1
|
Balla H, Borsodi K, Őrsy P, Horváth B, Molnár PJ, Lénárt Á, Kosztelnik M, Ruisanchez É, Wess J, Offermanns S, Nyirády P, Benyó Z. Intracellular signaling pathways of muscarinic acetylcholine receptor-mediated detrusor muscle contractions. Am J Physiol Renal Physiol 2023; 325:F618-F628. [PMID: 37675459 PMCID: PMC11905796 DOI: 10.1152/ajprenal.00261.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 08/10/2023] [Accepted: 08/27/2023] [Indexed: 09/08/2023] Open
Abstract
Acetylcholine plays an essential role in the regulation of detrusor muscle contractions, and antimuscarinics are widely used in the management of overactive bladder syndrome. However, several adverse effects limit their application and patients' compliance. Thus, this study aimed to further analyze the signal transduction of M2 and M3 receptors in the murine urinary bladder to eventually find more specific therapeutic targets. Experiments were performed on adult male wild-type, M2, M3, M2/M3, or Gαq/11 knockout (KO), and pertussis toxin (PTX)-treated mice. Contraction force and RhoA activity were measured in the urinary bladder smooth muscle (UBSM). Our results indicate that carbamoylcholine (CCh)-induced contractions were associated with increased activity of RhoA and were reduced in the presence of the Rho-associated kinase (ROCK) inhibitor Y-27632 in UBSM. CCh-evoked contractile responses and RhoA activation were markedly reduced in detrusor strips lacking either M2 or M3 receptors and abolished in M2/M3 KO mice. Inhibition of Gαi-coupled signaling by PTX treatment shifted the concentration-response curve of CCh to the right and diminished RhoA activation. CCh-induced contractile responses were markedly decreased in Gαq/11 KO mice; however, RhoA activation was unaffected. In conclusion, cholinergic detrusor contraction and RhoA activation are mediated by both M2 and M3 receptors. Furthermore, whereas both Gαi and Gαq/11 proteins mediate UBSM contraction, the activation at the RhoA-ROCK pathway appears to be linked specifically to Gαi. These findings may aid the identification of more specific therapeutic targets for bladder dysfunctions.NEW & NOTEWORTHY Muscarinic acetylcholine receptors are of utmost importance in physiological regulation of micturition and also in the development of voiding disorders. We demonstrate that the RhoA-Rho-associated kinase (ROCK) pathway plays a crucial role in contractions induced by cholinergic stimulation in detrusor muscle. Activation of RhoA is mediated by both M2 and M3 receptors as well as by Gi but not Gq/11 proteins. The Gi-RhoA-ROCK pathway may provide a novel therapeutic target for overactive voiding disorders.
Collapse
MESH Headings
- Animals
- Muscle Contraction/drug effects
- Signal Transduction/drug effects
- Urinary Bladder/drug effects
- Urinary Bladder/metabolism
- Male
- Mice, Knockout
- Receptor, Muscarinic M3/metabolism
- Receptor, Muscarinic M3/genetics
- Muscle, Smooth/drug effects
- Muscle, Smooth/metabolism
- rhoA GTP-Binding Protein/metabolism
- rho-Associated Kinases/metabolism
- rho-Associated Kinases/antagonists & inhibitors
- Receptor, Muscarinic M2/metabolism
- Receptor, Muscarinic M2/genetics
- Mice
- Mice, Inbred C57BL
- GTP-Binding Protein alpha Subunits, Gq-G11/genetics
- GTP-Binding Protein alpha Subunits, Gq-G11/metabolism
- Carbachol/pharmacology
Collapse
Affiliation(s)
- Helga Balla
- Institute of Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Kinga Borsodi
- Institute of Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Petra Őrsy
- Institute of Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Béla Horváth
- Institute of Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Péter József Molnár
- Institute of Translational Medicine, Semmelweis University, Budapest, Hungary
- Department of Urology, Semmelweis University, Budapest, Hungary
| | - Ádám Lénárt
- Institute of Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Mónika Kosztelnik
- Institute of Translational Medicine, Semmelweis University, Budapest, Hungary
- HUN-REN-SE Cerebrosvascular and Neurodegenerative Disease Research Group, Budapest, Hungary
| | - Éva Ruisanchez
- Institute of Translational Medicine, Semmelweis University, Budapest, Hungary
- HUN-REN-SE Cerebrosvascular and Neurodegenerative Disease Research Group, Budapest, Hungary
| | - Jürgen Wess
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland, United States
| | - Stefan Offermanns
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Péter Nyirády
- Department of Urology, Semmelweis University, Budapest, Hungary
| | - Zoltán Benyó
- Institute of Translational Medicine, Semmelweis University, Budapest, Hungary
- HUN-REN-SE Cerebrosvascular and Neurodegenerative Disease Research Group, Budapest, Hungary
| |
Collapse
|
2
|
Severin S, Consonni A, Chicanne G, Allart S, Payrastre B, Gratacap MP. SHIP1 Controls Internal Platelet Contraction and α IIbβ 3 Integrin Dynamics in Early Platelet Activation. Int J Mol Sci 2023; 24:ijms24020958. [PMID: 36674478 PMCID: PMC9860818 DOI: 10.3390/ijms24020958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/20/2022] [Accepted: 12/21/2022] [Indexed: 01/06/2023] Open
Abstract
The Src homology 2 domain-containing inositol 5-phosphatase 1 (SHIP1) is known to dephosphorylate PtdIns(3,4,5)P3 into PtdIns(3,4)P2 and to interact with several signaling proteins though its docking functions. It has been shown to negatively regulate platelet adhesion and spreading on a fibrinogen surface and to positively regulate thrombus growth. In the present study, we have investigated its role during the early phase of platelet activation. Using confocal-based morphometric analysis, we found that SHIP1 is involved in the regulation of cytoskeletal organization and internal contractile activity in thrombin-activated platelets. The absence of SHIP1 has no significant impact on thrombin-induced Akt or Erk1/2 activation, but it selectively affects the RhoA/Rho-kinase pathway and myosin IIA relocalization to the cytoskeleton. SHIP1 interacts with the spectrin-based membrane skeleton, and its absence induces a loss of sustained association of integrins to this network together with a decrease in αIIbβ3 integrin clustering following thrombin stimulation. This αIIbβ3 integrin dynamics requires the contractile cytoskeleton under the control of SHIP1. RhoA activation, internal platelet contraction, and membrane skeleton integrin association were insensitive to the inhibition of PtdIns(3,4,5)P3 synthesis or SHIP1 phosphatase activity, indicating a role for the docking properties of SHIP1 in these processes. Altogether, our data reveal a lipid-independent function for SHIP1 in the regulation of the contractile cytoskeleton and integrin dynamics in platelets.
Collapse
Affiliation(s)
- Sonia Severin
- Institut des Maladies Métabolique et Cardiovasculaire (I2MC), Inserm and Université Toulouse III Paul-Sabatier (UMR-1297), 1 Avenue J. Poulhes, CEDEX 4, 31432 Toulouse, France
- Correspondence: (S.S.); (M.-P.G.); Tel.: +33-5-31-22-41-43 (S.S.); +33-5-31-22-41-50 (M.-P.G.)
| | - Alessandra Consonni
- Institut des Maladies Métabolique et Cardiovasculaire (I2MC), Inserm and Université Toulouse III Paul-Sabatier (UMR-1297), 1 Avenue J. Poulhes, CEDEX 4, 31432 Toulouse, France
- Laboratory of Biochemistry, Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, 27100 Pavia, Italy
| | - Gaëtan Chicanne
- Institut des Maladies Métabolique et Cardiovasculaire (I2MC), Inserm and Université Toulouse III Paul-Sabatier (UMR-1297), 1 Avenue J. Poulhes, CEDEX 4, 31432 Toulouse, France
| | - Sophie Allart
- Institut Toulousain des Maladies Infectieuses et Inflammatoires (Infinity), Université Toulouse III Paul-Sabatier and Inserm (UMR-1291) and CNRS (UMR-5051), Centre Hospitalier Universitaire Purpan, CEDEX 3, 31024 Toulouse, France
| | - Bernard Payrastre
- Institut des Maladies Métabolique et Cardiovasculaire (I2MC), Inserm and Université Toulouse III Paul-Sabatier (UMR-1297), 1 Avenue J. Poulhes, CEDEX 4, 31432 Toulouse, France
- Laboratoire d’Hématologie, Centre de Référence des Pathologies Plaquettaires, Centre Hospitalier Universitaire Rangueil, CEDEX 4, 31432 Toulouse, France
| | - Marie-Pierre Gratacap
- Institut des Maladies Métabolique et Cardiovasculaire (I2MC), Inserm and Université Toulouse III Paul-Sabatier (UMR-1297), 1 Avenue J. Poulhes, CEDEX 4, 31432 Toulouse, France
- Correspondence: (S.S.); (M.-P.G.); Tel.: +33-5-31-22-41-43 (S.S.); +33-5-31-22-41-50 (M.-P.G.)
| |
Collapse
|
3
|
Comer S, Nagy Z, Bolado A, von Kriegsheim A, Gambaryan S, Walter U, Pagel O, Zahedi RP, Jurk K, Smolenski A. The RhoA regulators Myo9b and GEF-H1 are targets of cyclic nucleotide-dependent kinases in platelets. J Thromb Haemost 2020; 18:3002-3012. [PMID: 32692911 DOI: 10.1111/jth.15028] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 06/15/2020] [Accepted: 07/13/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND Circulating platelets are maintained in an inactive state by the endothelial lining of the vasculature. Endothelium-derived prostacyclin and nitric oxide stimulate cAMP- and cGMP-dependent kinases, PKA and PKG, to inhibit platelets. PKA and PKG effects include the inhibition of the GTPase RhoA, which has been suggested to involve the direct phosphorylation of RhoA on serine 188. OBJECTIVES We wanted to confirm RhoA S188 phosphorylation by cyclic nucleotide-dependent kinases and to identify possible alternative mechanisms of RhoA regulation in platelets. METHODS Phosphoproteomics data of human platelets were used to identify candidate PKA and PKG substrates. Phosphorylation of individual proteins was studied by Western blotting and Phos-tag gel electrophoresis in human platelets and transfected HEK293T cells. Pull-down assays were performed to analyze protein interaction and function. RESULTS Our data indicate that RhoA is not phosphorylated by PKA in platelets. Instead, we provide evidence that cyclic nucleotide effects are mediated through the phosphorylation of the RhoA-specific GTPase-activating protein Myo9b and the guanine nucleotide exchange factor GEF-H1. We identify Myo9b S1354 and guanine nucleotide exchange factor-H1 (GEF-H1) S886 as PKA and PKG phosphorylation sites. Myo9b S1354 phosphorylation enhances its GTPase activating protein function leading to reduced RhoA-GTP levels. GEF-H1 S886 phosphorylation stimulates binding of 14-3-3β and has been shown to inhibit GEF function by facilitating binding of GEF-H1 to microtubules. Microtubule disruption increases RhoA-GTP levels confirming the importance of GEF-H1 in platelets. CONCLUSION Phosphorylation of RhoA regulatory proteins Myo9b and GEF-H1, but not RhoA itself, is involved in cyclic nucleotide-mediated control of RhoA in human platelets.
Collapse
Affiliation(s)
- Shane Comer
- UCD School of Medicine and Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
- Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Zoltan Nagy
- UCD School of Medicine and Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
- Institute of Experimental Biomedicine, University Hospital and Rudolf Virchow Center, University of Würzburg, Würzburg, Germany
| | - Alfonso Bolado
- Cancer Research UK Edinburgh Centre, University of Edinburgh, Edinburgh, UK
| | | | - Stepan Gambaryan
- Sechenov Institute for Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St. Petersburg, Russia
| | - Ulrich Walter
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany
| | - Oliver Pagel
- Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., Dortmund, Germany
| | - René P Zahedi
- Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., Dortmund, Germany
- Segal Cancer Proteomics Centre, Lady Davis Institute, Jewish General Hospital, McGill University, Montreal, QC, Canada
| | - Kerstin Jurk
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany
| | - Albert Smolenski
- UCD School of Medicine and Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
- Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| |
Collapse
|
4
|
Signal profiling of the β 1AR reveals coupling to novel signalling pathways and distinct phenotypic responses mediated by β 1AR and β 2AR. Sci Rep 2020; 10:8779. [PMID: 32471984 PMCID: PMC7260363 DOI: 10.1038/s41598-020-65636-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 05/07/2020] [Indexed: 12/22/2022] Open
Abstract
A comprehensive understanding of signalling downstream of GPCRs requires a broad approach to capture novel signalling modalities in addition to established pathways. Here, using an array of sixteen validated BRET-based biosensors, we analyzed the ability of seven different β-adrenergic ligands to engage five distinct signalling pathways downstream of the β1-adrenergic receptor (β1AR). In addition to generating signalling signatures and capturing functional selectivity for the different ligands toward these pathways, we also revealed coupling to signalling pathways that have not previously been ascribed to the βAR. These include coupling to Gz and G12 pathways. The signalling cascade linking the β1AR to calcium mobilization was also characterized using a combination of BRET-based biosensors and CRISPR-engineered HEK 293 cells lacking the Gαs subunit or with pharmacological or genetically engineered pathway inhibitors. We show that both Gs and G12 are required for the full calcium response. Our work highlights the power of combining signal profiling with genome editing approaches to capture the full complement of GPCR signalling activities in a given cell type and to probe their underlying mechanisms.
