road, Heping District, 300070, Tianjin, China
| | - Huanan Yu
- State Key Laboratory of Experimental Hematology & The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics & International Joint Laboratory of Ocular Diseases (Ministry of Education), School of Medical Technology & School of Basic Medical Sciences, Tianjin Medical University, Qixiangtai Road, Heping District, 300070, Tianjin, China
| | - Chao Liang
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, 518055, Shenzhen, China
| | - Ke Wen
- Department of Pharmacology, Tianjin Key Laboratory of Inflammatory Biology, School of Basic Medical Sciences, Tianjin Medical University, 300070, Tianjin, China
| | - Yiqi Seow
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis St, Genome, Singapore, 138672, Republic of Singapore
| | - HaiFang Yin
- State Key Laboratory of Experimental Hematology & The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics & International Joint Laboratory of Ocular Diseases (Ministry of Education), School of Medical Technology & School of Basic Medical Sciences, Tianjin Medical University, Qixiangtai Road, Heping District, 300070, Tianjin, China.
- Department of Clinical Laboratory, Tianjin Medical University General Hospital, 300052, Tianjin, China.
| |
Collapse
|
7
|
Tang X, Liao R, Zhou L, Yi T, Ran M, Luo J, Huang F, Wu A, Mei Q, Wang L, Huang X, Wu J. Genistin: A Novel Estrogen Analogue Targeting ERβ to Alleviate Thrombocytopenia. Int J Biol Sci 2024; 20:2236-2260. [PMID: 38617546 PMCID: PMC11008259 DOI: 10.7150/ijbs.90483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 02/14/2024] [Indexed: 04/16/2024] Open
Abstract
Thrombocytopenia, a prevalent hematologic challenge, correlates directly with the mortality of numerous ailments. Current therapeutic avenues for thrombocytopenia are not without limitations. Here, we identify genistin, an estrogen analogue, as a promising candidate for thrombocytopenia intervention, discovered through AI-driven compound library screening. While estrogen's involvement in diverse biological processes is recognized, its role in thrombopoiesis remains underexplored. Our findings elucidate genistin's ability to enhance megakaryocyte differentiation, thereby augmenting platelet formation and production. In vivo assessments further underscore genistin's remedial potential against radiation-induced thrombocytopenia. Mechanistically, genistin's efficacy is attributed to its direct interaction with estrogen receptor β (ERβ), with subsequent activation of both ERK1/2 and the Akt signaling pathways membrane ERβ. Collectively, our study positions genistin as a prospective therapeutic strategy for thrombocytopenia, shedding light on novel interplays between platelet production and ERβ.
Collapse
Affiliation(s)
- Xiaoqin Tang
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou,646000, China
- School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China
| | - Rui Liao
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou,646000, China
- School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China
| | - Ling Zhou
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou,646000, China
- School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China
| | - Taian Yi
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Mei Ran
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou,646000, China
| | - Jiesi Luo
- School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China
| | - Feihong Huang
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou,646000, China
| | - Anguo Wu
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou,646000, China
| | - Qibing Mei
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou,646000, China
| | - Long Wang
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou,646000, China
| | - Xinwu Huang
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou,646000, China
| | - Jianming Wu
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou,646000, China
- School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China
- Education Ministry Key Laboratory of Medical Electrophysiology, Southwest Medical University, Luzhou 646000, China
| |
Collapse
|
8
|
Singh N, Kulkarni PP, Tripathi P, Agarwal V, Dash D. Nanogold-coated stent facilitated non-invasive photothermal ablation of stent thrombosis and restoration of blood flow. NANOSCALE ADVANCES 2024; 6:1497-1506. [PMID: 38419863 PMCID: PMC10898437 DOI: 10.1039/d3na00751k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 01/23/2024] [Indexed: 03/02/2024]
Abstract
In-stent restenosis (ISR) and stent thrombosis (ST) are the most serious complications of coronary angioplasty and stenting. Although the evolution of drug-eluting stents (DES) has significantly restricted the incidence of ISR, they are associated with an enhanced risk of ST. In the present study, we explore the photothermal ablation of a thrombus using a nano-enhanced thermogenic stent (NETS) as a modality for revascularization following ST. The photothermal activity of NETS, fabricated by coating bare metal stents with gold nanorods generating a thin plasmonic film of gold, was found to be effective in rarefying clots formed within the stent lumen in various in vitro assays including those under conditions mimicking blood flow. NETS implanted in the rat common carotid artery generated heat following exposure to a NIR-laser that led to effective restoration of blood flow within the occluded vessel in a model of ferric chloride-induced thrombosis. Our results present a proof-of-concept for a novel photothermal ablation approach by employing coated stents in the non-invasive management of ST.
Collapse
Affiliation(s)
- Nitesh Singh
- Centre for Advanced Research on Platelet Signaling and Thrombosis Biology, Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University Varanasi-221005 India
| | - Paresh P Kulkarni
- Centre for Advanced Research on Platelet Signaling and Thrombosis Biology, Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University Varanasi-221005 India
| | - Prashant Tripathi
- School of Physical Sciences, Jawaharlal Nehru University New Mehrauli Road New Delhi Delhi-110067 India
| | - Vikas Agarwal
- Department of Cardiology, Institute of Medical Sciences, Banaras Hindu University Varanasi-221005 India
| | - Debabrata Dash
- Centre for Advanced Research on Platelet Signaling and Thrombosis Biology, Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University Varanasi-221005 India
| |
Collapse
|
9
|
Yang M, Silverstein RL. Targeting Cysteine Oxidation in Thrombotic Disorders. Antioxidants (Basel) 2024; 13:83. [PMID: 38247507 PMCID: PMC10812781 DOI: 10.3390/antiox13010083] [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: 12/05/2023] [Revised: 12/29/2023] [Accepted: 01/05/2024] [Indexed: 01/23/2024] Open
Abstract
Oxidative stress increases the risk for clinically significant thrombotic events, yet the mechanisms by which oxidants become prothrombotic are unclear. In this review, we provide an overview of cysteine reactivity and oxidation. We then highlight recent findings on cysteine oxidation events in oxidative stress-related thrombosis. Special emphasis is on the signaling pathway induced by a platelet membrane protein, CD36, in dyslipidemia, and by protein disulfide isomerase (PDI), a member of the thiol oxidoreductase family of proteins. Antioxidative and chemical biology approaches to target cysteine are discussed. Lastly, the knowledge gaps in the field are highlighted as they relate to understanding how oxidative cysteine modification might be targeted to limit thrombosis.
Collapse
Affiliation(s)
- Moua Yang
- Division of Hemostasis and Thrombosis, Beth Israel Deaconess Medical Center, Harvard Medical School, 3 Blackfan Circle, CLS-924, Boston, MA 02115, USA
| | - Roy L. Silverstein
- Department of Medicine, Medical College of Wisconsin, Hub 8745, 8701 W Watertown Plank Rd., Milwaukee, WI 53226, USA
- Versiti Blood Research Institute, Milwaukee, WI 53226, USA
| |
Collapse
|
10
|
Li Z, Zhang J, Ma Z, Zhao G, He X, Yu X, Fu Q, Wu N, Ding Z, Sun H, Zhang X, Zhu Y, Chen L, He J. Endothelial YAP Mediates Hyperglycemia-Induced Platelet Hyperactivity and Arterial Thrombosis. Arterioscler Thromb Vasc Biol 2024; 44:254-270. [PMID: 37916416 DOI: 10.1161/atvbaha.123.319835] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 10/11/2023] [Indexed: 11/03/2023]
Abstract
BACKGROUND Hyperglycemia-a symptom that characterizes diabetes-is highly associated with atherothrombotic complications. However, the underlying mechanism by which hyperglycemia fuels platelet activation and arterial thrombus formation is still not fully understood. METHODS The profiles of polyunsaturated fatty acid metabolites in the plasma of patients with diabetes and healthy controls were determined with targeted metabolomics. FeCl3-induced carotid injury model was used to assess arterial thrombus formation in mice with endothelial cell (EC)-specific YAP (yes-associated protein) deletion or overexpression. Flow cytometry and clot retraction assay were used to evaluate platelet activation. RNA sequencing and multiple biochemical analyses were conducted to unravel the underlying mechanism. RESULTS The plasma PGE2 (prostaglandin E2) concentration was elevated in patients with diabetes with thrombotic complications and positively correlated with platelet activation. The PGE2 synthetases COX-2 (cyclooxygenase-2) and mPGES-1 (microsomal prostaglandin E synthase-1) were found to be highly expressed in ECs but not in other type of vessel cells in arteries from both patients with diabetes and hyperglycemic mice, compared with nondiabetic individuals and control mice, respectively. A combination of RNA sequencing and ingenuity pathway analyses indicated the involvement of YAP signaling. EC-specific deletion of YAP limited platelet activation and arterial thrombosis in hyperglycemic mice, whereas EC-specific overexpression of YAP in mice mimicked the prothrombotic state of diabetes, without affecting hemostasis. Mechanistically, we found that hyperglycemia/high glucose-induced endothelial YAP nuclear translocation and subsequently transcriptional expression of COX-2 and mPGES-1 contributed to the elevation of PGE2 and platelet activation. Blockade of EP3 (prostaglandin E receptor 3) activation by oral administration of DG-041 reversed the hyperactivity of platelets and delayed thrombus formation in both EC-specific YAP-overexpressing and hyperglycemic mice. CONCLUSIONS Collectively, our data suggest that hyperglycemia-induced endothelial YAP activation aggravates platelet activation and arterial thrombus formation via PGE2/EP3 signaling. Targeting EP3 with DG-041 might be therapeutic for diabetes-related thrombosis.
Collapse
Affiliation(s)
- Zhiyu Li
- Tianjin Key Laboratory of Metabolic Diseases, Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Physiology and Pathophysiology (Z.L., J.Z., G.Z., X.H., X.Z., Y.Z., J.H.), Tianjin Medical University, China
| | - Jiachen Zhang
- Tianjin Key Laboratory of Metabolic Diseases, Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Physiology and Pathophysiology (Z.L., J.Z., G.Z., X.H., X.Z., Y.Z., J.H.), Tianjin Medical University, China
| | - Zejun Ma
- Tianjin Key Laboratory of Metabolic Diseases, Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Physiology and Pathophysiology (Z.L., J.Z., G.Z., X.H., X.Z., Y.Z., J.H.), Tianjin Medical University, China
- National Humanities Center Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology (Z.M., H.S., L.C.), Tianjin Medical University, China
| | - Guobing Zhao
- Tianjin Key Laboratory of Metabolic Diseases, Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Physiology and Pathophysiology (Z.L., J.Z., G.Z., X.H., X.Z., Y.Z., J.H.), Tianjin Medical University, China
| | - Xue He
- Tianjin Key Laboratory of Metabolic Diseases, Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Physiology and Pathophysiology (Z.L., J.Z., G.Z., X.H., X.Z., Y.Z., J.H.), Tianjin Medical University, China
| | - Xuefang Yu
- Departments of Cardiology (X.Y.), Tianjin Medical University General Hospital, China
| | - Qiang Fu
- Cardiovascular Surgery (Q.F., N.W.), Tianjin Medical University General Hospital, China
| | - Naishi Wu
- Cardiovascular Surgery (Q.F., N.W.), Tianjin Medical University General Hospital, China
| | - Zhongren Ding
- School of Pharmacy (Z.D.), Tianjin Medical University, China
| | - Haipeng Sun
- National Humanities Center Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology (Z.M., H.S., L.C.), Tianjin Medical University, China
| | - Xu Zhang
- Tianjin Key Laboratory of Metabolic Diseases, Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Physiology and Pathophysiology (Z.L., J.Z., G.Z., X.H., X.Z., Y.Z., J.H.), Tianjin Medical University, China
| | - Yi Zhu
- Tianjin Key Laboratory of Metabolic Diseases, Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Physiology and Pathophysiology (Z.L., J.Z., G.Z., X.H., X.Z., Y.Z., J.H.), Tianjin Medical University, China
| | - Liming Chen
- National Humanities Center Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology (Z.M., H.S., L.C.), Tianjin Medical University, China
| | - Jinlong He
- Tianjin Key Laboratory of Metabolic Diseases, Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Physiology and Pathophysiology (Z.L., J.Z., G.Z., X.H., X.Z., Y.Z., J.H.), Tianjin Medical University, China
| |
Collapse
|
11
|
Han X, Knauss EA, de la Fuente M, Li W, Conlon RA, LePage DF, Jiang W, Renna SA, McKenzie SE, Nieman MT. A Mouse Model of the Protease Activated Receptor 4 (PAR4) Pro310Leu Variant has Reduced Platelet Reactivity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.01.569075. [PMID: 38077081 PMCID: PMC10705540 DOI: 10.1101/2023.12.01.569075] [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] [Indexed: 12/17/2023]
Abstract
Background Protease activated receptor 4 (PAR4) mediates thrombin signaling on platelets and other cells. Our recent structural studies demonstrated a single nucleotide polymorphism in extracellular loop 3 (ECL3), PAR4-P310L (rs2227376) leads to a hypo-reactive receptor. Objectives The goal of this study was to determine how the hypo-reactive PAR4 variant in ECL3 impacts platelet function in vivo using a novel knock-in mouse model (PAR4-322L). Methods A point mutation was introduced into the PAR4 gene, F2rl3, via CRISPR/Cas9 to create PAR4-P322L, the mouse homolog to human PAR4-P310L. Platelet response to PAR4 activation peptide (AYPGKF), thrombin, ADP, and convulxin was monitored by αIIbβ3 integrin activation and P-selectin translocation using flow cytometry or platelet aggregation. In vivo responses were determined by the tail bleeding assay and the ferric chloride-induced carotid artery injury model. Results PAR4-P/L and PAR4-L/L platelets had a reduced response to AYPGKF and thrombin measured by P-selectin translocation or αIIbβ3 activation. The response to ADP and convulxin was unchanged among genotypes. In addition, both PAR4-P/L and PAR4-L/L platelets showed a reduced response to thrombin in aggregation studies. There was an increase in the tail bleeding time for PAR4-L/L mice. The PAR4-P/L and PAR4-L/L mice both showed an extended time to arterial thrombosis. Conclusions PAR4-322L significantly reduced platelet responsiveness to AYPGKF and thrombin, which is in agreement with our previous structural and cell signaling studies. In addition, PAR4-322L had prolonged arterial thrombosis time. Our mouse model provides a foundation to further evaluate the role of PAR4 in other pathophysiological contexts.
