1
|
Malamud M, Brown GD. The Dectin-1 and Dectin-2 clusters: C-type lectin receptors with fundamental roles in immunity. EMBO Rep 2024:10.1038/s44319-024-00296-2. [PMID: 39482490 DOI: 10.1038/s44319-024-00296-2] [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: 07/22/2024] [Revised: 09/24/2024] [Accepted: 10/14/2024] [Indexed: 11/03/2024] Open
Abstract
The ability of myeloid cells to recognize and differentiate endogenous or exogenous ligands rely on the presence of different transmembrane protein receptors. C-type lectin receptors (CLRs), defined by the presence of a conserved structural motif called C-type lectin-like domain (CTLD), are a crucial family of receptors involved in this process, being able to recognize a diverse range of ligands from glycans to proteins or lipids and capable of initiating an immune response. The Dectin-1 and Dectin-2 clusters involve two groups of CLRs, with genes genomically linked within the natural killer cluster of genes in both humans and mice, and all characterized by the presence of a single extracellular CTLD. Fundamental immune cell functions such as antimicrobial effector mechanisms as well as internalization and presentation of antigens are induced and/or regulated through activatory, or inhibitory signalling pathways triggered by these receptors after ligand binding. In this review, we will discuss the most recent concepts regarding expression, ligands, signaling pathways and functions of each member of the Dectin clusters of CLRs, highlighting the importance and diversity of their functions.
Collapse
Affiliation(s)
- Mariano Malamud
- Medical Research Council (MRC) Centre for Medical Mycology, University of Exeter, Exeter, UK.
| | - Gordon D Brown
- Medical Research Council (MRC) Centre for Medical Mycology, University of Exeter, Exeter, UK.
| |
Collapse
|
2
|
Laspa Z, Dicenta-Baunach V, Schaale D, Sigle M, Hochuli R, Castor T, Bayrak A, Harm T, Müller KAL, Pillaiyar T, Laufer S, Rohlfing AK, Gawaz MP. Hemin-induced platelet activation is regulated by the ACKR3 chemokine surface receptor and has implications for passivation of vulnerable atherosclerotic plaques. FEBS J 2024. [PMID: 39387619 DOI: 10.1111/febs.17294] [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: 04/12/2024] [Revised: 07/11/2024] [Accepted: 09/25/2024] [Indexed: 10/15/2024]
Abstract
In vulnerable atherosclerotic plaques, intraplaque hemorrhages (IPH) result in hemolysis of red blood cells and release of hemoglobin and free hemin. Hemin activates platelets and leads to thrombosis. Agonism of the inhibitory platelet receptor ACKR3 inhibits hemin-dependent platelet activation and thrombus formation. To characterize the effect of hemin and ACKR3 agonism on isolated human platelets, multi-color flow cytometry and classical experimental setup such as light transmission aggregometry and a flow chamber assay were used. Hemin induces platelet aggregation and ex vivo platelet-dependent thrombus formation on immobilized collagen under a low shear rate of 500 s-1, indicating that free hemin is a strong activator of platelet-dependent thrombosis. Recently, we described that ACKR3 is a prominent inhibitory receptor of platelet activation. Specific ACKR3 agonists but not conventional antiplatelet compounds such as COX-1 inhibitor (indometacin), ADP-receptor blocker (cangrelor), or PAR1 inhibitor (ML161) inhibit both hemin-dependent aggregation and thrombus formation. To further characterize the effect of hemin on platelet subpopulations, we established a multi-color flow cytometry assay. We found that hemin induces procoagulant (CD42bpos/PAC-1neg/AnnexinVpos), aggregatory (CD42bpos/PAC-1pos/AnnexinVneg), and inflammatory (CD42bpos/CXCR4pos/ACKR3pos/AnnexinVpos) platelet subpopulations. Treatment with ACKR3 agonists significantly decreased the formation of procoagulant and ACKR3pos platelets in response to hemin. We conclude that hemin is a strong activator for the formation of procoagulant platelets and thrombus formation which is dependent on the function of ACKR3. Activation of ACKR3 using specific agonists may offer a therapeutic strategy to regulate the vulnerability of atherosclerotic plaques in areas of IPH.
Collapse
Affiliation(s)
- Zoi Laspa
- Department of Cardiology and Angiology, University Hospital Tübingen, University Tübingen, Germany
| | - Valerie Dicenta-Baunach
- Department of Cardiology and Angiology, University Hospital Tübingen, University Tübingen, Germany
| | - David Schaale
- Department of Cardiology and Angiology, University Hospital Tübingen, University Tübingen, Germany
| | - Manuel Sigle
- Department of Cardiology and Angiology, University Hospital Tübingen, University Tübingen, Germany
| | - Ravi Hochuli
- Department of Cardiology and Angiology, University Hospital Tübingen, University Tübingen, Germany
| | - Tatsiana Castor
- Department of Cardiology and Angiology, University Hospital Tübingen, University Tübingen, Germany
| | - Alp Bayrak
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmaceutical Sciences, University Tübingen, Germany
| | - Tobias Harm
- Department of Cardiology and Angiology, University Hospital Tübingen, University Tübingen, Germany
| | - Karin Anne Lydia Müller
- Department of Cardiology and Angiology, University Hospital Tübingen, University Tübingen, Germany
| | - Thanigaimalai Pillaiyar
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmaceutical Sciences, University Tübingen, Germany
- Tübingen Center for Academic Drug Discovery & Development (TüCAD2), Germany
| | - Stefan Laufer
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmaceutical Sciences, University Tübingen, Germany
- Tübingen Center for Academic Drug Discovery & Development (TüCAD2), Germany
- iFIT Cluster of Excellence EXC 2180 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Germany
| | - Anne-Katrin Rohlfing
- Department of Cardiology and Angiology, University Hospital Tübingen, University Tübingen, Germany
| | - Meinrad Paul Gawaz
- Department of Cardiology and Angiology, University Hospital Tübingen, University Tübingen, Germany
| |
Collapse
|
3
|
Lefèbre J, Falk T, Ning Y, Rademacher C. Secondary Sites of the C-type Lectin-Like Fold. Chemistry 2024; 30:e202400660. [PMID: 38527187 DOI: 10.1002/chem.202400660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 03/23/2024] [Accepted: 03/25/2024] [Indexed: 03/27/2024]
Abstract
C-type lectins are a large superfamily of proteins involved in a multitude of biological processes. In particular, their involvement in immunity and homeostasis has rendered them attractive targets for diverse therapeutic interventions. They share a characteristic C-type lectin-like domain whose adaptability enables them to bind a broad spectrum of ligands beyond the originally defined canonical Ca2+-dependent carbohydrate binding. Together with variable domain architecture and high-level conformational plasticity, this enables C-type lectins to meet diverse functional demands. Secondary sites provide another layer of regulation and are often intricately linked to functional diversity. Located remote from the canonical primary binding site, secondary sites can accommodate ligands with other physicochemical properties and alter protein dynamics, thus enhancing selectivity and enabling fine-tuning of the biological response. In this review, we outline the structural determinants allowing C-type lectins to perform a large variety of tasks and to accommodate the ligands associated with it. Using the six well-characterized Ca2+-dependent and Ca2+-independent C-type lectin receptors DC-SIGN, langerin, MGL, dectin-1, CLEC-2 and NKG2D as examples, we focus on the characteristics of non-canonical interactions and secondary sites and their potential use in drug discovery endeavors.
Collapse
Affiliation(s)
- Jonathan Lefèbre
- Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria
- Vienna Doctoral School of Pharmaceutical, Nutritional and Sport, Sciences, University of Vienna, Vienna, Austria
- Department of Microbiology, Immunology and Genetics, University of Vienna, Max F. Perutz Labs, Vienna, Austria
| | - Torben Falk
- Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria
- Vienna Doctoral School of Pharmaceutical, Nutritional and Sport, Sciences, University of Vienna, Vienna, Austria
- Department of Microbiology, Immunology and Genetics, University of Vienna, Max F. Perutz Labs, Vienna, Austria
| | - Yunzhan Ning
- Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria
- Vienna Doctoral School of Pharmaceutical, Nutritional and Sport, Sciences, University of Vienna, Vienna, Austria
- Department of Microbiology, Immunology and Genetics, University of Vienna, Max F. Perutz Labs, Vienna, Austria
| | - Christoph Rademacher
- Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria
- Department of Microbiology, Immunology and Genetics, University of Vienna, Max F. Perutz Labs, Vienna, Austria
| |
Collapse
|
4
|
Sun L, Wang Z, Liu Z, Mu G, Cui Y, Xiang Q. C-type lectin-like receptor 2: roles and drug target. Thromb J 2024; 22:27. [PMID: 38504248 PMCID: PMC10949654 DOI: 10.1186/s12959-024-00594-8] [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: 01/06/2024] [Accepted: 03/07/2024] [Indexed: 03/21/2024] Open
Abstract
C-type lectin-like receptor-2 (CLEC-2) is a member of the C-type lectin superfamily of cell surface receptors. The first confirmed endogenous and exogenous ligands of CLEC-2 are podoplanin and rhodocytin, respectively. CLEC-2 is expressed on the surface of platelets, which participates in platelet activation and aggregation by binding with its ligands. CLEC-2 and its ligands are involved in pathophysiological processes, such as atherosclerosis, cancer, inflammatory thrombus status, maintenance of vascular wall integrity, and cancer-related thrombosis. In the last 5 years, different anti- podoplanin antibody types have been developed for the treatment of cancers, such as glioblastoma and lung cancer. New tests and new diagnostics targeting CLEC-2 are also discussed. CLEC-2 mediates thrombosis in various pathological states, but CLEC-2-specific deletion does not affect normal hemostasis, which would provide a new therapeutic tool for many thromboembolic diseases. The CLEC-2-podoplanin interaction is a target for cancer treatment. CLEC-2 may be applied in clinical practice and play a therapeutic role.
Collapse
Affiliation(s)
- Lan Sun
- Department of Pharmacy, Peking University First Hospital, No. 6, Da Hong Luo Chang Street, Xicheng District, Beijing, 100034, China
- Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing, China
| | - Zhe Wang
- Department of Pharmacy, Peking University First Hospital, No. 6, Da Hong Luo Chang Street, Xicheng District, Beijing, 100034, China
- Institute of Clinical Pharmacology, Peking University, Beijing, China
| | - Zhiyan Liu
- Department of Pharmacy, Peking University First Hospital, No. 6, Da Hong Luo Chang Street, Xicheng District, Beijing, 100034, China
- Institute of Clinical Pharmacology, Peking University, Beijing, China
| | - Guangyan Mu
- Department of Pharmacy, Peking University First Hospital, No. 6, Da Hong Luo Chang Street, Xicheng District, Beijing, 100034, China
- Institute of Clinical Pharmacology, Peking University, Beijing, China
| | - Yimin Cui
- Department of Pharmacy, Peking University First Hospital, No. 6, Da Hong Luo Chang Street, Xicheng District, Beijing, 100034, China
- Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing, China
- Institute of Clinical Pharmacology, Peking University, Beijing, China
| | - Qian Xiang
- Department of Pharmacy, Peking University First Hospital, No. 6, Da Hong Luo Chang Street, Xicheng District, Beijing, 100034, China.
- Institute of Clinical Pharmacology, Peking University, Beijing, China.
| |
Collapse
|
5
|
Zayed A, Al-Saedi DA, Mensah EO, Kanwugu ON, Adadi P, Ulber R. Fucoidan's Molecular Targets: A Comprehensive Review of Its Unique and Multiple Targets Accounting for Promising Bioactivities Supported by In Silico Studies. Mar Drugs 2023; 22:29. [PMID: 38248653 PMCID: PMC10820140 DOI: 10.3390/md22010029] [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: 11/22/2023] [Revised: 12/28/2023] [Accepted: 12/29/2023] [Indexed: 01/23/2024] Open
Abstract
Fucoidan is a class of multifunctional polysaccharides derived from marine organisms. Its unique and diversified physicochemical and chemical properties have qualified them for potential and promising pharmacological uses in human diseases, including inflammation, tumors, immunity disorders, kidney diseases, and diabetes. Physicochemical and chemical properties are the main contributors to these bioactivities. The previous literature has attributed such activities to its ability to target key enzymes and receptors involved in potential disease pathways, either directly or indirectly, where the anionic sulfate ester groups are mainly involved in these interactions. These findings also confirm the advantageous pharmacological uses of sulfated versus non-sulfated polysaccharides. The current review shall highlight the molecular targets of fucoidans, especially enzymes, and the subsequent responses via either the upregulation or downregulation of mediators' expression in various tissue abnormalities. In addition, in silico studies will be applied to support the previous findings and show the significant contributors. The current review may help in understanding the molecular mechanisms of fucoidan. Also, the findings of this review may be utilized in the design of specific oligomers inspired by fucoidan with the purpose of treating life-threatening human diseases effectively.
Collapse
Affiliation(s)
- Ahmed Zayed
- Institute of Bioprocess Engineering, Rheinland-Pfälzische Technische Universität Kaiserslautern-Landau, Gottlieb-Daimler-Straße 49, 67663 Kaiserslautern, Germany
- Department of Pharmacognosy, College of Pharmacy, Tanta University, El-Guish Street (Medical Campus), Tanta 31527, Egypt
| | - Dalal A. Al-Saedi
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
| | - Emmanuel Ofosu Mensah
- Faculty of Ecotechnology, ITMO University, Lomonosova Street 9, Saint Petersburg 191002, Russia;
| | - Osman Nabayire Kanwugu
- Institute of Chemical Engineering, Ural Federal University, Mira Street 28, Yekaterinburg 620002, Russia;
- ARC Centre of Excellence in Synthetic Biology, School of Natural Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Parise Adadi
- Department of Food Science, University of Otago, Dunedin 9054, New Zealand;
| | - Roland Ulber
- Institute of Bioprocess Engineering, Rheinland-Pfälzische Technische Universität Kaiserslautern-Landau, Gottlieb-Daimler-Straße 49, 67663 Kaiserslautern, Germany
| |
Collapse
|
6
|
Yokomori R, Shirai T, Tsukiji N, Oishi S, Sasaki T, Takano K, Suzuki-Inoue K. C-type lectin-like receptor-2 (CLEC-2) is a key regulator of kappa-carrageenan-induced tail thrombosis model in mice. Platelets 2023; 34:2281941. [PMID: 38010137 DOI: 10.1080/09537104.2023.2281941] [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: 05/18/2023] [Accepted: 11/03/2023] [Indexed: 11/29/2023]
Abstract
Kappa-carrageenan (KCG), which is used to induce thrombosis in laboratory animals for antithrombotic drug screening, can trigger platelet aggregation. However, the cell-surface receptor and related signaling pathways remain unclear. In this study, we investigated the molecular basis of KCG-induced platelet activation using light-transmittance aggregometry, flow cytometry, western blotting, and surface plasmon resonance assays using platelets from platelet receptor-deficient mice and recombinant proteins. KCG-induced tail thrombosis was also evaluated in mice lacking the platelet receptor. We found that KCG induces platelet aggregation with α-granule secretion, activated integrin αIIbβ3, and phosphatidylserine exposure. As this aggregation was significantly inhibited by the Src family kinase inhibitor and spleen tyrosine kinase (Syk) inhibitor, a tyrosine kinase-dependent pathway is required. Platelets exposed to KCG exhibited intracellular tyrosine phosphorylation of Syk, linker activated T cells, and phospholipase C gamma 2. KCG-induced platelet aggregation was abolished in platelets from C-type lectin-like receptor-2 (CLEC-2)-deficient mice, but not in platelets pre-treated with glycoprotein VI-blocking antibody, JAQ1. Surface plasmon resonance assays showed a direct association between murine/human recombinant CLEC-2 and KCG. KCG-induced thrombosis and thrombocytopenia were significantly inhibited in CLEC-2-deficient mice. Our findings show that KCG induces platelet activation via CLEC-2.