Collapse
|
5
|
Green SM, Padula MP, Marks DC, Johnson L. The Lipid Composition of Platelets and the Impact of Storage: An Overview. Transfus Med Rev 2020; 34:108-116. [PMID: 31987597 DOI: 10.1016/j.tmrv.2019.12.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 12/01/2019] [Accepted: 12/07/2019] [Indexed: 02/07/2023]
Abstract
Lipids and bioactive lipid mediators are essential for platelet function. The lipid profile of platelets is highly dynamic due to free exchange of lipids with the plasma, release of extracellular vesicles, and both enzymatic and nonenzymatic lipid conversion. The lipidome of platelets changes in response to activation to accommodate the functional requirements of platelets, particularly for maintenance of hemostasis. Furthermore, when stored at room temperature as a component for transfusion, the lipid profile of platelets is altered. Although there is a growing interest in alternate storage conditions, such as refrigeration and cryopreservation, few contemporary studies have examined the impact of these storage modes on the lipid profile. However, evidence exists that bioactive lipid mediators produced over the storage of blood products may have functional implications once these products are transfused. As such, there is a need to determine the changes occurring to the lipid profile of these products over storage. This review outlines the role of lipids in platelets and discusses the current state of lipidomics for studying platelet components for transfusion in an effort to highlight the necessity for additional transfusion-focused investigations.
Collapse
Affiliation(s)
- Sarah M Green
- Research & Development, Australian Red Cross Blood Service, Alexandria, NSW, Australia; School of Life Sciences, University of Technology Sydney, Sydney, NSW, Australia
| | - Matthew P Padula
- School of Life Sciences, University of Technology Sydney, Sydney, NSW, Australia
| | - Denese C Marks
- Research & Development, Australian Red Cross Blood Service, Alexandria, NSW, Australia; Sydney Medical School, The University of Sydney, Camperdown, NSW, Australia
| | - Lacey Johnson
- Research & Development, Australian Red Cross Blood Service, Alexandria, NSW, Australia.
| |
Collapse
|
6
|
Qasim H, Karim ZA, Hernandez KR, Lozano D, Khasawneh FT, Alshbool FZ. Arhgef1 Plays a Vital Role in Platelet Function and Thrombogenesis. J Am Heart Assoc 2019; 8:e011712. [PMID: 30994039 PMCID: PMC6512111 DOI: 10.1161/jaha.118.011712] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 03/25/2019] [Indexed: 01/09/2023]
Abstract
Background Platelets are the cellular mediators of hemostasis and thrombosis, and their function is regulated by a number of molecular mediators, such as small GTP ases. These small GTP ases are themselves regulated by guanine nucleotide exchange factors such as Arhgefs, several of which are found in platelets, including the highly expressed Arhgef1. However, the role of Arhgef1 in platelets has not yet been investigated. Methods and Results We employed mice with genetic deletion of Arhgef1 (ie, Arhgef1-/-) and investigated their platelet phenotype by employing a host of in vivo and in vitro platelet assays. Our results indicate that Arhgef1-/- mice had prolonged carotid artery occlusion and tail bleeding times. Moreover, platelets from these mice exhibited defective aggregation, dense and α granule secretion, α II bβ3 integrin activation, clot retraction and spreading, in comparison to their wild-type littermates. Finally, we also found that the mechanism by which Arhgef1 regulates platelets is mediated in part by a defect in the activation of the RhoA-Rho-associated kinase axis, but not Rap1b. Conclusions Our data demonstrate, for the first time, that Arhgef1 plays a critical role in platelet function, in vitro and in vivo.
Collapse
Affiliation(s)
- Hanan Qasim
- Department of Pharmaceutical SciencesSchool of PharmacyThe University of Texas El PasoEl PasoTX
| | - Zubair A. Karim
- Department of Pharmaceutical SciencesSchool of PharmacyThe University of Texas El PasoEl PasoTX
| | - Keziah R. Hernandez
- Department of Pharmaceutical SciencesSchool of PharmacyThe University of Texas El PasoEl PasoTX
| | | | - Fadi T. Khasawneh
- Department of Pharmaceutical SciencesSchool of PharmacyThe University of Texas El PasoEl PasoTX
| | - Fatima Z. Alshbool
- Department of Pharmaceutical SciencesSchool of PharmacyThe University of Texas El PasoEl PasoTX
| |
Collapse
|
7
|
Nagy Z, Smolenski A. Cyclic nucleotide-dependent inhibitory signaling interweaves with activating pathways to determine platelet responses. Res Pract Thromb Haemost 2018; 2:558-571. [PMID: 30046761 PMCID: PMC6046581 DOI: 10.1002/rth2.12122] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 04/20/2018] [Indexed: 12/22/2022] Open
Abstract
Platelets are regulated by extracellular cues that impact on intracellular signaling. The endothelium releases prostacyclin and nitric oxide which stimulate the synthesis of cyclic nucleotides cAMP and cGMP leading to platelet inhibition. Other inhibitory mechanisms involve immunoreceptor tyrosine-based inhibition motif-containing receptors, intracellular receptors and receptor desensitization. Inhibitory cyclic nucleotide pathways are traditionally thought to represent a passive background system keeping platelets in a quiescent state. In contrast, cyclic nucleotides are increasingly seen to be dynamically involved in most aspects of platelet regulation. This review focuses on crosstalk between activating and cyclic nucleotide-mediated inhibitory pathways highlighting emerging new hub structures and signaling mechanisms. In particular, interactions of plasma membrane receptors like P2Y12 and GPIb/IX/V with the cyclic nucleotide system are described. Furthermore, differential regulation of the RGS18 complex, second messengers, protein kinases, and phosphatases are presented, and control over small G-proteins by guanine-nucleotide exchange factors and GTPase-activating proteins are outlined. Possible clinical implications of signaling crosstalk are discussed.
Collapse
Affiliation(s)
- Zoltan Nagy
- Institute of Cardiovascular SciencesCollege of Medical and Dental SciencesUniversity of BirminghamBirminghamUK
| | - Albert Smolenski
- UCD School of MedicineUniversity College DublinDublinIreland
- UCD Conway InstituteUniversity College DublinDublinIreland
- Irish Centre for Vascular BiologyRoyal College of Surgeons in IrelandDublinIreland
| |
Collapse
|
8
|
Xu H, Lu H, Zhu X, Wang W, Zhang Z, Fu H, Ma S, Luo Y, Fu J. Inhibitory effects of luteolin‑4'‑O‑β‑D‑glucopyranoside on P2Y12 and thromboxane A2 receptor‑mediated amplification of platelet activation in vitro. Int J Mol Med 2018; 42:615-624. [PMID: 29693158 DOI: 10.3892/ijmm.2018.3634] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 03/27/2018] [Indexed: 11/06/2022] Open
Abstract
Platelet activation and subsequent accumulation at sites of vascular injury are central to thrombus formation, which is considered to be a trigger of several cardiovascular diseases. Callicarpa nudiflora (C. nudiflora) Hook is a traditional Chinese medicinal herb for promoting blood circulation by removing blood stasis. In our previous study, several compounds extracted from this herb, including luteolin‑4'‑O‑β‑D‑glucopyranoside (LGP), were revealed to exert inhibitory effects on adenosine diphosphate (ADP)‑induced platelet aggregation. The aim of present study was to confirm these antiplatelet effects and elucidate the potential mechanisms. Using a platelet‑aggregation assay, it was revealed that LGP significantly inhibited platelet aggregation induced by ADP, U46619 and arachidonic acid. It was also found that LGP exhibited marked inhibitory effects on the activation of αIIbβ3 integrin, the secretion of serotonin from granules, and the synthesis of thromboxane A2. In addition, the results showed that LGP suppressed Ras homolog family member A and phosphoinositide 3‑kinase/Akt/glycogen synthase kinase 3β signal transduction. Data from a radiolabeled ligand‑binding assay indicated that LGP exhibited apparent competing effects on thromboxane receptor (TP) and P2Y12 receptors. In conclusion, the data presented here demonstrated that LGP, a natural compound from C. nudiflora Hook, inhibited the development of platelet aggregation and amplification of platelet activation. These inhibitory effects may be associated with its dual‑receptor inhibition on P2Y12 and TP receptors.
Collapse
Affiliation(s)
- Huanjun Xu
- Department of Pharmacology, School of Pharmacy, Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi 330004, P.R. China
| | - Hong Lu
- Network and Educational Technology Center, Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi 330004, P.R. China
| | - Xiaocui Zhu
- Department of Pharmacology, School of Pharmacy, Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi 330004, P.R. China
| | - Wei Wang
- Department of Pharmacology, School of Pharmacy, Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi 330004, P.R. China
| | - Zhoumiao Zhang
- Department of Pharmacology, School of Pharmacy, Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi 330004, P.R. China
| | - Huizheng Fu
- Jiangxi Provincial Institute for Drug Control, Nanchang, Jiangxi 330029, P.R. China
| | - Shuangcheng Ma
- National Institutes for Food and Drug Control, Beijing 100050, P.R. China
| | - Yuehua Luo
- Jiangxi Provincial Institute for Drug Control, Nanchang, Jiangxi 330029, P.R. China
| | - Jianjiang Fu
- Department of Pharmacology, School of Pharmacy, Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi 330004, P.R. China
| |
Collapse
|
9
|
Makhoul S, Walter E, Pagel O, Walter U, Sickmann A, Gambaryan S, Smolenski A, Zahedi RP, Jurk K. Effects of the NO/soluble guanylate cyclase/cGMP system on the functions of human platelets. Nitric Oxide 2018; 76:71-80. [PMID: 29550521 DOI: 10.1016/j.niox.2018.03.008] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 03/03/2018] [Accepted: 03/12/2018] [Indexed: 02/07/2023]
Abstract
Platelets are circulating sentinels of vascular integrity and are activated, inhibited, or modulated by multiple hormones, vasoactive substances or drugs. Endothelium- or drug-derived NO strongly inhibits platelet activation via activation of the soluble guanylate cyclase (sGC) and cGMP elevation, often in synergy with cAMP-elevation by prostacyclin. However, the molecular mechanisms and diversity of cGMP effects in platelets are poorly understood and sometimes controversial. Recently, we established the quantitative human platelet proteome, the iloprost/prostacyclin/cAMP/protein kinase A (PKA)-regulated phosphoproteome, and the interactions of the ADP- and iloprost/prostacyclin-affected phosphoproteome. We also showed that the sGC stimulator riociguat is in vitro a highly specific inhibitor, via cGMP, of various functions of human platelets. Here, we review the regulatory role of the cGMP/protein kinase G (PKG) system in human platelet function, and our current approaches to establish and analyze the phosphoproteome after selective stimulation of the sGC/cGMP pathway by NO donors and riociguat. Present data indicate an extensive and diverse NO/riociguat/cGMP phosphoproteome, which has to be compared with the cAMP phosphoproteome. In particular, sGC/cGMP-regulated phosphorylation of many membrane proteins, G-proteins and their regulators, signaling molecules, protein kinases, and proteins involved in Ca2+ regulation, suggests that the sGC/cGMP system targets multiple signaling networks rather than a limited number of PKG substrate proteins.