Collapse
Affiliation(s)
- Xu Han
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH United States
| | - Elizabeth A. Knauss
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH United States
| | - Maria de la Fuente
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH United States
| | - Wei Li
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine at Marshall University, Huntington, WV United States
| | - Ronald A Conlon
- Case Transgenic and Targeting Facility, Case Western Reserve University, Cleveland, OH United States
| | - David F. LePage
- Case Transgenic and Targeting Facility, Case Western Reserve University, Cleveland, OH United States
| | - Weihong Jiang
- Case Transgenic and Targeting Facility, Case Western Reserve University, Cleveland, OH United States
| | - Stephanie A. Renna
- Department of Medicine, The Cardeza Foundation for Hematologic Research, Thomas Jefferson University, Philadelphia, PA United States
| | - Steven E. McKenzie
- Department of Medicine, The Cardeza Foundation for Hematologic Research, Thomas Jefferson University, Philadelphia, PA United States
| | - Marvin T. Nieman
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH United States
| |
Collapse
|
12
|
Wan Y, Wei Y, Zhang C, Liu Y, Xu L, Gu C, Yu Z, Yin J, Zhang Q, Deng W. A novel role of acellular hemoglobin in hemolytic thrombosis. Thromb Res 2023; 228:33-41. [PMID: 37267672 DOI: 10.1016/j.thromres.2023.05.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 05/15/2023] [Accepted: 05/24/2023] [Indexed: 06/04/2023]
Abstract
BACKGROUND Hemolytic thrombosis has been associated with acellular hemoglobin released from damaged red blood cells during hemolysis. However, the precise molecular mechanism underlying acellular hemoglobin-induced thrombosis remains arguable. In this study, we examined the interaction between hemoglobin and the A1 domain of von Willebrand factor (VWF), which is a critical mediator of platelet activation. METHODS Previous studies have suggested that the interaction between hemoglobin and the A1 domain of VWF enhances VWF's hemostatic activity. We employed a multidisciplinary investigation to re-examine this interaction, and identified significant differences in binding affinity between the active and inactive forms of A1. RESULTS We found that hemoglobin binds more strongly to the active A1 than the inactive form. Using hydrogen‑deuterium exchange mass spectrometry, we identified the specific residues involved in this interaction, which are located on the α1-β2 and β3-α2 loops that are typically covered by the "autoinhibitory module" in the inactive A1. This observation provides a structural explanation for the differential binding affinity between the active and inactive forms of A1. We demonstrated that the binding of hemoglobin to A1 blocks the interaction between GPIbα and VWF, and inhibits VWF-mediated thrombosis in vivo. Furthermore, we found that administration of hemoglobin led to similar levels of thrombocytopenia and microthrombosis in both wildtype and VWF-deficient mice, indicating that the mechanism underlying acellular hemoglobin-induced thrombosis is VWF-independent. CONCLUSIONS These findings challenge the previous theory that hemoglobin-induced thrombosis occurs solely through binding with VWF, and provide evidence supporting a novel role for hemoglobin in hemolytic thrombosis.
Collapse
Affiliation(s)
- Yan Wan
- Cyrus Tang Medical Institute and Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, Jiangsu Province, China
| | - Yaxuan Wei
- Cyrus Tang Medical Institute and Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, Jiangsu Province, China
| | - Canhe Zhang
- Cyrus Tang Medical Institute and Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, Jiangsu Province, China
| | - Yuanyuan Liu
- Cyrus Tang Medical Institute and Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, Jiangsu Province, China
| | - Linru Xu
- Cyrus Tang Medical Institute and Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, Jiangsu Province, China
| | - Chengyuan Gu
- The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| | - Ziqiang Yu
- The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| | - Jie Yin
- The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| | - Qing Zhang
- State Key Laboratory of Biocontrol School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Wei Deng
- Cyrus Tang Medical Institute and Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, Jiangsu Province, China.
| |
Collapse
|
13
|
He F, Hou W, Lan Y, Gao W, Zhou M, Li J, Liu S, Yang B, Zhang J. High Contrast Detection of Carotid Neothrombus with Strong Near-Infrared Absorption Selenium Nanosphere Enhanced Photoacoustic Imaging. Int J Nanomedicine 2023; 18:4043-4054. [PMID: 37520300 PMCID: PMC10377622 DOI: 10.2147/ijn.s404743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 07/04/2023] [Indexed: 08/01/2023] Open
Abstract
Background Carotid artery thrombosis is the leading cause of stroke. Since there are no apparent symptoms in the early stages of carotid atherosclerosis onset, it causes a more significant clinical diagnosis. Photoacoustic (PA) imaging provides high contrast and good depth information, which has been used for the early detection and diagnosis of many diseases. Methods We investigated thrombus formation by using 20% ferric chloride (FeCl3) in the carotid arteries of KM mice for the thrombosis model. The near-infrared selenium/polypyrrole (Se@PPy) nanomaterials are easy to synthesize and have excellent optical absorption in vivo, which can be used as PA contrast agents to obtain thrombosis information. Results In vitro experiments showed that Se@PPy nanocomposites have fulfilling PA ability in the 700 nm to 900 nm wavelength range. In the carotid atherosclerosis model, maximum PA signal enhancement up to 3.44, 4.04, and 5.07 times was observed by injection of Se@PPy nanomaterials, which helped to diagnose the severity of carotid atherosclerosis. Conclusion The superior PA signal of Se@PPy nanomaterials can identify the extent of atherosclerotic carotid lesions, demonstrating the feasibility of PA imaging technology in diagnosing carotid thrombosis lesion formation. This study demonstrates nanocomposites and PA techniques for imaging and diagnosing carotid thrombosis in vivo.
Collapse
Affiliation(s)
- Fengbing He
- Qingyuan People’s Hospital, The Sixth Affiliated Hospital of Guangzhou Medical University, School of Biomedical Engineering, Guangzhou Medical University, Guangdong, People’s Republic of China
| | - Wenzhong Hou
- Qingyuan People’s Hospital, The Sixth Affiliated Hospital of Guangzhou Medical University, School of Biomedical Engineering, Guangzhou Medical University, Guangdong, People’s Republic of China
| | - Yintao Lan
- Qingyuan People’s Hospital, The Sixth Affiliated Hospital of Guangzhou Medical University, School of Biomedical Engineering, Guangzhou Medical University, Guangdong, People’s Republic of China
- Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangdong, People’s Republic of China
| | - Weijian Gao
- Qingyuan People’s Hospital, The Sixth Affiliated Hospital of Guangzhou Medical University, School of Biomedical Engineering, Guangzhou Medical University, Guangdong, People’s Republic of China
| | - Mengyu Zhou
- Qingyuan People’s Hospital, The Sixth Affiliated Hospital of Guangzhou Medical University, School of Biomedical Engineering, Guangzhou Medical University, Guangdong, People’s Republic of China
| | - Jinghang Li
- Qingyuan People’s Hospital, The Sixth Affiliated Hospital of Guangzhou Medical University, School of Biomedical Engineering, Guangzhou Medical University, Guangdong, People’s Republic of China
| | - Shutong Liu
- Qingyuan People’s Hospital, The Sixth Affiliated Hospital of Guangzhou Medical University, School of Biomedical Engineering, Guangzhou Medical University, Guangdong, People’s Republic of China
| | - Bin Yang
- Qingyuan People’s Hospital, The Sixth Affiliated Hospital of Guangzhou Medical University, School of Biomedical Engineering, Guangzhou Medical University, Guangdong, People’s Republic of China
| | - Jian Zhang
- Qingyuan People’s Hospital, The Sixth Affiliated Hospital of Guangzhou Medical University, School of Biomedical Engineering, Guangzhou Medical University, Guangdong, People’s Republic of China
| |
Collapse
|
14
|
Zeng L, Hu S, Zeng L, Chen R, Li H, Yu J, Yang H. Animal Models of Ischemic Stroke with Different Forms of Middle Cerebral Artery Occlusion. Brain Sci 2023; 13:1007. [PMID: 37508939 PMCID: PMC10377124 DOI: 10.3390/brainsci13071007] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 06/24/2023] [Accepted: 06/26/2023] [Indexed: 07/30/2023] Open
Abstract
Ischemic stroke is a common type of stroke that significantly affects human well-being and quality of life. In order to further characterize the pathophysiology of ischemic stroke and develop new treatment strategies, ischemic stroke models with controllable and consistent response to potential clinical treatments are urgently needed. The middle cerebral artery occlusion (MCAO) model is currently the most widely used animal model of ischemic stroke. This review discusses various methods for constructing the MCAO model and compares their advantages and disadvantages in order to provide better approaches for studying ischemic stroke.
Collapse
Affiliation(s)
- Lang Zeng
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Shengqi Hu
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Lingcheng Zeng
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Rudong Chen
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Hua Li
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Jiasheng Yu
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Hongkuan Yang
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
| |
Collapse
|
15
|
Du L, Yue H, Rorabaugh BR, Li OQY, DeHart AR, Toloza‐Alvarez G, Hong L, Denvir J, Thompson E, Li W. Thymidine Phosphorylase Deficiency or Inhibition Preserves Cardiac Function in Mice With Acute Myocardial Infarction. J Am Heart Assoc 2023; 12:e028023. [PMID: 36974758 PMCID: PMC10122909 DOI: 10.1161/jaha.122.028023] [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] [Received: 08/31/2022] [Accepted: 02/22/2023] [Indexed: 03/29/2023]
Abstract
Background Ischemic cardiovascular disease is the leading cause of death worldwide. Current pharmacologic therapy has multiple limitations, and patients remain symptomatic despite maximal medical therapies. Deficiency or inhibition of thymidine phosphorylase (TYMP) in mice reduces thrombosis, suggesting that TYMP could be a novel therapeutic target for patients with acute myocardial infarction (AMI). Methods and Results A mouse AMI model was established by ligation of the left anterior descending coronary artery in C57BL/6J wild-type and TYMP-deficient (Tymp-/-) mice. Cardiac function was monitored by echocardiography or Langendorff assay. TYMP-deficient hearts had lower baseline contractility. However, cardiac function, systolic left ventricle anterior wall thickness, and diastolic wall strain were significantly greater 4 weeks after AMI compared with wild-type hearts. TYMP deficiency reduced microthrombus formation after AMI. TYMP deficiency did not affect angiogenesis in either normal or infarcted myocardium but increased arteriogenesis post-AMI. TYMP deficiency enhanced the mobilization of bone marrow stem cells and promoted mesenchymal stem cell (MSC) proliferation, migration, and resistance to inflammation and hypoxia. TYMP deficiency increased the number of larger MSCs and decreased matrix metalloproteinase-2 expression, resulting in a high homing capability. TYMP deficiency induced constitutive AKT phosphorylation in MSCs but reduced expression of genes associated with retinoid-interferon-induced mortality-19, a molecule that enhances cell death. Inhibition of TYMP with its selective inhibitor, tipiracil, phenocopied TYMP deficiency, improved post-AMI cardiac function and systolic left ventricle anterior wall thickness, attenuated diastolic stiffness, and reduced infarct size. Conclusions This study demonstrated that TYMP plays an adverse role after AMI. Targeting TYMP may be a novel therapy for patients with AMI.
Collapse
Affiliation(s)
- Lili Du
- Department of Biomedical SciencesJoan C. Edwards School of Medicine at Marshall UniversityHuntingtonWVUSA
- Department of PathophysiologyCollege of Basic Medical Science, China Medical UniversityShenyangLiaoningChina
| | - Hong Yue
- Department of Biomedical SciencesJoan C. Edwards School of Medicine at Marshall UniversityHuntingtonWVUSA
| | - Boyd R. Rorabaugh
- Department of Biomedical SciencesJoan C. Edwards School of Medicine at Marshall UniversityHuntingtonWVUSA
- Department of Pharmaceutical SciencesSchool of Pharmacy at Marshall UniversityHuntingtonWVUSA
| | - Oliver Q. Y. Li
- Department of Biomedical SciencesJoan C. Edwards School of Medicine at Marshall UniversityHuntingtonWVUSA
| | - Autumn R. DeHart
- Department of Biomedical SciencesJoan C. Edwards School of Medicine at Marshall UniversityHuntingtonWVUSA
| | - Gretel Toloza‐Alvarez
- Department of Biomedical SciencesJoan C. Edwards School of Medicine at Marshall UniversityHuntingtonWVUSA
| | - Liang Hong
- Department of Biomedical SciencesJoan C. Edwards School of Medicine at Marshall UniversityHuntingtonWVUSA
| | - James Denvir
- Department of Biomedical SciencesJoan C. Edwards School of Medicine at Marshall UniversityHuntingtonWVUSA
| | - Ellen Thompson
- Department of MedicineJoan C. Edwards School of Medicine at Marshall UniversityHuntingtonWVUSA
| | - Wei Li
- Department of Biomedical SciencesJoan C. Edwards School of Medicine at Marshall UniversityHuntingtonWVUSA
| |
Collapse
|
16
|
Fang M, Cha JH, Wang HC, Ye P, Chen B, Chen M, Yang WH, Yan X. An undefined cystatin CsCPI1 from tea plant Camellia sinensis harbors antithrombotic activity. Biomed Pharmacother 2023; 159:114285. [PMID: 36706630 DOI: 10.1016/j.biopha.2023.114285] [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: 12/06/2022] [Revised: 01/17/2023] [Accepted: 01/18/2023] [Indexed: 01/27/2023] Open
Abstract
Tea consumption has been linked to a decreased risk of cardiovascular disease (CVD) mortality, which imposes a heavy burden on the healthcare system; however, which components in tea cause this beneficial effect is not fully understood. Here we uncovered a cystatin (namely CsCPI1), which is a cysteine proteinase inhibitor (CPI) of the tea plant (Camellia sinensis) that promotes antithrombotic activity. Since thrombosis is a common pathogenesis of fatal CVDs, we investigated the effects of CsCPI1, which showed good therapeutic effects in mouse models of thrombotic disease and ischemic stroke. CsCPI1 significantly increases endothelial cell production of nitric oxide (NO) and inhibits platelet aggregation. Notably, CsCPI1 exhibited no cytotoxicity or resistance to pH and temperature changes, which indicates that CsCPI1 might be a potent antithrombotic agent that contributes to the therapeutic effects of tea consumption against CVD. Specifically, the antithrombotic effects of CsCPI1 are distinct from the classical function of plant cystatins against herbivorous insects. Therefore, our study proposes a new potential role of cystatins in CVD prevention and treatment, which requires further study.