Collapse
Affiliation(s)
- Ryohei Yokomori
- Department of Clinical and Laboratory Medicine, Faculty of Medicine, University of Yamanashi, Kofu, Japan
| | - Toshiaki Shirai
- Department of Clinical and Laboratory Medicine, Faculty of Medicine, University of Yamanashi, Kofu, Japan
| | - Nagaharu Tsukiji
- Department of Clinical and Laboratory Medicine, Faculty of Medicine, University of Yamanashi, Kofu, Japan
| | - Saori Oishi
- Department of Clinical and Laboratory Medicine, Faculty of Medicine, University of Yamanashi, Kofu, Japan
| | - Tomoyuki Sasaki
- Department of Clinical and Laboratory Medicine, Faculty of Medicine, University of Yamanashi, Kofu, Japan
| | - Katsuhiro Takano
- Department of Transfusion and Cell Therapy, University of Yamanashi Hospital, Chuo, Japan
| | - Katsue Suzuki-Inoue
- Department of Clinical and Laboratory Medicine, Faculty of Medicine, University of Yamanashi, Kofu, Japan
- Department of Transfusion and Cell Therapy, University of Yamanashi Hospital, Chuo, Japan
| |
Collapse
|
7
|
Dangelmaier CA, Patchin M, Vajipayajula DN, Vari HR, Singh PK, Wright MN, Kostyak JC, Tsygankov AY, Kunapuli SP. Phosphorylation of spleen tyrosine kinase at Y346 negatively regulates ITAM-mediated signaling and function in platelets. J Biol Chem 2023; 299:104865. [PMID: 37268160 PMCID: PMC10320515 DOI: 10.1016/j.jbc.2023.104865] [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: 11/17/2022] [Revised: 04/29/2023] [Accepted: 05/02/2023] [Indexed: 06/04/2023] Open
Abstract
Spleen tyrosine kinase (Syk) is expressed in a variety of hemopoietic cells. Upon phosphorylation of the platelet immunoreceptor-based activation motif of the glycoprotein VI (GPVI)/Fc receptor gamma chain collagen receptor, both the tyrosine phosphorylation and activity of Syk are increased leading to downstream signaling events. Although it has been established that the activity of Syk is regulated by tyrosine phosphorylation, the specific roles of individual phosphorylation sites remain to be elucidated. We observed that Syk Y346 in mouse platelets was still phosphorylated when GPVI-induced Syk activity was inhibited. We then generated Syk Y346F mice and analyzed the effect this mutation exerts on platelet responses. Syk Y346F mice bred normally, and their blood cell count was unaltered. We did observe potentiation of GPVI-induced platelet aggregation and ATP secretion as well as increased phosphorylation of other tyrosines on Syk in the Syk Y346F mouse platelets when compared to WT littermates. This phenotype was specific for GPVI-dependent activation, since it was not seen when AYPGKF, a PAR4 agonist, or 2-MeSADP, a purinergic receptor agonist, was used to activate platelets. Despite a clear effect of Syk Y346F on GPVI-mediated signaling and cellular responses, there was no effect of this mutation on hemostasis as measured by tail-bleeding times, although the time to thrombus formation determined using the ferric chloride injury model was reduced. Thus, our results indicate a significant effect of Syk Y346F on platelet activation and responses in vitro and reveal its complex nature manifesting itself by the diversified translation of platelet activation into physiological responses.
Collapse
Affiliation(s)
- Carol A Dangelmaier
- Department of Cardiovascular Sciences, Sol Sherry Thrombosis Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Margaret Patchin
- Department of Cardiovascular Sciences, Sol Sherry Thrombosis Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Dhruv N Vajipayajula
- Department of Cardiovascular Sciences, Sol Sherry Thrombosis Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Hymavathi Reddy Vari
- Department of Cardiovascular Sciences, Sol Sherry Thrombosis Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Pankaj K Singh
- Department of Cardiovascular Sciences, Sol Sherry Thrombosis Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Monica N Wright
- Department of Cardiovascular Sciences, Sol Sherry Thrombosis Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - John C Kostyak
- Department of Cardiovascular Sciences, Sol Sherry Thrombosis Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Alexander Y Tsygankov
- Department of Cardiovascular Sciences, Sol Sherry Thrombosis Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Satya P Kunapuli
- Department of Cardiovascular Sciences, Sol Sherry Thrombosis Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
8
|
Pereira L, Cotas J. Therapeutic Potential of Polyphenols and Other Micronutrients of Marine Origin. Mar Drugs 2023; 21:323. [PMID: 37367648 DOI: 10.3390/md21060323] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 05/22/2023] [Accepted: 05/25/2023] [Indexed: 06/28/2023] Open
Abstract
Polyphenols are compounds found in various plants and foods, known for their antioxidant and anti-inflammatory properties. Recently, researchers have been exploring the therapeutic potential of marine polyphenols and other minor nutrients that are found in algae, fish and crustaceans. These compounds have unique chemical structures and exhibit diverse biological properties, including anti-inflammatory, antioxidant, antimicrobial and antitumor action. Due to these properties, marine polyphenols are being investigated as possible therapeutic agents for the treatment of a wide variety of conditions, such as cardiovascular disease, diabetes, neurodegenerative diseases and cancer. This review focuses on the therapeutic potential of marine polyphenols and their applications in human health, and also, in marine phenolic classes, the extraction methods, purification techniques and future applications of marine phenolic compounds.
Collapse
Affiliation(s)
- Leonel Pereira
- MARE-Marine and Environmental Sciences Centre/ARNET-Aquatic Research Network, IATV-Institute of Environment, Technology and Life, Department of Life Sciences, University of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal
- Instituto do Ambiente Tecnologia e Vida, Faculdade de Ciências e Tecnologia, Rua Sílvio Lima, 3030-790 Coimbra, Portugal
| | - João Cotas
- MARE-Marine and Environmental Sciences Centre/ARNET-Aquatic Research Network, IATV-Institute of Environment, Technology and Life, Department of Life Sciences, University of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal
| |
Collapse
|
9
|
Fink A, Rohlfing AK, Dicenta V, Schaale D, Kremser M, Laspa Z, Sigle M, Fu X, Pelzer A, Fischer M, Münzer P, Castor T, Müller KAL, Borst O, Lämmerhofer M, Gawaz MP. The Subtilisin-Like Protease Furin Regulates Hemin-Dependent Ectodomain Shedding of Glycoprotein VI. Thromb Haemost 2023. [PMID: 37037200 DOI: 10.1055/s-0043-1768057] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2023]
Abstract
INTRODUCTION Hemolysis results in release of free hemoglobin and hemin liberation from erythrocytes. Hemin has been described to induce platelet activation and to trigger thrombosis. METHODS We evaluated the effect of hemin on platelet function and surface expression of the platelet collagen receptor glycoprotein VI (GPVI). Isolated platelets were stimulated with increasing concentrations of hemin. RESULTS We found that hemin strongly enhanced platelet activation, aggregation, and aggregate formation on immobilized collagen under flow. In contrast, we found that surface expression of GPVI was significantly reduced upon hemin stimulation with high hemin concentrations indicating that hemin-induced loss of surface GPVI does not hinder platelet aggregation. Loss of hemin-induced surface expression of GPVI was caused by shedding of the ectodomain of GPVI as verified by immunoblotting and is independent of the GPVI or CLEC-2 mediated ITAM (immunoreceptor-tyrosine-based-activation-motif) signaling pathway as inhibitor studies revealed. Hemin-induced GPVI shedding was independent of metalloproteinases such as ADAM10 or ADAM17, which were previously described to regulate GPVI degradation. Similarly, concentration-dependent shedding of CD62P was also induced by hemin. Unexpectedly, we found that the subtilisin-like proprotein convertase furin controls hemin-dependent GPVI shedding as shown by inhibitor studies using the specific furin inhibitors SSM3 and Hexa-D-arginine. In the presence of SSM3 and Hexa-D-arginine, hemin-associated GPVI degradation was substantially reduced. Further, SSM3 inhibited hemin-induced but not CRP-XL-induced platelet aggregation and thrombus formation, indicating that furin controls specifically hemin-associated platelet functions. CONCLUSION In summary, we describe a novel mechanism of hemin-dependent GPVI shedding and platelet function mediated by furin.
Collapse
Affiliation(s)
- Annalena Fink
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Anne-Katrin Rohlfing
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Valerie Dicenta
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany
| | - David Schaale
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Marcel Kremser
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Zoi Laspa
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Manuel Sigle
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Xiaoqing Fu
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmaceutical Sciences, University of Tübingen, Tübingen, Germany
| | - Andreas Pelzer
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Melina Fischer
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Patrick Münzer
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Tatsiana Castor
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Karin Anne Lydia Müller
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Oliver Borst
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Michael Lämmerhofer
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmaceutical Sciences, University of Tübingen, Tübingen, Germany
| | - Meinrad Paul Gawaz
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany
| |
Collapse
|
10
|
Shen C, Mackeigan DT, Shoara AA, Xu R, Bhoria P, Karakas D, Ma W, Cerenzia E, Chen Z, Hoard B, Lin L, Lei X, Zhu G, Chen P, Johnson PE, Ni H. Dual roles of fucoidan-GPIbα interaction in thrombosis and hemostasis: implications for drug development targeting GPIbα. JOURNAL OF THROMBOSIS AND HAEMOSTASIS : JTH 2023; 21:1274-1288. [PMID: 36732162 DOI: 10.1016/j.jtha.2022.12.030] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 12/14/2022] [Accepted: 12/27/2022] [Indexed: 01/11/2023]
Abstract
BACKGROUND Platelet GPIbα-von Willebrand factor (VWF) interaction initiates platelet adhesion, activation, and thrombus growth, especially under high shear conditions. Therefore, the GPIb-VWF axis has been suggested as a promising target against arterial thrombosis. The polysaccharide fucoidan has been reported to have opposing prothrombotic and antithrombotic effects; however, its binding mechanism with platelets has not been adequately studied. OBJECTIVE The objective of this study was to explore the mechanism of fucoidan and its hydrolyzed products in thrombosis and hemostasis. METHODS Natural fucoidan was hydrolyzed by using hydrochloric acid and was characterized by using size-exclusion chromatography, UV-visible spectroscopy, and fluorometry techniques. The effects of natural and hydrolyzed fucoidan on platelet aggregation were examined by using platelets from wild-type, VWF and fibrinogen-deficient, GPIbα-deficient, and IL4Rα/GPIbα-transgenic and αIIb-deficient mice and from human beings. Platelet activation markers (P-selectin expression, PAC-1, and fibrinogen binding) and platelet-VWF A1 interaction were measured by using flow cytometry. GPIbα-VWF A1 interaction was evaluated by using enzyme-linked immunosorbent assay. GPIb-IX-induced signal transduction was detected by using western blot. Heparinized whole blood from healthy donors was used to test thrombus formation and growth in a perfusion chamber. RESULTS We found that GPIbα is critical for fucoidan-induced platelet activation. Fucoidan interacted with the extracellular domain of GPIbα and blocked its interaction with VWF but itself could lead to GPIbα-mediated signal transduction and, subsequently, αIIbβ3 activation and platelet aggregation. Conversely, low-molecular weight fucoidan inhibited GPIb-VWF-mediated platelet aggregation, spreading, and thrombus growth at high shear. CONCLUSION Fucoidan-GPIbα interaction may have unique therapeutic potential against bleeding disorders in its high-molecular weight state and protection against arterial thrombosis by blocking GPIb-VWF interaction after fucoidan is hydrolyzed.
Collapse
Affiliation(s)
- Chuanbin Shen
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, M5S 1A1, ON, Canada; Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Shandong, China; Department of Laboratory Medicine, LKSKI-Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Toronto, Canada
| | - Daniel T Mackeigan
- Department of Laboratory Medicine, LKSKI-Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Toronto, Canada; Department of Physiology, University of Toronto, Toronto, Canada
| | - Aron A Shoara
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, M5S 1A1, ON, Canada; Department of Laboratory Medicine, LKSKI-Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Toronto, Canada
| | - Runjia Xu
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, M5S 1A1, ON, Canada; Department of Laboratory Medicine, LKSKI-Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Toronto, Canada
| | - Preeti Bhoria
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, M5S 1A1, ON, Canada; Department of Laboratory Medicine, LKSKI-Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Toronto, Canada; CCOA Therapeutics Inc Toronto, Canada
| | - Danielle Karakas
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, M5S 1A1, ON, Canada; Department of Laboratory Medicine, LKSKI-Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Toronto, Canada
| | - Wenjing Ma
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, M5S 1A1, ON, Canada; Department of Laboratory Medicine, LKSKI-Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Toronto, Canada; CCOA Therapeutics Inc Toronto, Canada
| | - Eric Cerenzia
- Department of Laboratory Medicine, LKSKI-Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Toronto, Canada; Department of Physiology, University of Toronto, Toronto, Canada
| | - ZiYan Chen
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, M5S 1A1, ON, Canada; Department of Laboratory Medicine, LKSKI-Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Toronto, Canada
| | - Brock Hoard
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, M5S 1A1, ON, Canada; Department of Laboratory Medicine, LKSKI-Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Toronto, Canada
| | - Lisha Lin
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, M5S 1A1, ON, Canada; Department of Laboratory Medicine, LKSKI-Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Toronto, Canada; State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, China
| | - Xi Lei
- Department of Laboratory Medicine, LKSKI-Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Toronto, Canada; CCOA Therapeutics Inc Toronto, Canada
| | - Guangheng Zhu
- Department of Laboratory Medicine, LKSKI-Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Toronto, Canada; CCOA Therapeutics Inc Toronto, Canada
| | - Pingguo Chen
- Department of Laboratory Medicine, LKSKI-Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Toronto, Canada; Canadian Blood Services Centre for Innovation, Toronto, Canada
| | - Philip E Johnson
- Department of Chemistry and Centre for Research on Biomolecular Interactions, York University, Toronto, Canada
| | - Heyu Ni
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, M5S 1A1, ON, Canada; Department of Laboratory Medicine, LKSKI-Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Toronto, Canada; Department of Physiology, University of Toronto, Toronto, Canada; CCOA Therapeutics Inc Toronto, Canada; Canadian Blood Services Centre for Innovation, Toronto, Canada; Department of Medicine, University of Toronto, Toronto, Canada.
