1
|
Striesow J, Wesche J, McKitterick N, Busch LM, von Woedtke T, Greinacher A, Bekeschus S, Wende K. Gas plasma-induced platelet activation corresponds to reactive species profiles and lipid oxidation. Free Radic Biol Med 2023; 207:212-225. [PMID: 37490986 DOI: 10.1016/j.freeradbiomed.2023.07.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 07/17/2023] [Accepted: 07/19/2023] [Indexed: 07/27/2023]
Abstract
Surgical-induced hemostasis is a critical step in the closure of incisions, which is frequently achieved via electrocauterization and subsequent tissue necrotization. The latter is associated with postoperative complications. Recent in vivo work suggested reactive species-producing gas plasma technology as a pro-homeostatic agent acting via platelet activation. However, it remained elusive how platelet activation is linked to lipid and protein oxidation and the reactive species compositions. A direct relation between the reactive species composition and platelet activation was revealed by assessing the production of several reactive species and by using antioxidants. In addition, platelet lipidome and proteome analysis identified significantly regulated key lipids in the platelet activation pathway, such as diacylglycerols and phosphatidylinositol as well as oxylipins like thromboxanes. Lipid oxidation products mainly derived from phosphatidylethanolamine and phosphatidylserine species were observed at modest levels. In addition, oxidative post-translational modifications were identified on key proteins of the hemostasis machinery. This study provides new insights into oxidation-induced platelet activation in general and suggests a potential role of those processes in gas plasma-mediated hemostasis in particular.
Collapse
Affiliation(s)
- Johanna Striesow
- ZIK plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Felix-Hausdorff-Str. 2, 17489, Greifswald, Germany
| | - Jan Wesche
- Institute of Transfusion Medicine, Greifswald University Medical Center, Sauerbruchstr., 17475, Greifswald, Germany
| | - Nicholas McKitterick
- ZIK plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Felix-Hausdorff-Str. 2, 17489, Greifswald, Germany
| | - Larissa M Busch
- Interfaculty Institute for Genetics and Functional Genomics, Greifswald University, Felix-Hausdorff-Str. 8, 17475, Greifswald, Germany
| | - Thomas von Woedtke
- ZIK plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Felix-Hausdorff-Str. 2, 17489, Greifswald, Germany; Institute for Hygiene and Environmental Medicine, Greifswald University Medical Center, Sauerbruchstr., 17475, Greifswald, Germany
| | - Andreas Greinacher
- Institute of Transfusion Medicine, Greifswald University Medical Center, Sauerbruchstr., 17475, Greifswald, Germany
| | - Sander Bekeschus
- ZIK plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Felix-Hausdorff-Str. 2, 17489, Greifswald, Germany; Clinic and Policlinic for Dermatology and Venerology, Rostock University Medical Center, Strempelstr. 13, 18057, Rostock, Germany.
| | - Kristian Wende
- ZIK plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Felix-Hausdorff-Str. 2, 17489, Greifswald, Germany.
| |
Collapse
|
2
|
Lin S, Liu X, Sun A, Liang H, Li Z, Ye S, Ma H, Fan W, Shen C, Jin M, He Q. Qilong capsule alleviates ponatinib-induced ischemic stroke in a zebrafish model by regulating coagulation, inflammation and apoptosis. JOURNAL OF ETHNOPHARMACOLOGY 2023; 314:116397. [PMID: 37086871 DOI: 10.1016/j.jep.2023.116397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 02/23/2023] [Accepted: 03/14/2023] [Indexed: 05/03/2023]
Abstract
ETHOPHARMACOLOGICAL RELEVANCE Qilong capsule (QLC) is a compound traditional Chinese medicine commonly used to treat ischemic stroke (IS). QLC is made of eight kinds of medicinal materials. It has two kinds of monarch medicine and six kinds of minister medicine. However, the pharmacodynamic mechanism of QLC is still unknown. AIM OF THE STUDY The aim of this paper was to evaluate the pharmacology mechanism of QLC against ischemic stroke through coagulation, inflammation and apoptosis. MATERIALS AND METHODS We used an existing zebrafish model to explore the protective mechanism of QLC on IS. We treated normally-developing zebrafish larvae with QLC and ponatinib 2 days post fertilization (dpf), and used the area of cerebral vascular thrombosis, red blood cell staining intensity, and brain cell apoptosis to quantitate QLC efficacy against IS. Evaluation of brain inflammation in zebrafish by observing macrophage aggregation and migration. In addition, we also explored the effect of QLC on zebrafish angiogenesis. Quantitative polymerase chain reaction (qPCR) was used to detect changes in the expression of genes involved in coagulation, inflammation, vascular endothelium, and apoptosis. RESULTS We found that QLC reduced the area affected by ponatinib-induced cerebral vascular embolism, erythrocyte staining intensity, and the number of apoptotic brain cells. For inflammation, QLC can improve the aggregation and migration of macrophages. QLC can significantly promote the formation of blood vessels in zebrafish. qPCR showed that QLC inhibited the expression of genes related to coagulation, inflammation, and apoptosis. CONCLUSION Qilong capsule had a significant protective efficacy in ponatinib-induced IS.