Collapse
Affiliation(s)
- Stephanie Makhoul
- University Medical Center Mainz, Center for Thrombosis and Hemostasis (CTH), Mainz, Germany
| | - Elena Walter
- University Medical Center Mainz, Center for Thrombosis and Hemostasis (CTH), Mainz, Germany
| | - Oliver Pagel
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e. V., Dortmund, Germany
| | - Ulrich Walter
- University Medical Center Mainz, Center for Thrombosis and Hemostasis (CTH), Mainz, Germany
| | - Albert Sickmann
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e. V., Dortmund, Germany; Ruhr Universität Bochum, Medizinisches Proteom Center, Medizinische Fakultät, Bochum, Germany; Department of Chemistry, College of Physical Sciences, University of Aberdeen, Aberdeen, UK
| | - Stepan Gambaryan
- University Medical Center Mainz, Center for Thrombosis and Hemostasis (CTH), Mainz, Germany; Russian Academy of Sciences, Sechenov Institute of Evolutionary Physiology and Biochemistry, St. Petersburg, Russia; St. Petersburg State University, Department of Cytology and Histology, St. Petersburg, Russia
| | - Albert Smolenski
- Conway Institute of Biomolecular & Biomedical Research, Univ. College Dublin, Dublin, Ireland; Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - René P Zahedi
- Gerald Bronfman Department of Oncology, Jewish General Hospital, McGill University , Montreal, Quebec H4A 3T2, Canada; Segal Cancer Proteomics Centre, Lady Davis Institute, Jewish General Hospital, McGill University , Montreal, Quebec H3T 1E2, Canada
| | - Kerstin Jurk
- University Medical Center Mainz, Center for Thrombosis and Hemostasis (CTH), Mainz, Germany.
| |
Collapse
|
10
|
Nagy Z, Wynne K, von Kriegsheim A, Gambaryan S, Smolenski A. Cyclic Nucleotide-dependent Protein Kinases Target ARHGAP17 and ARHGEF6 Complexes in Platelets. J Biol Chem 2015; 290:29974-83. [PMID: 26507661 DOI: 10.1074/jbc.m115.678003] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Indexed: 01/01/2023] Open
Abstract
Endothelial cells release prostacyclin (PGI2) and nitric oxide (NO) to inhibit platelet functions. PGI2 and NO effects are mediated by cyclic nucleotides, cAMP- and cGMP-dependent protein kinases (PKA, PKG), and largely unknown PKA and PKG substrate proteins. The small G-protein Rac1 plays a key role in platelets and was suggested to be a target of cyclic nucleotide signaling. We confirm that PKA and PKG activation reduces Rac1-GTP levels. Screening for potential mediators of this effect resulted in the identification of the Rac1-specific GTPase-activating protein ARHGAP17 and the guanine nucleotide exchange factor ARHGEF6 as new PKA and PKG substrates in platelets. We mapped the PKA/PKG phosphorylation sites to serine 702 on ARHGAP17 using Phos-tag gels and to serine 684 on ARHGEF6. We show that ARHGAP17 binds to the actin-regulating CIP4 protein in platelets and that Ser-702 phosphorylation interferes with this interaction. Reduced CIP4 binding results in enhanced inhibition of cell migration by ARHGAP17. Furthermore, we show that ARHGEF6 is constitutively linked to GIT1, a GAP of Arf family small G proteins, and that ARHGEF6 phosphorylation enables binding of the 14-3-3 adaptor protein to the ARHGEF6/GIT1 complex. PKA and PKG induced rearrangement of ARHGAP17- and ARHGEF6-associated protein complexes might contribute to Rac1 regulation and platelet inhibition.
Collapse
Affiliation(s)
- Zoltan Nagy
- From the UCD Conway Institute and the School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| | - Kieran Wynne
- Mass Spectrometry Resource, UCD Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | | | - Stepan Gambaryan
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, 44 Thorez Prospect, St. Petersburg, 194223 Russia
| | - Albert Smolenski
- From the UCD Conway Institute and the School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland,
| |
Collapse
|
11
|
Elvers M. RhoGAPs and Rho GTPases in platelets. Hamostaseologie 2015; 36:168-77. [PMID: 25639730 DOI: 10.5482/hamo-14-09-0046] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 01/13/2015] [Indexed: 01/03/2023] Open
Abstract
Platelet cytoskeletal reorganization is essential for platelet adhesion and thrombus formation in hemostasis and thrombosis. The Rho GTPases RhoA, Rac1 and Cdc42 are the main players in cytoskeletal dynamics of platelets responsible for the formation of filopodia and lamellipodia to strongly increase the platelet surface upon activation. They are involved in platelet activation and aggregate formation including platelet secretion, integrin activation and arterial thrombus formation. The activity of Rho GTPases is tightly controlled by different proteins such as GTPase-activating proteins (GAPs). GAPs stimulate GTP hydrolysis to terminate Rho signaling. The role and impact of GAPs in platelets is not well-defined and many of the RhoGAPs identified are not known to be present in platelets or to have any function in platelets. The recently identified RhoGAPs Oligophrenin1 (OPHN1) and Nadrin regulate the activity of RhoA, Rac1 and Cdc42 and subsequent platelet cytoskeletal reorganization, platelet activation and thrombus formation. In the last years, the analysis of genetically modified mice helped to gain the understanding of Rho GTPases and their regulators in cytoskeletal rearrangements and other Rho mediated cellular processes in platelets.
Collapse
Affiliation(s)
- Margitta Elvers
- Margitta Elvers, Ph.D., Department of Clinical and Experimental Hemostasis, Hemotherapy and Transfusion Medicine, Heinrich-Heine-University Duesseldorf, Moorenstr. 5, 40225 Duesseldorf, Germany, Tel. +49/(0)211/81-08851, Fax -17498., E-mail:
| |
Collapse
|
12
|
Kassassir H, Siewiera K, Sychowski R, Watała C. Can the antiplatelet effects of cangrelor be reliably studied in mice under in vivo and in vitro conditions using flow cytometry? Pharmacol Rep 2014; 65:870-83. [PMID: 24145081 DOI: 10.1016/s1734-1140(13)71068-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2012] [Revised: 02/05/2013] [Indexed: 11/17/2022]
Abstract
BACKGROUND The effects of blood platelet inhibitors are often not quite equivalent under in vivo and in vitro conditions. Amongst various models of human pathology using laboratory animals, mice offer several benefits that make them convenient tools for studying the putative therapeutic value of various compounds. However, despite its advantages, the mouse model has methodological limitations concerning the small amount of blood available and technical difficulties with its collection. Among the variety of available methods used to study blood platelet activation and/or reactivity, flow cytometry seems an attractive technique that largely minimizes the constraints of using small rodents and enables outcomes of laboratory research to be transferred successfully to clinical practice. In this study we aimed at a critical evaluation of the optimal discriminative flow cytometric protocol, useful for reliable studies of the effect of cangrelor, a P2Y12 receptor antagonist, on mouse platelets under in vitro and in vivo conditions. METHODS Blood samples were drawn from two-month-old female BALB/c mice. Protocols differing in methods of anesthesia, blood withdrawal, anticoagulation, gating antibodies, blood preparation and fixation were tested to optimize the one best suited to discrimination between resting and activated platelets. The antiplatelet capabilities of cangrelor were tested in vitro (140 μM in whole blood) and in vivo (7.8 mg/kg b.w. administered once, directly into the bloodstream through the vena cava of the anesthetized animal, 15 min prior to blood withdrawal). Expressions of P-selectin, activated α(IIb)β3 complex and GPIba were monitored using two-color flow cytometry. RESULTS "Washed blood" anticoagulated with low molecular weight heparin demonstrated the best discrimination between circulating (resting) platelets and upon their in vitro response to thrombin, collagen or ADP in freshly-stained unfixed cell suspensions. Cangrelor inhibited the expression of the active form of the integrin a(IIb)β3 to approximately the same extent under in vitro and in vivo conditions (84.5 ± 7.7% vs. 75.4 ± 19.5% for the in vitro and in vivo approaches, respectively, n.s.). CONCLUSIONS The agreement between the in vivo and in vitro approaches with respect to cangrelor-inhibited hallmarks of blood platelet activation and reactivity supports our proposal that flow cytometry is useful and reliable for determining the effects of antiplatelet agents on the activation of circulating platelets in the mouse model, as well as the in vitro response of platelets to agonists.
Collapse
Affiliation(s)
- Hassan Kassassir
- Laboratory of Animal Experimental Models, Department of Haemostasis and Haemostatic Disorders, Medical University of Lodz, Veterans' Central Hospital, Żeromskiego 113, PL 90-549 Łódź, Poland.
| | | | | | | |
Collapse
|
13
|
Kim S, Dangelmaier C, Bhavanasi D, Meng S, Wang H, Goldfinger LE, Kunapuli SP. RhoG protein regulates glycoprotein VI-Fc receptor γ-chain complex-mediated platelet activation and thrombus formation. J Biol Chem 2013; 288:34230-34238. [PMID: 24106269 DOI: 10.1074/jbc.m113.504928] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We investigated the mechanism of activation and functional role of a hitherto uncharacterized signaling molecule, RhoG, in platelets. We demonstrate for the first time the expression and activation of RhoG in platelets. Platelet aggregation, integrin αIIbβ3 activation, and α-granule and dense granule secretion in response to the glycoprotein VI (GPVI) agonists collagen-related peptide (CRP) and convulxin were significantly inhibited in RhoG-deficient platelets. In contrast, 2-MeSADP- and AYPGKF-induced platelet aggregation and secretion were minimally affected in RhoG-deficient platelets, indicating that the function of RhoG in platelets is GPVI-specific. CRP-induced phosphorylation of Syk, Akt, and ERK, but not SFK (Src family kinase), was significantly reduced in RhoG-deficient platelets. CRP-induced RhoG activation was consistently abolished by a pan-SFK inhibitor but not by Syk or PI3K inhibitors. Interestingly, unlike CRP, platelet aggregation and Syk phosphorylation induced by fucoidan, a CLEC-2 agonist, were unaffected in RhoG-deficient platelets. Finally, RhoG(-/-) mice had a significant delay in time to thrombotic occlusion in cremaster arterioles compared with wild-type littermates, indicating the important in vivo functional role of RhoG in platelets. Our data demonstrate that RhoG is expressed and activated in platelets, plays an important role in GPVI-Fc receptor γ-chain complex-mediated platelet activation, and is critical for thrombus formation in vivo.
Collapse
Affiliation(s)
- Soochong Kim
- Department of Physiology, Temple University School of Medicine, Philadelphia, Pennsylvania 19140; Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania 19140
| | - Carol Dangelmaier
- Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania 19140
| | - Dheeraj Bhavanasi
- Department of Physiology, Temple University School of Medicine, Philadelphia, Pennsylvania 19140; Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania 19140
| | - Shu Meng
- Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania 19140; Department of Pharmacology, Temple University School of Medicine, Philadelphia, Pennsylvania 19140
| | - Hong Wang
- Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania 19140; Department of Pharmacology, Temple University School of Medicine, Philadelphia, Pennsylvania 19140
| | - Lawrence E Goldfinger
- Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania 19140; Department of Anatomy and Cell Biology, Temple University School of Medicine, Philadelphia, Pennsylvania 19140.
| | - Satya P Kunapuli
- Department of Physiology, Temple University School of Medicine, Philadelphia, Pennsylvania 19140; Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania 19140; Department of Pharmacology, Temple University School of Medicine, Philadelphia, Pennsylvania 19140.
| |
Collapse
|
14
|
cAMP signaling regulates platelet myosin light chain (MLC) phosphorylation and shape change through targeting the RhoA-Rho kinase-MLC phosphatase signaling pathway. Blood 2013; 122:3533-45. [PMID: 24100445 DOI: 10.1182/blood-2013-03-487850] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Cyclic adenosine monophosphate (cAMP)-dependent signaling modulates platelet shape change through unknown mechanisms. We examined the effects of cAMP signaling on platelet contractile machinery. Prostaglandin E1 (PGE1)-mediated inhibition of thrombin-stimulated shape change was accompanied by diminished phosphorylation of myosin light chain (MLC). Since thrombin stimulates phospho-MLC through RhoA/Rho-associated, coiled-coil containing protein kinase (ROCK)-dependent inhibition of MLC phosphatase (MLCP), we examined the effects of cAMP on this pathway. Thrombin stimulated the membrane localization of RhoA and the formation of a signaling complex of RhoA/ROCK2/myosin phosphatase-targeting subunit 1 (MYPT1). This resulted in ROCK-mediated phosphorylation of MYPT1 on threonine 853 (thr(853)), the disassociation of the catalytic subunit protein phosphatase 1δ (PP1δ) from MYPT1 and inhibition of basal MLCP activity. Treatment of platelets with PGE1 prevented thrombin-induced phospho-MYPT1-thr(853) in a protein kinase A (PKA)-dependent manner. Examination of the molecular mechanisms revealed that PGE1 induced the phosphorylation of RhoA on serine(188) through a pathway requiring cAMP and PKA. This event inhibited the membrane relocalization of RhoA, prevented the association of RhoA with ROCK2 and MYPT1, attenuated the dissociation of PP1δ from MYPT1, and thereby restored basal MLCP activity leading to a decrease in phospho-MLC. These data reveal a new mechanism by which the cAMP-PKA signaling pathway regulates platelet function.