| |
Collapse
|
11
|
Morán LA, Di Y, Sowa MA, Hermida-Nogueira L, Barrachina MN, Martin E, Clark JC, Mize TH, Eble JA, Moreira D, Pollitt AY, Loza MI, Domínguez E, Watson SP, García Á. Katacine Is a New Ligand of CLEC-2 that Acts as a Platelet Agonist. Thromb Haemost 2022; 122:1361-1368. [PMID: 35170009 PMCID: PMC9393086 DOI: 10.1055/a-1772-1069] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2022]
Abstract
BACKGROUND CLEC-2 is a platelet receptor with an important role in thromboinflammation but a minor role in hemostasis. Two endogenous ligands of CLEC-2 have been identified, the transmembrane protein podoplanin and iron-containing porphyrin hemin, which is formed following hemolysis from red blood cells. Other exogenous ligands such as rhodocytin have contributed to our understanding of the role of CLEC-2. OBJECTIVES To identify novel CLEC-2 small-molecule ligands to aid therapeutic targeting of CLEC-2. METHODS ALPHA screen technology has been used for the development of a high-throughput screening (HTS) assay recapitulating the podoplanin-CLEC-2 interaction. Light transmission aggregometry was used to evaluate platelet aggregation. Immunoprecipitation and western blot were used to evaluate direct phosphorylation of CLEC-2 and downstream protein phosphorylation. Autodock vina software was used to predict the molecular binding site of katacine and mass spectrometry to determine the polymeric nature of the ligand. RESULTS AND CONCLUSION We developed a CLEC-2-podoplanin interaction assay in a HTS format and screened 5,016 compounds from a European Union-open screen library. We identified katacine, a mixture of polymers of proanthocyanidins, as a novel ligand for CLEC-2 and showed that it induces platelet aggregation and CLEC-2 phosphorylation via Syk and Src kinases. Platelet aggregation induced by katacine is inhibited by the anti-CLEC-2 monoclonal antibody fragment AYP1 F(ab)'2. Katacine is a novel nonprotein ligand of CLEC-2 that could contribute to a better understanding of CLEC-2 activation in human platelets.
Collapse
Affiliation(s)
- Luis A Morán
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidade de Santiago de Compostela, and Instituto de Investigación Sanitaria de Santiago (IDIS), Santiago de Compostela, Spain.,Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Ying Di
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Marcin A Sowa
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidade de Santiago de Compostela, and Instituto de Investigación Sanitaria de Santiago (IDIS), Santiago de Compostela, Spain.,Institute for Cardiovascular and Metabolic Research (ICMR), School of Biological Sciences, University of Reading, Reading, United Kingdom
| | - Lidia Hermida-Nogueira
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidade de Santiago de Compostela, and Instituto de Investigación Sanitaria de Santiago (IDIS), Santiago de Compostela, Spain
| | - María N Barrachina
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidade de Santiago de Compostela, and Instituto de Investigación Sanitaria de Santiago (IDIS), Santiago de Compostela, Spain
| | - Eleyna Martin
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Joanne C Clark
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Todd H Mize
- Advanced Mass Spectrometry Facility, Biosciences, College of Life and Environmental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Johannes A Eble
- Institute for Physiological Chemistry and Pathobiochemistry, University of Münster, Münster, Germany
| | - David Moreira
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidade de Santiago de Compostela, and Instituto de Investigación Sanitaria de Santiago (IDIS), Santiago de Compostela, Spain
| | - Alice Y Pollitt
- Institute for Cardiovascular and Metabolic Research (ICMR), School of Biological Sciences, University of Reading, Reading, United Kingdom
| | - María I Loza
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidade de Santiago de Compostela, and Instituto de Investigación Sanitaria de Santiago (IDIS), Santiago de Compostela, Spain
| | - Eduardo Domínguez
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidade de Santiago de Compostela, and Instituto de Investigación Sanitaria de Santiago (IDIS), Santiago de Compostela, Spain
| | - Steve P Watson
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Ángel García
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidade de Santiago de Compostela, and Instituto de Investigación Sanitaria de Santiago (IDIS), Santiago de Compostela, Spain
| |
Collapse
|
12
|
Kostyak JC, Mauri B, Dangelmaier C, Vari HR, Patel A, Wright M, Reddy H, Tsygankov AY, Kunapuli SP. Phosphorylation on Syk Y342 is important for both ITAM and hemITAM signaling in platelets. J Biol Chem 2022; 298:102189. [PMID: 35753354 PMCID: PMC9287148 DOI: 10.1016/j.jbc.2022.102189] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 06/09/2022] [Accepted: 06/10/2022] [Indexed: 11/29/2022] Open
Abstract
Immune cells express receptors bearing an immune tyrosine activation motif (ITAM) containing two YXXL motifs or hemITAMs containing only one YXXL motif. Phosphorylation of the ITAM/hemITAM is mediated by Src family kinases allowing for the binding and activation of spleen tyrosine kinase (Syk). It is believed that Syk must be phosphorylated on tyrosine residues for activation, and Tyr342, а conserved tyrosine in the interdomain B region, has been shown to be critical for regulating Syk in FcεR1-activated mast cells. Syk is a key mediator of signaling pathways downstream of several platelet pathways including the ITAM bearing glycoprotein VI (GPVI)/Fc receptor gamma chain collagen receptor and the hemITAM containing C-type lectin-like receptor-2 (CLEC-2). Since platelet activation is a crucial step in both hemostasis and thrombosis, we evaluated the importance of Syk Y342 in these processes by producing an Syk Y342F knock-in mouse. When using a CLEC-2 antibody as an agonist, reduced aggregation and secretion were observed in Syk Y342F mouse platelets when compared with control mouse platelets. Platelet reactivity was also reduced in response to the GPVI agonist collagen-related peptide. Signaling initiated by either GPVI or CLEC-2 was also greatly inhibited, including Syk Y519/520 phosphorylation. Hemostasis, as measured by tail bleeding time, was not altered in Syk Y342F mice, but thrombus formation in response to FeCl3 injury was prolonged in Syk Y342F mice. These data demonstrate that phosphorylation of Y342 on Syk following stimulation of either GPVI or CLEC-2 receptors is important for the ability of Syk to transduce a signal.
Collapse
Affiliation(s)
- John C Kostyak
- Sol Sherry Thrombosis Research Center and Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Benjamin Mauri
- Sol Sherry Thrombosis Research Center and Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Carol Dangelmaier
- Sol Sherry Thrombosis Research Center and Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Hymavathi Reddy Vari
- Sol Sherry Thrombosis Research Center and Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Akruti Patel
- Sol Sherry Thrombosis Research Center and Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Monica Wright
- Sol Sherry Thrombosis Research Center and Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Haritha Reddy
- Sol Sherry Thrombosis Research Center and Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Alexander Y Tsygankov
- Sol Sherry Thrombosis Research Center and Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Satya P Kunapuli
- Sol Sherry Thrombosis Research Center and Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
13
|
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
|
14
|
Zhang L, Zhu Y, Wei X, Chen X, Li Y, Zhu Y, Xia J, Huang Y, Huang Y, Wang J, Pang Z. Nanoplateletsomes restrain metastatic tumor formation through decoy and active targeting in a preclinical mouse model. Acta Pharm Sin B 2022; 12:3427-3447. [PMID: 35967283 PMCID: PMC9366539 DOI: 10.1016/j.apsb.2022.01.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 12/16/2021] [Accepted: 12/30/2021] [Indexed: 11/28/2022] Open
Abstract
Platelets buoy up cancer metastasis via arresting cancer cells, enhancing their adhesion, and facilitating their extravasation through the vasculature. When deprived of intracellular and granular contents, platelet decoys could prevent metastatic tumor formation. Inspired by these, we developed nanoplatesomes by fusing platelet membranes with lipid membranes (P-Lipo) to restrain metastatic tumor formation more efficiently. It was shown nanoplateletsomes bound with circulating tumor cells (CTC) efficiently, interfered with CTC arrest by vessel endothelial cells, CTC extravasation through endothelial layers, and epithelial-mesenchymal transition of tumor cells as nanodecoys. More importantly, in the mouse breast tumor metastasis model, nanoplateletsomes could decrease CTC survival in the blood and counteract metastatic tumor growth efficiently by inhibiting the inflammation and suppressing CTC escape. Therefore, nanoplatelesomes might usher in a new avenue to suppress lung metastasis.
Collapse
Affiliation(s)
- Longlong Zhang
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China
| | - Yuefei Zhu
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Xunbin Wei
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Xing Chen
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China
| | - Yang Li
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China
| | - Ying Zhu
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Jiaxuan Xia
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yiheng Huang
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yongzhuo Huang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jianxin Wang
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China
- Institute of Materia Medica, Academy of Chinese and Western Integrative Medicine, Fudan University, Shanghai 201203, China
- Corresponding authors.
| | - Zhiqing Pang
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China
- Corresponding authors.
| |
Collapse
|
15
|
Montague SJ, Patel P, Martin EM, Slater A, Quintanilla LG, Perrella G, Kardeby C, Nagy M, Mezzano D, Mendes PM, Watson SP. Platelet activation by charged ligands and nanoparticles: platelet glycoprotein receptors as pattern recognition receptors. Platelets 2021; 32:1018-1030. [PMID: 34266346 DOI: 10.1080/09537104.2021.1945571] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 06/10/2021] [Accepted: 06/15/2021] [Indexed: 02/08/2023]
Abstract
Charge interactions play a critical role in the activation of the innate immune system by damage- and pathogen-associated molecular pattern receptors. The ability of these receptors to recognize a wide spectrum of ligands through a common mechanism is critical in host defense. In this article, we argue that platelet glycoprotein receptors that signal through conserved tyrosine-based motifs function as pattern recognition receptors (PRRs) for charged endogenous and exogenous ligands, including sulfated polysaccharides, charged proteins and nanoparticles. This is exemplified by GPVI, CLEC-2 and PEAR1 which are activated by a wide spectrum of endogenous and exogenous ligands, including diesel exhaust particles, sulfated polysaccharides and charged surfaces. We propose that this mechanism has evolved to drive rapid activation of platelets at sites of injury, but that under some conditions it can drive occlusive thrombosis, for example, when blood comes into contact with infectious agents or toxins. In this Opinion Article, we discuss mechanisms behind charge-mediated platelet activation and opportunities for designing nanoparticles and related agents such as dendrimers as novel antithrombotics.
Collapse
Affiliation(s)
- Samantha J Montague
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Pushpa Patel
- School of Chemical Engineering, College of Engineering and Physical Sciences, University of Birmingham, Birmingham, UK
- Centre of Membrane Proteins and Receptors, Universities of Birmingham and Nottingham, UK
| | - Eleyna M Martin
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Alexandre Slater
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Lourdes Garcia Quintanilla
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Gina Perrella
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- Departments of Biochemistry and Internal Medicine, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands
| | - Caroline Kardeby
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Magdolna Nagy
- Departments of Biochemistry and Internal Medicine, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands
| | - Diego Mezzano
- Laboratorio de Trombosis y Hemostasia, School of Medicine, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Paula M Mendes
- School of Chemical Engineering, College of Engineering and Physical Sciences, University of Birmingham, Birmingham, UK
- Centre of Membrane Proteins and Receptors, Universities of Birmingham and Nottingham, UK
| | - Steve P Watson
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- Centre of Membrane Proteins and Receptors, Universities of Birmingham and Nottingham, UK
| |
Collapse
|
16
|
Yao Y, Yim EKF. Fucoidan for cardiovascular application and the factors mediating its activities. Carbohydr Polym 2021; 270:118347. [PMID: 34364596 PMCID: PMC10429693 DOI: 10.1016/j.carbpol.2021.118347] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 06/12/2021] [Accepted: 06/12/2021] [Indexed: 12/17/2022]
Abstract
Fucoidan is a sulfated polysaccharide with various bioactivities. The application of fucoidan in cancer treatment, wound healing, and food industry has been extensively studied. However, the therapeutic value of fucoidan in cardiovascular diseases has been less explored. Increasing number of investigations in the past years have demonstrated the effects of fucoidan on cardiovascular system. In this review, we will focus on the bioactivities related to cardiovascular applications, for example, the modulation functions of fucoidan on coagulation system, inflammation, and vascular cells. Factors mediating those activities will be discussed in detail. Current therapeutic strategies and future opportunities and challenges will be provided to inspire and guide further research.
Collapse
Affiliation(s)
- Yuan Yao
- Department of Chemical Engineering, University of Waterloo, 200 University Avenue West, Waterloo, ON N2L 3G1, Canada.
| | - Evelyn K F Yim
- Department of Chemical Engineering, University of Waterloo, 200 University Avenue West, Waterloo, ON N2L 3G1, Canada; Waterloo Institute for Nanotechnology, University of Waterloo, 200 University Avenue West, Waterloo, ON N2L 3G1, Canada; Center for Biotechnology and Bioengineering, University of Waterloo, 200 University Avenue West, Waterloo, ON N2L 3G1, Canada.
| |
Collapse
|
17
|
Martin EM, Zuidscherwoude M, Morán LA, Di Y, García A, Watson SP. The structure of CLEC-2: mechanisms of dimerization and higher-order clustering. Platelets 2021; 32:733-743. [PMID: 33819136 DOI: 10.1080/09537104.2021.1906407] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 03/04/2021] [Accepted: 03/16/2021] [Indexed: 02/07/2023]
Abstract
The platelet C-type lectin-like receptor CLEC-2 drives inflammation-driven venous thrombosis in mouse models of thrombo-inflammatory disease with a minimal effect on hemostasis identifying it as a target for a new class of antiplatelet agent. Here, we discuss how the protein structure and dynamic arrangement of CLEC-2 on the platelet membrane helps the receptor, which has a single YxxL motif (known as a hemITAM), to trigger intracellular signaling. CLEC-2 exists as a monomer and homo-dimer within resting platelets and forms higher-order oligomers following ligand activation, a process that is mediated by the multivalent nature of its ligands and the binding of the tandem SH2 domains of Syk to the phosphorylated hemITAM and concomitantly to PIP2 or PIP3 to localize it to the membrane. We propose that a low level of active Syk is present at the membrane in resting platelets due to phosphorylation by Src family kinases and that clustering of receptors disturbs the equilibrium between kinases and phosphatases, triggering phosphorylation of the CLEC-2 hemITAM and recruitment of Syk. Knowledge of the structure of CLEC-2 and the mechanism of platelet activation has important implications for development of therapeutics.