Collapse
Affiliation(s)
- Mingqian Fang
- Affiliated Cancer Institute & Hospital and Key Laboratory for Cell Homeostasis and Cancer Research of Guangdong Higher Education Institutes, Guangzhou Medical University, Guangzhou 910095, Guangdong, China; Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences and Key Laboratory of Bioactive Peptides of Yunnan Province, Institute of Zoology, Kunming 650107, Yunnan, China
| | - Jong-Ho Cha
- Department of Biomedical Science and Engineering, Graduate School, Inha University, Incheon 22212, the Republic of Korea; Department of Biomedical Sciences, College of Medicine, Inha University, Incheon 22212, the Republic of Korea
| | - Hao-Ching Wang
- Graduate Institute of Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan
| | - Peng Ye
- Affiliated Cancer Institute & Hospital and Key Laboratory for Cell Homeostasis and Cancer Research of Guangdong Higher Education Institutes, Guangzhou Medical University, Guangzhou 910095, Guangdong, China
| | - Bi Chen
- Affiliated Cancer Institute & Hospital and Key Laboratory for Cell Homeostasis and Cancer Research of Guangdong Higher Education Institutes, Guangzhou Medical University, Guangzhou 910095, Guangdong, China
| | - Mengrou Chen
- Product Development Department, Nanjing Legend Biotech Co., Ltd., Nanjing 211100, Jiangsu, China
| | - Wen-Hao Yang
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan.
| | - Xiuwen Yan
- Affiliated Cancer Institute & Hospital and Key Laboratory for Cell Homeostasis and Cancer Research of Guangdong Higher Education Institutes, Guangzhou Medical University, Guangzhou 910095, Guangdong, China.
| |
Collapse
|
17
|
Lin X, Wang H, Huang S, Chen L, Yang S, Zhao P, Lin Z, Yang J, Ruan L, Ni H, Wang K, Wen M, Jin K, Zhuge Q. A Reliable Nonhuman Primate Model of Ischemic Stroke with Reproducible Infarct Size and Long-term Sensorimotor Deficits. Aging Dis 2023; 14:245-255. [PMID: 36818571 PMCID: PMC9937702 DOI: 10.14336/ad.2022.0722] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 07/22/2022] [Indexed: 11/18/2022] Open
Abstract
A nonhuman primate model of ischemic stroke is considered as an ideal preclinical model to replicate various aspects of human stroke because of their similarity to humans in genetics, neuroanatomy, physiology, and immunology. However, it remains challenging to produce a reliable and reproducible stroke model in nonhuman primates due to high mortality and variable outcomes. Here, we developed a focal cerebral ischemic model induced by topical application of 50% ferric chloride (FeCl3) onto the MCA-M1 segment through a cranial window in the cynomolgus monkeys. We found that FeCl3 rapidly produced a stable intraarterial thrombus that caused complete occlusion of the MCA, leading to the quick decrease of the regional cerebral blood flow in 10 min. A typical cortical infarct was detected 24 hours by magnetic resonance imaging (MRI) and was stable at least for 1 month after surgery. The sensorimotor deficit assessed by nonhuman primate stroke scale was observed at 1 day and up to 3 months after ischemic stroke. No spontaneous revascularization or autolysis of thrombus was observed, and vital signs were not affected. All operated cynomolgus monkeys survived. Our data suggested that FeCl3-induced stroke in nonhuman primates was a replicable and reliable model that is necessary for the correct prediction of the relevance of experimental therapeutic approaches in human beings.
Collapse
Affiliation(s)
- Xiao Lin
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, Wenzhou Medical University, Wenzhou, China.
| | - Hua Wang
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, Wenzhou Medical University, Wenzhou, China.
| | - Shengwei Huang
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, Wenzhou Medical University, Wenzhou, China.
| | - Lefu Chen
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, Wenzhou Medical University, Wenzhou, China.
| | - Su Yang
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, Wenzhou Medical University, Wenzhou, China.
| | - Peiqi Zhao
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, Wenzhou Medical University, Wenzhou, China.
| | - Zhongxiao Lin
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, Wenzhou Medical University, Wenzhou, China.
| | - Jianjing Yang
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, Wenzhou Medical University, Wenzhou, China.
| | - Linhui Ruan
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, Wenzhou Medical University, Wenzhou, China.
| | - Haoqi Ni
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, Wenzhou Medical University, Wenzhou, China.
| | - Kankai Wang
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, Wenzhou Medical University, Wenzhou, China.
| | - Min Wen
- Department of Neurology, Guangzhou First People's Hospital, Guangzhou, China.
| | - Kunlin Jin
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Qichuan Zhuge
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, Wenzhou Medical University, Wenzhou, China.,Correspondence should be addressed to: Dr. Qichuan Zhuge, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China. .
| |
Collapse
|
18
|
Yang X, Leng M, Yang L, Peng Y, Wang J, Wang Q, Wu K, Zou J, Wan W, Li L, Ye Y, Meng Z. Effect of Evodiamine on Collagen-Induced Platelet Activation and Thrombosis. BIOMED RESEARCH INTERNATIONAL 2022; 2022:4893859. [PMID: 35937403 PMCID: PMC9348926 DOI: 10.1155/2022/4893859] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 07/02/2022] [Accepted: 07/05/2022] [Indexed: 12/03/2022]
Abstract
Evodia rutaecarpa has multiple pharmacological effects and is widely used in the prevention and treatment of migraine, diabetes, cardiovascular disease, cancer, and other chronic diseases; however, the pharmacological effects of its active compound evodiamine (Evo) have not been thoroughly investigated. The purpose of this study was to investigate the effects of Evo on antiplatelet activation and thrombosis. We discovered that Evo effectively inhibited collagen-induced platelet activation but had no effect on platelet aggregation caused by activators such as thrombin, ADP, and U46619. Second, we found that Evo effectively inhibited the release of platelet granules induced by collagen. Finally, evodiamine inhibits the transduction of the SFKs/Syk/Akt/PLCγ2 activation pathway in platelets. According to in vivo studies, Evo significantly prolonged the mesenteric thromboembolism induced by ferric chloride and had no discernible effect on the coagulation function of mice. In conclusion, the antiplatelet and thrombotic effects of Evo discovered in this study provide an experimental basis for the investigation of the pharmacological mechanisms of Evo and the development of antiplatelet drugs.
Collapse
Affiliation(s)
- Xiaona Yang
- Laboratory of Molecular Cardiology, Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Min Leng
- Laboratory of Molecular Cardiology, Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Lihong Yang
- Laboratory of Molecular Cardiology, Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yunzhu Peng
- Laboratory of Molecular Cardiology, Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Jing Wang
- Laboratory of Molecular Cardiology, Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Qian Wang
- Laboratory of Molecular Cardiology, Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Kun Wu
- Laboratory of Molecular Cardiology, Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Junhua Zou
- Laboratory of Molecular Cardiology, Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Wen Wan
- Laboratory of Molecular Cardiology, Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Longjun Li
- Laboratory of Molecular Cardiology, Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yujia Ye
- Laboratory of Molecular Cardiology, Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Zhaohui Meng
- Laboratory of Molecular Cardiology, Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
19
|
Zhang G, Pan Y, Cheng H, Gong S, Chu Q, Chen P. Theaflavin: a natural candidate to restrain thrombosis. Food Funct 2022; 13:7572-7581. [PMID: 35815842 DOI: 10.1039/d2fo00152g] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Many clinical studies have demonstrated the beneficial effects of black tea on cardiovascular diseases. However, the antiplatelet and antithrombotic activities of theaflavin (TF-1) remain unknown. In this study, we aimed to investigate the beneficial effects of TF-1 on platelet activation and thrombosis formation both in vitro and in vivo. Firstly, the in vitro antiplatelet activity of TF-1 was analyzed using platelets isolated from human blood via aggregometry, flow cytometry, the ELISA kit, western blot and fluorescence microscopy. Subsequently, the in vivo analysis of the hemostatic state and thrombosis formation was carried out in C57BL/6 mice based on the tail bleeding time and an FeCl3-induced arterial thrombus model. The results showed that TF-1 could prominently inhibit platelet aggregation in a dose-dependent manner, and attenuate P-selectin expression, fibrinogen binding, spreading and thromboxane A2 (TxA2) formation. Western blot analysis showed that TF-1 potently inhibited spleen tyrosine kinase (Syk) and Akt (ser473/474) phosphorylation. The in vivo data further confirmed the inhibition of platelet activation by TF-1 with a prolonged arterial occlusion time (from 15.0 ± 1.1 minutes to 40.0 ± 5.4 minutes). All the results indicated that TF-1 is a powerful inhibitor of platelet activation and thrombosis formation in C57BL/6 mice, and could be developed as a novel food-based inhibitor of thrombotic disorders.
Collapse
Affiliation(s)
- Gang Zhang
- Department of Tea Science, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, China.
| | - Yani Pan
- Department of Tea Science, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, China.
| | - Hao Cheng
- Department of Tea Science, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, China.
| | - Shuying Gong
- Department of Tea Science, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, China.
| | - Qiang Chu
- Department of Tea Science, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, China.
| | - Ping Chen
- Department of Tea Science, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, China.
| |
Collapse
|
20
|
Saito MS, Zatta KC, Sathler PC, Furtado PS, C O Miguel N, Frattani FF, Berger M, Lavayen V, Pohlmann AR, Guterres SS. Therapeutic implementation in arterial thrombosis with pulmonary administration of fucoidan microparticles containing acetylsalicylic acid. Int J Pharm 2022; 622:121841. [PMID: 35623486 DOI: 10.1016/j.ijpharm.2022.121841] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 05/13/2022] [Accepted: 05/15/2022] [Indexed: 11/16/2022]
Abstract
Several antithrombotic drugs are available to treat cardiovascular diseases due to its high mortality and morbidity worldwide. Despite these, severe adverse effects that can lead to treatment withdrawal have been described, highlighting the importance of new therapies. Thus, this work describes the development of fucoidan microparticles containing acetylsalicylic acid (MP/F4M) for pulmonary delivery and in vitro, ex vivo, and in vivo evaluation. Microparticles were prepared via spray-drying and characterized in vitro (mucoadhesive properties, coagulation time, platelet aggregation, adhesion, and hemolysis) followed by ex vivo platelet aggregation, in vivo arterial thrombosis, and hemorrhagic profile. The formulation physicochemical characterization showed suitable characteristics along with delayed drug release, increased breathable particle fraction, and high washability resistance as well as antiplatelet activity and enhanced platelet adhesion in vitro. In in vivo assays, MP/F4M protected against arterial thrombosis, without changes in the hemorrhagic profile. Finally, no lung changes were observed after prolonged pulmonary administration, whereas isolated ASA led to an inflammatory response. In conclusion, pulmonary administration of fucoidan microparticles with an antiplatelet drug may be an alternative therapy to treat cardiovascular diseases, opening the field for different formulations.
Collapse
Affiliation(s)
- Max S Saito
- Postgraduate Program in Pharmaceutical Sciences, Federal University of Rio Grande do Sul, RS, Brazil.
| | - Kelly C Zatta
- Postgraduate Program in Pharmaceutical Sciences, Federal University of Rio Grande do Sul, RS, Brazil
| | - Plínio C Sathler
- Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Federal University of Rio de Janeiro
| | - Priscila S Furtado
- Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Federal University of Rio de Janeiro
| | - Nádia C O Miguel
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Flávia F Frattani
- Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Federal University of Rio de Janeiro
| | - Markus Berger
- Laboratory of Biochemical Pharmacology, Experimental Research Center, Hospital de Clinicas de Porto Alegre, Federal University of Rio Grande do Sul
| | - Vladimir Lavayen
- Postgraduate Program in Chemistry, Federal University of Rio Grande do Sul, RS, Brazil
| | - Adriana R Pohlmann
- Postgraduate Program in Pharmaceutical Sciences, Federal University of Rio Grande do Sul, RS, Brazil
| | - Sílvia S Guterres
- Postgraduate Program in Pharmaceutical Sciences, Federal University of Rio Grande do Sul, RS, Brazil
| |
Collapse
|
21
|
Memory-like response in platelet attenuates platelet hyperactivation in arterial thrombosis. Biochem Biophys Res Commun 2022; 612:154-161. [DOI: 10.1016/j.bbrc.2022.04.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 04/03/2022] [Accepted: 04/06/2022] [Indexed: 11/18/2022]
|
22
|
Diindolylmethane ameliorates platelet aggregation and thrombosis: In silico, in vitro, and in vivo studies. Eur J Pharmacol 2022; 919:174812. [DOI: 10.1016/j.ejphar.2022.174812] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 01/21/2022] [Accepted: 02/08/2022] [Indexed: 01/01/2023]
|
23
|
Erythrocyte Fraction in Thrombi Is Increased with Serum Iron by Influencing Fibrin Networks via Oxidative Stress. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2021:3673313. [PMID: 34976298 PMCID: PMC8719990 DOI: 10.1155/2021/3673313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 11/12/2021] [Accepted: 12/04/2021] [Indexed: 11/18/2022]
Abstract
Objective This study investigated whether the erythrocyte fraction in thrombi would be increased with serum iron via oxidative stress. Methods This study retrospectively enrolled patients with acute ischemic stroke treated using endovascular treatment in a single stroke center from October to December 2019. We examined the relationship between serum iron and erythrocyte-rich thrombi and the correlation of serum iron and the erythrocyte fraction in thrombi using clinical samples. Experiments in vivo and in vitro were performed to investigate the influence of oxidative stress on the correlation between serum iron concentration and erythrocyte fraction in thrombi. Results We found from the clinical samples that serum iron concentration was related to erythrocyte-rich thrombi and positively associated with the erythrocyte fraction in thrombi in vivo. Further, the tightness of the fibrin networks regulating the erythrocyte fraction in thrombi was increased with serum iron concentration in vivo. Additionally, the oxidative stress level was increased with serum iron concentration in vivo. Moreover, we found that the tightness of the fibrin networks increased with higher oxidative stress levels in vitro. Lastly, experiments in vivo with inhibiting oxidative stress showed that the erythrocyte fraction in thrombi and the tightness of fibrin networks significantly increased in the iron group than those in the iron with oxidative stress inhibitor group and control group. Conclusions Oxidative stress played a role in the process that the erythrocyte fraction in thrombi was increased with serum iron by influencing fibrin networks.