Collapse
|
15
|
El-Haibi CP, Sharma P, Singh R, Gupta P, Taub DD, Singh S, Lillard JW. Differential G protein subunit expression by prostate cancer cells and their interaction with CXCR5. Mol Cancer 2013; 12:64. [PMID: 23773523 PMCID: PMC3720210 DOI: 10.1186/1476-4598-12-64] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2013] [Accepted: 06/05/2013] [Indexed: 11/16/2022] Open
Abstract
Background Prostate cancer (PCa) cell lines and tissues differentially express CXCR5, which positively correlate with PCa progression, and mediate PCa cell migration and invasion following interaction with CXCL13. However, the differential expression of G protein α, β, and γ subunits by PCa cell lines and the precise combination of these proteins with CXCR5 has not been elucidated. Methods We examined differences in G protein expression of normal prostate (RWPE-1) and PCa cell lines (LNCaP, C4-2B, and PC3) by western blot analysis. Further, we immunoprecipitated CXCR5 with different G protein subunits, and CXCR4, following CXCL13 stimulation. To investigate constitutive coupling of CXCR5 with CXCR4 and PAR-1 we performed invasion assay in PCa cells transfected with Gαq/i2 or Gα13 siRNA, following CXCL13 treatment. We also investigated Rac and RhoA activity by G-LISA activation assay in PCa cells following CXCL13/thrombin stimulation. Result Of the 22 G proteins studied, Gαi1-3, Gβ1-4, Gγ5, Gγ7, and Gγ10 were expressed by both normal and PCa cell lines. Gαs was moderately expressed in C4-2B and PC3 cell lines, Gαq/11 was only present in RWPE-1 and LNCaP cell lines, while Gα12 and Gα13 were expressed in C4-2B and PC3 cell lines. Gγ9 was expressed only in PCa cell lines. Gα16, Gβ5, Gγ1-4, and Gγ13 were not detected in any of the cell lines studied. Surprisingly, CXCR4 co-immunoprecipitated with CXCR5 in PCa cell lines irrespective of CXCL13 treatment. We also identified specific G protein isoforms coupled to CXCR5 in its resting and active states. Gαq/11/Gβ3/Gγ9 in LNCaP and Gαi2/Gβ3/Gγ9 in C4-2B and PC3 cell lines, were coupled to CXCR5 and disassociated following CXCL13 stimulation. Interestingly, Gα13 co-immunoprecipitated with CXCR5 in CXCL13-treated, but not in untreated PCa cell lines. Inhibition of Gαq/i2 significantly decreased the ability of cells to invade, whereas silencing Gα13 did not affect CXCL13-dependent cell invasion. Finally, CXCL13 treatment significantly increased Rac activity in Gαq/i2 dependent manner, but not RhoA activity, in PCa cell lines. Conclusions These findings offer insight into molecular mechanisms of PCa progression and can help to design some therapeutic strategies involving CXCR5 and/or CXCL13 blockade and specific G protein inhibition to abrogate PCa metastasis.
Collapse
|
16
|
Abstract
Phosphatidylinositol and its phosphorylated derivatives, phosphoinositides, are minor constituents of phospholipids at the cellular membrane level. Nevertheless, phosphatidylinositol and phosphoinositides represent essential components of intracellular signaling that regulate diverse cellular processes, including platelet plug formation. Accumulating evidence indicates that the metabolism of phosphoinositides is temporally and spatially modulated by the opposing effects of specific phosphoinositide-metabolizing enzymes, including lipid kinases, lipid phosphatases, and phospholipases. Each of these enzymes generates a selective phosphoinositide or second messenger within precise cellular compartments. Intriguingly, phosphoinositide-metabolizing enzymes exist in different isoforms, which all produce the same phosphoinositide products. Recent studies using isoform-specific mouse models and chemical inhibitors have elucidated that the different isoforms of phosphoinositide-metabolizing enzymes have nonredundant functions and provide an additional layer of complexity to the temporo-spatial organization of intracellular signaling events. In this review, we will discuss recent advances in our understanding of phosphoinositide organization during platelet activation.
Collapse
|
17
|
Abstract
Lysophosphatidic acid (LPA) is a potent bioactive phospholipid. As many other biological active lipids, LPA is an autacoid: it is formed locally on demand, and it acts locally near its site of synthesis. LPA has a plethora of biological activities on blood cells (platelets, monocytes) and cells of the vessel wall (endothelial cells, smooth muscle cells, macrophages) that are all key players in atherosclerotic and atherothrombotic processes. The specific cellular actions of LPA are determined by its multifaceted molecular structures, the expression of multiple G-protein coupled LPA receptors at the cell surface and their diverse coupling to intracellular signalling pathways. Numerous studies have now shown that LPA has thrombogenic and atherogenic actions. Here, we aim to provide a comprehensive, yet concise, thoughtful and critical review of this exciting research area and to pinpoint potential pharmacological targets for inhibiting thrombogenic and atherogenic activities of LPA. We hope that the review will serve to accelerate knowledge of basic and clinical science, and to foster drug development in the field of LPA and atherosclerotic/atherothrombotic diseases.
Collapse
Affiliation(s)
- Andreas Schober
- Institute for Molecular Cardiovascular Research, RWTH Aachen University, Aachen, Germany
| | | |
Collapse
|
18
|
Abstract
The Rho family of GTP binding proteins, also commonly referred to as the Rho GTPases, are master regulators of the platelet cytoskeleton and platelet function. These low-molecular-weight or 'small' GTPases act as signaling switches in the spatial and temporal transduction, and amplification of signals from platelet cell surface receptors to the intracellular signaling pathways that drive platelet function. The Rho GTPase family members RhoA, Cdc42 and Rac1 have emerged as key regulators in the dynamics of the actin cytoskeleton in platelets and play key roles in platelet aggregation, secretion, spreading and thrombus formation. Rho GTPase regulators, including GEFs and GAPs and downstream effectors, such as the WASPs, formins and PAKs, may also regulate platelet activation and function. In this review, we provide an overview of Rho GTPase signaling in platelet physiology. Previous studies of Rho GTPases and platelets have had a shared history, as platelets have served as an ideal, non-transformed cellular model to characterize Rho function. Likewise, recent studies of the cell biology of Rho GTPase family members have helped to build an understanding of the molecular regulation of platelet function and will continue to do so through the further characterization of Rho GTPases as well as Rho GAPs, GEFs, RhoGDIs and Rho effectors in actin reorganization and other Rho-driven cellular processes.
Collapse
Affiliation(s)
- J E Aslan
- Department of Biomedical Engineering and Cell & Developmental Biology, School of Medicine, Oregon Health & Science University, Portland, OR 97239, USA.
| | | |
Collapse
|
19
|
Steele BM, Harper MT, Smolenski AP, Alkazemi N, Poole AW, Fitzgerald DJ, Maguire PB. WNT-3a modulates platelet function by regulating small GTPase activity. FEBS Lett 2012; 586:2267-72. [PMID: 22705156 DOI: 10.1016/j.febslet.2012.05.060] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2012] [Revised: 05/04/2012] [Accepted: 05/23/2012] [Indexed: 12/13/2022]
Abstract
Here we provide evidence that WNT-3a modulates platelet function by regulating the activity of four key GTPase proteins: Rap1, Cdc42, Rac1 and RhoA. We observe WNT-3a to differentially regulate small GTPase activity in platelets, promoting the GDP-bound form of Rap1b to inhibit integrin-α(IIb)β(3) adhesion, while concomitantly increasing Cdc42 and Rac1-GTP levels thereby disrupting normal platelet spreading. We demonstrate that Daam-1 interacts with Dishevelled upon platelet activation, which correlates with increased RhoA-GTP levels. Upon pre-treatment with WNT-3a, this complex disassociates, concurrent with a reduction in RhoA-GTP. Together these data implicate WNT-3a as a novel upstream regulator of small GTPase activity in platelets.
Collapse
Affiliation(s)
- Brian M Steele
- UCD Conway Institute, University College Dublin, Belfield, Dublin D4, Ireland.
| | | | | | | | | | | | | |
Collapse
|
20
|
Abstract
Endothelial prostacyclin and nitric oxide potently inhibit platelet functions. Prostacyclin and nitric oxide actions are mediated by platelet adenylyl and guanylyl cyclases, which synthesize cyclic AMP (cAMP) and cyclic GMP (cGMP), respectively. Cyclic nucleotides stimulate cAMP-dependent protein kinase (protein kinase A [PKA]I and PKAII) and cGMP-dependent protein kinase (protein kinase G [PKG]I) to phosphorylate a broad panel of substrate proteins. Substrate phosphorylation results in the inactivation of small G-proteins of the Ras and Rho families, inhibition of the release of Ca(2+) from intracellular stores, and modulation of actin cytoskeleton dynamics. Thus, PKA/PKG substrates translate prostacyclin and nitric oxide signals into a block of platelet adhesion, granule release, and aggregation. cAMP and cGMP are degraded by phosphodiesterases, which might restrict signaling to specific subcellular compartments. An emerging principle of cyclic nucleotide signaling in platelets is the high degree of interconnection between activating and cAMP/cGMP-dependent inhibitory signaling pathways at all levels, including cAMP/cGMP synthesis and breakdown, and PKA/PKG-mediated substrate phosphorylation. Furthermore, defects in cAMP/cGMP pathways might contribute to platelet hyperreactivity in cardiovascular disease. This article focuses on recent insights into the regulation of the cAMP/cGMP signaling network and on new targets of PKA and PKG in platelets.
Collapse
Affiliation(s)
- A Smolenski
- UCD Conway Institute, UCD School of Medicine and Medical Science, University College Dublin, Belfield, Dublin, Ireland.
| |
Collapse
|
21
|
Megakaryocyte-specific RhoA deficiency causes macrothrombocytopenia and defective platelet activation in hemostasis and thrombosis. Blood 2012; 119:1054-63. [DOI: 10.1182/blood-2011-08-372193] [Citation(s) in RCA: 124] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Abstract
Vascular injury initiates rapid platelet activation that is critical for hemostasis, but it also may cause thrombotic diseases, such as myocardial infarction or ischemic stroke. Reorganizations of the platelet cytoskeleton are crucial for platelet shape change and secretion and are thought to involve activation of the small GTPase RhoA. In this study, we analyzed the in vitro and in vivo consequences of megakaryocyte- and platelet-specific RhoA gene deletion in mice. We found a pronounced macrothrombocytopenia in RhoA-deficient mice, with platelet counts of approximately half that of wild-type controls. The mutant cells displayed an altered shape but only a moderately reduced life span. Shape change of RhoA-deficient platelets in response to G13-coupled agonists was abolished, and it was impaired in response to Gq stimulation. Similarly, RhoA was required for efficient secretion of α and dense granules downstream of G13 and Gq. Furthermore, RhoA was essential for integrin-mediated clot retraction but not for actomyosin rearrangements and spreading of activated platelets on fibrinogen. In vivo, RhoA deficiency resulted in markedly prolonged tail bleeding times but also significant protection in different models of arterial thrombosis and in a model of ischemic stroke. Together, these results establish RhoA as an important regulator of platelet function in thrombosis and hemostasis.
Collapse
|
22
|
Fediuk J, Gutsol A, Nolette N, Dakshinamurti S. Thromboxane-induced actin polymerization in hypoxic pulmonary artery is independent of Rho. Am J Physiol Lung Cell Mol Physiol 2012; 302:L13-26. [DOI: 10.1152/ajplung.00016.2011] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Actin polymerization (APM), regulated by Rho GTPases, promotes myocyte force generation. Hypoxia is known to impede postnatal disassembly of the actin cytoskeleton in pulmonary arterial (PA) myocytes. We compared basal and agonist-induced APM in myocytes from PA and descending aorta (Ao), under hypoxic and normoxic conditions. We also examined effects of thromboxane challenge on force generation and cytoskeletal assembly in resistance PA and renal arteries from neonatal swine with persistent pulmonary hypertension (PPHN) induced by 72-h normobaric hypoxia, compared with age-matched controls. Synthetic and contractile phenotype myocytes from neonatal porcine PA or Ao were grown in hypoxia (10% O2) or normoxia (21% O2) for 7 days, then challenged with 10−6 M thromboxane mimetic U46619. F/G actin ratio was quantified by laser-scanning cytometry and by cytoskeletal fractionation. Thromboxane receptor (TP) G protein coupling was measured by immunoprecipitation and probing for Gαq, G12, or G13, RhoA activation by Rhotekin-RBD affinity precipitation, and LIM kinase (LIMK) and cofilin phosphorylation by Western blot. Isometric force to serial concentrations of U46619 was measured in muscular pulmonary and renal arteries from PPHN and control swine; APM was quantified in fixed contracted vessels. Contractile PA myocytes exhibit marked Rho-dependent APM in hypoxia, with increased active RhoA and LIMK phosphorylation. Their additional APM response to U46619 challenge is independent of RhoA, reflecting decreased TP association with G12/13 in favor of Gαq. In contrast, hypoxic contractile Ao myocytes polymerize actin modestly and depolymerize to U46619. Both basal APM and the APM response to U46619 are increased in PPHN PA. APM corresponds with increased force generation to U46619 challenge in PPHN PA but not renal arteries.