Collapse
Affiliation(s)
- Eleyna M Martin
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham
| | - Malou Zuidscherwoude
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham
| | - Luis A Morán
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham
- Centre for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidade De Santiago De Compostela, Spain
| | - Ying Di
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham
| | - Angel García
- Centre for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidade De Santiago De Compostela, Spain
| | - Steve P Watson
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham
- Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, The Midlands
| |
Collapse
|
18
|
Pérez-Campos Mayoral L, Hernández-Huerta MT, Papy-García D, Barritault D, Zenteno E, Sánchez Navarro LM, Pérez-Campos Mayoral E, Matias Cervantes CA, Martínez Cruz M, Mayoral Andrade G, López Cervantes M, Vázquez Martínez G, López Sánchez C, Pina Canseco S, Martínez Cruz R, Pérez-Campos E. Immunothrombotic dysregulation in chagas disease and COVID-19: a comparative study of anticoagulation. Mol Cell Biochem 2021; 476:3815-3825. [PMID: 34110554 PMCID: PMC8190527 DOI: 10.1007/s11010-021-04204-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 06/03/2021] [Indexed: 12/27/2022]
Abstract
Chagas and COVID-19 are diseases caused by Trypanosoma cruzi and SARS-CoV-2, respectively. These diseases present very different etiological agents despite showing similarities such as susceptibility/risk factors, pathogen-associated molecular patterns (PAMPs), recognition of glycosaminoglycans, inflammation, vascular leakage hypercoagulability, microthrombosis, and endotheliopathy; all of which suggest, in part, treatments with similar principles. Here, both diseases are compared, focusing mainly on the characteristics related to dysregulated immunothrombosis. Given the in-depth investigation of molecules and mechanisms related to microthrombosis in COVID-19, it is necessary to reconsider a prompt treatment of Chagas disease with oral anticoagulants.
Collapse
Affiliation(s)
- Laura Pérez-Campos Mayoral
- Centro de Investigación Facultad de Medicina UNAM-UABJO, Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca, 68020, México
| | | | | | | | - Edgar Zenteno
- Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, 04360, México
| | | | - Eduardo Pérez-Campos Mayoral
- Centro de Investigación Facultad de Medicina UNAM-UABJO, Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca, 68020, México
| | | | | | - Gabriel Mayoral Andrade
- Centro de Investigación Facultad de Medicina UNAM-UABJO, Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca, 68020, México
| | | | | | - Claudia López Sánchez
- Tecnológico Nacional de México / Instituto Tecnológico de Oaxaca, Oaxaca, 68030, México
| | - Socorro Pina Canseco
- Centro de Investigación Facultad de Medicina UNAM-UABJO, Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca, 68020, México
| | - Ruth Martínez Cruz
- Centro de Investigación Facultad de Medicina UNAM-UABJO, Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca, 68020, México
| | - Eduardo Pérez-Campos
- Tecnológico Nacional de México / Instituto Tecnológico de Oaxaca, Oaxaca, 68030, México. .,Laboratorio de Patología Clínica "Eduardo Pérez Ortega", Oaxaca, 68000, México.
| |
Collapse
|
19
|
Meng D, Luo M, Liu B. The Role of CLEC-2 and Its Ligands in Thromboinflammation. Front Immunol 2021; 12:688643. [PMID: 34177942 PMCID: PMC8220156 DOI: 10.3389/fimmu.2021.688643] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 05/24/2021] [Indexed: 12/17/2022] Open
Abstract
C-type lectin-like receptor 2 (CLEC-2, also known as CLEC-1b) is expressed on platelets, Kupffer cells and other immune cells, and binds to various ligands including the mucin-like protein podoplanin (PDPN). The role of CLEC-2 in infection and immunity has become increasingly evident in recent years. CLEC-2 is involved in platelet activation, tumor cell metastasis, separation of blood/lymphatic vessels, and cerebrovascular patterning during embryonic development. In this review, we have discussed the role of CLEC-2 in thromboinflammation, and focused on the recent research.
Collapse
Affiliation(s)
- Danyang Meng
- Department of Neurology, Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Man Luo
- Department of Neurology, Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Beibei Liu
- Department of Central Laboratory, Affiliated Hospital of Jiaxing University, Jiaxing, China
| |
Collapse
|
20
|
Kardeby C, Damaskinaki FN, Sun Y, Watson SP. Is the endogenous ligand for PEAR1 a proteoglycan: clues from the sea. Platelets 2020; 32:779-785. [PMID: 33356751 DOI: 10.1080/09537104.2020.1863938] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Platelet Endothelial Aggregation Receptor 1 (PEAR1) is an orphan receptor of unknown function which mediates powerful activation of platelets and endothelial cells in response to crosslinking by antibodies and sulfated polysaccharides belonging to the dextran and fucoidan families. PEAR1 is a single transmembrane protein composed of 15 epidermal growth factor-like repeat sequences and with a conserved binding motif, YXXM, which when phosphorylated binds to phosphoinositide 3-kinase (PI3K). The 13th of the repeats has a heparin-binding sequence that is the site of interaction with the sulfated fucoidans and the only known endogenous ligand FcεRIα. Crosslinking of PEAR1 drives Src family kinase phosphorylation of the cytosolic tail leading to binding and activation of PI3K. In this Opinion Article, we summarize the literature on PEAR1 expression, structure and signaling, and the search for further endogenous ligands. We highlight one article in which phosphorylation of a 150 kDa platelet protein by heparin-containing ligands has been reported and propose that PEAR1 is a receptor for one or more glycosaminoglycan-conjugated proteins (proteoglycans). The up-regulation of PEAR1 at sites of inflammation in the vasculature and its role in angiogenesis suggests a role in the interplay of inflammation, platelets, coagulation, and thromboinflammation. We speculate that this may explain the link between single nucleotide variants in PEAR1 and cardiovascular disease.
Collapse
Affiliation(s)
- Caroline Kardeby
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, West Midlands, UK
| | - Foteini-Nafsika Damaskinaki
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, West Midlands, UK.,Biodiscovery Institute, University of Nottingham, University Park, Nottingham, East Midlands, UK.,Centre of Membrane Proteins and Receptors, Universities of Birmingham and Nottingham, The Midlands, UK
| | - Yi Sun
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, West Midlands, UK.,Centre of Membrane Proteins and Receptors, Universities of Birmingham and Nottingham, The Midlands, UK
| | - Stephen P Watson
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, West Midlands, UK.,Centre of Membrane Proteins and Receptors, Universities of Birmingham and Nottingham, The Midlands, UK
| |
Collapse
|
21
|
Wu S, Yang W, Cheng C, Lin K, Sampurna BP, Chan S, Yuh C. Low molecular weight fucoidan inhibits hepatocarcinogenesis and nonalcoholic fatty liver disease in zebrafish via ASGR/STAT3/HNF4A signaling. Clin Transl Med 2020; 10:e252. [PMID: 33377648 PMCID: PMC7752165 DOI: 10.1002/ctm2.252] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 11/03/2020] [Accepted: 11/28/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Hepatocellular carcinoma ranks fourth in cancer-related mortality currently lacks effective therapeutics. Fucoidan is sulfated polysaccharide that is mainly found in brown seaweeds. In this study, we investigated the effects and mechanisms of low molecular weight fucoidan (i.e. oligo-fucoidan [OF]) preventing hepatocarcinogenesis. METHODS We used [HBx,src], [HBx,src,p53-/+ ], and [CD36] transgenic zebrafish liver cancer model treated with OF, and performed molecular and histopathological analysis. Transcriptomic and pathways analysis was performed. RESULTS Decreased expression of lipogenic enzymes, fibrosis markers, and cell cycle/proliferation markers by OF in [HBx,src] and [HBx,src,p53-/+ ] transgenic fish. Liver fibrosis was decreased as revealed by Sirius Red staining, and the liver cancer formation was eventually reduced by feeding OF. OF was also found to be capable of reducing lipid accumulation and cancer formation in non-B non-C Hepatocellular carcinoma (HCC) model in CD36 transgenic zebrafish. Whole-genome expression analysis showed that 661 genes were up-regulated, and 451 genes were downregulated by feeding OF. Upregulated genes were mostly found in protein transporter activity, and downregulated genes were enriched with response to extracellular stimulus and metal binding in gene ontology analysis. The driver gene was HNF4A revealed by NetworkAnalyst from OF differential regulated genes at various insults. OF is able to bind the asialoglycoprotein receptor (ASGR) in hepatoma cells, and increased the phosphorylation of signal transducer and activator of transcription 3 (STAT3) in both hepatoma cells and [HBx,src,p53-/+ ] transgenic fish liver cancer model. Using chromatin-immunoprecipitation, we found pSTAT3 could associate with the P1 promoter of HNF4A. Knockdown of either ASGR or HNF4A reversed OF mediated anti-cancer cell proliferation. CONCLUSIONS Taken together, we provide evidence that OF exhibits the anti-HCC, anti-steatosis, and anti-fibrosis effect for liver in zebrafish models, and the anti-cancer potential of OF attributed to the binding to ASGR and activation of STAT3/HNF4A signaling. OF might be potentially valuable for the management of HCC.
Collapse
Affiliation(s)
- Szu‐Yuan Wu
- Department of Food Nutrition and Health Biotechnology, College of Medical and Health ScienceAsia UniversityTaichung41354Taiwan
- Big Data Center, Lo‐Hsu Medical FoundationLotung Poh‐Ai HospitalYilan26546Taiwan
- Division of Radiation Oncology, Lo‐Hsu Medical FoundationLotung Poh‐Ai HospitalYilan26546Taiwan
- Department of Healthcare Administration, College of Medical and Health ScienceAsia UniversityTaichungTaiwan
- Graduate Institute of Business AdministrationFu Jen Catholic UniversityTaipei24205Taiwan
| | - Wan‐Yu Yang
- Institute of Molecular and Genomic MedicineNational Health Research InstitutesZhunan35053Taiwan
| | - Chun‐Chia Cheng
- Institute of Molecular and Genomic MedicineNational Health Research InstitutesZhunan35053Taiwan
- Radiation Biology Research Center, Institute for Radiological ResearchChang Gung University/Chang Gung Memorial Hospital at LinkouTaoyuan33302Taiwan
| | - Kuan‐Hao Lin
- Institute of Molecular and Genomic MedicineNational Health Research InstitutesZhunan35053Taiwan
| | | | - Suat‐Ming Chan
- Institute of Molecular and Genomic MedicineNational Health Research InstitutesZhunan35053Taiwan
| | - Chiou‐Hwa Yuh
- Institute of Molecular and Genomic MedicineNational Health Research InstitutesZhunan35053Taiwan
- Institute of Bioinformatics and Structural BiologyNational Tsing‐Hua UniversityHsinchu30013Taiwan
- Department of Biological Science & TechnologyNational Chiao Tung UniversityHsinchu30010Taiwan
- PhD Program in Environmental and Occupational MedicineKaohsiung Medical UniversityKaohsiung80708Taiwan
| |
Collapse
|
22
|
Koukouraki P, Tsoupras A, Sotiroudis G, Demopoulos CA, Sotiroudis TG. Antithrombotic properties of Spirulina extracts against platelet-activating factor and thrombin. FOOD BIOSCI 2020. [DOI: 10.1016/j.fbio.2020.100686] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
23
|
Lin L, Yang L, Chen J, Zhou L, Li S, Gao N, Zhao J. High-molecular-weight fucosylated glycosaminoglycan induces human platelet aggregation depending on αIIbβ3 and platelet secretion. Platelets 2020; 32:975-983. [PMID: 32970503 DOI: 10.1080/09537104.2020.1820976] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Affiliation(s)
- Lisha Lin
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lian Yang
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China
| | - Jun Chen
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China
| | - Lutan Zhou
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- School of Chemical Science and Technology, School of Chemical Science and Technology, Yunnan University, Kunming, Yunnan Province 650091, China
| | - Sujuan Li
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Na Gao
- School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan 430074, China
| | - Jinhua Zhao
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China
- School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan 430074, China
| |
Collapse
|
24
|
Lin Z, Tan X, Zhang Y, Li F, Luo P, Liu H. Molecular Targets and Related Biologic Activities of Fucoidan: A Review. Mar Drugs 2020; 18:E376. [PMID: 32707775 PMCID: PMC7459501 DOI: 10.3390/md18080376] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 07/17/2020] [Accepted: 07/20/2020] [Indexed: 02/06/2023] Open
Abstract
Fucoidan-a marine natural active polysaccharide derived from brown algae with a variety of medicinal activities and low toxicity-has been used as clinical drug for renal diseases for nearly 20 years. The pharmacological mechanism of fucoidan has been well-investigated, based on target molecules and downstream signaling pathways. This review summarizes some important molecular targets of fucoidan and its related biologic activities, including scavenger receptor (SR), Toll-like receptors (TLRs), C-type lectin (CLEC) and some newly found target molecules, which may be beneficial for further understanding the pharmacological mechanism of fucoidan and discovering its new functions, as well as developing related clinical or adjuvant drugs and functional preparations.
Collapse
Affiliation(s)
| | | | | | | | | | - Huazhong Liu
- Faculty of Chemistry & Environment Sciences, Guangdong Ocean University, Zhanjiang 524088, China; (Z.L.); (X.T.); (Y.Z.); (F.L.); (P.L.)
| |
Collapse
|
25
|
Wu SY, Yang WY, Cheng CC, Hsiao MC, Tsai SL, Lin HK, Lin KH, Yuh CH. Low Molecular Weight Fucoidan Prevents Radiation-Induced Fibrosis and Secondary Tumors in a Zebrafish Model. Cancers (Basel) 2020; 12:cancers12061608. [PMID: 32570707 PMCID: PMC7353073 DOI: 10.3390/cancers12061608] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 06/12/2020] [Accepted: 06/16/2020] [Indexed: 12/15/2022] Open
Abstract
Radiotherapy often causes unwanted side effects such as radiation-induced fibrosis and second malignancies. Fucoidan, a sulfated polysaccharide extracted from brown seaweed, has many biological effects including anti-inflammation and anti-tumor. In the present study, we investigated the radioprotective effect of Oligo-Fucoidan (OF) using a zebrafish animal model. Adult zebrafish of wild-type and transgenic fish with hepatocellular carcinoma were orally fed with Oligo-Fucoidan before irradiation. Quantitative PCR, Sirius red stain, hematoxylin, and eosin stain were used for molecular and pathological analysis. Whole genomic microarrays were used to discover the global program of gene expression after Oligo-Fucoidan treatment and identified distinct classes of up- and downregulated genes/pathways during this process. Using Oligo-Fucoidan oral gavage in adult wild-type zebrafish, we found Oligo-Fucoidan pretreatment decreased irradiation-induced fibrosis in hepatocyte. Using hepatitis B virus X antigen (HBx), Src and HBx, Src, p53−/+ transgenic zebrafish liver cancer model, we found that Oligo-Fucoidan pretreatment before irradiation could lower the expression of lipogenic factors and enzymes, fibrosis, and cell cycle/proliferation markers, which eventually reduced formation of liver cancer compared to irradiation alone. Gene ontology analysis revealed that Oligo-Fucoidan pretreatment increased the expression of genes involved in oxidoreductase activity in zebrafish irradiation. Oligo-Fucoidan also decreased the expression of genes involved in transferase activity in wild-type fish without irradiation (WT), nuclear outer membrane-endoplasmic reticulum membrane network, and non-homologous end-joining (NHEJ) in hepatocellular carcinoma (HCC) transgenic fish. Rescue of those genes can prevent liver cancer formation. Conclusions: Our results provide evidence for the ability of Oligo-Fucoidan to prevent radiation-induced fibrosis and second malignancies in zebrafish.