Collapse
|
24
|
Pan N, Li ZC, Li ZH, Chen SH, Jiang MH, Yang HY, Liu YS, Hu R, Zeng YW, Dai LH, Liu L, Wang GL. Antiplatelet and Antithrombotic Effects of Isaridin E Isolated from the Marine-Derived Fungus via Downregulating the PI3K/Akt Signaling Pathway. Mar Drugs 2021; 20:23. [PMID: 35049878 PMCID: PMC8780978 DOI: 10.3390/md20010023] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/21/2021] [Accepted: 12/21/2021] [Indexed: 02/07/2023] Open
Abstract
Isaridin E, a cyclodepsipeptide isolated from the marine-derived fungus Amphichorda felina (syn. Beauveria felina) SYSU-MS7908, has been demonstrated to possess anti-inflammatory and insecticidal activities. Here, we first found that isaridin E concentration-dependently inhibited ADP-induced platelet aggregation, activation, and secretion in vitro, but did not affect collagen- or thrombin-induced platelet aggregation. Furthermore, isaridin E dose-dependently reduced thrombosis formation in an FeCl3-induced mouse carotid model without increasing the bleeding time. Mechanistically, isaridin E significantly decreased the ADP-mediated phosphorylation of PI3K and Akt. In conclusion, these results suggest that isaridin E exerts potent antithrombotic effects in vivo without increasing the risk of bleeding, which may be due to its important role in inhibiting ADP-induced platelet activation, secretion and aggregation via the PI3K/Akt pathways.
Collapse
Affiliation(s)
- Ni Pan
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; (N.P.); (Z.-C.L.); (Z.-H.L.); (H.-Y.Y.); (Y.-S.L.); (R.H.); (Y.-W.Z.)
- Institute of Pediatrics, Guangzhou Women and Children’s Medical Centre, Guangzhou Medical University, Guangzhou 510080, China
| | - Zi-Cheng Li
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; (N.P.); (Z.-C.L.); (Z.-H.L.); (H.-Y.Y.); (Y.-S.L.); (R.H.); (Y.-W.Z.)
| | - Zhi-Hong Li
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; (N.P.); (Z.-C.L.); (Z.-H.L.); (H.-Y.Y.); (Y.-S.L.); (R.H.); (Y.-W.Z.)
| | - Sen-Hua Chen
- School of Marine Sciences, Sun Yat-sen University, Guangzhou 510080, China; (S.-H.C.); (M.-H.J.)
| | - Ming-Hua Jiang
- School of Marine Sciences, Sun Yat-sen University, Guangzhou 510080, China; (S.-H.C.); (M.-H.J.)
| | - Han-Yan Yang
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; (N.P.); (Z.-C.L.); (Z.-H.L.); (H.-Y.Y.); (Y.-S.L.); (R.H.); (Y.-W.Z.)
| | - Yao-Sheng Liu
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; (N.P.); (Z.-C.L.); (Z.-H.L.); (H.-Y.Y.); (Y.-S.L.); (R.H.); (Y.-W.Z.)
| | - Rui Hu
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; (N.P.); (Z.-C.L.); (Z.-H.L.); (H.-Y.Y.); (Y.-S.L.); (R.H.); (Y.-W.Z.)
| | - Yu-Wei Zeng
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; (N.P.); (Z.-C.L.); (Z.-H.L.); (H.-Y.Y.); (Y.-S.L.); (R.H.); (Y.-W.Z.)
| | - Le-Hui Dai
- Department of Basic Medical Sciences, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China;
| | - Lan Liu
- School of Marine Sciences, Sun Yat-sen University, Guangzhou 510080, China; (S.-H.C.); (M.-H.J.)
| | - Guan-Lei Wang
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; (N.P.); (Z.-C.L.); (Z.-H.L.); (H.-Y.Y.); (Y.-S.L.); (R.H.); (Y.-W.Z.)
| |
Collapse
|
25
|
Sharma C, Osmolovskiy A, Singh R. Microbial Fibrinolytic Enzymes as Anti-Thrombotics: Production, Characterisation and Prodigious Biopharmaceutical Applications. Pharmaceutics 2021; 13:1880. [PMID: 34834294 PMCID: PMC8625737 DOI: 10.3390/pharmaceutics13111880] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 10/23/2021] [Accepted: 10/29/2021] [Indexed: 12/19/2022] Open
Abstract
Cardiac disorders such as acute myocardial infarction, embolism and stroke are primarily attributed to excessive fibrin accumulation in the blood vessels, usually consequential in thrombosis. Numerous methodologies including the use of anti-coagulants, anti-platelet drugs, surgical operations and fibrinolytic enzymes are employed for the dissolution of fibrin clots and hence ameliorate thrombosis. Microbial fibrinolytic enzymes have attracted much more attention in the management of cardiovascular disorders than typical anti-thrombotic strategies because of the undesirable after-effects and high expense of the latter. Fibrinolytic enzymes such as plasminogen activators and plasmin-like proteins hydrolyse thrombi with high efficacy with no significant after-effects and can be cost effectively produced on a large scale with a short generation time. However, the hunt for novel fibrinolytic enzymes necessitates complex purification stages, physiochemical and structural-functional attributes, which provide an insight into their mechanism of action. Besides, strain improvement and molecular technologies such as cloning, overexpression and the construction of genetically modified strains for the enhanced production of fibrinolytic enzymes significantly improve their thrombolytic potential. In addition, the unconventional applicability of some fibrinolytic enzymes paves their way for protein hydrolysis in addition to fibrin/thrombi, blood pressure regulation, anti-microbials, detergent additives for blood stain removal, preventing dental caries, anti-inflammatory and mucolytic expectorant agents. Therefore, this review article encompasses the production, biochemical/structure-function properties, thrombolytic potential and other surplus applications of microbial fibrinolytic enzymes.
Collapse
Affiliation(s)
- Chhavi Sharma
- Amity Institute of Microbial Technology, Amity University Uttar Pradesh, Noida 201313, India;
| | - Alexander Osmolovskiy
- Department of Microbiology, Faculty of Biology, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Rajni Singh
- Amity Institute of Microbial Technology, Amity University Uttar Pradesh, Noida 201313, India;
| |
Collapse
|
26
|
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: 2.7] [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
|
27
|
Annarapu GK, Nolfi-Donegan D, Reynolds M, Wang Y, Shiva S. Mitochondrial reactive oxygen species scavenging attenuates thrombus formation in a murine model of sickle cell disease. J Thromb Haemost 2021; 19:2256-2262. [PMID: 33724688 DOI: 10.1111/jth.15298] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 03/10/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND Sickle cell disease (SCD) is characterized by hemolysis-associated platelet dysfunction that leads to increased risk of thrombosis and plays a role in the high morbidity and mortality of the disease. The mechanisms by which hemolysis induces platelet activation remain unclear. We recently demonstrated that patients with SCD showed increased platelet mitochondrial reactive oxygen species (mtROS) production that correlates with markers of hemolysis and platelet activation. Experiments in isolated platelets demonstrated that mtROS stimulated platelet activation. However, the role of hemolysis-induced mtROS in thrombus formation in vivo remains unclear. OBJECTIVES Here, we hypothesize that scavenging of mtROS attenuates the propensity for thrombosis in mouse models of hemolysis. METHODS We used models of hemolysate infusion into wildtype mice as well as the Berkley transgenic mouse model of SCD, a chronic mode of hemolysis, to test the effect of hemolysis on platelet mtROS production and thrombosis. RESULTS We show that infusion of hemolysate in wildtype mice induces platelet mtROS production and decreases time to vessel occlusion in a model of ferric chloride-induced carotid artery thrombosis. Increased mtROS and propensity for thrombosis was also observed in the Berkley transgenic mouse model of SCD. Notably, treatment with mtROS scavengers decreased platelet mtROS levels and attenuated the propensity for thrombus formation in both models. CONCLUSIONS These data demonstrate that mtROS significantly contribute to the mechanism of hemolysis-induced thrombosis in vivo and suggest a potential role for mitochondrially targeted antioxidant therapy in hemolysis and SCD-related thrombosis.
Collapse
Affiliation(s)
- Gowtham K Annarapu
- Heart, Lung, Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Deirdre Nolfi-Donegan
- Heart, Lung, Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Pediatrics, Division of Hematology/Oncology, Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Michael Reynolds
- Heart, Lung, Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Yinna Wang
- Heart, Lung, Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Sruti Shiva
- Heart, Lung, Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
28
|
Sun B, Hettie KS, Zhu S. Near-infrared Fluorophores for Thrombosis Diagnosis and Therapy. ADVANCED THERAPEUTICS 2021; 4:2000278. [PMID: 33997270 PMCID: PMC8115206 DOI: 10.1002/adtp.202000278] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Indexed: 12/23/2022]
Abstract
Thrombosis is an adverse physiological event wherein the resulting thrombus and thrombus-induced diseases collectively result in high morbidity and mortality rates. Currently, nano-medicines that incorporate fluorophores emitting in the near-infrared-I (NIR-I, 700-900 nm) spectral region into their systems have been adopted to afford thrombosis theranostics. However, several unsolved problems such as limited penetration depth and image quality severely impede further applications of such nano-medicine systems. Fortunately, the ability to incorporate fluorophores emitting in the NIR-II (1000-1700 nm) window into nano-medicine systems can unambiguously identify biological processes with high signal-to-noise, deep tissue penetration depth, and high image resolution. Considering the inherently favorable properties of NIR-II fluorophores, we believe such have enormous potential to quickly become incorporated into nano-medicine systems for thrombosis theranostics. In this review, we i) discuss the development of NIR fluorescence as an imaging modality and fluorescent agents; ii) comprehensively summarize the recent development of NIR-I fluorophore-based nano-medicine systems for thrombosis theranostics; iii) highlight the state-of-the-art NIR-II fluorophores that have been designed for the specific purpose of affording thrombotic diagnosis; iv) speculate on possible forward avenues for the use of NIR-II fluorophores towards thrombosis diagnosis and therapy; and v) discuss the potential for their clinical translation.
Collapse
Affiliation(s)
- Bin Sun
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, The First Hospital of Jilin University, Changchun, 130061, P.R. China; State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P.R. China
| | - Kenneth S Hettie
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Shoujun Zhu
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, The First Hospital of Jilin University, Changchun, 130061, P.R. China; State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P.R. China
| |
Collapse
|
29
|
Bocquet O, Wahart A, Sarazin T, Vincent E, Schneider C, Fougerat A, Gayral S, Henry A, Blaise S, Romier-Crouzet B, Boulagnon C, Jaisson S, Gillery P, Bennasroune A, Sartelet H, Laffargue M, Martiny L, Duca L, Maurice P. Adverse Effects of Oseltamivir Phosphate Therapy on the Liver of LDLR-/- Mice Without Any Benefit on Atherosclerosis and Thrombosis. J Cardiovasc Pharmacol 2021; 77:660-672. [PMID: 33760798 DOI: 10.1097/fjc.0000000000001002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 02/10/2021] [Indexed: 02/06/2023]
Abstract
ABSTRACT Desialylation, governed by sialidases or neuraminidases, is strongly implicated in a wide range of human disorders, and accumulative data show that inhibition of neuraminidases, such as neuraminidases 1 sialidase, may be useful for managing atherosclerosis. Several studies have reported promising effects of oseltamivir phosphate, a widely used anti-influenza sialidase inhibitor, on human cancer cells, inflammation, and insulin resistance. In this study, we evaluated the effects of oseltamivir phosphate on atherosclerosis and thrombosis and potential liver toxicity in LDLR-/- mice fed with high-fat diet. Our results showed that oseltamivir phosphate significantly decreased plasma levels of LDL cholesterol and elastin fragmentation in aorta. However, no effect was observed on both atherosclerotic plaque size in aortic roots and chemically induced thrombosis in carotid arteries. Importantly, oseltamivir phosphate administration had adverse effects on the liver of mice and significantly increased messenger RNA expression levels of F4/80, interleukin-1β, transforming growth factor-β1, matrix metalloproteinase-12, and collagen. Taken together, our findings suggest that oseltamivir phosphate has limited benefits on atherosclerosis and carotid thrombosis and may lead to adverse side effects on the liver with increased inflammation and fibrosis.