Collapse
Affiliation(s)
- Jena Fediuk
- Departments of 1Physiology and
- Biology of Breathing Group, Manitoba Institute of Child Health, Winnipeg, Manitoba, Canada
| | - Alexey Gutsol
- Biology of Breathing Group, Manitoba Institute of Child Health, Winnipeg, Manitoba, Canada
| | - Nora Nolette
- Biology of Breathing Group, Manitoba Institute of Child Health, Winnipeg, Manitoba, Canada
| | - Shyamala Dakshinamurti
- Departments of 1Physiology and
- Pediatrics, University of Manitoba
- Biology of Breathing Group, Manitoba Institute of Child Health, Winnipeg, Manitoba, Canada
| |
Collapse
|
23
|
Qian F, Le Breton GC, Chen J, Deng J, Christman JW, Wu D, Ye RD. Role for the guanine nucleotide exchange factor phosphatidylinositol-3,4,5-trisphosphate-dependent rac exchanger 1 in platelet secretion and aggregation. Arterioscler Thromb Vasc Biol 2011; 32:768-77. [PMID: 22207728 DOI: 10.1161/atvbaha.111.243675] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
OBJECTIVE Recent studies have shown a role for Rac1 in regulating platelet functions, but how Rac1 is activated in platelets remains unclear. Phosphatidylinositol-3,4,5-trisphosphate-dependent Rac exchanger 1 (P-Rex1) was originally identified in neutrophils that regulates phagocyte functions. We sought to examine whether P-Rex1 plays a role in platelet activation. METHODS AND RESULTS Western blotting showed P-Rex1 expression in mouse and human platelets. Mice lacking P-Rex1 exhibited prolonged bleeding time and increased rebleeding. When challenged with low doses of the G protein-coupled receptor (GPCR) agonists U46619 and thrombin, P-Rex1-/- platelets displayed significantly reduced secretion and aggregation compared with wild-type platelets. Increasing the concentration of these agonists could overcome the defect. Platelet aggregation induced by collagen, a non-GPCR agonist, was also compromised in the absence of P-Rex1. Along with these phenotypic changes were impaired Rac1 activation; reduced ATP secretion; and decreased phosphorylation of Akt, c-Jun N-terminal kinase, and p38 mitogen-activated protein kinase in P-Rex1-/- platelets on agonist stimulation. CONCLUSION These results demonstrate for the first time the presence of P-Rex1 in platelets as well as its role in platelet secretion and aggregation induced by low-dose agonists for GPCR and by collagen.
Collapse
Affiliation(s)
- Feng Qian
- Department of Pharmacology, University of Illinois College of Medicine, 835 S Wolcott Ave, Chicago, IL 60612, USA
| | | | | | | | | | | | | |
Collapse
|
24
|
Stefanini L, Boulaftali Y, Ouellette TD, Holinstat M, Désiré L, Leblond B, Andre P, Conley PB, Bergmeier W. Rap1-Rac1 circuits potentiate platelet activation. Arterioscler Thromb Vasc Biol 2011; 32:434-41. [PMID: 22075250 DOI: 10.1161/atvbaha.111.239194] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE The goal of this study was to investigate the potential crosstalk between Rap1 and Rac1, 2 small GTPases central to platelet activation, particularly downstream of the collagen receptor GPVI. METHODS AND RESULTS We compared the activation response of platelets with impaired Rap signaling (double knock-out; deficient in both the guanine nucleotide exchange factor, CalDAG-GEFI, and the Gi-coupled receptor for ADP, P2Y12), to that of wild-type platelets treated with a small-molecule Rac inhibitor, EHT 1864 (wild-type /EHT). We found that Rac1 is sequentially activated downstream of Rap1 on stimulation via GPVI. In return, Rac1 provides important feedback for both CalDAG-GEFI- and P2Y12-dependent activation of Rap1. When analyzing platelet responses controlled by Rac1, we observed (1) impaired lamellipodia formation, clot retraction, and granule release in both double knock-out and EHT 1864-treated wild-type platelets; and (2) reduced calcium store release in EHT 1864-treated wild-type but not double knock-out platelets. Consistent with the latter finding, we identified 2 pools of Rac1, one activated immediately downstream of GPVI and 1 activated downstream of Rap1. CONCLUSIONS We demonstrate important crosstalk between Rap1 and Rac1 downstream of GPVI. Whereas Rap1 signaling directly controls sustained Rac1 activation, Rac1 affects CalDAG-GEFI- and P2Y12-dependent Rap1 activation via its role in calcium mobilization and granule/ADP release, respectively.
Collapse
Affiliation(s)
- Lucia Stefanini
- Dept. of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, 98 Manning Drive, 306a Mary Ellen Jones Building, Chapel Hill, NC 27599, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Rac1-mediated signaling plays a central role in secretion-dependent platelet aggregation in human blood stimulated by atherosclerotic plaque. J Transl Med 2010; 8:128. [PMID: 21134286 PMCID: PMC3018435 DOI: 10.1186/1479-5876-8-128] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2010] [Accepted: 12/06/2010] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Platelet activation requires rapid remodeling of the actin cytoskeleton which is regulated by small GTP-binding proteins. By using the Rac1-specific inhibitor NSC23766, we have recently found that Rac1 is a central component of a signaling pathway that regulates dephosphorylation and activation of the actin-dynamising protein cofilin, dense and α-granule secretion, and subsequent aggregation of thrombin-stimulated washed platelets. OBJECTIVES To study whether NSC23766 inhibits stimulus-induced platelet secretion and aggregation in blood. METHODS Human platelet aggregation and ATP-secretion were measured in hirudin-anticoagulated blood and platelet-rich plasma (PRP) by using multiple electrode aggregometry and the Lumi-aggregometer. Platelet P-selectin expression was quantified by flow cytometry. RESULTS NSC23766 (300 μM) inhibited TRAP-, collagen-, atherosclerotic plaque-, and ADP-induced platelet aggregation in blood by 95.1%, 93.4%, 92.6%, and 70%, respectively. The IC50 values for inhibition of TRAP-, collagen-, and atherosclerotic plaque-, were 50 ± 18 μM, 64 ± 35 μM, and 50 ± 30 μM NSC23766 (mean ± SD, n = 3-7), respectively. In blood containing RGDS to block integrin αIIbβ3-mediated platelet aggregation, NSC23766 (300 μM) completely inhibited P-selectin expression and reduced ATP-secretion after TRAP and collagen stimulation by 73% and 85%, respectively. In ADP-stimulated PRP, NSC23766 almost completely inhibited P-selectin expression, in contrast to aspirin, which was ineffective. Moreover, NSC23766 (300 μM) decreased plaque-stimulated platelet adhesion/aggregate formation under arterial flow conditions (1500s-1) by 72%. CONCLUSIONS Rac1-mediated signaling plays a central role in secretion-dependent platelet aggregation in blood stimulated by a wide array of platelet agonists including atherosclerotic plaque. By specifically inhibiting platelet secretion, the pharmacological targeting of Rac1 could be an interesting approach in the development of future antiplatelet drugs.
Collapse
|
26
|
Getz TM, Dangelmaier CA, Jin J, Daniel JL, Kunapuli SP. Differential phosphorylation of myosin light chain (Thr)18 and (Ser)19 and functional implications in platelets. J Thromb Haemost 2010; 8:2283-93. [PMID: 20670370 PMCID: PMC2965805 DOI: 10.1111/j.1538-7836.2010.04000.x] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
BACKGROUND Myosin IIA is an essential platelet contractile protein that is regulated by phosphorylation of its regulatory light chain (MLC) on residues (Thr)18 and (Ser)19 via the myosin light chain kinase (MLCK). OBJECTIVE The present study was carried out to elucidate the mechanisms regulating MLC (Ser)19 and (Thr)18 phosphorylation and the functional consequence of each phosphorylation event in platelets. RESULTS Induction of 2MeSADP-induced shape change occurs within 5s along with robust phosphorylation of MLC (Ser)19 with minimal phosphorylation of MLC (Thr)18. Selective activation of G(12/13) produces both slow shape change and comparably slow MLC (Thr)18 and (Ser)19 phosphorylation. Stimulation with agonists that trigger ATP secretion caused rapid MLC (Ser)19 phosphorylation while MLC (Thr)18 phosphorylation was coincident with secretion. Platelets treated with p160(ROCK) inhibitor Y-27632 exhibited a partial inhibition in secretion and had a substantial inhibition in MLC (Thr)18 phosphorylation without effecting MLC (Ser)19 phosphorylation. These data suggest that phosphorylation of MLC (Ser)19 is downstream of Gq/Ca(2+) -dependent mechanisms and sufficient for shape change, whereas MLC (Thr)18 phosphorylation is substantially downstream of G(12/13) -regulated Rho kinase pathways and necessary, probably in concert with MLC (Ser)19 phosphorylation, for full contractile activity leading to dense granule secretion. Overall, we suggest that the amplitude of the platelet contractile response is differentially regulated by a least two different signaling pathways, which lead to different phosphorylation patterns of the myosin light chain, and this mechanism results in a graded response rather than a simple on/off switch.
Collapse
Affiliation(s)
- Todd M. Getz
- Department of Physiology, Temple University School of Medicine, Philadelphia, PA, U.S.A
- Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, PA, U.S.A
| | - Carol A. Dangelmaier
- Department of Physiology, Temple University School of Medicine, Philadelphia, PA, U.S.A
| | - Jianguo Jin
- Department of Physiology, Temple University School of Medicine, Philadelphia, PA, U.S.A
| | - James L. Daniel
- Department of Pharmacology, Temple University School of Medicine, Philadelphia, PA, U.S.A
- Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, PA, U.S.A
| | - Satya P. Kunapuli
- Department of Physiology, Temple University School of Medicine, Philadelphia, PA, U.S.A
- Department of Pharmacology, Temple University School of Medicine, Philadelphia, PA, U.S.A
- Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, PA, U.S.A
| |
Collapse
|
27
|
Ting HJ, Khasawneh FT. Platelet function and Isoprostane biology. Should isoprostanes be the newest member of the orphan-ligand family? J Biomed Sci 2010; 17:24. [PMID: 20370921 PMCID: PMC2854111 DOI: 10.1186/1423-0127-17-24] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2010] [Accepted: 04/06/2010] [Indexed: 12/19/2022] Open
Abstract
While there have been many reports investigating the biological activity and signaling mechanisms of isoprostanes, their role in biology, particularly in platelets, appears to still be underestimated. Moreover, whether these lipids have their own receptors is still debated, despite multiple reports that discrete receptors for isporpstanes do exist on platelets, vascular tissues, amongst others. This paper provides a review of the important literature of isoprostanes and provides reasoning that isoprostanes should be classified as orphan ligands until their receptor(s) is/are identified.
Collapse
Affiliation(s)
- Harold J Ting
- Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, California 91766, USA
| | | |
Collapse
|
28
|
Borensztajn K, Peppelenbosch MP, Spek CA. Coagulation Factor Xa inhibits cancer cell migration via LIMK1-mediated cofilin inactivation. Thromb Res 2010; 125:e323-8. [PMID: 20347121 DOI: 10.1016/j.thromres.2010.02.018] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2009] [Revised: 02/19/2010] [Accepted: 02/26/2010] [Indexed: 01/21/2023]
Abstract
Previously, we showed that activated coagulation factor X (FXa) inhibits migration of breast, lung and colon cancer cells. We showed that the effect of FXa on migration was protease-activated receptor (PAR)-1-dependent, but the subsequent cellular signaling routes remained elusive. In the current manuscript, we show that both the Rho/ROCK and Src/FAK/paxillin pathways are required for FXa-mediated inhibition of breast cancer cell migration. FXa induced pronounced stress fiber formation that was partially inhibited by pre-treatment with specific ROCK or Src inhibitors. Downstream of Rho/ROCK and Src/FAK/paxillin, FXa induced myosin light chain phosphorylation and LIMK1 activation resulting in cofilin inactivation. Knocking-down LIMK1 expression abolished FXa-induced inhibition of cell invasion. Our results reveal that FXa-mediated sustained cofilin inactivation leads to stabilization of actin filaments incompatible with migration. Overall we confirm that, beyond its role in blood coagulation, FXa plays a key role in cell migration and we unravel a new mechanism of PAR-1-mediated inhibition of migration via Rho and Src dependent pathways.