Collapse
Affiliation(s)
- Szu-Yuan Wu
- Department of Food Nutrition and Health Biotechnology, College of Medical and Health Science, Asia University, Taichung 42354, Taiwan;
- Division of Radiation Oncology, Department of Medicine, Lo-Hsu Medical Foundation, Lotung Poh-Ai Hospital, Yilan 265, Taiwan
- Big Data Center, Lo-Hsu Medical Foundation, Lotung Poh-Ai Hospital, Yilan 265, Taiwan
- Department of Healthcare Administration, College of Medical and Health Science, Asia University, Taichung 41354, Taiwan
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Wan-Yu Yang
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan; (W.-Y.Y.); (C.-C.C.); (S.-L.T.); (H.-K.L.); (K.-H.L.)
| | - Chun-Chia Cheng
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan; (W.-Y.Y.); (C.-C.C.); (S.-L.T.); (H.-K.L.); (K.-H.L.)
- Radiation Biology Research Center, Institute for Radiological Research, Chang Gung University/Chang Gung Memorial Hospital at Linkou, Taoyuan 33302, Taiwan
| | - Ming-Chen Hsiao
- Research and Development Center, Hi-Q Marine Biotech International Ltd., Songshan District, Taipei 10561, Taiwan;
| | - Shin-Lin Tsai
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan; (W.-Y.Y.); (C.-C.C.); (S.-L.T.); (H.-K.L.); (K.-H.L.)
| | - Hua-Kuo Lin
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan; (W.-Y.Y.); (C.-C.C.); (S.-L.T.); (H.-K.L.); (K.-H.L.)
| | - Kuan-Hao Lin
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan; (W.-Y.Y.); (C.-C.C.); (S.-L.T.); (H.-K.L.); (K.-H.L.)
| | - Chiou-Hwa Yuh
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan; (W.-Y.Y.); (C.-C.C.); (S.-L.T.); (H.-K.L.); (K.-H.L.)
- Institute of Bioinformatics and Structural Biology, National Tsing-Hua University, Hsinchu 30013, Taiwan
- Department of Biological Science & Technology, National Chiao Tung University, Hsinchu 30010, Taiwan
- Program in Environmental and Occupational Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Correspondence: ; Tel.: +886-37-246-166 (ext. 3538); Fax: +886-37-586-459
| |
Collapse
|
26
|
Transcriptomically Revealed Oligo-Fucoidan Enhances the Immune System and Protects Hepatocytes via the ASGPR/STAT3/HNF4A Axis. Biomolecules 2020; 10:biom10060898. [PMID: 32545625 PMCID: PMC7355575 DOI: 10.3390/biom10060898] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 06/09/2020] [Accepted: 06/10/2020] [Indexed: 12/12/2022] Open
Abstract
Oligo-fucoidan, a sulfated polysaccharide extracted from brown seaweed, exhibits anti-inflammatory and anti-tumor effects. However, the knowledge concerning the detailed mechanism of oligo-fucoidan on liver cells is obscure. In this study, we investigate the effect of oligo-fucoidan in normal hepatocytes by transcriptomic analysis. Using an oligo-fucoidan oral gavage in wild-type adult zebrafish, we find that oligo-fucoidan pretreatment enhances the immune system and anti-viral genes in hepatocytes. Oligo-fucoidan pretreatment also decreases the expression of lipogenic enzymes and liver fibrosis genes. Using pathway analysis, we identify hepatocyte nuclear factor 4 alpha (HNF4A) to be the potential driver gene. We further investigate whether hepatocyte nuclear factor 4 alpha (HNF4A) could be induced by oligo-fucoidan and the underlying mechanism. Therefore, a normal hepatocyte clone 9 cell as an in vitro model was used. We demonstrate that oligo-fucoidan increases cell viability, Cyp3a4 activity, and Hnf4a expression in clone 9 cells. We further demonstrate that oligo-fucoidan might bind to asialoglycoprotein receptors (ASGPR) in normal hepatocytes through both in vitro and in vivo competition assays. This binding, consequently activating the signal transducer and activator of transcription 3 (STAT3), increases the expression of the P1 isoform of HNF4A. According to our data, we suggest that oligo-fucoidan not only enhances the gene expression associated with anti-viral ability and immunity, but also increases P1-HNF4A levels through ASGPR/STAT3 axis, resulting in protecting hepatocytes.
Collapse
|
27
|
Martyanov AA, Balabin FA, Dunster JL, Panteleev MA, Gibbins JM, Sveshnikova AN. Control of Platelet CLEC-2-Mediated Activation by Receptor Clustering and Tyrosine Kinase Signaling. Biophys J 2020; 118:2641-2655. [PMID: 32396849 DOI: 10.1016/j.bpj.2020.04.023] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 03/06/2020] [Accepted: 04/13/2020] [Indexed: 02/07/2023] Open
Abstract
Platelets are blood cells responsible for vascular integrity preservation. The activation of platelet receptor C-type lectin-like receptor II-type (CLEC-2) could partially mediate the latter function. Although this receptor is considered to be of importance for hemostasis, the rate-limiting steps of CLEC-2-induced platelet activation are not clear. Here, we aimed to investigate CLEC-2-induced platelet signal transduction using computational modeling in combination with experimental approaches. We developed a stochastic multicompartmental computational model of CLEC-2 signaling. The model described platelet activation beginning with CLEC-2 receptor clustering, followed by Syk and Src family kinase phosphorylation, determined by the cluster size. Active Syk mediated linker adaptor for T cell protein phosphorylation and membrane signalosome formation, which resulted in the activation of Bruton's tyrosine kinase, phospholipase and phosphoinositide-3-kinase, calcium, and phosphoinositide signaling. The model parameters were assessed from published experimental data. Flow cytometry, total internal reflection fluorescence and confocal microscopy, and western blotting quantification of the protein phosphorylation were used for the assessment of the experimental dynamics of CLEC-2-induced platelet activation. Analysis of the model revealed that the CLEC-2 receptor clustering leading to the membrane-based signalosome formation is a critical element required for the accurate description of the experimental data. Both receptor clustering and signalosome formation are among the rate-limiting steps of CLEC-2-mediated platelet activation. In agreement with these predictions, the CLEC-2-induced platelet activation, but not activation mediated by G-protein-coupled receptors, was strongly dependent on temperature conditions and cholesterol depletion. Besides, the model predicted that CLEC-2-induced platelet activation results in cytosolic calcium spiking, which was confirmed by single-platelet total internal reflection fluorescence microscopy imaging. Our results suggest a refined picture of the platelet signal transduction network associated with CLEC-2. We show that tyrosine kinase activation is not the only rate-limiting step in CLEC-2-induced activation of platelets. Translocation of receptor-agonist complexes to the signaling region and linker adaptor for T cell signalosome formation in this region are limiting CLEC-2-induced activation as well.
Collapse
Affiliation(s)
- Alexey A Martyanov
- Center for Theoretical Problems of Physico-chemical Pharmacology, Russian Academy of Sciences, Moscow, Russia; Dmitry Rogachev National Medical Research Centre of Pediatric Hematology, Oncology and Immunology, Moscow, Russia; Institute for Biochemical Physics, Russian Academy of Sciences, Moscow, Russia; Faculty of Physics, Lomonosov Moscow State University, Moscow, Russia
| | - Fedor A Balabin
- Center for Theoretical Problems of Physico-chemical Pharmacology, Russian Academy of Sciences, Moscow, Russia; Dmitry Rogachev National Medical Research Centre of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Joanne L Dunster
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, Harborne Building, University of Reading, Whiteknights, Reading, United Kingdom
| | - Mikhail A Panteleev
- Center for Theoretical Problems of Physico-chemical Pharmacology, Russian Academy of Sciences, Moscow, Russia; Dmitry Rogachev National Medical Research Centre of Pediatric Hematology, Oncology and Immunology, Moscow, Russia; Faculty of Physics, Lomonosov Moscow State University, Moscow, Russia; Faculty of Biological and Medical Physics, Moscow Institute of Physics and Technology, Dolgoprudnyi, Russia
| | - Jonathan M Gibbins
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, Harborne Building, University of Reading, Whiteknights, Reading, United Kingdom
| | - Anastasia N Sveshnikova
- Center for Theoretical Problems of Physico-chemical Pharmacology, Russian Academy of Sciences, Moscow, Russia; Dmitry Rogachev National Medical Research Centre of Pediatric Hematology, Oncology and Immunology, Moscow, Russia; Faculty of Physics, Lomonosov Moscow State University, Moscow, Russia; Department of Normal Physiology, Sechenov First Moscow State Medical University, Moscow, Russia.
| |
Collapse
|
28
|
Ishihara M, Nakamura S, Sato Y, Takayama T, Fukuda K, Fujita M, Murakami K, Yokoe H. Heparinoid Complex-Based Heparin-Binding Cytokines and Cell Delivery Carriers. Molecules 2019; 24:molecules24244630. [PMID: 31861225 PMCID: PMC6943580 DOI: 10.3390/molecules24244630] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 12/10/2019] [Accepted: 12/11/2019] [Indexed: 12/20/2022] Open
Abstract
Heparinoid is the generic term that is used for heparin, heparan sulfate (HS), and heparin-like molecules of animal or plant origin and synthetic derivatives of sulfated polysaccharides. Various biological activities of heparin/HS are attributed to their specific interaction and regulation with various heparin-binding cytokines, antithrombin (AT), and extracellular matrix (ECM) biomolecules. Specific domains with distinct saccharide sequences in heparin/HS mediate these interactions are mediated and require different highly sulfated saccharide sequences with different combinations of sulfated groups. Multivalent and cluster effects of the specific sulfated sequences in heparinoids are also important factors that control their interactions and biological activities. This review provides an overview of heparinoid-based biomaterials that offer novel means of engineering of various heparin-binding cytokine-delivery systems for biomedical applications and it focuses on our original studies on non-anticoagulant heparin-carrying polystyrene (NAC-HCPS) and polyelectrolyte complex-nano/microparticles (N/MPs), in addition to heparin-coating devices.
Collapse
Affiliation(s)
- Masayuki Ishihara
- Division of Biomedical Engineering, Research Institute, National Defense Medical College, 3-2 Namiki, Tokorazawa, Saitama 359-8513, Japan; (S.N.); (Y.S.); (K.F.)
- Correspondence: ; Tel.: +81-429-95-1211 (ext. 2610)
| | - Shingo Nakamura
- Division of Biomedical Engineering, Research Institute, National Defense Medical College, 3-2 Namiki, Tokorazawa, Saitama 359-8513, Japan; (S.N.); (Y.S.); (K.F.)
| | - Yoko Sato
- Division of Biomedical Engineering, Research Institute, National Defense Medical College, 3-2 Namiki, Tokorazawa, Saitama 359-8513, Japan; (S.N.); (Y.S.); (K.F.)
| | - Tomohiro Takayama
- Department of Oral and Maxillofacial Surgery, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama 359-8513, Japan; (T.T.); (K.M.); (H.Y.)
| | - Koichi Fukuda
- Division of Biomedical Engineering, Research Institute, National Defense Medical College, 3-2 Namiki, Tokorazawa, Saitama 359-8513, Japan; (S.N.); (Y.S.); (K.F.)
| | - Masanori Fujita
- Division of Environmental Medicine, Research Institute, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama 359-1324, Japan;
| | - Kaoru Murakami
- Department of Oral and Maxillofacial Surgery, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama 359-8513, Japan; (T.T.); (K.M.); (H.Y.)
| | - Hidetaka Yokoe
- Department of Oral and Maxillofacial Surgery, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama 359-8513, Japan; (T.T.); (K.M.); (H.Y.)
| |
Collapse
|
29
|
Sung PS, Hsieh SL. CLEC2 and CLEC5A: Pathogenic Host Factors in Acute Viral Infections. Front Immunol 2019; 10:2867. [PMID: 31867016 PMCID: PMC6909378 DOI: 10.3389/fimmu.2019.02867] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 11/22/2019] [Indexed: 12/11/2022] Open
Abstract
The protective roles of endosomal toll-like receptors (TLRs) and cytosolic nucleic acid sensors are well elucidated, but the pathogenic host factors during viral infections remain unclear. Spleen tyrosine kinase (Syk)-coupled C-type lectins (CLECs) CLEC2 and CLEC5A are highly expressed on platelets and myeloid cells, respectively. CLEC2 has been shown to recognize snake venom aggretin and the endogenous ligand podoplanin and acts as a critical regulator in the development and immunothrombosis. Although CLEC2 has been reported to interact with type I immunodeficiency virus (HIV-1), its role in viral infections is still unclear. CLEC5A binds to fucose and mannose moieties of dengue virus membrane glycans, as well as to N-acetylglucosamine (GlcNAc)/N-acetylmuramic acid (MurNAc) disaccharides that form the backbone of L. monocytogenes peptidoglycans. Recently, we demonstrated that both CLEC2 and CLEC5A are critical in microbe-induced “neutrophil extracellular trap” (NET) formation and proinflammatory cytokine production. Moreover, activation of CLEC2 by dengue virus (DV) and H5N1 influenza virus (IAV) induces the release of extracellular vesicles (EVs), which further enhance NETosis and proinflammatory cytokine production via CLEC5A and Toll-like receptor 2 (TLR2). These findings not only illustrate the immunomodulatory effects of EVs during platelet-leukocyte interactions, but also demonstrate the critical roles of CLEC2 and CLEC5A in acute viral infections.