Collapse
Affiliation(s)
- Olivier Bocquet
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes et Naturelles, Reims, France
| | - Amandine Wahart
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes et Naturelles, Reims, France
| | - Thomas Sarazin
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes et Naturelles, Reims, France
| | - Elise Vincent
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes et Naturelles, Reims, France
| | - Christophe Schneider
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes et Naturelles, Reims, France
| | - Anne Fougerat
- INSERM UMR1048 I2MC, Université Paul Sabatier, Toulouse, France
| | | | - Aubéri Henry
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes et Naturelles, Reims, France
| | - Sébastien Blaise
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes et Naturelles, Reims, France
| | - Béatrice Romier-Crouzet
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes et Naturelles, Reims, France
| | - Camille Boulagnon
- Laboratoire d'anatomie et de Cytologie Pathologique, Hôpital Robert Debré, CHU de Reims; and
| | - Stéphane Jaisson
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes et Naturelles, Reims, France
- Department of Biochemistry-Pharmacology-Toxicology, University Hospital of Reims, Reims, France
| | - Philippe Gillery
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes et Naturelles, Reims, France
- Department of Biochemistry-Pharmacology-Toxicology, University Hospital of Reims, Reims, France
| | - Amar Bennasroune
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes et Naturelles, Reims, France
| | - Hervé Sartelet
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes et Naturelles, Reims, France
| | | | - Laurent Martiny
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes et Naturelles, Reims, France
| | - Laurent Duca
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes et Naturelles, Reims, France
| | - Pascal Maurice
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes et Naturelles, Reims, France
| |
Collapse
|
30
|
Njoroge W, Hernández ACH, Musa FI, Butler R, Harper AGS, Yang Y. The Combination of Tissue-Engineered Blood Vessel Constructs and Parallel Flow Chamber Provides a Potential Alternative to In Vivo Drug Testing Models. Pharmaceutics 2021; 13:pharmaceutics13030340. [PMID: 33807995 PMCID: PMC7998107 DOI: 10.3390/pharmaceutics13030340] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/22/2021] [Accepted: 02/27/2021] [Indexed: 11/16/2022] Open
Abstract
Cardiovascular disease is a major cause of death globally. This has led to significant efforts to develop new anti-thrombotic therapies or re-purpose existing drugs to treat cardiovascular diseases. Due to difficulties of obtaining healthy human blood vessel tissues to recreate in vivo conditions, pre-clinical testing of these drugs currently requires significant use of animal experimentation, however, the successful translation of drugs from animal tests to use in humans is poor. Developing humanised drug test models that better replicate the human vasculature will help to develop anti-thrombotic therapies more rapidly. Tissue-engineered human blood vessel (TEBV) models were fabricated with biomimetic matrix and cellular components. The pro- and anti-aggregatory properties of both intact and FeCl3-injured TEBVs were assessed under physiological flow conditions using a modified parallel-plate flow chamber. These were perfused with fluorescently labelled human platelets and endothelial progenitor cells (EPCs), and their responses were monitored in real-time using fluorescent imaging. An endothelium-free TEBV exhibited the capacity to trigger platelet activation and aggregation in a shear stress-dependent manner, similar to the responses observed in vivo. Ketamine is commonly used as an anaesthetic in current in vivo models, but this drug significantly inhibited platelet aggregation on the injured TEBV. Atorvastatin was also shown to enhance EPC attachment on the injured TEBV. The TEBV, when perfused with human blood or blood components under physiological conditions, provides a powerful alternative to current in vivo drug testing models to assess their effects on thrombus formation and EPC recruitment.
Collapse
Affiliation(s)
- Wanjiku Njoroge
- School of Pharmacy and Bioengineering, Keele University, Stoke-on-Trent ST4 7QB, UK; (W.N.); (A.C.H.H.); (F.I.M.)
| | | | - Faiza Idris Musa
- School of Pharmacy and Bioengineering, Keele University, Stoke-on-Trent ST4 7QB, UK; (W.N.); (A.C.H.H.); (F.I.M.)
| | - Robert Butler
- Department of Cardiology, Royal Stoke Hospital, Stoke-on-Trent ST4 6QG, UK;
| | - Alan G. S. Harper
- School of Medicine, Keele University, Staffs ST5 5BG, UK
- Correspondence: (A.G.S.H.); (Y.Y.); Tel.: +44-17-8273-4654 (A.G.S.H.); +44-17-8267-4386 (Y.Y.)
| | - Ying Yang
- School of Pharmacy and Bioengineering, Keele University, Stoke-on-Trent ST4 7QB, UK; (W.N.); (A.C.H.H.); (F.I.M.)
- Correspondence: (A.G.S.H.); (Y.Y.); Tel.: +44-17-8273-4654 (A.G.S.H.); +44-17-8267-4386 (Y.Y.)
| |
Collapse
|
31
|
Belcher A, Zulfiker AHM, Li OQ, Yue H, Gupta AS, Li W. Targeting Thymidine Phosphorylase With Tipiracil Hydrochloride Attenuates Thrombosis Without Increasing Risk of Bleeding in Mice. Arterioscler Thromb Vasc Biol 2021; 41:668-682. [PMID: 33297751 PMCID: PMC8105268 DOI: 10.1161/atvbaha.120.315109] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Current antiplatelet medications increase the risk of bleeding, which leads to a clear clinical need in developing novel mechanism-based antiplatelet drugs. TYMP (Thymidine phosphorylase), a cytoplasm protein that is highly expressed in platelets, facilitates multiple agonist-induced platelet activation, and enhances thrombosis. Tipiracil hydrochloride (TPI), a selective TYMP inhibitor, has been approved by the Food and Drug Administration for clinical use. We tested the hypothesis that TPI is a safe antithrombotic medication. Approach and Results: By coexpression of TYMP and Lyn, GST (glutathione S-transferase) tagged Lyn-SH3 domain or Lyn-SH2 domain, we showed the direct evidence that TYMP binds to Lyn through both SH3 and SH2 domains, and TPI diminished the binding. TYMP deficiency significantly inhibits thrombosis in vivo in both sexes. Pretreatment of platelets with TPI rapidly inhibited collagen- and ADP-induced platelet aggregation. Under either normal or hyperlipidemic conditions, treating wild-type mice with TPI via intraperitoneal injection, intravenous injection, or gavage feeding dramatically inhibited thrombosis without inducing significant bleeding. Even at high doses, TPI has a lower bleeding side effect compared with aspirin and clopidogrel. Intravenous delivery of TPI alone or combined with tissue plasminogen activator dramatically inhibited thrombosis. Dual administration of a very low dose of aspirin and TPI, which had no antithrombotic effects when used alone, significantly inhibited thrombosis without disturbing hemostasis. CONCLUSIONS This study demonstrated that inhibition of TYMP, a cytoplasmic protein, attenuated multiple signaling pathways that mediate platelet activation, aggregation, and thrombosis. TPI can be used as a novel antithrombotic medication without the increase in risk of bleeding.
Collapse
Affiliation(s)
- Adam Belcher
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine of Marshall University, Huntington, WV, 25755, USA
| | - Abu Hasanat Md Zulfiker
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine of Marshall University, Huntington, WV, 25755, USA
| | - Oliver Qiyue Li
- Marshall Institute for Interdisciplinary Research; Huntington, WV, 25701, USA
| | - Hong Yue
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine of Marshall University, Huntington, WV, 25755, USA
| | - Anirban Sen Gupta
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland OH 44106, USA
| | - Wei Li
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine of Marshall University, Huntington, WV, 25755, USA
| |
Collapse
|
32
|
Bouck EG, de la Fuente M, Zunica ER, Li W, Mumaw MM, Nieman MT. Murine cadherin-6 mediates thrombosis in vivo in a platelet-independent manner. Res Pract Thromb Haemost 2021; 5:125-131. [PMID: 33537536 PMCID: PMC7845066 DOI: 10.1002/rth2.12458] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 10/26/2020] [Indexed: 11/07/2022] Open
Abstract
BACKGROUND Platelet adhesion is the critical process mediating stable thrombus formation. Previous reports of cadherin-6 on human platelets have demonstrated its role in platelet aggregation and thrombus formation. OBJECTIVES We aimed to further characterize the importance of cadherin-6 in thrombosis in vivo. METHODS Cadherin-6 platelet expression was evaluated by western blotting, flow cytometry, and immunoprecipitation. Thrombosis was evaluated using the FeCl3 and Rose Bengal carotid artery models in C57Bl6 mice treated with anti-cadherin-6 or IgG and wild-type or Cdh6-/- mice. Platelet function was compared in wild-type and Cdh6-/- mice using tail-clip assays, aggregometry, and flow cytometry. RESULTS Human platelet expression of cadherin-6 was confirmed at ~3000 copies per platelet. Cdh6-/- mice or those treated with anti-cadherin-6 antibody showed an increased time to occlusion in both thrombosis models. Cadherin-6 was not expressed on mouse platelets, and there were no differences in tail bleeding times, platelet aggregation, or platelet activation in wild-type versus Cdh6-/- mice. CONCLUSIONS Cadherin-6 plays an essential role in thrombosis in vivo. However, cadherin-6 is not expressed on murine platelets. These data are in contrast to human platelets, which express a functional cadherin-6/catenin complex. The essential, platelet-independent role for cadherin-6 in hemostasis may allow it to be an effective and safe therapeutic target.
Collapse
Affiliation(s)
- Emma G. Bouck
- Department of PharmacologyCase Western Reserve UniversityClevelandOHUSA
| | | | | | - Wei Li
- Deparmtent of Biomedical SciencesMarshall University Joan C. Edwards School of MedicineHuntingtonWVUSA
| | - Michele M. Mumaw
- Department of PharmacologyCase Western Reserve UniversityClevelandOHUSA
| | - Marvin T. Nieman
- Department of PharmacologyCase Western Reserve UniversityClevelandOHUSA
| |
Collapse
|
33
|
Jeanne A, Sarazin T, Charlé M, Kawecki C, Kauskot A, Hedtke T, Schmelzer CEH, Martiny L, Maurice P, Dedieu S. Towards the Therapeutic Use of Thrombospondin 1/CD47 Targeting TAX2 Peptide as an Antithrombotic Agent. Arterioscler Thromb Vasc Biol 2021; 41:e1-e17. [PMID: 33232198 DOI: 10.1161/atvbaha.120.314571] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OBJECTIVE TSP-1 (thrombospondin 1) is one of the most expressed proteins in platelet α-granules and plays an important role in the regulation of hemostasis and thrombosis. Interaction of released TSP-1 with CD47 membrane receptor has been shown to regulate major events leading to thrombus formation, such as, platelet adhesion to vascular endothelium, nitric oxide/cGMP (cyclic guanosine monophosphate) signaling, platelet activation as well as aggregation. Therefore, targeting TSP-1:CD47 axis may represent a promising antithrombotic strategy. Approach and Results: A CD47-derived cyclic peptide was engineered, namely TAX2, that targets TSP-1 and selectively prevents TSP-1:CD47 interaction. Here, we demonstrate for the first time that TAX2 peptide strongly decreases platelet aggregation and interaction with collagen under arterial shear conditions. TAX2 also delays time for complete thrombotic occlusion in 2 mouse models of arterial thrombosis following chemical injury, while Thbs1-/- mice recapitulate TAX2 effects. Importantly, TAX2 administration is not associated with increased bleeding risk or modification of hematologic parameters. CONCLUSIONS Overall, this study sheds light on the major contribution of TSP-1:CD47 interaction in platelet activation and thrombus formation while putting forward TAX2 as an innovative antithrombotic agent with high added-value.
Collapse
Affiliation(s)
- Albin Jeanne
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes et Naturelles, Reims, France (A.J., T.S., M.C., C.K., L.M., P.M., S.D.)
- SATT Nord, Lille, France (A.J.)
- Apmonia Therapeutics, Reims, France (A.J., S.D.)
| | - Thomas Sarazin
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes et Naturelles, Reims, France (A.J., T.S., M.C., C.K., L.M., P.M., S.D.)
| | - Magalie Charlé
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes et Naturelles, Reims, France (A.J., T.S., M.C., C.K., L.M., P.M., S.D.)
| | - Charlotte Kawecki
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes et Naturelles, Reims, France (A.J., T.S., M.C., C.K., L.M., P.M., S.D.)
| | - Alexandre Kauskot
- HITh, UMR_S 1176, INSERM Univ. Paris-Sud, Université Paris-Saclay, France (A.K.)
| | - Tobias Hedtke
- Fraunhofer Institute for Microstructure of Materials and Systems IMWS, Halle (Saale), Germany (T.H., C.E.H.S.)
| | - Christian E H Schmelzer
- Fraunhofer Institute for Microstructure of Materials and Systems IMWS, Halle (Saale), Germany (T.H., C.E.H.S.)
| | - Laurent Martiny
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes et Naturelles, Reims, France (A.J., T.S., M.C., C.K., L.M., P.M., S.D.)
| | - Pascal Maurice
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes et Naturelles, Reims, France (A.J., T.S., M.C., C.K., L.M., P.M., S.D.)
| | - Stéphane Dedieu
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes et Naturelles, Reims, France (A.J., T.S., M.C., C.K., L.M., P.M., S.D.)