Collapse
Affiliation(s)
- Keren Borensztajn
- Center for Experimental and Molecular Medicine, Academic Medical Center, Meibergdreef 9, NL-1105 AZ, Amsterdam, The Netherlands.
| | | | | |
Collapse
|
29
|
Offermanns S. Thrombozytäre Signaltransduktion. Hamostaseologie 2010. [DOI: 10.1007/978-3-642-01544-1_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
30
|
Holinstat M, Preininger AM, Milne SB, Hudson WJ, Brown HA, Hamm HE. Irreversible platelet activation requires protease-activated receptor 1-mediated signaling to phosphatidylinositol phosphates. Mol Pharmacol 2009; 76:301-13. [PMID: 19483102 PMCID: PMC2713123 DOI: 10.1124/mol.109.056622] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2009] [Accepted: 05/29/2009] [Indexed: 01/25/2023] Open
Abstract
Thrombin induces platelet activation through an early, reversible stage of platelet aggregation, which is followed by a later, irreversible stage of platelet aggregation. Without intervention, events leading to pathological platelet activation can result in vessel occlusion, acute coronary syndrome, and stroke. Therefore, a better understanding of events leading to platelet-mediated clot formation may provide insight into new therapeutic targets. Once activated, protease activated receptors (PARs) are essential in regulating events leading to platelet aggregation. We have determined a signaling cascade through PAR1, which involves phosphatidylinositol (PI) kinases, phosphatidylinositol bisphosphate (PIP(2)), and Rap1 activation (independent of P2Y12) in the formation of a stable platelet aggregate. The putative phosphatidylinositol-3 kinase (PI3K) inhibitor LY294002 was found to reduce basal and PAR-stimulated PIP(2) levels by mass spectrometry and to inhibit PAR1-mediated stable platelet aggregation. Rap1 activation in platelets (during time points corresponding to the late, irreversible phase of aggregation) was found to require the PI signaling pathway. Perturbation of PI3K signaling by isoform-selective inhibitors had differential effects on Rap1 activation through PAR1 and PAR4. Hence, it is possible to disrupt lipid signaling pathways involved in stable clot formation without inhibiting early clot formation, offering a new potential target for antiplatelet therapy.
Collapse
Affiliation(s)
- Michael Holinstat
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232-6600, USA
| | | | | | | | | | | |
Collapse
|
31
|
Unraveling a novel Rac1-mediated signaling pathway that regulates cofilin dephosphorylation and secretion in thrombin-stimulated platelets. Blood 2009; 114:415-24. [DOI: 10.1182/blood-2008-10-183582] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Abstract
In platelets stimulated by thrombin to secrete and aggregate, cofilin is rapidly dephosphorylated leading to its activation. Cofilin by severing existing actin filaments and stimulating F-actin polymerization on newly created barbed ends dynamizes the actin cytoskeleton. We previously found that cofilin dephosphorylation is Ca2+-dependent and occurs upstream of degranulation in stimulated platelets. We report now in thrombin-stimulated platelets that Rac1 and class II PAKs (PAK4/5/6) were rapidly (within 5 seconds) activated, whereas PAK1/2 (class I PAKs) phosphorylation was slower. The Rac1-specific inhibitor NSC23766 blocked phosphorylation of class II PAKs, but not PAK1/2. Moreover, inhibition of the Ca2+/calmodulin-dependent phosphatase calcineurin inhibited Rac1 activation and class II PAKs phosphorylation. Prevention of Rac1 activation by calcineurin inhibition or NSC23766 also blocked cofilin dephosphorylation and platelet granule secretion indicating that a calcineurin/Rac1/class II PAKs pathway regulates cofilin dephosphorylation leading to secretion. We further found that PI3-kinases were activated downstream of Rac1, but were not involved in regulating cofilin dephosphorylation and secretion in thrombin-stimulated platelets. Our study unravels a Ca2+-dependent pathway of secretion in stimulated platelets as a signaling pathway linking Rac1 activation to actin dynamics: calcineurin→Rac1→class II PAKs→cofilin activation. We further demonstrate that this pathway is separate and independent of the protein kinase C (PKC) pathway mediating secretion.
Collapse
|
32
|
Herroeder S, Reichardt P, Sassmann A, Zimmermann B, Jaeneke D, Hoeckner J, Hollmann MW, Fischer KD, Vogt S, Grosse R, Hogg N, Gunzer M, Offermanns S, Wettschureck N. Guanine Nucleotide-Binding Proteins of the G12 Family Shape Immune Functions by Controlling CD4+ T Cell Adhesiveness and Motility. Immunity 2009; 30:708-20. [DOI: 10.1016/j.immuni.2009.02.010] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2008] [Revised: 01/09/2009] [Accepted: 02/23/2009] [Indexed: 12/22/2022]
|
33
|
Jin J, Mao Y, Thomas D, Kim S, Daniel JL, Kunapuli SP. RhoA downstream of G(q) and G(12/13) pathways regulates protease-activated receptor-mediated dense granule release in platelets. Biochem Pharmacol 2008; 77:835-44. [PMID: 19073150 DOI: 10.1016/j.bcp.2008.11.017] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2008] [Revised: 11/13/2008] [Accepted: 11/14/2008] [Indexed: 11/17/2022]
Abstract
Platelet secretion is an important physiological event in hemostasis. The protease-activated receptors, PAR 1 and PAR 4, and the thromboxane receptor activate the G(12/13) pathways, in addition to the G(q) pathways. Here, we investigated the contribution of G(12/13) pathways to platelet dense granule release. 2MeSADP, which does not activate G(12/13) pathways, does not cause dense granule release in aspirin-treated platelets. However, supplementing 2MeSADP with YFLLRNP (60muM), as selective activator of G(12/13) pathways, resulted in dense granule release. Similarly, supplementing PLC activation with G(12/13) stimulation also leads to dense granule release. These results demonstrate that supplemental signaling from G(12/13) is required for G(q)-mediated dense granule release and that ADP fails to cause dense granule release because the platelet P2Y receptors, although activate PLC, do not activate G(12/13) pathways. When RhoA, downstream signaling molecule in G(12/13) pathways, is blocked, PAR-mediated dense granule release is inhibited. Furthermore, ADP activated RhoA downstream of G(q) and upstream of PLC. Finally, RhoA regulated PKCdelta T505 phosphorylation, suggesting that RhoA pathways contribute to platelet secretion through PKCdelta activation. We conclude that G(12/13) pathways, through RhoA, regulate dense granule release and fibrinogen receptor activation in platelets.
Collapse
Affiliation(s)
- Jianguo Jin
- Department of Physiology, Temple University School of Medicine, Philadelphia, PA, USA
| | | | | | | | | | | |
Collapse
|
34
|
Nürnberg A, Braüer AU, Wettschureck N, Offermanns S. Antagonistic regulation of neurite morphology through Gq/G11 and G12/G13. J Biol Chem 2008; 283:35526-31. [PMID: 18854320 DOI: 10.1074/jbc.m804972200] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The induction of neurite retraction and growth cone collapse via G-protein-coupled receptors is involved in developmental as well as regenerative processes. The role of individual G-protein-mediated signaling processes in the regulation of neurite morphology is still incompletely understood. Using primary neurons from brains lacking Galpha(q)/Galpha(11) or Galpha(12)/Galpha(13), we show here that G(12)/G(13)-mediated signaling is absolutely required for neurite retraction and growth cone collapse induced by the blood-borne factors lysophosphatidic acid and thrombin. Interestingly, the effects of lysophosphatidic acid were mediated mainly by G(13), whereas thrombin effects required G(12). Surprisingly, lack of Galpha(q)/Galpha(11) resulted in overshooting responses to both stimuli, indicating that G(q)/G(11)-mediated signaling most likely via activation of Rac antagonizes the effects of G(12)/G(13).
Collapse
Affiliation(s)
- Alexander Nürnberg
- Institute of Pharmacology, University of Heidelberg, Im Neuenheimer Feld 366, D-69120 Heidelberg, Germany
| | | | | | | |
Collapse
|
35
|
Loss of PIP5KIbeta demonstrates that PIP5KI isoform-specific PIP2 synthesis is required for IP3 formation. Proc Natl Acad Sci U S A 2008; 105:14064-9. [PMID: 18772378 DOI: 10.1073/pnas.0804139105] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The three isoforms of PIP5KI (alpha, beta, and gamma) synthesize PI4,5P(2) (PIP(2)) by phosphorylating PI4P. Therefore, it is not clear why platelets, like all eukaryotic cells, have more than one isoform. To test the hypothesis that PIP5KI isoforms have nonoverlapping functions, we generated a murine line containing a null mutation of PIP5KIbeta and analyzed the effect on platelet signaling. PIP5KIbeta-null mice had normal platelet counts. In contrast to platelets lacking PIP5KIalpha, platelets lacking PIP5KIbeta exhibited impaired aggregation accompanied by disaggregation. Although platelets lacking PIP5KIbeta had only a moderate deficiency of PIP(2) under basal conditions, they had a striking deficiency in PIP(2) synthesis and IP(3) formation after thrombin stimulation. We have also observed that platelets lacking both PIP5KIalpha and PIP5KIbeta have a complete loss of thrombin-induced IP(3) synthesis even though they still contain PIP5KIgamma, the predominant PIP5KI isoform in platelets. These results demonstrate that PIP5KIbeta, like PIP5KIalpha, contributes to the rapid synthesis of a pool of PIP(2) that is required for second-messenger formation, whereas the pool of PIP(2) synthesized by PIP5KIgamma does not contribute to this process. Additionally, we found that PIP5KIbeta-null platelets failed to form arterial thrombi properly in vivo. Together, these data demonstrate that PIP5KIbeta is required for rapid PIP(2) synthesis, second-messenger production, and stable platelet adhesion under shear in vivo. These results also demonstrate that after stimulation of a G protein-coupled receptor, IP(3) is completely derived from a rapidly synthesized discrete pool of PIP(2) synthesized by PIP5KIalpha and PIP5KIbeta.
Collapse
|
36
|
Rac1 is essential for phospholipase C-gamma2 activation in platelets. Pflugers Arch 2008; 457:1173-85. [PMID: 18704487 DOI: 10.1007/s00424-008-0573-7] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2008] [Revised: 07/17/2008] [Accepted: 08/01/2008] [Indexed: 10/21/2022]
Abstract
Platelet activation at sites of vascular injury is triggered through different signaling pathways leading to activation of phospholipase (PL) Cbeta or PLCgamma2. Active PLCs trigger Ca(2+) mobilization and entry, which is a prerequisite for adhesion, secretion, and thrombus formation. PLCbeta isoenzymes are activated downstream of G protein-coupled receptors (GPCRs), whereas PLCgamma2 is activated downstream of immunoreceptor tyrosine-based activation motif (ITAM)-coupled receptors, such as the major platelet collagen receptor glycoprotein (GP) VI or CLEC-2. The mechanisms underlying PLC regulation are not fully understood. An involvement of small GTPases of the Rho family (Rho, Rac, Cdc42) in PLC activation has been proposed but this has not been investigated in platelets. We here show that murine platelets lacking Rac1 display severely impaired GPVI- or CLEC-2-dependent activation and aggregation. This defect was associated with impaired production of inositol 1,4,5-trisphosphate (IP(3)) and intracellular calcium mobilization suggesting inappropriate activation of PLCgamma2 despite normal tyrosine phosphorylation of the enzyme. Rac1 ( -/- ) platelets displayed defective thrombus formation on collagen under flow conditions which could be fully restored by co-infusion of ADP and the TxA(2) analog U46619, indicating that impaired GPVI-, but not G-protein signaling, was responsible for the observed defect. In line with this, Rac1 ( -/- ) mice were protected in two collagen-dependent arterial thrombosis models. Together, these results demonstrate that Rac1 is essential for ITAM-dependent PLCgamma2 activation in platelets and that this is critical for thrombus formation in vivo.
Collapse
|
37
|
Wikström K, Kavanagh DJ, Reid HM, Kinsella BT. Differential regulation of RhoA-mediated signaling by the TPalpha and TPbeta isoforms of the human thromboxane A2 receptor: independent modulation of TPalpha signaling by prostacyclin and nitric oxide. Cell Signal 2008; 20:1497-512. [PMID: 18502100 PMCID: PMC2681257 DOI: 10.1016/j.cellsig.2008.04.006] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2008] [Revised: 04/02/2008] [Accepted: 04/07/2008] [Indexed: 01/16/2023]
Abstract
In humans, thromboxane (TX) A2 signals through the
TPα and TPβ isoforms of the TXA2 receptor that exhibit common
and distinct roles. For example, Gq/phospholipase (PL)Cβ signaling by TPα is
directly inhibited by the vasodilators prostacyclin and nitric oxide (NO)
whereas that signaling by TPβ is unaffected. Herein, we investigated whether TPα
and/or TPβ regulate G12/Rho activation and whether that
signaling might be differentially regulated by prostacyclin and/or NO. Both TPα
and TPβ independently regulated RhoA activation and signaling in clonal cells
over-expressing TPα or TPβ and in primary human aortic smooth muscle cells (1°
AoSMCs). While RhoA-signaling by TPα was directly impaired by prostacyclin and
NO through protein kinase (PK)A- and PKG-dependent phosphorylation,
respectively, signaling by TPβ was not directly affected by either agent.
Collectively, while TPα and TPβ contribute to RhoA activation, our findings
support the hypothesis that TPα is involved in the dynamic regulation of
haemostasis and vascular tone, such as in response to prostacyclin and NO.