Collapse
Affiliation(s)
- Pei-Shan Sung
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Shie-Liang Hsieh
- Genomics Research Center, Academia Sinica, Taipei, Taiwan.,Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan.,Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan.,Institute for Cancer Biology and Drug Discovery, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
30
|
Xu L, Liu F, Li C, Li S, Wu H, Guo B, Gu J, Wang L. Fucoidan suppresses the gastric cancer cell malignant phenotype and production of TGF-β1 via CLEC-2. Glycobiology 2019; 30:301-311. [DOI: 10.1093/glycob/cwz097] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 11/12/2019] [Accepted: 11/12/2019] [Indexed: 12/12/2022] Open
Abstract
Abstract
The sulfated polysaccharide fucoidan displays excellent anticancer properties with low toxicity in many kinds of cancers. However, its detailed pharmacological effect and mechanism of action in gastric carcinoma remains unclear. In this study, we found that fucoidan could suppress gastric cancer (GC) cell growth, as well as cell migration and invasion. A cytokine expression screen demonstrated that transforming growth factor beta 1 (TGF-β1) secretion was decreased in fucoidan-treated cells. Fucoidan has been reported to be a platelet agonist for the C-type lectin-like receptor 2 (CLEC-2), and our previous research found that upregulation of CLEC-2 inhibited GC progression. Here, we confirmed that fucoidan, combined with CLEC-2, significantly increased CLEC-2 expression in GC cells via the transcription factor caudal type homeobox transcription factor 2, an important regulator of gut homeostasis. In addition, the inhibitory effect of fucoidan on the GC cell malignant phenotype and TGF-β1 secretion could be restored by knocking down CLEC-2. Thus, our data suggest that fucoidan targets CLEC-2 to exert antitumorigenesis and antimetastatic activity, suggesting that fucoidan is a promising treatment for gastric carcinoma.
Collapse
Affiliation(s)
- Ling Xu
- NHC Key Laboratory of Glycoconjugate Research, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Fenglin Liu
- Department of General Surgery, Zhongshan Hospital, 180 Fenglin Road, Fudan University, Shanghai 20032, China
| | - Can Li
- NHC Key Laboratory of Glycoconjugate Research, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Shuxuan Li
- NHC Key Laboratory of Glycoconjugate Research, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Hao Wu
- NHC Key Laboratory of Glycoconjugate Research, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Bao Guo
- NHC Key Laboratory of Glycoconjugate Research, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Jianxin Gu
- NHC Key Laboratory of Glycoconjugate Research, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Lan Wang
- NHC Key Laboratory of Glycoconjugate Research, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| |
Collapse
|
31
|
Shimazu T, Borjigin L, Katoh K, Roh SG, Kitazawa H, Abe K, Suda Y, Saito H, Kunii H, Nihei K, Uemoto Y, Aso H, Suzuki K. Addition of Wakame seaweed (Undaria pinnatifida) stalk to animal feed enhances immune response and improves intestinal microflora in pigs. Anim Sci J 2019; 90:1248-1260. [PMID: 31321863 DOI: 10.1111/asj.13274] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 06/05/2019] [Accepted: 06/11/2019] [Indexed: 11/28/2022]
Abstract
This study was conducted to evaluate the effects of supplementation of Wakame seaweed stalks on the immunity and intestinal microflora of pigs. Three separate experiments were performed: Relatively young (start at 20-30 kg; Experiments 1 and 2) and fattening period (70 kg; Experiment 3). All pigs (including the control group) were fed the same commercial feed, free from antibiotic additives, but in the feed for the treatment groups, 1% seaweed powder was added. There were no group differences observed in daily weight gain and feed intake in Experiments 1 and 2 between groups; however, daily weight gain was significantly higher in the treatment group compared to the control group in Experiment 3. The percentage of peripheral blood natural killer cells of the treatment group was significantly higher than that of the control group in all experiments. Although addition of seaweed changed the gene expression of cytokine and toll-like receptors of the small intestinal Peyer's patches slightly, seaweed seems to alter intestinal microflora preferentially, for instance, there was an increase in Lactobacillus and a decrease of Escherichia coli observed. These results suggest that Wakame seaweed can be used as supplement for pig feed to improve the gut health and immunity of pigs.
Collapse
Affiliation(s)
- Tomoyuki Shimazu
- Department of Food, Agriculture and Environment, Miyagi University, Sendai, Japan
| | - Liushiqi Borjigin
- Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Kazuo Katoh
- Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Sang-Gun Roh
- Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Haruki Kitazawa
- Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Keietsu Abe
- Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Yoshihito Suda
- Department of Food, Agriculture and Environment, Miyagi University, Sendai, Japan
| | - Hayato Saito
- Miyagi Prefecture Animal Industry Experiment Station, Oosaki, Japan
| | - Hiroshi Kunii
- Miyagi Prefecture Animal Industry Experiment Station, Oosaki, Japan
| | - Ken Nihei
- Miyagi Prefecture Fishery Cooperation Union, Ishinomaki, Japan
| | - Yoshinobu Uemoto
- Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Hisashi Aso
- Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Keiichi Suzuki
- Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| |
Collapse
|
32
|
Martyanov AA, Kaneva VN, Panteleev MA, Sveshnikova AN. [CLEC-2 induced signalling in blood platelets]. BIOMEDIT︠S︡INSKAI︠A︡ KHIMII︠A︡ 2019; 64:387-396. [PMID: 30378555 DOI: 10.18097/pbmc20186405387] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Platelet activating receptor CLEC-2 has been identified on platelet surface a decade ago. The only confirmed endogenous CLEC-2 agonist is podoplanin. Podoplanin is a transmembrane protein expressed by lymphatic endothelial cells, reticular fibroblastic cells in lymph nodes, kidney podocytes and by cells of certain tumors. CLEC-2 and podoplanin are involved in the processes of embryonic development (blood-lymph vessel separation and angiogenesis), maintaining of vascular integrity of small vessels during inflammation and prevention of blood-lymphatic mixing in high endothelial venules. However, CLEC-2 and podoplanin are contributing to tumor methastasis progression, Salmonella sepsis, deep-vein thrombosis. CLEC-2 signalling cascade includes tyrosine-kinases (Syk, SFK, Btk) as well as adapter LAT and phospholipase Cg2, which induces calcium signalling. CLEC-2, podoplanin and proteins, participating in CLEC-2 signalling cascade, are perspective targets for antithrombotic therapy.
Collapse
Affiliation(s)
- A A Martyanov
- Faculty of Physics, Lomonosov Moscow State University, Moscow, Russia; Center for Theoretical Problems of Physicochemical Pharmacology, Moscow, Russia
| | - V N Kaneva
- Faculty of Physics, Lomonosov Moscow State University, Moscow, Russia; Rogachev National Scientific and Practical Centre of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - M A Panteleev
- Center for Theoretical Problems of Physicochemical Pharmacology, Moscow, Russia; Rogachev National Scientific and Practical Centre of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - A N Sveshnikova
- Faculty of Physics, Lomonosov Moscow State University, Moscow, Russia; Center for Theoretical Problems of Physicochemical Pharmacology, Moscow, Russia
| |
Collapse
|
33
|
Kardeby C, Fälker K, Haining EJ, Criel M, Lindkvist M, Barroso R, Påhlsson P, Ljungberg LU, Tengdelius M, Rainger GE, Watson S, Eble JA, Hoylaerts MF, Emsley J, Konradsson P, Watson SP, Sun Y, Grenegård M. Synthetic glycopolymers and natural fucoidans cause human platelet aggregation via PEAR1 and GPIbα. Blood Adv 2019; 3:275-287. [PMID: 30700416 PMCID: PMC6373755 DOI: 10.1182/bloodadvances.2018024950] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 12/27/2018] [Indexed: 12/14/2022] Open
Abstract
Fucoidans are sulfated fucose-based polysaccharides that activate platelets and have pro- and anticoagulant effects; thus, they may have therapeutic value. In the present study, we show that 2 synthetic sulfated α-l-fucoside-pendant glycopolymers (with average monomeric units of 13 and 329) and natural fucoidans activate human platelets through a Src- and phosphatidylinositol 3-kinase (PI3K)-dependent and Syk-independent signaling cascade downstream of the platelet endothelial aggregation receptor 1 (PEAR1). Synthetic glycopolymers and natural fucoidan stimulate marked phosphorylation of PEAR1 and Akt, but not Syk. Platelet aggregation and Akt phosphorylation induced by natural fucoidan and synthetic glycopolymers are blocked by a monoclonal antibody to PEAR1. Direct binding of sulfated glycopolymers to epidermal like growth factor (EGF)-like repeat 13 of PEAR1 was shown by avidity-based extracellular protein interaction screen technology. In contrast, synthetic glycopolymers and natural fucoidans activate mouse platelets through a Src- and Syk-dependent pathway regulated by C-type lectin-like receptor 2 (CLEC-2) with only a minor role for PEAR1. Mouse platelets lacking the extracellular domain of GPIbα and human platelets treated with GPIbα-blocking antibodies display a reduced aggregation response to synthetic glycopolymers. We found that synthetic sulfated glycopolymers bind directly to GPIbα, substantiating that GPIbα facilitates the interaction of synthetic glycopolymers with CLEC-2 or PEAR1. Our results establish PEAR1 as the major signaling receptor for natural fucose-based polysaccharides and synthetic glycopolymers in human, but not in mouse, platelets. Sulfated α-l-fucoside-pendant glycopolymers are unique tools for further investigation of the physiological role of PEAR1 in platelets and beyond.
Collapse
Affiliation(s)
- Caroline Kardeby
- Cardiovascular Research Centre, School of Medical Sciences, Örebro University, Örebro, Sweden
| | - Knut Fälker
- Cardiovascular Research Centre, School of Medical Sciences, Örebro University, Örebro, Sweden
| | - Elizabeth J Haining
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Maarten Criel
- Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Madelene Lindkvist
- Cardiovascular Research Centre, School of Medical Sciences, Örebro University, Örebro, Sweden
| | - Ruben Barroso
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
- Centre of Membrane Proteins and Receptors, Universities of Birmingham and Nottingham, The Midlands, United Kingdom
| | - Peter Påhlsson
- Division of Cell Biology, Department of Clinical and Experimental Medicine, and
| | - Liza U Ljungberg
- Cardiovascular Research Centre, School of Medical Sciences, Örebro University, Örebro, Sweden
| | | | - G Ed Rainger
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Stephanie Watson
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Johannes A Eble
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Münster, Germany; and
| | - Marc F Hoylaerts
- Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Jonas Emsley
- Centre of Membrane Proteins and Receptors, Universities of Birmingham and Nottingham, The Midlands, United Kingdom
- Division of Biomolecular Science and Medicinal Chemistry, Centre for Biomolecular Sciences, School of Pharmacy, University of Nottingham, Nottingham, United Kingdom
| | - Peter Konradsson
- Division of Organic Chemistry, Linköping University, Linköping, Sweden
| | - Steve P Watson
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
- Centre of Membrane Proteins and Receptors, Universities of Birmingham and Nottingham, The Midlands, United Kingdom
| | - Yi Sun
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
- Centre of Membrane Proteins and Receptors, Universities of Birmingham and Nottingham, The Midlands, United Kingdom
| | - Magnus Grenegård
- Cardiovascular Research Centre, School of Medical Sciences, Örebro University, Örebro, Sweden
| |
Collapse
|
34
|
Rayes J, Watson SP, Nieswandt B. Functional significance of the platelet immune receptors GPVI and CLEC-2. J Clin Invest 2019; 129:12-23. [PMID: 30601137 DOI: 10.1172/jci122955] [Citation(s) in RCA: 208] [Impact Index Per Article: 41.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Although platelets are best known for their role in hemostasis, they are also crucial in development, host defense, inflammation, and tissue repair. Many of these roles are regulated by the immune-like receptors glycoprotein VI (GPVI) and C-type lectin receptor 2 (CLEC-2), which signal through an immunoreceptor tyrosine-based activation motif (ITAM). GPVI is activated by collagen in the subendothelial matrix, by fibrin and fibrinogen in the thrombus, and by a remarkable number of other ligands. CLEC-2 is activated by the transmembrane protein podoplanin, which is found outside of the vasculature and is upregulated in development, inflammation, and cancer, but there is also evidence for additional ligands. In this Review, we discuss the physiological and pathological roles of CLEC-2 and GPVI and their potential as targets in thrombosis and thrombo-inflammatory disorders (i.e., disorders in which inflammation plays a critical role in the ensuing thrombosis) relative to current antiplatelet drugs.
Collapse
Affiliation(s)
- Julie Rayes
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Steve P Watson
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom.,Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, United Kingdom
| | - Bernhard Nieswandt
- Institute of Experimental Biomedicine, University Hospital and Rudolf Virchow Center, University of Würzburg, Würzburg, Germany
| |
Collapse
|
35
|
Martyanov AA, Kaneva VN, Panteleev MA, Sveshnikova AN. Physiological and pathophysiological aspects of blood platelet activation through CLEC-2 receptor. ONCOHEMATOLOGY 2018. [DOI: 10.17650/1818-8346-2018-13-3-83-90] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Platelet activating receptor CLEC-2 has been identified on platelet surface a decade ago. The only confirmed endogenous CLEC-2 agonist is podoplanin. Podoplanin is a transmembrane protein expressed by lymphatic endothelial cells, reticular fibroblastic cells in lymph nodes, kidney podocytes and by cells of certain tumors. Association of CLEC-2 with podoplanin is involved in processes of embryonic development (blood-lymph vessel separation and angiogenesis), maintaining of vascular integrity of small vessels during inflammation and prevention of blood-lymphatic mixing in high endothelial venules. However, CLEC-2 and podoplanin are contributing to tumor metastasis progression, Salmonella sepsis and deep-vein thrombosis. This makes CLEC-2 and podoplanin a perspective target for pharmacological treatment. Aspirin and Ibrutinib are considered to be perspective for abrogation of podoplanin-induced platelet activation via CLEC-2. The present review discusses already known pathological and physiological roles of CLEC-2 and possibilities of a targeted therapy for CLEC-2 associated diseases.
Collapse
Affiliation(s)
- A. A. Martyanov
- Dmitry Rogachev National Medical Research Centre of Pediatric Hematology, Oncology and Immunology; Lomonosov Moscow State University, Faculty of Physics; Center for Theoretical Problems of Physicochemical Pharmacology, Russian Academy of Sciences
| | - V. N. Kaneva
- Dmitry Rogachev National Medical Research Centre of Pediatric Hematology, Oncology and Immunology; Lomonosov Moscow State University, Faculty of Physics
| | - M. A. Panteleev
- Dmitry Rogachev National Medical Research Centre of Pediatric Hematology, Oncology and Immunology; Lomonosov Moscow State University, Faculty of Physics; Center for Theoretical Problems of Physicochemical Pharmacology, Russian Academy of Sciences; Moscow Institute of Physics and Technology (State University), Faculty of Biological and Medical Physics
| | - A. N. Sveshnikova
- Dmitry Rogachev National Medical Research Centre of Pediatric Hematology, Oncology and Immunology; Lomonosov Moscow State University, Faculty of Physics; Center for Theoretical Problems of Physicochemical Pharmacology, Russian Academy of Sciences
| |
Collapse
|
36
|
Burchell SR, Tang J, Zhang JH. Hematoma Expansion Following Intracerebral Hemorrhage: Mechanisms Targeting the Coagulation Cascade and Platelet Activation. Curr Drug Targets 2018; 18:1329-1344. [PMID: 28378693 DOI: 10.2174/1389450118666170329152305] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Revised: 07/20/2016] [Accepted: 03/14/2017] [Indexed: 01/04/2023]
Abstract
Hematoma expansion (HE), defined as a greater than 33% increase in intracerebral hemorrhage (ICH) volume within the first 24 hours, results in significant neurological deficits, and enhancement of ICH-induced primary and secondary brain injury. An escalation in the use of oral anticoagulants has led to a surge in the incidences of oral anticoagulation-associated ICH (OAT-ICH), which has been associated with a greater risk for HE and worse functional outcomes following ICH. The oral anticoagulants in use include vitamin K antagonists, and direct thrombin and factor Xa inhibitors. Fibrinolytic agents are also frequently administered. These all act via differing mechanisms and thus have varying degrees of impact on HE and ICH outcome. Additionally, antiplatelet medications have also been increasingly prescribed, and result in increased bleeding risks and worse outcomes after ICH. Aspirin, thienopyridines, and GPIIb/IIIa receptor blockers are some of the most common agents in use clinically, and also have different effects on ICH and hemorrhage growth, based on their mechanisms of action. Recent studies have found that reduced platelet activity may be more effective in predicting ICH risk, hemorrhage expansion, and outcomes, than antiplatelet agents, and activating platelets may thus be a novel target for ICH therapy. This review explores how dysfunctions or alterations in the coagulation and platelet cascades can lead to, and/or exacerbate, hematoma expansion following intracerebral hemorrhage, and describe the mechanisms behind these effects and the drugs that induce them. We also discuss potential future therapy aimed at increasing platelet activity after ICH.