- Apmonia Therapeutics, Reims, France (A.J., S.D.)
| |
Collapse
|
34
|
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: 55] [Impact Index Per Article: 13.8] [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
|
35
|
GPR56/ADGRG1 is a platelet collagen-responsive GPCR and hemostatic sensor of shear force. Proc Natl Acad Sci U S A 2020; 117:28275-28286. [PMID: 33097663 PMCID: PMC7668045 DOI: 10.1073/pnas.2008921117] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
We identified the known collagen receptor GPR56/ADGRG1 on platelets. GPR56 is an adhesion G protein-coupled receptor that becomes activated following forced dissociation of its N-terminal fragment and C-terminal fragment or seven-transmembrane spanning domain (7TM). Fragment dissociation reveals the cryptic stalk of the 7TM, which acts as a tethered peptide agonist, and for GPR56, this activates platelet G13 signaling. GPR56 pharmacological probes activated platelets to undergo shape change and aggregation, which are critical for the formation of hemostatic plugs. Gpr56−/− mice exhibit prolonged bleeding, defective platelet plug formation in vessel injury assays, and delayed thrombotic vessel occlusion. Shear-force dependency of platelet adhesion to immobilized collagen was found to be GPR56 dependent. Circulating platelets roll along exposed collagen at vessel injury sites and respond with filipodia protrusion, shape change, and surface area expansion to facilitate platelet adhesion and plug formation. Various glycoproteins were considered to be both collagen responders and mediators of platelet adhesion, yet the signaling kinetics emanating from these receptors do not fully account for the rapid platelet cytoskeletal changes that occur in blood flow. We found the free N-terminal fragment of the adhesion G protein-coupled receptor (GPCR) GPR56 in human plasma and report that GPR56 is the platelet receptor that transduces signals from collagen and blood flow-induced shear force to activate G protein 13 signaling for platelet shape change. Gpr56−/− mice have prolonged bleeding, defective platelet plug formation, and delayed thrombotic occlusion. Human and mouse blood perfusion studies demonstrated GPR56 and shear-force dependence of platelet adhesion to immobilized collagen. Our work places GPR56 as an initial collagen responder and shear-force transducer that is essential for platelet shape change during hemostasis.
Collapse
|
36
|
Yang M, Li W, Harberg C, Chen W, Yue H, Ferreira RB, Wynia-Smith SL, Carroll KS, Zielonka J, Flaumenhaft R, Silverstein RL, Smith BC. Cysteine sulfenylation by CD36 signaling promotes arterial thrombosis in dyslipidemia. Blood Adv 2020; 4:4494-4507. [PMID: 32946569 PMCID: PMC7509873 DOI: 10.1182/bloodadvances.2020001609] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 08/03/2020] [Indexed: 12/20/2022] Open
Abstract
Arterial thrombosis in the setting of dyslipidemia promotes clinically significant events, including myocardial infarction and stroke. Oxidized lipids in low-density lipoproteins (oxLDL) are a risk factor for athero-thrombosis and are recognized by platelet scavenger receptor CD36. oxLDL binding to CD36 promotes platelet activation and thrombosis by promoting generation of reactive oxygen species. The downstream signaling events initiated by reactive oxygen species in this setting are poorly understood. In this study, we report that CD36 signaling promotes hydrogen peroxide flux in platelets. Using carbon nucleophiles that selectively and covalently modify cysteine sulfenic acids, we found that hydrogen peroxide generated through CD36 signaling promotes cysteine sulfenylation of platelet proteins. Specifically, cysteines were sulfenylated on Src family kinases, which are signaling transducers that are recruited to CD36 upon recognition of its ligands. Cysteine sulfenylation promoted activation of Src family kinases and was prevented by using a blocking antibody to CD36 or by enzymatic degradation of hydrogen peroxide. CD36-mediated platelet aggregation and procoagulant phosphatidylserine externalization were inhibited in a concentration-dependent manner by a panel of sulfenic acid-selective carbon nucleophiles. At the same concentrations, these probes did not inhibit platelet aggregation induced by the purinergic receptor agonist adenosine diphosphate or the collagen receptor glycoprotein VI agonist collagen-related peptide. Selective modification of cysteine sulfenylation in vivo with a benzothiazine-based nucleophile rescued the enhanced arterial thrombosis seen in dyslipidemic mice back to control levels. These findings suggest that CD36 signaling generates hydrogen peroxide to oxidize cysteines within platelet proteins, including Src family kinases, and lowers the threshold for platelet activation in dyslipidemia.
Collapse
Affiliation(s)
- Moua Yang
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI
- Blood Research Institute, Versiti Blood Center of Wisconsin, Milwaukee, WI
- Division of Hemostasis and Thrombosis, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA
| | - Wei Li
- Department of Biomedical Sciences, Marshall University Joan C. Edwards School of Medicine, Huntington, WV
| | - Calvin Harberg
- Medical School, Medical College of Wisconsin, Milwaukee, WI
| | - Wenjing Chen
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI
| | - Hong Yue
- Department of Biomedical Sciences, Marshall University Joan C. Edwards School of Medicine, Huntington, WV
| | - Renan B Ferreira
- Department of Chemistry, Scripps Research Institute, Jupiter, FL; and
| | | | - Kate S Carroll
- Department of Chemistry, Scripps Research Institute, Jupiter, FL; and
| | | | - Robert Flaumenhaft
- Division of Hemostasis and Thrombosis, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA
| | - Roy L Silverstein
- Blood Research Institute, Versiti Blood Center of Wisconsin, Milwaukee, WI
- Department of Medicine, and
| | - Brian C Smith
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI
- Program in Chemical Biology, Medical College of Wisconsin, Milwaukee, WI
| |
Collapse
|
37
|
Ortillon J, Hézard N, Belmokhtar K, Kawecki C, Terryn C, Fritz G, Kauskot A, Schmidt AM, Rieu P, Nguyen P, Maurice P, Touré F. Receptor for Advanced Glycation End Products is Involved in Platelet Hyperactivation and Arterial Thrombosis during Chronic Kidney Disease. Thromb Haemost 2020; 120:1300-1312. [PMID: 32726852 DOI: 10.1055/s-0040-1714101] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Chronic kidney disease (CKD) is associated with a high cardiovascular mortality due to increased rates of vascular lesions and thrombotic events, as well as serum accumulation of uremic toxins. A subgroup of these toxins (advanced glycation end products [AGEs] and S100 proteins) can interact with the receptor for AGEs (RAGE). In this study, we analyzed the impact of CKD on platelet function and arterial thrombosis, and the potential role of RAGE in this process. METHODS Twelve weeks after induction of CKD in mice, platelet function and time to complete carotid artery occlusion were analyzed in four groups of animals (sham-operated, CKD, apolipoprotein E [Apoe]-/-, and Apoe-/-/Ager-/- mice). RESULTS Analysis of platelet function from whole blood and platelet-rich plasma showed hyperactivation of platelets only in CKD Apoe-/- mice. There was no difference when experiments were done on washed platelets. However, preincubation of such platelets with AGEs or S100 proteins induced RAGE-mediated platelet hyperactivation. In vivo, CKD significantly reduced carotid occlusion times of Apoe-/- mice (9.2 ± 1.1 vs. 11.1 ± 0.6 minutes for sham, p < 0.01). In contrast, CKD had no effect on occlusion times in Apoe-/-/Ager-/- mice. Moreover, carotid occlusion in Apoe-/- CKD mice occurred significantly faster than in Apoe-/-/Ager-/- CKD mice (p < 0.0001). CONCLUSION Our results show that CKD induces platelet hyperactivation, accelerates thrombus formation in a murine model of arterial thrombosis, and that RAGE deletion has a protective role. We propose that RAGE ligands binding to RAGE is involved in CKD-induced arterial thrombosis.
Collapse
Affiliation(s)
- Jérémy Ortillon
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Team 2 "Matrix Aging and Vascular Remodelling," Université de Reims Champagne Ardenne, Reims, France
| | - Nathalie Hézard
- Hémostase et Remodelage Vasculaire Post-Ischémique, Laboratoire d'Hématologie, Faculté de Médecine & CHU Reims, Hôpital Robert Debré, Reims, France
| | - Karim Belmokhtar
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Team 2 "Matrix Aging and Vascular Remodelling," Université de Reims Champagne Ardenne, Reims, France
| | - Charlotte Kawecki
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Team 2 "Matrix Aging and Vascular Remodelling," Université de Reims Champagne Ardenne, Reims, France
| | - Christine Terryn
- PICT Platform, Université de Reims Champagne Ardenne, Reims, France
| | - Guenter Fritz
- Institute of Neuropathology, Neurozentrum, University of Freiburg, Freiburg, Germany
| | - Alexandre Kauskot
- HITh, UMR_S 1176, INSERM Université Paris-Sud, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Ann Marie Schmidt
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University School of Medicine, New York, New York, United States
| | - Philippe Rieu
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Team 2 "Matrix Aging and Vascular Remodelling," Université de Reims Champagne Ardenne, Reims, France.,Division of Nephrology, CHU Reims, Reims, France
| | - Philippe Nguyen
- Hémostase et Remodelage Vasculaire Post-Ischémique, Laboratoire d'Hématologie, Faculté de Médecine & CHU Reims, Hôpital Robert Debré, Reims, France
| | - Pascal Maurice
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Team 2 "Matrix Aging and Vascular Remodelling," Université de Reims Champagne Ardenne, Reims, France
| | - Fatouma Touré
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Team 2 "Matrix Aging and Vascular Remodelling," Université de Reims Champagne Ardenne, Reims, France.,Division of Nephrology, CHU Limoges, Limoges, France
| |
Collapse
|
38
|
Choi E, Oh J, Sung GH. Antithrombotic and Antiplatelet Effects of Cordyceps militaris. MYCOBIOLOGY 2020; 48:228-232. [PMID: 37970560 PMCID: PMC10635111 DOI: 10.1080/12298093.2020.1763115] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Revised: 04/20/2020] [Accepted: 04/26/2020] [Indexed: 11/17/2023]
Abstract
Cordyceps is a genus of ascomycete fungi and is well known as one of the important medical fungi in Chinese, Korea, and other Asian countries, because of its various beneficial effects on human health. The pharmacological activities of Cordyceps extract are mainly focused on anti-cancer, anti-metastatic, and immune modulating effects. In the present study, we investigated whether the antiplatelet effect of ethanol extract of cultured Cordyceps militaris (CMEE) with FeCl3-induced arterial thrombosis model. We observed that CMEE exhibited a significant inhibitory effect against ADP and collagen-induced platelet aggregation. However, there were no significant differences in prothrombin time (PT) and activated partial thromboplastin time (aPTT). These results suggest that antithrombotic activity of CMEE is related to antiplatelet effect rather than anticoagulation effect, and CMEE may be a positive effect on improving blood circulation against vessel injury and occlusion.
Collapse
Affiliation(s)
- Eunhyun Choi
- Translational Research Division, Biomedical Institute of Mycological Resource, International St. Mary’s Hospital and College of Medicine, Catholic Kwandong University, Incheon, Republic of Korea
| | - Junsang Oh
- Translational Research Division, Biomedical Institute of Mycological Resource, International St. Mary’s Hospital and College of Medicine, Catholic Kwandong University, Incheon, Republic of Korea
| | - Gi-Ho Sung
- Translational Research Division, Biomedical Institute of Mycological Resource, International St. Mary’s Hospital and College of Medicine, Catholic Kwandong University, Incheon, Republic of Korea
- Department of Microbiology, College of Medicine, Catholic Kwandong University, Gangneung-si, Republic of Korea
| |
Collapse
|
39
|
Tsuji S, Mukai T, Hirano H, Morita Y. In vivo analysis of thrombus formation in arthritic mice. Mod Rheumatol 2020; 31:498-503. [PMID: 32149538 DOI: 10.1080/14397595.2020.1740401] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
OBJECTIVES Rheumatoid arthritis (RA) is characterized by inflammation in multiple joints. In addition to causing joint destruction, the persistent systemic inflammation with RA increases the risk of cardiovascular disease. Although there are in vitro studies showing the prothrombotic effect of inflammatory cytokines, especially TNF, in vivo experimental evidence is lacking due to the complexity of in vivo modeling and observation. In this study, we aimed to model in vivo thrombus formation in arthritic mice and to determine whether the arthritic condition would further promote thrombotic formation. METHODS Human TNF-transgenic mice were used as the arthritis model. Thrombus formation was observed on the testicular arterioles. Thrombus formation was induced by reactive oxygen species generated from hematoporphyrin under laser irradiation. RESULTS Platelet thrombus formation was observed in real-time using a laser confocal microscopy in both wild-type and arthritic mice. Quantitative analyses revealed that no significant differences were observed in thrombus formation, represented by platelet attachment time and vascular obstruction time, in our experimental setting. CONCLUSION Although we confirmed the usefulness of this novel technique for in vivo studies, further investigation is required to conclude the possible mechanism of prothrombotic phenotypes under inflammatory conditions.
Collapse
Affiliation(s)
- Shoko Tsuji
- Department of Rheumatology, Kawasaki Medical School, Kurashiki, Okayama, Japan
| | - Tomoyuki Mukai
- Department of Rheumatology, Kawasaki Medical School, Kurashiki, Okayama, Japan
| | - Hiroyasu Hirano
- Department of Rheumatology, Kawasaki Medical School, Kurashiki, Okayama, Japan
| | - Yoshitaka Morita
- Department of Rheumatology, Kawasaki Medical School, Kurashiki, Okayama, Japan
| |
Collapse
|
40
|
Yu Y, Yu M, Liu J, Ding N, Huang J, Wan D, Zhao Y, Ma Z. In vivo monitoring of thrombosis in mice by optical coherence tomography. JOURNAL OF BIOPHOTONICS 2019; 12:e201900105. [PMID: 31339664 DOI: 10.1002/jbio.201900105] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 06/17/2019] [Accepted: 07/21/2019] [Indexed: 06/10/2023]
Abstract
The objective of this study is to establish a novel method for continuously monitoring thrombus progression with various outcome measures and to assess the efficacy of antithrombotic drugs in murine thrombosis model in mice. In the study, thrombus was induced in the femoral vein of mice by FeCl3 and monitored over time by spectral-domain optical coherence tomography (OCT). Three-dimensional images of thrombi with or without heparin as an antithrombotic agent were obtained from OCT angiography. In addition, several parameters of thrombi were analyzed and compared between control and anticoagulant groups. By using OCT, we were able to trace thrombus generation in the same mouse in real time. We found that in our model heparin reduced thrombus size by ~60% and thrombus cross-sectional area by 50%. OCT results also show that both time to thrombus size (>0.02mm3 ) and time to occlusion (>30%) were significantly reduced after heparin addition. This study demonstrates that OCT reliably monitors thrombus generation and progression from various aspects including thrombus size. This enables us to measure the kinetic of thrombosis more accurately, and effectively evaluate the efficacy and activities of antithrombotic drugs. This model may represent a useful tool in antithrombotic drug discoveries in preclinical studies.