Conversely, the role of TPβ in such processes remains unsolved. Data herein
provide essential new insights into the physiologic roles of TPα and TPβ and,
through studies in AoSMCs, reveal an additional mode of regulation of VSM
contractile responses by TXA2.
Collapse
Affiliation(s)
- Katarina Wikström
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| | | | | | | |
Collapse
|
38
|
Calaminus SDJ, Auger JM, McCarty OJT, Wakelam MJO, Machesky LM, Watson SP. MyosinIIa contractility is required for maintenance of platelet structure during spreading on collagen and contributes to thrombus stability. J Thromb Haemost 2007; 5:2136-45. [PMID: 17645784 DOI: 10.1111/j.1538-7836.2007.02696.x] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND MyosinIIs are adenosine triphosphate-driven molecular motors that form part of a cell's contractile machinery. They are activated by phosphorylation of their light chains, by either activation of myosin light chain (MLC) kinase or inhibition of MLC phosphatase via Rho kinase (ROCK). MyosinIIa phosphorylation underlies platelet rounding and stress fiber formation. OBJECTIVE To identify the functional significance of myosinIIa in platelet spreading and thrombus formation on collagen using inhibitors of ROCK (Y27632) and myosinII (blebbistatin). RESULTS Stress fiber formation on collagen is inhibited by both Y27632 and blebbistatin. A substantial proportion of spread platelets generate internal holes or splits on collagen, presumably because of a reduction in contractile strength. Platelet integrity, however, is maintained. In an in vitro model, thrombus embolization on collagen is increased in the presence of Y27632 and blebbistatin at intermediate shear, leading to a reduction in platelet aggregate growth. Moreover, Y27632 causes a marked reduction in thrombus formation in an in vivo laser-injury model. CONCLUSIONS MyosinIIa contractility is required for maintenance of platelet structure during spreading on collagen and contributes to thrombus stability.
Collapse
Affiliation(s)
- S D J Calaminus
- Centre for Cardiovascular Sciences, Institute of Biomedical Research, The Medical School, University of Birmingham, Birmingham, UK.
| | | | | | | | | | | |
Collapse
|
39
|
Akbar H, Kim J, Funk K, Cancelas JA, Shang X, Chen L, Johnson JF, Williams DA, Zheng Y. Genetic and pharmacologic evidence that Rac1 GTPase is involved in regulation of platelet secretion and aggregation. J Thromb Haemost 2007; 5:1747-55. [PMID: 17663742 DOI: 10.1111/j.1538-7836.2007.02646.x] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND Rac1 GTPase, a member of the Ras-related Rho GTPase family, is the major Rac isoform present in platelets and has been shown to be involved in cell actin cytoskeleton reorganization and adhesion. Agonists that induce platelet secretion and aggregation also activate Rac1 GTPase, raising the possibility that Rac1 GTPase may be involved in regulation of platelet function. OBJECTIVES To rigorously define the role of Rac1 in platelet regulation. METHODS We have used a dual approach of gene targeting in mice and pharmacologic inhibition of Rac1 by NSC23766, a rationally designed specific small molecule inhibitor, to study the role of Rac1 in platelet function. RESULTS Platelets from mice as well as human platelets treated with NSC23766 exhibited a significant decrease in: (i) active Rac1 species and phosphorylation of the Rac effector, p21-activated kinase; (ii) expression of P-selectin and secretion of adenosine triphosphate induced by thrombin or U46619; and (iii) aggregation induced by adenosine 5'-diphosphate, collagen, thrombin and U46619, a stable analog of thromboxane A(2). NSC23766 did not alter the cAMP or cGMP levels in platelets. Consistent with the requirement of Rac1 for normal platelet function, the bleeding times in Rac1(-/-) mice or mice given NSC23766 were significantly prolonged. CONCLUSIONS Our data show that deficiency or inhibition of Rac1 GTPase blocks platelet secretion. The inhibition of secretion, at least in part, is responsible for diminished platelet aggregation and prolonged bleeding times observed in Rac1 knockout or Rac1 inhibitor-treated mice.
Collapse
Affiliation(s)
- H Akbar
- Division of Experimental Hematology, Cincinnati Children's Research Foundation, University of Cincinnati, Cincinnati, OH, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Pandey D, Goyal P, Siess W. Lysophosphatidic acid stimulation of platelets rapidly induces Ca2+-dependent dephosphorylation of cofilin that is independent of dense granule secretion and aggregation. Blood Cells Mol Dis 2007; 38:269-79. [PMID: 17321765 DOI: 10.1016/j.bcmd.2007.01.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2006] [Revised: 01/16/2007] [Accepted: 01/17/2007] [Indexed: 10/23/2022]
Abstract
Cofilin is an actin dynamizing protein and inactivated after Ser3 phosphorylation by LIM-kinases (LIMKs). We studied whether in platelets stimulated by lysophosphatidic acid (LPA), Rho-kinase or p21-activated kinase (PAK) mediates LIMK-1 activation leading to subsequent phosphorylation and inactivation of cofilin and the increase of F-actin. During LPA (0.1 microM)-induced shape change, a rapid Rho-kinase activation and a slower activation of PAK were observed. Rho-kinase activation led to rapid LIMK-1 (Thr508) phosphorylation. Despite of LIMK-1 activation, cofilin net phosphorylation was not increased. Cofilin rapidly associated with F-actin and preceded the F-actin increase. Pretreatment with the Rho-kinase inhibitor Y-27632 inhibited LIMK-1 phosphorylation, unmasked cofilin dephosphorylation and inhibited the reversible F-actin increase during shape change. In the presence of fibrinogen, LPA (10 microM) induced ATP-secretion from dense granules and aggregation, and cofilin was rapidly dephosphorylated and then rephosphorylated in a Rho-kinase/LIMK-1-dependent manner. In the absence of fibrinogen, cofilin de- and rephosphorylation after LPA (10 microM) was unchanged, but secretion and aggregation were absent. Cofilin dephosphorylation was completely blocked by BAPTA-AM indicating that it was mediated by an increase of cytosolic Ca(2+). We conclude that in LPA-stimulated platelets, Rho-kinase-dependent LIMK-1 activation mediates the F-actin increase during shape change without enhancing cofilin net phosphorylation. However, a rapid dephosphorylation of cofilin occurs during secretion and aggregation, which is Ca(2+)-dependent, upstream of secretion and aggregation and might regulate these platelet responses.
Collapse
Affiliation(s)
- Dharmendra Pandey
- Institute for Prevention of Cardiovascular Diseases, Klinikum Innenstadt, University of Munich, Pettenkoferstrasse 9, D-80336 Munich, Germany
| | | | | |
Collapse
|
41
|
Oude Weernink PA, López de Jesús M, Schmidt M. Phospholipase D signaling: orchestration by PIP2 and small GTPases. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2007; 374:399-411. [PMID: 17245604 PMCID: PMC2020506 DOI: 10.1007/s00210-007-0131-4] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2006] [Accepted: 12/20/2006] [Indexed: 11/12/2022]
Abstract
Hydrolysis of phosphatidylcholine by phospholipase D (PLD) leads to the generation of the versatile lipid second messenger, phosphatidic acid (PA), which is involved in fundamental cellular processes, including membrane trafficking, actin cytoskeleton remodeling, cell proliferation and cell survival. PLD activity can be dramatically stimulated by a large number of cell surface receptors and is elaborately regulated by intracellular factors, including protein kinase C isoforms, small GTPases of the ARF, Rho and Ras families and, particularly, by the phosphoinositide, phosphatidylinositol 4,5-bisphosphate (PIP(2)). PIP(2) is well known as substrate for the generation of second messengers by phospholipase C, but is now also understood to recruit and/or activate a variety of actin regulatory proteins, ion channels and other signaling proteins, including PLD, by direct interaction. The synthesis of PIP(2) by phosphoinositide 5-kinase (PIP5K) isoforms is tightly regulated by small GTPases and, interestingly, by PA as well, and the concerted formation of PIP(2) and PA has been shown to mediate receptor-regulated cellular events. This review highlights the regulation of PLD by membrane receptors, and describes how the close encounter of PLD and PIP5K isoforms with small GTPases permits the execution of specific cellular functions.
Collapse
Affiliation(s)
| | | | - Martina Schmidt
- />Department of Molecular Pharmacology, University of Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
| |
Collapse
|
42
|
Moraes LA, Swales KE, Wray JA, Damazo A, Gibbins JM, Warner TD, Bishop-Bailey D. Nongenomic signaling of the retinoid X receptor through binding and inhibiting Gq in human platelets. Blood 2007; 109:3741-4. [PMID: 17213293 PMCID: PMC2612640 DOI: 10.1182/blood-2006-05-022566] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Retinoid X receptors (RXRs) are important transcriptional nuclear hormone receptors, acting as either homodimers or the binding partner for at least one fourth of all the known human nuclear receptors. Functional nongenomic effects of nuclear receptors are poorly understood; however, recently peroxisome proliferator-activated receptor (PPAR) γ, PPARβ, and the glucocorticoid receptor have all been found active in human platelets. Human platelets express RXRα and RXRβ. RXR ligands inhibit platelet aggregation and TXA2 release to ADP and the TXA2 receptors, but only weakly to collagen. ADP and TXA2 both signal via the G protein, Gq. RXR rapidly binds Gq but not Gi/z/o/t/gust in a ligand-dependent manner and inhibits Gq-induced Rac activation and intracellular calcium release. We propose that RXR ligands may have beneficial clinical actions through inhibition of platelet activation. Furthermore, our results demonstrate a novel nongenomic mode for nuclear receptor action and a functional cross-talk between G-protein and nuclear receptor signaling families.
Collapse
Affiliation(s)
- Leonardo A Moraes
- Cardiac, Vascular and Inflammation Research, William Harvey Research Institute, Barts and the London, Queen Mary University of London, Charterhouse Square, London, UK
| | | | | | | | | | | | | |
Collapse
|
43
|
Abstract
Because of their ability to become rapidly activated at places of vascular injury, platelets are important players in primary hemostasis as well as in arterial thrombosis. In addition, they are also involved in chronic pathological processes including the atherosclerotic remodeling of the vascular system. Although primary adhesion of platelets to the vessel wall is largely independent of G protein-mediated signaling, the subsequent recruitment of additional platelets into a growing platelet thrombus requires mediators such as ADP, thromboxane A(2), or thrombin, which act through G protein-coupled receptors. Platelet activation via G protein-coupled receptors involves 3 major G protein-mediated signaling pathways that are initiated by the activation of the G proteins G(q), G(13), and G(i). This review summarizes recent progress in understanding the mechanisms underlying platelet activation and thrombus extension via G protein-mediated signaling pathways.
Collapse
Affiliation(s)
- Stefan Offermanns
- Institute of Pharmacology, University of Heidelberg, Im Neuenheimer Feld 366, 69120 Heidelberg, Germany.
| |
Collapse
|
44
|
Minuz P, Fumagalli L, Gaino S, Tommasoli R, Degan M, Cavallini C, Lecchi A, Cattaneo M, Lechi Santonastaso C, Berton G. Rapid stimulation of tyrosine phosphorylation signals downstream of G-protein-coupled receptors for thromboxane A2 in human platelets. Biochem J 2006; 400:127-34. [PMID: 16859489 PMCID: PMC1635449 DOI: 10.1042/bj20061015] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Signals ensuing from trimeric G-protein-coupled receptors synergize to induce platelet activation. At low doses, the thromboxane A2 analogue U46619 does not activate integrin alphaIIbbeta3 or trigger platelet aggregation, but it induces shape changes. In the present study, we addressed whether low doses of U46619 trigger tyrosine phosphorylation independently of integrin alphaIIbbeta3 activation and ADP secretion, and synergize with adrenaline (epinephrine) to induce aggregation in acetylsalicylic acid (aspirin)-treated platelets. Low doses of U46619 triggered tyrosine phosphorylation of different proteins, including FAK (focal adhesion kinase), Src and Syk, independently of signals ensuing from integrin alphaIIbbeta3 or ADP receptors engaged by secreted ADP. The G(12/13)-mediated Rho/Rho-kinase pathway was also increased by low doses of U46619; however, this pathway was not upstream of tyrosine phosphorylation, because this occurred in the presence of the Rho-kinase inhibitor Y-27632. Although low doses of U46619 or adrenaline alone were unable to trigger platelet aggregation and integrin alphaIIbbeta3 activation, the combination of the two stimuli effectively induced these responses. PP2, a tyrosine kinase inhibitor, and Y-27632 inhibited platelet activation induced by low doses of U46619 plus adrenaline and, when used in combination, totally suppressed this platelet response. In addition, the two inhibitors selectively blocked tyrosine kinases and the Rho/Rho-kinase pathway respectively. These findings suggest that both tyrosine phosphorylation and the Rho/Rho-kinase pathway are required to activate platelet aggregation via G(12/13) plus G(z) signalling.