Collapse
Affiliation(s)
- Sherrefa R Burchell
- Department of Physiology, Loma Linda University School of Medicine, Loma Linda CA, USA.,Center for Neuroscience Research, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Jiping Tang
- Department of Physiology, Loma Linda University School of Medicine, Loma Linda CA, USA.,Center for Neuroscience Research, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - John H Zhang
- Department of Physiology, Loma Linda University School of Medicine, Loma Linda CA, USA.,Center for Neuroscience Research, Loma Linda University School of Medicine, Loma Linda, CA, USA.,Department of Anesthesiology, Loma Linda University School of Medicine, Loma Linda CA, USA
| |
Collapse
|
37
|
Janapati S, Wurtzel J, Dangelmaier C, Manne BK, Bhavanasi D, Kostyak JC, Kim S, Holinstat M, Kunapuli SP, Goldfinger LE. TC21/RRas2 regulates glycoprotein VI-FcRγ-mediated platelet activation and thrombus stability. J Thromb Haemost 2018; 16:S1538-7836(22)02217-6. [PMID: 29883056 PMCID: PMC6286703 DOI: 10.1111/jth.14197] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Indexed: 12/27/2022]
Abstract
Essentials RAS proteins are expressed in platelets but their functions are largely uncharacterized. TC21/RRas2 is required for glycoprotein VI-induced platelet responses and for thrombus stability in vivo. TC21 regulates platelet aggregation by control of αIIb β3 integrin activation, via crosstalk with Rap1b. This is the first indication of functional importance of a proto-oncogenic RAS protein in platelets. SUMMARY Background Many RAS family small GTPases are expressed in platelets, including RAC, RHOA, RAP, and HRAS/NRAS/RRAS1, but most of their signaling and cellular functions remain poorly understood. Like RRAS1, TC21/RRAS2 reverses HRAS-induced suppression of integrin activation in CHO cells. However, a role for TC21 in platelets has not been explored. Objectives To determine TC21 expression in platelets, TC21 activation in response to platelet agonists, and roles of TC21 in platelet function in in vitro and in vivo thrombosis. Results We demonstrate that TC21 is expressed in human and murine platelets, and is activated in response to agonists for the glycoprotein (GP) VI-FcRγ immunoreceptor tyrosine-based activation motif (ITAM)-containing collagen receptor, in an Src-dependent manner. GPVI-induced platelet aggregation, integrin αIIb β3 activation, and α-granule and dense granule secretion, as well as phosphorylation of Syk, phospholipase Cγ2, AKT, and extracellular signal-regulated kinase, were inhibited in TC21-deficient platelets ex vivo. In contrast, these responses were normal in TC21-deficient platelets following stimulation with P2Y, protease-activated receptor 4 and C-type lectin receptor 2 receptor agonists, indicating that the function of TC21 in platelets is GPVI-FcRγ-ITAM-specific. TC21 was required for GPVI-induced activation of Rap1b. TC21-deficient mice did not show a significant delay in injury-induced thrombosis as compared with wild-type controls; however, thrombi were unstable. Hemostatic responses showed similar effects. Conclusions TC21 is essential for GPVI-FcRγ-mediated platelet activation and for thrombus stability in vivo via control of Rap1b and integrins.
Collapse
Affiliation(s)
- S Janapati
- The Sol Sherry Thrombosis Research Center and Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - J Wurtzel
- The Sol Sherry Thrombosis Research Center and Department of Anatomy & Cell Biology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - C Dangelmaier
- The Sol Sherry Thrombosis Research Center and Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - B K Manne
- The Sol Sherry Thrombosis Research Center and Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - D Bhavanasi
- The Sol Sherry Thrombosis Research Center and Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - J C Kostyak
- The Sol Sherry Thrombosis Research Center and Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - S Kim
- The Sol Sherry Thrombosis Research Center and Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - M Holinstat
- Department of Pharmacology, Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, MI, USA
| | - S P Kunapuli
- The Sol Sherry Thrombosis Research Center and Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - L E Goldfinger
- The Sol Sherry Thrombosis Research Center and Department of Anatomy & Cell Biology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| |
Collapse
|
38
|
Manne BK, Münzer P, Badolia R, Walker-Allgaier B, Campbell RA, Middleton E, Weyrich AS, Kunapuli SP, Borst O, Rondina MT. PDK1 governs thromboxane generation and thrombosis in platelets by regulating activation of Raf1 in the MAPK pathway. J Thromb Haemost 2018; 16:1211-1225. [PMID: 29575487 PMCID: PMC5984143 DOI: 10.1111/jth.14005] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Indexed: 01/02/2023]
Abstract
Essentials Phosphoinositide 3-kinase and MAPK pathways crosstalk via PDK1. PDK1 is required for adenosine diphosphate-induced platelet activation and thromboxane generation. PDK1 regulates RAF proto-oncogene Ser/Thr kinase (Raf1) activation in the MAPK pathway. Genetic ablation of PDK1 protects against platelet-dependent thrombosis in vivo. SUMMARY Background Platelets are dynamic effector cells with functions that span hemostatic, thrombotic and inflammatory continua. Phosphoinositide-dependent protein kinase 1 (PDK1) regulates protease-activated receptor 4-induced platelet activation and thrombus formation through glycogen synthase kinase3β. However, whether PDK1 also signals through the ADP receptor and its functional importance in vivo remain unknown. Objective To establish the mechanism of PDK1 in ADP-induced platelet activation and thrombosis. Methods We assessed the role of PDK1 on 2MeSADP-induced platelet activation by measuring aggregation, thromboxane generation and phosphorylation events in the presence of BX-795, which inhibits PDK1, or by using platelet-specific PDK1 knockout mice and performing western blot analysis. PDK1 function in thrombus formation was assessed with an in vivo pulmonary embolism model. Results PDK1 inhibition with BX-795 reduced 2-methylthio-ADP (2MeSADP)-induced aggregation of human and murine platelets by abolishing thromboxane generation. Similar results were observed in pdk1-/- mice. PDK1 was also necessary for the phosphorylation of mitogen-activated protein kinase kinase 1/2 (MEK1/2), extracellular signal-regulated kinase 1/2, and cytosolic phospholipase A2, indicating that PDK1 regulates an upstream kinase in the mitogen-activated protein kinase (MAPK) pathway. We next determined that this upstream kinase is Raf-1, a serine/threonine kinase that is necessary for the phosphorylation of MEK1/2, as pharmacological inhibition and genetic ablation of PDK1 were sufficient to prevent Raf1 phosphorylation. Furthermore, in vivo inhibition or genetic ablation of PDK1 protected mice from collagen/epinephrine-induced pulmonary embolism. Conclusion PDK1 governs thromboxane generation and thrombosis in platelets that are stimulated with 2MeSADP by regulating activation of the MAPK pathway.
Collapse
Affiliation(s)
- Bhanu Kanth Manne
- Department of Internal Medicine, Molecular Medicine Program, University of Utah, Salt Lake City, UT, 84112 USA
| | - Patrick Münzer
- Department of Cardiology and Cardiovascular Medicine, University of Tübingen, Tübingen, 72076 Germany
| | - Rachit Badolia
- Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, PA, 19140 USA
| | - Britta Walker-Allgaier
- Department of Cardiology and Cardiovascular Medicine, University of Tübingen, Tübingen, 72076 Germany
| | - Robert A Campbell
- Department of Internal Medicine, Molecular Medicine Program, University of Utah, Salt Lake City, UT, 84112 USA
| | - Elizabeth Middleton
- Department of Internal Medicine, Molecular Medicine Program, University of Utah, Salt Lake City, UT, 84112 USA
| | - Andrew S Weyrich
- Department of Internal Medicine, Molecular Medicine Program, University of Utah, Salt Lake City, UT, 84112 USA
| | - Satya P Kunapuli
- Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, PA, 19140 USA
| | - Oliver Borst
- Department of Cardiology and Cardiovascular Medicine, University of Tübingen, Tübingen, 72076 Germany
| | - Matthew T. Rondina
- Department of Internal Medicine, Molecular Medicine Program, University of Utah, Salt Lake City, UT, 84112 USA
- Department of Internal Medicine, GRECC, George E. Wahlen VAMC, Salt Lake City, UT, 84148
| |
Collapse
|
39
|
Badolia R, Inamdar V, Manne BK, Dangelmaier C, Eble JA, Kunapuli SP. G q pathway regulates proximal C-type lectin-like receptor-2 (CLEC-2) signaling in platelets. J Biol Chem 2017; 292:14516-14531. [PMID: 28705934 DOI: 10.1074/jbc.m117.791012] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 07/09/2017] [Indexed: 11/06/2022] Open
Abstract
Platelets play a key role in the physiological hemostasis or pathological process of thrombosis. Rhodocytin, an agonist of the C-type lectin-like receptor-2 (CLEC-2), elicits powerful platelet activation signals in conjunction with Src family kinases (SFKs), spleen tyrosine kinase (Syk), and phospholipase γ2 (PLCγ2). Previous reports have shown that rhodocytin-induced platelet aggregation depends on secondary mediators such as thromboxane A2 (TxA2) and ADP, which are agonists for G-protein-coupled receptors (GPCRs) on platelets. How the secondary mediators regulate CLEC-2-mediated platelet activation in terms of signaling is not clearly defined. In this study, we report that CLEC-2-induced Syk and PLCγ2 phosphorylation is potentiated by TxA2 and that TxA2 plays a critical role in the most proximal event of CLEC-2 signaling, i.e. the CLEC-2 receptor tyrosine phosphorylation. We show that the activation of other GPCRs, such as the ADP receptors and protease-activated receptors, can also potentiate CLEC-2 signaling. By using the specific Gq inhibitor, UBO-QIC, or Gq knock-out murine platelets, we demonstrate that Gq signaling, but not other G-proteins, is essential for GPCR-induced potentiation of Syk phosphorylation downstream of CLEC-2. We further elucidated the signaling downstream of Gq and identified an important role for the PLCβ-PKCα pathway, possibly regulating activation of SFKs, which are crucial for initiation of CLEC-2 signaling. Together, these results provide evidence for novel Gq-PLCβ-PKCα-mediated regulation of proximal CLEC-2 signaling by Gq-coupled receptors.
Collapse
Affiliation(s)
- Rachit Badolia
- From the Department of Physiology.,Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania 19140 and
| | - Vaishali Inamdar
- From the Department of Physiology.,Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania 19140 and
| | - Bhanu Kanth Manne
- From the Department of Physiology.,Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania 19140 and
| | - Carol Dangelmaier
- From the Department of Physiology.,Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania 19140 and
| | - Johannes A Eble
- the Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Waldeyerstrasse 15, 48149 Münster, Germany
| | - Satya P Kunapuli
- From the Department of Physiology, .,Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania 19140 and
| |
Collapse
|
40
|
Abstract
Platelets are small, anucleate circulating cells that possess a dynamic repertoire of functions spanning the hemostatic, inflammatory, and immune continuum. Once thought to be merely cell fragments with responses limited primarily to acute hemostasis and vascular wall repair, platelets are now increasingly recognized as key sentinels and effector cells regulating host responses to many inflammatory and infectious cues. Platelet granules, including α-granules and dense-granules, store hundreds of factors and secrete these mediators in response to activating signals. The cargo packaged and stored within platelet granules orchestrates communication between platelets and other circulating cells, augments host defense mechanisms to invading pathogens and tumor cells, and - in some settings - drives dysregulated and injurious responses. This focused review will highlight several of the established and emerging mechanisms and roles of platelet secretion in inflammatory and infectious diseases.
Collapse
Affiliation(s)
- Bhanu K Manne
- a The University of Utah Molecular Medicine Program , Salt Lake City , Utah , USA
| | | | - Matthew T Rondina
- a The University of Utah Molecular Medicine Program , Salt Lake City , Utah , USA.,c Department of Internal Medicine , Salt Lake City , Utah , USA.,d The GRECC, George E. Wahlen Salt Lake City VAMC , Salt Lake City , Utah , USA
| |
Collapse
|
41
|
Tengdelius M, Kardeby C, Fälker K, Griffith M, Påhlsson P, Konradsson P, Grenegård M. Fucoidan-Mimetic Glycopolymers as Tools for Studying Molecular and Cellular Responses in Human Blood Platelets. Macromol Biosci 2016; 17. [DOI: 10.1002/mabi.201600257] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Revised: 08/15/2016] [Indexed: 11/09/2022]
Affiliation(s)
- Mattias Tengdelius
- Division of Organic Chemistry; Department of Physics; Biology and Chemistry (IFM); Linköping University; SE-581 83 Linköping Sweden
| | - Caroline Kardeby
- Cardiovascular Research Centre; School of Medical Sciences; Örebro University; SE-701 82 Örebro Sweden
| | - Knut Fälker
- Cardiovascular Research Centre; School of Medical Sciences; Örebro University; SE-701 82 Örebro Sweden
| | - May Griffith
- Division of Cell Biology; Department of Clinical and Experimental Medicine (IKE); Linköping University; SE-581 83 Linköping Sweden
| | - Peter Påhlsson
- Division of Cell Biology; Department of Clinical and Experimental Medicine (IKE); Linköping University; SE-581 83 Linköping Sweden
| | - Peter Konradsson
- Division of Organic Chemistry; Department of Physics; Biology and Chemistry (IFM); Linköping University; SE-581 83 Linköping Sweden
| | - Magnus Grenegård
- Cardiovascular Research Centre; School of Medical Sciences; Örebro University; SE-701 82 Örebro Sweden
| |
Collapse
|
42
|
Tanino Y, Hashimoto T, Ojima T, Mizuno M. F-fucoidan from Saccharina japonica is a novel inducer of galectin-9 and exhibits anti-allergic activity. J Clin Biochem Nutr 2016; 59:25-30. [PMID: 27499575 PMCID: PMC4933687 DOI: 10.3164/jcbn.15-144] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Accepted: 01/08/2016] [Indexed: 01/03/2023] Open
Abstract
Fucoidan is a sulfated polysaccharide from brown sea algae. In the present study, it was demonstrated that oral administration of F-fucoidan from Saccharina japonica possessed anti-allergic effects using the passive cutaneous anaphylaxis reaction, but not by intraperitoneal administration. The inhibitory mechanism was dependent on galectin-9, which belongs to a soluble lectin family that recognizes β-galactoside and prevents IgE binding to mast cells. The anti-allergy properties of F-fucoidan were cancelled by an intravenous dose of anti-galectin-9 antibody or lactose, which bind competitively with galectin-9 before the passive cutaneous anaphylaxis reaction. F-fucoidan increased the expression level of galectin-9 mRNA in intestinal epithelial cells and serum galectin-9 levels. Oral treatment with F-fucoidan suppressed allergic symptoms through the induction of galectin-9. This is the first report that F-fucoidan can induce the secretion of galectin-9.