Collapse
Affiliation(s)
- Yao Yu
- School of Control Engineering, Northeastern University at Qinhuangdao, Qinhuangdao, China
| | - Menghan Yu
- School of Control Engineering, Northeastern University at Qinhuangdao, Qinhuangdao, China
| | - Jian Liu
- School of Control Engineering, Northeastern University at Qinhuangdao, Qinhuangdao, China
| | - Ning Ding
- School of Sino-Dutch Biomedical and Information Engineering, Northeastern University, Shenyang, China
| | - Jiangmei Huang
- Department of Pathology, the First Hospital of Qinhuangdao, Qinhuangdao, China
| | - Dong Wan
- School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Yuliang Zhao
- School of Control Engineering, Northeastern University at Qinhuangdao, Qinhuangdao, China
| | - Zhenhe Ma
- School of Control Engineering, Northeastern University at Qinhuangdao, Qinhuangdao, China
| |
Collapse
|
41
|
Burnouf T, Chen CH, Tan SJ, Tseng CL, Lu KY, Chang LH, Nyambat B, Huang SC, Jheng PR, Aditya RN, Mi FL, Chuang EY. A bioinspired hyperthermic macrophage-based polypyrrole-polyethylenimine (Ppy-PEI) nanocomplex carrier to prevent and disrupt thrombotic fibrin clots. Acta Biomater 2019; 96:468-479. [PMID: 31260820 DOI: 10.1016/j.actbio.2019.06.053] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 06/17/2019] [Accepted: 06/26/2019] [Indexed: 11/16/2022]
Abstract
Fibrinolytic treatments for venous or arterial thrombotic syndromes using systemic administration of thrombolytics, such as streptokinase, can induce life-threatening bleeding complications. In this study, we offer the first proof of concept for a targeted photothermal fibrin clot prevention and reduction technology using macrophages loaded with polypyrrole-polyethylenimine nanocomplexes (Ppy-PEI NCs) and subjected to near-infrared radiation (NIR). We first show that the developed Ppy-PEI NCs could be taken up by defensive macrophages in vitro through endocytosis. The Ppy-PEI NCs generated local hyperthermia upon NIR treatment, which appeared to produce reactive oxygen species in Ppy-PEI NC-loaded macrophages. Preliminary evidence of efficacy as an antithrombotic tool is provided, in vitro, using fibrinogen-converted fibrin clots, and in vivo, in a rat femoral vascular thrombosis model generated by exposure to ferric chloride substance. The in vivo biocompatibility, photothermal behavior, biodistribution, and histological observation of cellular interactions with the Ppy-PEI NCs in the rat model provide rationale in support of further preclinical studies. This Ppy-PEI NC/NIR-based method, which uses a unique macrophage-guided targeting approach to prevent and lyse fibrin clots, may potentially overcome some of the disadvantages of current thrombolytic treatments. STATEMENT OF SIGNIFICANCE: Fibrinolytic treatments for venous or arterial thrombotic syndromes using systemic administration of thrombolytics, such as streptokinase, can induce life-threatening bleeding complications. In this study, we offer the first proof of concept for a targeted photothermal fibrin clot reduction technology using macrophages loaded with polypyrrole-polyethylenimine nanocomplexes (Ppy-PEI NCs) and subjected to near-infrared radiation (NIR). We first show that the developed Ppy-PEI NCs can be taken up by defensive macrophages in vitro through endocytosis. The Ppy-PEI NCs generated local hyperthermia upon NIR treatment, which appeared to produce reactive oxygen species in Ppy-PEI NC-loaded macrophages. Preliminary evidence of efficacy as an antithrombotic tool is provided, in vitro, using fibrinogen-converted fibrin clots, and in vivo, in a rat femoral vascular thrombosis model generated by exposure to ferric chloride substance. The in vivo biocompatibility, photothermal behavior, biodistribution, and histological observation of cellular interactions with the Ppy-PEI NCs in the rat model provide rationale in support of further preclinical studies. This Ppy-PEI NC/NIR-based method, which uses a unique macrophage-guided targeting approach to disintegrate fibrin clots, may potentially overcome some of the disadvantages of current thrombolytic treatments.
Collapse
Affiliation(s)
- Thierry Burnouf
- Graduate Institute of Biomedical Materials and Tissue Engineering, and International PhD Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan, ROC
| | - Chih-Hwa Chen
- Department of Orthopedics, Shuang Ho Hospital, Taipei Medical University, Taipei, Taiwan, ROC; School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan, ROC; School of Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan, ROC
| | - Shun-Jen Tan
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Taipei Medical University Hospital, Taipei, Taiwan, ROC; Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan, ROC; Department of Obstetrics and Gynecology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan, ROC; Department of Obstetrics and Gynecology, School of Medicine, National Defense Medical Center, Taiwan, ROC
| | - Ching-Li Tseng
- Graduate Institute of Biomedical Materials and Tissue Engineering, and International PhD Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan, ROC
| | - Kun-Ying Lu
- Graduate Institute of Biomedical Materials and Tissue Engineering, and International PhD Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan, ROC; Department of Biochemistry and Molecular Cell Biology, School of Medicine, Graduate Institute of Medical Sciences, College of Medicine, Graduate Institute of Biomedical Materials and Tissue Engineering, Graduate Institute of Nanomedicine and Medical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan, ROC
| | - Lee-Hsin Chang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan, ROC
| | - Batzaya Nyambat
- Graduate Institute of Biomedical Materials and Tissue Engineering, and International PhD Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan, ROC
| | - Shao-Chan Huang
- Graduate Institute of Biomedical Materials and Tissue Engineering, and International PhD Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan, ROC
| | - Pei-Ru Jheng
- Graduate Institute of Biomedical Materials and Tissue Engineering, and International PhD Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan, ROC
| | - Robby Nur Aditya
- Graduate Institute of Biomedical Materials and Tissue Engineering, and International PhD Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan, ROC
| | - Fwu-Long Mi
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, Graduate Institute of Medical Sciences, College of Medicine, Graduate Institute of Biomedical Materials and Tissue Engineering, Graduate Institute of Nanomedicine and Medical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan, ROC.
| | - Er-Yuan Chuang
- Graduate Institute of Biomedical Materials and Tissue Engineering, and International PhD Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan, ROC.
| |
Collapse
|
42
|
Yang M, Silverstein RL. CD36 and ERK5 link dyslipidemia to apoptotic-like platelet procoagulant function. Curr Opin Hematol 2019; 26:357-365. [PMID: 31261174 PMCID: PMC9308374 DOI: 10.1097/moh.0000000000000522] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
PURPOSE OF REVIEW Metabolic diseases, including dyslipidemia, diabetes mellitus, and chronic inflammation are risk factors for clinically significant thrombotic events. Thrombosis in these settings is multifaceted with coordinated mechanisms between platelet activation and the hemostatic pathways. This review focuses on recent advances in platelet procoagulant and apoptotic signaling with emphasis on the pathophysiologic mechanisms induced by platelet CD36 in dyslipidemia, and the key unaddressed questions relating to the field. RECENT FINDINGS CD36 promotes platelet activation and increases the risk for thrombosis through signaling events. These include generation of reactive oxygen species, activation of redox-sensitive MAP kinase ERK5, and promotion of a pro-thrombotic phenotype. CD36 promotes phosphatidylserine externalization leading to a procoagulant function downstream from MAP kinase ERK5 that is separate from a pro-aggregatory function. Phosphatidylserine externalization requires maladaptive caspase activation, promotes assembly of the factor tenase and prothrombinase complex, and promotes fibrin formation. It is distinct from the canonical pathways mediating platelet procoagulant function by strong physiologic stimuli or by the platelet apoptotic-like Bak/Bax-mediated pathway for cellular clearance. SUMMARY Understanding CD36 signaling in the context of dyslipidemia, or other metabolic diseases will identify important and novel signaling hubs that could be potential therapeutic targets for intervention without impacting hemostasis.
Collapse
Affiliation(s)
- Moua Yang
- Department of Biochemistry, Medical College of Wisconsin
- Blood Research Institute, Versiti Blood Center of Wisconsin
| | - Roy L. Silverstein
- Blood Research Institute, Versiti Blood Center of Wisconsin
- Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| |
Collapse
|
43
|
Synthetic Flavonoids as Novel Modulators of Platelet Function and Thrombosis. Int J Mol Sci 2019; 20:ijms20123106. [PMID: 31242657 PMCID: PMC6627635 DOI: 10.3390/ijms20123106] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 06/21/2019] [Accepted: 06/23/2019] [Indexed: 01/27/2023] Open
Abstract
Cardiovascular diseases represent a major cause of mortality and morbidity in the world, and specifically, thrombotic conditions such as heart attacks and strokes are caused by unwarranted activation of platelets and subsequent formation of blood clots (thrombi) within the blood vessels during pathological circumstances. Therefore, platelets act as a primary therapeutic target to treat and prevent thrombotic conditions. Current treatments are limited due to intolerance, and they are associated with severe side effects such as bleeding complications. Hence, the development of novel therapeutic strategies for thrombotic diseases is an urgent priority. Flavonoids are naturally occurring plant-derived molecules that exert numerous beneficial effects in humans through modulating the functions of distinct cell types. However, naturally occurring flavonoids suffer from several issues such as poor solubility, lipophilicity, and bioavailability, which hinder their efficacy and potency. Despite these, flavonoids act as versatile templates for the design and synthesis of novel molecules for various therapeutic targets. Indeed, several synthetic flavonoids have recently been developed to improve their stability, bioavailability, and efficacy, including for the modulation of platelet function. Here, we provide insight into the actions of certain natural flavonoids along with the advantages of synthetic flavonoids in the modulation of platelet function, haemostasis, and thrombosis.
Collapse
|
44
|
Yang M, Silverstein RL. CD36 signaling in vascular redox stress. Free Radic Biol Med 2019; 136:159-171. [PMID: 30825500 PMCID: PMC6488418 DOI: 10.1016/j.freeradbiomed.2019.02.021] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 01/29/2019] [Accepted: 02/18/2019] [Indexed: 12/20/2022]
Abstract
Scavenger receptor CD36 is a multifunctional membrane protein that promotes thrombosis in conditions of oxidative stress such as metabolic disorders including dyslipidemia, diabetes mellitus, and chronic inflammation. In these conditions, specific reactive oxidant species are generated that are context and cell dependent. In the vasculature, CD36 signaling in smooth muscle cells and endothelial cells promotes generation of reactive oxygen species, genetic downregulation of antioxidant genes, and impaired smooth muscle and endothelial function. In hematopoietic cells, CD36 signaling enhances platelet dysfunction thus decreasing the threshold for platelet activation and accelerating arterial thrombosis, whereas in macrophages, CD36 promotes lipid-laden foam cell formation and atherosclerosis. These clinically significant processes are mediated through complex redox regulated signaling mechanisms that include Src-family kinases, MAP kinases and other downstream effectors. We provide an overview of CD36 signaling in vascular redox stress highlighting the role in oxidant generation in vascular and hematopoietic cells, but with special emphasis on platelets and dyslipidemia.
Collapse
Affiliation(s)
- Moua Yang
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, USA; Blood Research Institute, BloodCenter of Wisconsin, Milwaukee, WI, USA
| | - Roy L Silverstein
- Blood Research Institute, BloodCenter of Wisconsin, Milwaukee, WI, USA; Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA.
| |
Collapse
|
45
|
Kawano T, Shimamura M, Nakagami H, Iso T, Koriyama H, Takeda S, Baba K, Sasaki T, Sakaguchi M, Morishita R, Mochizuki H. Therapeutic Vaccine Against S100A9 (S100 Calcium-Binding Protein A9) Inhibits Thrombosis Without Increasing the Risk of Bleeding in Ischemic Stroke in Mice. Hypertension 2019; 72:1355-1364. [PMID: 30571223 DOI: 10.1161/hypertensionaha.118.11316] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Decreased adherence to daily ingestion of antiplatelet drugs is a critical issue, increasing mortality and morbidity in poststroke patients. As vaccination could be a promising approach to solving this, we designed an antiplatelet vaccine that inhibited S100A9 (S100 calcium-binding protein A9)/CD36 (cluster of differentiation 36) signaling in platelets, which was reported to be a key signal in arterial thrombosis, but not hemostasis. Immunization with this vaccine induced a sustainable increase in the anti-S100A9 antibody titer for >2 months and an additional booster immunization enhanced the antibody production further. The middle cerebral artery occlusion time was successfully prolonged in the vaccinated mice, which was comparable to that in mice treated with clopidogrel. The antithrombotic effect lasted for 84 days after the last vaccination, as well as after the booster immunization. Importantly, the bleeding time was not affected in the immunized mice. The antithrombotic effect was also observed in the common carotid artery, which was similar to that found in CD36-/- mice. Vascular injury increased the expression of S100A9 in the serum and phosphorylation of JNK (c-Jun N-terminal kinase) and VAV1 in the platelets, but these increases were inhibited in the immunized mice. Moreover, the S100A9 vaccine did not induce cell-mediated autoimmunity, as demonstrated by the enzyme-linked immunosorbent spot assay. Thus, immunization with the S100A9 vaccine resulted in long-term inhibition of thrombus formation through inhibition of increased S100A9/CD36 signaling without risk of bleeding or adverse autoimmune responses. Vaccination against S100A9 might be a novel therapy to prevent recurrent ischemic stroke.