Collapse
Affiliation(s)
- Pietro Minuz
- *Department of Biomedical and Surgical Sciences, University of Verona, Verona, Italy
| | - Laura Fumagalli
- †Department of Pathology, Section of General Pathology, University of Verona, Verona, Italy
| | - Stefania Gaino
- ‡Department of Morphological and Biomedical Sciences, University of Verona, Verona, Italy
| | - Rosa M. Tommasoli
- ‡Department of Morphological and Biomedical Sciences, University of Verona, Verona, Italy
| | - Maurizio Degan
- ‡Department of Morphological and Biomedical Sciences, University of Verona, Verona, Italy
| | - Chiara Cavallini
- ‡Department of Morphological and Biomedical Sciences, University of Verona, Verona, Italy
| | - Anna Lecchi
- §Angelo Bianchi Bonomi Haemophila and Thrombosis Centre, IRCCS Maggiore Hospital, University of Milan, Milan, Italy
| | - Marco Cattaneo
- ∥Unit of Haematology and Thrombosis, San Paolo Hospital, DMCO-University of Milan, Milan, Italy
| | | | - Giorgio Berton
- †Department of Pathology, Section of General Pathology, University of Verona, Verona, Italy
- To whom correspondence should be addressed (email )
| |
Collapse
|
45
|
Santarius M, Lee C, Anderson R. Supervised membrane swimming: small G-protein lifeguards regulate PIPK signalling and monitor intracellular PtdIns(4,5)P2 pools. Biochem J 2006; 398:1-13. [PMID: 16856876 PMCID: PMC1525017 DOI: 10.1042/bj20060565] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Regulation of PIPK (phosphatidylinositol phosphate kinase) and PtdIns(4,5)P2 signalling by small G-proteins and their effectors is key to many biological functions. Through selective recruitment and activation of different PIPK isoforms, small G-proteins such as Rho, Rac and Cdc42 modulate actin dynamics and cytoskeleton-dependent cellular events in response to extracellular signalling. These activities affect a number of processes, including endocytosis, bacterial penetration into host cells and cytolytic granule-mediated targeted cell killing. Small G-proteins and their modulators are also regulated by phosphoinositides through translocation and conformational changes. Arf family small G-proteins act at multiple sites as regulators of membrane trafficking and actin cytoskeletal remodelling, and regulate a feedback loop comprising phospholipase D, phosphatidic acid, PIPKs and PtdIns(4,5)P2, contributing to enhancement of PtdIns(4,5)P2-mediated cellular events and receptor signalling. Na+, Kir (inwardly rectifying K+), Ca2+ and TRP (transient receptor potential) ion channels are regulated by small G-proteins and membrane pools of PtdIns(4,5)P2. Yeast phosphatidylinositol 4-phosphate 5-kinases Mss4 and Its3 are involved in resistance against disturbance of sphingolipid biosynthesis and maintenance of cell integrity through the synthesis of PtdIns(4,5)P2 and downstream signalling through the Rom2/Rho2 and Rgf1/Rho pathways. Here, we review models for regulated intracellular targeting of PIPKs by small G-proteins and other modulators in response to extracellular signalling. We also describe the spatial and temporal cross-regulation of PIPKs and small G-proteins that is critical for a number of cellular functions.
Collapse
Affiliation(s)
- Megan Santarius
- *Program in Molecular and Cellular Pharmacology, University of Wisconsin-Madison, 1300 University Avenue, Madison, WI 53706, U.S.A
| | - Chang Ho Lee
- †Department of Pharmacology, College of Medicine, Hanyang University, 17 Hengdang-dong, Seongdong-ku, Seoul, 133-791, South Korea
- To whom correspondence should be addressed (email )
| | - Richard A. Anderson
- *Program in Molecular and Cellular Pharmacology, University of Wisconsin-Madison, 1300 University Avenue, Madison, WI 53706, U.S.A
- ‡Department of Pharmacology, University of Wisconsin Medical School, 1300 University Avenue, Madison, WI 53706, U.S.A
| |
Collapse
|
46
|
Ling K, Schill NJ, Wagoner MP, Sun Y, Anderson RA. Movin' on up: the role of PtdIns(4,5)P2 in cell migration. Trends Cell Biol 2006; 16:276-84. [PMID: 16616849 DOI: 10.1016/j.tcb.2006.03.007] [Citation(s) in RCA: 119] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2005] [Revised: 02/17/2006] [Accepted: 03/23/2006] [Indexed: 11/28/2022]
Abstract
Cell migration requires the coordination of many biochemical events, including cell-matrix contact turnover and cytoskeletal restructuring. Recent advances further implicate phosphatidylinositol(4,5)-bisphosphate [PtdIns(4,5)P(2)] in the control of these events. Many proteins that are crucial to the assembly of the migration machinery are regulated by PtdIns(4,5)P(2). Coordinated synthesis of PtdIns(4,5)P(2) at these sites is dependent on the precise targeting of the type I phosphatidylinositol phosphate kinases (PIPKs). Two PIPKI isoforms target to, and generate, PtdIns(4,5)P(2) at membrane ruffles and focal adhesions during cell migration. Here, we discuss our current understanding of PtdIns(4,5)P(2) in the regulation of cell responses to migratory stimuli and how the migrating cell controls PtdIns(4,5)P(2) availability.
Collapse
Affiliation(s)
- Kun Ling
- Program in Molecular and Cellular Pharmacology, University of Wisconsin-Madison, Department of Pharmacology, University of Wisconsin Medical School, 1300 University Ave, Madison, WI 53706, USA
| | | | | | | | | |
Collapse
|
47
|
McCarty OJT, Larson MK, Auger JM, Kalia N, Atkinson BT, Pearce AC, Ruf S, Henderson RB, Tybulewicz VLJ, Machesky LM, Watson SP. Rac1 is essential for platelet lamellipodia formation and aggregate stability under flow. J Biol Chem 2005; 280:39474-84. [PMID: 16195235 PMCID: PMC1395485 DOI: 10.1074/jbc.m504672200] [Citation(s) in RCA: 181] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The role of Rac family proteins in platelet spreading on matrix proteins under static and flow conditions has been investigated by using Rac-deficient platelets. Murine platelets form filopodia and undergo limited spreading on fibrinogen independent of Rac1 and Rac2. In the presence of thrombin, marked lamellipodia formation is observed on fibrinogen, which is abrogated in the absence of Rac1. However, Rac1 is not required for thrombin-induced aggregation or elevation of F-actin levels. Formation of lamellipodia on collagen and laminin is also Rac1-dependent. Analysis of platelet adhesion dynamics on collagen under flow conditions in vitro revealed that Rac1 is required for platelet aggregate stability at arterial rates of shear, as evidenced by a dramatic increase in platelet embolization. Furthermore, studies employing intravital microscopy demonstrated that Rac1 plays a critical role in the development of stable thrombi at sites of vascular injury in vivo. Thus, our data demonstrated that Rac1 is essential for lamellipodia formation in platelets and indicated that Rac1 is required for aggregate integrity leading to thrombus formation under physiologically relevant levels of shear both in vitro and in vivo.
Collapse
Affiliation(s)
- Owen J T McCarty
- Centre for Cardiovascular Sciences, the Institute of Biomedical Research, School of Biosciences, University of Birmingham, United Kingdom.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Pandey D, Goyal P, Bamburg JR, Siess W. Regulation of LIM-kinase 1 and cofilin in thrombin-stimulated platelets. Blood 2005; 107:575-83. [PMID: 16219803 PMCID: PMC1895613 DOI: 10.1182/blood-2004-11-4377] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Cofilin is a regulator of actin filament dynamics. We studied whether during platelet activation Rho kinase stimulates LIM kinase (LIMK) leading to subsequent phosphorylation and inactivation of cofilin. Platelet shape change and aggregation/secretion were induced by low and high concentrations of thrombin, respectively. We found that during these platelet responses Rho kinase activation was responsible for mediating rapid Thr508 phosphorylation and activation of LIMK-1 and for the F-actin increase during shape change and, in part, during secretion. Surprisingly, during shape change cofilin phosphorylation was unaltered, and during aggregation/secretion cofilin was first rapidly dephosphorylated by an okadaic acid-insensitive phosphatase and then slowly rephosphorylated by LIMK-1. LIMK-1 phosphorylation and cofilin dephosphorylation and rephosphorylation during aggregation were independent of integrin alpha(IIb)beta(3) engagement. Cofilin phosphorylation did not regulate cofilin association with F-actin and was unrelated to the F-actin increase in thrombin-activated platelets. Our study identifies LIMK-1 as being activated by Rho kinase in thrombin-stimulated platelets. Two counteracting pathways, a cofilin phosphatase and LIMK-1, are activated during platelet aggregation/secretion regulating cofilin phosphorylation sequentially and independently of integrin alpha(IIb)beta(3) engagement. Rho kinase-mediated F-actin increase during platelet shape change and secretion involves a mechanism other than LIMK-1-mediated cofilin phosphorylation, raising the possibility of another LIMK substrate regulating platelet actin assembly.
Collapse
Affiliation(s)
- Dharmendra Pandey
- Institute for Prevention of Cardiovascular Diseases, University of Munich, Germany
| | | | | | | |
Collapse
|
49
|
Soulet C, Hechler B, Gratacap MP, Plantavid M, Offermanns S, Gachet C, Payrastre B. A differential role of the platelet ADP receptors P2Y1 and P2Y12 in Rac activation. J Thromb Haemost 2005; 3:2296-306. [PMID: 16194206 DOI: 10.1111/j.1538-7836.2005.01588.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The dynamics of the actin cytoskeleton, largely controlled by the Rho family of small GTPases (Rho, Rac and Cdc42), is critical for the regulation of platelet responses such as shape change, adhesion, spreading and aggregation. Here, we investigated the role of adenosine diphosphate (ADP), a major co-activator of platelets, on the activation of Rac. ADP rapidly activated Rac in a dose-dependent manner and independently of GPIIb/IIIa and phosphoinositide 3-kinase. ADP alone, used as a primary agonist, activated Rac and its effector PAK via its P2Y1 receptor, through a G(q)-dependent pathway and independently of P2Y12. The P2Y12 receptor appeared unable to activate the GTPase per se as also observed for the adenosine triphosphate receptor P2X1. Conversely, secreted ADP strongly potentiated Rac activation induced by FcgammaRIIa clustering or TRAP via its P2Y12 receptor, the target of antithrombotic thienopyridines. Stimulation of the alpha(2A)-adrenergic receptor/G(z) pathway by epinephrine was able to replace the P2Y12/G(i)-mediated pathway to amplify Rac activation by FcgammaRIIa or by the thrombin receptor PAR-1. This co-activation appeared necessary to reach a full stimulation of Rac as well as PAK activation and actin polymerization and was blocked by a G-protein betagamma subunits scavenger peptide.
Collapse
Affiliation(s)
- C Soulet
- Inserm U563, Département d'Oncogenèse et signalisation dans les cellules hématopoïétiques, Hôpital Purpan, Toulouse, France
| | | | | | | | | | | | | |
Collapse
|
50
|
Falet H, Chang G, Brohard-Bohn B, Rendu F, Hartwig JH. Integrin αIIbβ3signals lead cofilin to accelerate platelet actin dynamics. Am J Physiol Cell Physiol 2005; 289:C819-25. [PMID: 15901596 DOI: 10.1152/ajpcell.00587.2004] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Cofilin, in its Ser3 dephosphorylated form, accelerates actin filament turnover in cells. We report here the role of cofilin in platelet actin assembly. Cofilin is primarily phosphorylated in the resting platelet as evidenced by a specific antibody directed against its Ser3 phosphorylated form. After stimulation with thrombin under nonstirring conditions, cofilin is reversibly dephosphorylated and transiently incorporates into the actin cytoskeleton. Its dephosphorylation is maximal 1–2 min after platelet stimulation, shortly after the peak of actin assembly occurs. Cofilin rephosphorylation begins 2 min after activation and exceeds resting levels by 5–10 min. Cofilin is dephosphorylated with identical kinetics but fails to become rephosphorylated when platelets are stimulated under stirring conditions. Cofilin is normally rephosphorylated when platelets are stimulated in the presence of Arg-Gly-Asp-Ser (RGDS) peptide or wortmannin to block αIIbβ3cross-linking and signaling or in platelets isolated from a patient with Glanzmann thrombasthenia, which express only 2–3% of normal αIIbβ3levels. Furthermore, actin assembly and Arp2/3 complex incorporation in the platelet actin cytoskeleton are decreased when αIIbβ3is engaged. Our results suggest that cofilin is essential for actin dynamics mediated by outside-in signals in activated platelets.
Collapse
Affiliation(s)
- Hervé Falet
- Division of Hematology, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, One Blackfan Circle, Karp 6, Boston, Massachusetts 02115, USA.
| | | | | | | | | |
Collapse
|