Collapse
Affiliation(s)
- Yuka Tanino
- Graduate School of Agricultural Science, Kobe University, 1-1 Rokkodai, Nada-ku, Kobe 657-8501, Japan
| | - Takashi Hashimoto
- Graduate School of Agricultural Science, Kobe University, 1-1 Rokkodai, Nada-ku, Kobe 657-8501, Japan
| | - Takao Ojima
- Faculty of Fisheries Sciences, Hokkaido University, 3-1-1 Minato, Hakodate 041-8611, Japan
| | - Masashi Mizuno
- Graduate School of Agricultural Science, Kobe University, 1-1 Rokkodai, Nada-ku, Kobe 657-8501, Japan
| |
Collapse
|
43
|
Abstract
In this chapter, a comprehensive overview of the known ligands for the C-type lectins (CTLs) is provided. Emphasis has been placed on the chemical structure of the glycans that bind to the different CTLs and the amount of structural variation (or overlap) that each CTL can tolerate. In this way, both the synthetic carbohydrate chemist and the immunologist can more readily gain insight into the existing structure-activity space for the CTL ligands and, ideally, see areas of synergy that will help identify and refine the ligands for these receptors.
Collapse
Affiliation(s)
- Sho Yamasaki
- Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| |
Collapse
|
44
|
Burchell SR, Iniaghe LO, Zhang JH, Tang J. Fucoidan from Fucus vesiculosus Fails to Improve Outcomes Following Intracerebral Hemorrhage in Mice. ACTA NEUROCHIRURGICA. SUPPLEMENT 2016; 121:191-8. [PMID: 26463947 DOI: 10.1007/978-3-319-18497-5_34] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Intracerebral hemorrhage (ICH) is the most fatal stroke subtype, with no effective therapies. Hematoma expansion and inflammation play major roles in the pathophysiology of ICH, contributing to primary and secondary brain injury, respectively. Fucoidan, a polysaccharide from the brown seaweed Fucus vesiculosus, has been reported to activate a platelet receptor that may function in limiting bleeding, and to exhibit anti-inflammatory effects. As such, the aim of the present study was to examine the effects of fucoidan on hemorrhaging and neurological outcomes after ICH. Male CD-1 mice were subjected to experimental ICH by infusion of bacterial collagenase. Animals were randomly divided into the following groups: sham, ICH + vehicle, ICH + 25 mg/kg fucoidan, ICH + 75 mg/kg fucoidan, and ICH + 100 mg/kg fucoidan. Brain water content, neurobehavioral outcomes, and hemoglobin content were evaluated at 24 h post ICH. Our findings show that fucoidan failed to attenuate the ICH-induced increase in BWC. The neurological deficits that result from ICH also did not differ in the treatment groups at all three doses. Finally, we found that fucoidan had no effect on the hemoglobin content after ICH. We postulate that fucoidan treatment did not improve the measured outcomes after ICH because we used crude fucoidan, which has a high molecular weight, in our study. High-molecular-weight fucoidans are reported to have less therapeutic potential than low molecular weight fucoidans. They have been shown to exhibit anti-coagulant and pro-apoptotic properties, which seem to outweigh their anti-inflammatory and potential procoagulant abilities. We propose that using a low-molecular-weight fucoidan, or fractionating the crude polysaccharide, may be effective in treating ICH. Future studies are needed to confirm this.
Collapse
Affiliation(s)
- Sherrefa R Burchell
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, USA
| | - Loretta O Iniaghe
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, USA
- Department of Pharmacology and Toxicology, University of Benin, Benin City, Nigeria
| | - John H Zhang
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, USA
- Department of Neurosurgery, Loma Linda University, Loma Linda, CA, USA
| | - Jiping Tang
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA.
| |
Collapse
|
45
|
Fitton JH, Stringer DN, Karpiniec SS. Therapies from Fucoidan: An Update. Mar Drugs 2015; 13:5920-46. [PMID: 26389927 PMCID: PMC4584361 DOI: 10.3390/md13095920] [Citation(s) in RCA: 235] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 09/02/2015] [Accepted: 09/06/2015] [Indexed: 12/30/2022] Open
Abstract
Fucoidans are a class of sulfated fucose-rich polysaccharides found in brown marine algae and echinoderms. Fucoidans have an attractive array of bioactivities and potential applications including immune modulation, cancer inhibition, and pathogen inhibition. Research into fucoidan has continued to gain pace over the last few years and point towards potential therapeutic or adjunct roles. The source, extraction, characterization and detection of fucoidan is discussed.
Collapse
Affiliation(s)
- Janet Helen Fitton
- Marinova Pty Ltd., 249 Kennedy Drive, Cambridge, Tasmania 7170, Australia.
| | - Damien N Stringer
- Marinova Pty Ltd., 249 Kennedy Drive, Cambridge, Tasmania 7170, Australia.
| | - Samuel S Karpiniec
- Marinova Pty Ltd., 249 Kennedy Drive, Cambridge, Tasmania 7170, Australia.
| |
Collapse
|
46
|
Activation of glycoprotein VI (GPVI) and C-type lectin-like receptor-2 (CLEC-2) underlies platelet activation by diesel exhaust particles and other charged/hydrophobic ligands. Biochem J 2015; 468:459-73. [PMID: 25849538 DOI: 10.1042/bj20150192] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Accepted: 04/07/2015] [Indexed: 01/29/2023]
Abstract
Platelets are activated by a range of stimuli that share little or no resemblance in structure to each other or to recognized ligands, including diesel exhaust particles (DEP), small peptides [4N1-1, Champs (computed helical anti-membrane proteins), LSARLAF (Leu-Ser-Ala-Arg-Leu-Ala-Phe)], proteins (histones) and large polysaccharides (fucoidan, dextran sulfate). This miscellaneous group stimulate aggregation of human and mouse platelets through the glycoprotein VI (GPVI)-FcR γ-chain complex and/or C-type lectin-like receptor-2 (CLEC-2) as shown using platelets from mice deficient in either or both of these receptors. In addition, all of these ligands stimulate tyrosine phosphorylation in GPVI/CLEC-2-double-deficient platelets, indicating that they bind to additional surface receptors, although only in the case of dextran sulfate does this lead to activation. DEP, fucoidan and dextran sulfate, but not the other agonists, activate GPVI and CLEC-2 in transfected cell lines as shown using a sensitive reporter assay confirming a direct interaction with the two receptors. We conclude that this miscellaneous group of ligands bind to multiple proteins on the cell surface including GPVI and/or CLEC-2, inducing activation. These results have pathophysiological significance in a variety of conditions that involve exposure to activating charged/hydrophobic agents.
Collapse
|
47
|
Bhatia S, Sharma K, Nagpal K, Bera T. Investigation of the factors influencing the molecular weight of porphyran and its associated antifungal activity. ACTA ACUST UNITED AC 2015. [DOI: 10.1016/j.bcdf.2015.03.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
48
|
Manne BK, Badolia R, Dangelmaier C, Eble JA, Ellmeier W, Kahn M, Kunapuli SP. Distinct pathways regulate Syk protein activation downstream of immune tyrosine activation motif (ITAM) and hemITAM receptors in platelets. J Biol Chem 2015; 290:11557-68. [PMID: 25767114 DOI: 10.1074/jbc.m114.629527] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Indexed: 11/06/2022] Open
Abstract
Tyrosine kinase pathways are known to play an important role in the activation of platelets. In particular, the GPVI and CLEC-2 receptors are known to activate Syk upon tyrosine phosphorylation of an immune tyrosine activation motif (ITAM) and hemITAM, respectively. However, unlike GPVI, the CLEC-2 receptor contains only one tyrosine motif in the intracellular domain. The mechanisms by which this receptor activates Syk are not completely understood. In this study, we identified a novel signaling mechanism in CLEC-2-mediated Syk activation. CLEC-2-mediated, but not GPVI-mediated, platelet activation and Syk phosphorylation were abolished by inhibition of PI3K, which demonstrates that PI3K regulates Syk downstream of CLEC-2. Ibrutinib, a Tec family kinase inhibitor, also completely abolished CLEC-2-mediated aggregation and Syk phosphorylation in human and murine platelets. Furthermore, embryos lacking both Btk and Tec exhibited cutaneous edema associated with blood-filled vessels in a typical lymphatic pattern similar to CLEC-2 or Syk-deficient embryos. Thus, our data show, for the first time, that PI3K and Tec family kinases play a crucial role in the regulation of platelet activation and Syk phosphorylation downstream of the CLEC-2 receptor.
Collapse
Affiliation(s)
- Bhanu Kanth Manne
- From the Department of Physiology, Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania 19140
| | - Rachit Badolia
- From the Department of Physiology, Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania 19140
| | - Carol Dangelmaier
- From the Department of Physiology, Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania 19140
| | - Johannes A Eble
- the Institute for Physiological Chemistry and Pathobiochemistry, University of Münster, 48149 Münster, Germany
| | - Wilfried Ellmeier
- the Division of Immunobiology, Institution of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, A-1090 Vienna, Austria, and
| | - Mark Kahn
- the Department of Medicine and Division of Cardiology, University of Pennsylvania, Philadelphia, Pennsylvania 19104-5159
| | - Satya P Kunapuli
- From the Department of Physiology, Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania 19140,
| |
Collapse
|
49
|
Manne BK, Badolia R, Dangelmaier CA, Kunapuli SP. C-type lectin like receptor 2 (CLEC-2) signals independently of lipid raft microdomains in platelets. Biochem Pharmacol 2014; 93:163-70. [PMID: 25462818 DOI: 10.1016/j.bcp.2014.11.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Revised: 11/05/2014] [Accepted: 11/12/2014] [Indexed: 10/24/2022]
Abstract
C-type lectin like receptor 2 (CLEC-2) has been reported to activate platelets through a lipid raft-dependent manner. Secreted ADP potentiates CLEC-2-mediated platelet aggregation. We have investigated whether the decrease in CLEC-2-mediated platelet aggregation, previously reported in platelets with disrupted rafts, is a result of the loss of agonist potentiation by ADP. We disrupted platelet lipid rafts with methyl-β-cyclodextrin (MβCD) and measured signaling events downstream of CLEC-2 activation. Lipid raft disruption decreases platelet aggregation induced by CLEC-2 agonists. The inhibition of platelet aggregation by the disruption of lipid rafts was rescued by the exogenous addition of epinephrine but not 2-methylthioadenosine diphosphate (2MeSADP), which suggests that lipid raft disruption effects P2Y12-mediated Gi activation but not Gz. Phosphorylation of Syk (Y525/526) and PLCγ2 (Y759), were not affected by raft disruption in CLEC-2 agonist-stimulated platelets. Furthermore, tyrosine phosphorylation of the CLEC-2 hemi-ITAM was not effected when MβCD disrupts lipid rafts. Lipid rafts do not directly contribute to CLEC-2 receptor activation in platelets. The effects of disruption of lipid rafts in in vitro assays can be attributed to inhibition of ADP feedback that potentiates CLEC-2 signaling.
Collapse
Affiliation(s)
- Bhanu Kanth Manne
- Department of Physiology, Temple University School of Medicine, Philadelphia, PA, USA; Sol Sherry Thrombosis Research Center and Temple University School of Medicine, Philadelphia, PA, USA
| | - Rachit Badolia
- Department of Physiology, Temple University School of Medicine, Philadelphia, PA, USA; Sol Sherry Thrombosis Research Center and Temple University School of Medicine, Philadelphia, PA, USA
| | - Carol A Dangelmaier
- Department of Physiology, Temple University School of Medicine, Philadelphia, PA, USA; Sol Sherry Thrombosis Research Center and Temple University School of Medicine, Philadelphia, PA, USA
| | - Satya P Kunapuli
- Department of Physiology, Temple University School of Medicine, Philadelphia, PA, USA; Sol Sherry Thrombosis Research Center and Temple University School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
50
|
Jin JO, Yu Q. Fucoidan delays apoptosis and induces pro-inflammatory cytokine production in human neutrophils. Int J Biol Macromol 2014; 73:65-71. [PMID: 25445688 DOI: 10.1016/j.ijbiomac.2014.10.059] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Revised: 10/25/2014] [Accepted: 10/30/2014] [Indexed: 01/19/2023]
Abstract
Although some immune modulatory effects of fucoidan have been elucidated, the effects of fucoidan on the apoptosis and activation of human neutrophils have not been investigated. In this study, we demonstrated that fucoidan purified from the brown seaweed Undaria pinnatifilda delays spontaneous apoptosis of human neutrophils and induces their activation. Fucoidan treatment inhibited apoptotic nuclei changes and phosphatidyl serine (PS) exposure on neutrophils cultured in vitro for 24h. The delay in neutrophil apoptosis mediated by fucoidan was associated with increased levels of the anti-apoptotic protein Mcl-1 and decreased levels of activated caspase-3. Screening of the signaling pathways by specific inhibitors indicated that fucoidan-induced delay in neutrophil apoptosis was dependent on the activation of PI3K/AKT signaling pathway, whereas MAPK signaling pathway was not critical. In addition, fucoidan enhanced the production of IL-6, IL-8 and TNF-α from neutrophils in an AKT-dependent manner. Taken together, these results demonstrated that fucoidan delays human neutrophil apoptosis and induces their production of pro-inflammatory cytokines. This knowledge could facilitate the development of novel therapeutic strategies for infectious diseases and neutropenia by controlling neutrophil homeostasis and function with fucoidan.
Collapse
Affiliation(s)
- Jun-O Jin
- Shanghai Public Health Clinical Center, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Qing Yu
- Department of Immunology and Infectios Diseases, The Forsyth Institute, Cambridge, MA, USA
| |
Collapse
|