Collapse
Affiliation(s)
- Tomohiro Kawano
- From the Department of Neurology (T.K., M.S., K.B., T.S., M.S., H.M.), Osaka University Graduate School of Medicine, Japan
- Department of Health Development and Medicine (T.K., M.S., H.N., H.K.), Osaka University Graduate School of Medicine, Japan
| | - Munehisa Shimamura
- From the Department of Neurology (T.K., M.S., K.B., T.S., M.S., H.M.), Osaka University Graduate School of Medicine, Japan
- Department of Health Development and Medicine (T.K., M.S., H.N., H.K.), Osaka University Graduate School of Medicine, Japan
| | - Hironori Nakagami
- Department of Health Development and Medicine (T.K., M.S., H.N., H.K.), Osaka University Graduate School of Medicine, Japan
| | - Tatsuya Iso
- Education and Research Support Center, Department of Cardiovascular Medicine, Gunma University Graduate School of Medicine, Japan (T.I.)
| | - Hiroshi Koriyama
- Department of Health Development and Medicine (T.K., M.S., H.N., H.K.), Osaka University Graduate School of Medicine, Japan
| | - Shuko Takeda
- Department of Clinical Gene Therapy (S.T., R.M.), Osaka University Graduate School of Medicine, Japan
| | - Kosuke Baba
- From the Department of Neurology (T.K., M.S., K.B., T.S., M.S., H.M.), Osaka University Graduate School of Medicine, Japan
| | - Tsutomu Sasaki
- From the Department of Neurology (T.K., M.S., K.B., T.S., M.S., H.M.), Osaka University Graduate School of Medicine, Japan
| | - Manabu Sakaguchi
- From the Department of Neurology (T.K., M.S., K.B., T.S., M.S., H.M.), Osaka University Graduate School of Medicine, Japan
| | - Ryuichi Morishita
- Department of Clinical Gene Therapy (S.T., R.M.), Osaka University Graduate School of Medicine, Japan
| | - Hideki Mochizuki
- From the Department of Neurology (T.K., M.S., K.B., T.S., M.S., H.M.), Osaka University Graduate School of Medicine, Japan
| |
Collapse
|
46
|
Sashindranath M, Sturgeon SA, French S, Craenmehr DDD, Selan C, Freddi S, Johnson C, Cody SH, Nesbitt WS, Hamilton JR, Nandurkar HH. The mode of anesthesia influences outcome in mouse models of arterial thrombosis. Res Pract Thromb Haemost 2019; 3:197-206. [PMID: 31011704 PMCID: PMC6462741 DOI: 10.1002/rth2.12184] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 12/22/2018] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Arterial thrombosis models are important for preclinical evaluation of antithrombotics but how anesthetic protocol can influence experimental results is not studied. OBJECTIVES We studied how three most commonly used rodent anesthetics affect the induction of thrombosis and thrombus resolution with antiplatelet agent integrilin (Eptifibatide). METHODS The Folts, electrolytic, and FeCl3 models of carotid artery thrombosis were evaluated. The extent of blood flow reduction required to elicit cyclic flow reductions (CFR) was examined in the Folts model. The occlusion time and stability following electrolytic or FeCl3 injury was assessed. The efficacy of Eptifibatide was studied in each cohort and clot composition following FeCl3 application was assessed histologically. RESULTS Isoflurane and ketamine-xylazine (ket-x) elicited higher basal blood flow velocities. For reliable CFR in the Folts model, a higher degree of blood flow reduction was required under ket-x and isoflurane. For the FeCl3 and electrolytic models, injury severity had to be increased in mice under ket-x anesthesia to achieve rapid occlusion. FeCl3-injured artery sections from ket-x and isoflurane-treated mice showed vessel dilatation and clots that were more fibrin/red-cell rich compared to pentobarbitone. Integrilin led to cycle abolishment for all three Folts-injury cohorts but for the electrolytic model a 2.5-fold higher dose was required to restore blood flow under pentobarbitone. Integrilin after FeCl3 arterial injury was partially ineffective in isoflurane-treated mice. CONCLUSIONS Anesthesia impacts rodent carotid artery occlusion experiments and alters integrilin efficacy. It is important to consider anesthetic protocols in animal experiments involving pharmacological agents for treatment of atherothrombosis.
Collapse
Affiliation(s)
- Maithili Sashindranath
- Australian Centre for Blood DiseasesCentral Clinical SchoolMonash UniversityAlfred HospitalMelbourneVic.Australia
| | - Sharelle A. Sturgeon
- Australian Centre for Blood DiseasesCentral Clinical SchoolMonash UniversityAlfred HospitalMelbourneVic.Australia
| | - Shauna French
- Australian Centre for Blood DiseasesCentral Clinical SchoolMonash UniversityAlfred HospitalMelbourneVic.Australia
| | - Daphne D. D. Craenmehr
- Australian Centre for Blood DiseasesCentral Clinical SchoolMonash UniversityAlfred HospitalMelbourneVic.Australia
| | - Carly Selan
- Australian Centre for Blood DiseasesCentral Clinical SchoolMonash UniversityAlfred HospitalMelbourneVic.Australia
| | - Susanna Freddi
- Australian Centre for Blood DiseasesCentral Clinical SchoolMonash UniversityAlfred HospitalMelbourneVic.Australia
| | - Chad Johnson
- Monash Micro ImagingMonash UniversityMelbourneVic.Australia
- Burnet InstituteMelbourneVic.Australia
| | | | - Warwick S. Nesbitt
- Australian Centre for Blood DiseasesCentral Clinical SchoolMonash UniversityAlfred HospitalMelbourneVic.Australia
- School of EngineeringRMIT UniversityMelbourneVICAustralia
| | - Justin R. Hamilton
- Australian Centre for Blood DiseasesCentral Clinical SchoolMonash UniversityAlfred HospitalMelbourneVic.Australia
| | - Harshal H. Nandurkar
- Australian Centre for Blood DiseasesCentral Clinical SchoolMonash UniversityAlfred HospitalMelbourneVic.Australia
| |
Collapse
|
47
|
He X, Drelich A, Yu S, Chang Q, Gong D, Zhou Y, Qu Y, Yuan Y, Su Z, Qiu Y, Tang SJ, Gaitas A, Ksiazek T, Xu Z, Zhou J, Feng Z, Wakamiya M, Lu F, Gong B. Exchange protein directly activated by cAMP plays a critical role in regulation of vascular fibrinolysis. Life Sci 2019; 221:1-12. [PMID: 30738042 DOI: 10.1016/j.lfs.2019.02.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Revised: 02/04/2019] [Accepted: 02/04/2019] [Indexed: 01/09/2023]
Abstract
Plasmin-mediated fibrinolysis at the surface of vascular endothelial cells (SVEC) plays a key role in maintaining vascular hemostasis, in which the cAMP pathway participates. After externalization to the SVEC, annexin A2 (ANXA2) serves as a platform for conversion of plasminogen to plasmin. Here we describe a regulatory role of the exchange protein directly activated by cAMP (EPAC) in ANXA2 externalization and vascular fibrinolysis. Knockout of EPAC1 in mice results in a decreased ANXA2 expression on the SVEC associated with increased fibrin deposition and fibrinolytic dysfunction. Reduced levels of EPAC1 are also found in endocardial tissues beneath atrial mural thrombi in patients. Notably, administration of recombinant ANXA2 ameliorates fibrinolytic dysfunction in the EPAC1-null mice. Mechanistically, EPAC1 regulates the SVEC plasminogen conversion depended on ANXA2. EPAC1 promotes tyrosine-23 phosphorylation of ANXA2, a prerequisite for its recruitment to the SVEC. Our data thus reveal a novel regulatory role for EPAC1 in vascular fibrinolysis.
Collapse
Affiliation(s)
- Xi He
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA; Department of Cardiovascular Surgery, Changhai Hospital, Shanghai 200433, China
| | - Aleksandra Drelich
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Shangyi Yu
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA; Department of Cardiovascular Surgery, Changhai Hospital, Shanghai 200433, China
| | - Qing Chang
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Dejun Gong
- Department of Cardiovascular Surgery, Changhai Hospital, Shanghai 200433, China
| | - Yixuan Zhou
- Department of Cardiovascular Surgery, Changhai Hospital, Shanghai 200433, China
| | - Yue Qu
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Yang Yuan
- Department of Cardiovascular Surgery, Changhai Hospital, Shanghai 200433, China
| | - Zhengchen Su
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Yuan Qiu
- Department of Mathematics and Statistics, Texas Tech University, Lubbock, TX 79409, USA
| | - Shao-Jun Tang
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Angelo Gaitas
- Department of Neurology, Icahn School of Medicine at Mount Sinai, NY 10029, USA
| | - Thomas Ksiazek
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Zhiyun Xu
- Department of Cardiovascular Surgery, Changhai Hospital, Shanghai 200433, China
| | - Jia Zhou
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Zongdi Feng
- Center for Vaccines and Immunity, The Research Institute at Nationwide Children's Hospital, Columbus, OH 43205, USA
| | - Maki Wakamiya
- Department of Neurology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Fanglin Lu
- Department of Cardiovascular Surgery, Changhai Hospital, Shanghai 200433, China.
| | - Bin Gong
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA.
| |
Collapse
|
48
|
Rezaeianpour S, Mosayebnia M, Moghimi A, Amidi S, Geramifar P, Kobarfard F, Shahhosseini S. [ 18F]FDG-Labeled CGPRPPC Peptide Serving as a Small Thrombotic Lesions Probe, Including a Comparison with [ 99mTc]-Labeled Form. Cancer Biother Radiopharm 2018; 33:438-444. [PMID: 30234382 DOI: 10.1089/cbr.2018.2515] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Purpose: Fibrin is a perfect target for specific imaging of all types of thrombotic lesions. Cyclic peptides were introduced as the best scaffolds out of the different types of probes for thrombi detection. This study was conducted to label previously synthesized peptide-targeting fibrin with [18F]FDG and its in vitro and in vivo assessments. Materials and Methods: CGPRPPC peptide functionalized with 6-hydrazinonicotinamide and Eei-NHS was synthesized and cyclized using air oxidation method. The cyclic sequences were labeled with [18F]FDG at 85°C within 30 min. The stability studies were performed in human plasma. Fibrin-binding and platelet aggregation tests were performed in vitro. Biodistribution and scintigraphy imaging in normal mice and carotid thrombotic rat model were considered as in vivo studies. Results: Radiolabeled peptides show a good stability in human plasma and also high-affinity binding for human fibrin. Platelet aggregation test confirmed specific binding of radiopeptides to fibrin. A key problem with the authors' previous research was inability to detect small-vessel thrombi. The results of positron emission tomography/computed tomography scanning show high specific uptake of [18F]FDG-labeled CGPRPPC in small-sized thrombosis. Conclusion: The experiment revealed that radiolabeling of cyclic peptide (CGPRPPC) with [18F]FDG enables us to detect small thrombotic lesions in small animal models with high resolution.
Collapse
Affiliation(s)
- Sedigheh Rezaeianpour
- Phytochemistry Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mona Mosayebnia
- Department of Radiopharmacy, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Abolghasem Moghimi
- Faculty of Chemistry, Tehran North Branch Islamic Azad University, Tehran, Iran
| | - Salimeh Amidi
- Department of Pharmaceutical Chemistry and Radiopharmacy, School of Pharmacy, Shahid Behesti University of Medical Sciences, Tehran, Iran
| | - Parham Geramifar
- Research Center for Nuclear Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Farzad Kobarfard
- Department of Pharmaceutical Chemistry and Radiopharmacy, School of Pharmacy, Phytochemistry Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Soraya Shahhosseini
- Department of Pharmaceutical Chemistry and Radiopharmacy, School of Pharmacy, and Protein Technology Research Center, Shahid Behesti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
49
|
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: 0.7] [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
|
50
|
Chen R, Jin G, Li W, McIntyre TM. Epidermal Growth Factor (EGF) Autocrine Activation of Human Platelets Promotes EGF Receptor-Dependent Oral Squamous Cell Carcinoma Invasion, Migration, and Epithelial Mesenchymal Transition. THE JOURNAL OF IMMUNOLOGY 2018; 201:2154-2164. [PMID: 30150285 DOI: 10.4049/jimmunol.1800124] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 07/25/2018] [Indexed: 12/11/2022]
Abstract
Activated platelets release functional, high m.w. epidermal growth factor (HMW-EGF). In this study, we show platelets also express epidermal growth factor (EGF) receptor (EGFR) protein, but not ErbB2 or ErbB4 coreceptors, and so might respond to HMW-EGF. We found HMW-EGF stimulated platelet EGFR autophosphorylation, PI3 kinase-dependent AKT phosphorylation, and a Ca2+ transient that were blocked by EGFR tyrosine kinase inhibition. Strong (thrombin) and weak (ADP, platelet-activating factor) G protein-coupled receptor agonists and non-G protein-coupled receptor collagen recruited EGFR tyrosine kinase activity that contributed to platelet activation because EGFR kinase inhibition reduced signal transduction and aggregation induced by each agonist. EGF stimulated ex vivo adhesion of platelets to collagen-coated microfluidic channels, whereas systemic EGF injection increased initial platelet deposition in FeCl3-damaged murine carotid arteries. EGFR signaling contributes to oral squamous cell carcinoma (OSCC) tumorigenesis, but the source of its ligand is not established. We find individual platelets were intercalated within OSCC tumors. A portion of these platelets expressed stimulation-dependent Bcl-3 and IL-1β and so had been activated. Stimulated platelets bound OSCC cells, and material released from stimulated platelets induced OSCC epithelial-mesenchymal transition and stimulated their migration and invasion through Matrigel barriers. Anti-EGF Ab or EGFR inhibitors abolished platelet-induced tumor cell phenotype transition, migration, and invasion; so the only factor released from activated platelets necessary for OSCC metastatic activity was HMW-EGF. These results establish HMW-EGF in platelet function and elucidate a previously unsuspected connection between activated platelets and tumorigenesis through rapid, and prolonged, autocrine-stimulated release of HMW-EGF by tumor-associated platelets.
Collapse
Affiliation(s)
- Rui Chen
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Ge Jin
- Case Western Reserve University School of Dental Medicine, Cleveland, OH 44106
| | - Wei Li
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195.,Department of Molecular Medicine, Case Western Reserve University, Cleveland, OH 44106; and.,Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44195
| | - Thomas M McIntyre
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195; .,Department of Molecular Medicine, Case Western Reserve University, Cleveland, OH 44106; and.,Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44195
| |
Collapse
|