Collapse
Affiliation(s)
- Shenghua Lin
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan, China
| | - Xin Liu
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan, China
| | - Aonan Sun
- School of Food Science and Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, China
| | - Huiliang Liang
- Shandong Jining Huaneng Pharmaceutical Factory, Jining, China
| | - Zhen Li
- Shandong Jining Huaneng Pharmaceutical Factory, Jining, China
| | - Suyan Ye
- Shandong Jining Huaneng Pharmaceutical Factory, Jining, China
| | - Honglin Ma
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan, China
| | - Wei Fan
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan, China
| | - Chuanlin Shen
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan, China
| | - Meng Jin
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan, China
| | - Qiuxia He
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan, China; Science and Technology Service Platform, Qilu University of Technology (Shandong Academy of Sciences), China.
| |
Collapse
|
3
|
Reversible Platelet Integrin αIIbβ3 Activation and Thrombus Instability. Int J Mol Sci 2022; 23:ijms232012512. [PMID: 36293367 PMCID: PMC9604507 DOI: 10.3390/ijms232012512] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 10/10/2022] [Accepted: 10/14/2022] [Indexed: 11/28/2022] Open
Abstract
Integrin αIIbβ3 activation is essential for platelet aggregation and, accordingly, for hemostasis and arterial thrombosis. The αIIbβ3 integrin is highly expressed on platelets and requires an activation step for binding to fibrinogen, fibrin or von Willebrand factor (VWF). A current model assumes that the process of integrin activation relies on actomyosin force-dependent molecular changes from a bent-closed and extended-closed to an extended-open conformation. In this paper we review the pathways that point to a functional reversibility of platelet αIIbβ3 activation and transient aggregation. Furthermore, we refer to mouse models indicating that genetic defects that lead to reversible platelet aggregation can also cause instable thrombus formation. We discuss the platelet agonists and signaling pathways that lead to a transient binding of ligands to integrin αIIbβ3. Our analysis points to the (autocrine) ADP P2Y1 and P2Y12 receptor signaling via phosphoinositide 3-kinases and Akt as principal pathways linked to reversible integrin activation. Downstream signaling events by protein kinase C, CalDAG-GEFI and Rap1b have not been linked to transient integrin activation. Insight into the functional reversibility of integrin activation pathways will help to better understand the effects of antiplatelet agents.
Collapse
|
4
|
Puerto-Camacho P, Díaz-Martín J, Olmedo-Pelayo J, Bolado-Carrancio A, Salguero-Aranda C, Jordán-Pérez C, Esteban-Medina M, Álamo-Álvarez I, Delgado-Bellido D, Lobo-Selma L, Dopazo J, Sastre A, Alonso J, Grünewald TGP, Bernabeu C, Byron A, Brunton VG, Amaral AT, Álava ED. Endoglin and MMP14 Contribute to Ewing Sarcoma Spreading by Modulation of Cell–Matrix Interactions. Int J Mol Sci 2022; 23:ijms23158657. [PMID: 35955799 PMCID: PMC9369355 DOI: 10.3390/ijms23158657] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 07/14/2022] [Accepted: 07/22/2022] [Indexed: 01/26/2023] Open
Abstract
Endoglin (ENG) is a mesenchymal stem cell (MSC) marker typically expressed by active endothelium. This transmembrane glycoprotein is shed by matrix metalloproteinase 14 (MMP14). Our previous work demonstrated potent preclinical activity of first-in-class anti-ENG antibody-drug conjugates as a nascent strategy to eradicate Ewing sarcoma (ES), a devastating rare bone/soft tissue cancer with a putative MSC origin. We also defined a correlation between ENG and MMP14 expression in ES. Herein, we show that ENG expression is significantly associated with a dismal prognosis in a large cohort of ES patients. Moreover, both ENG/MMP14 are frequently expressed in primary ES tumors and metastasis. To deepen in their functional relevance in ES, we conducted transcriptomic and proteomic profiling of in vitro ES models that unveiled a key role of ENG and MMP14 in cell mechano-transduction. Migration and adhesion assays confirmed that loss of ENG disrupts actin filament assembly and filopodia formation, with a concomitant effect on cell spreading. Furthermore, we observed that ENG regulates cell–matrix interaction through activation of focal adhesion signaling and protein kinase C expression. In turn, loss of MMP14 contributed to a more adhesive phenotype of ES cells by modulating the transcriptional extracellular matrix dynamics. Overall, these results suggest that ENG and MMP14 exert a significant role in mediating correct spreading machinery of ES cells, impacting the aggressiveness of the disease.
Collapse
Affiliation(s)
- Pilar Puerto-Camacho
- Institute of Biomedicine of Sevilla (IBiS), Virgen del Rocio University Hospital/CSIC/University of Sevilla/CIBERONC, Molecular Pathology of Sarcomas, 41013 Seville, Spain
| | - Juan Díaz-Martín
- Institute of Biomedicine of Sevilla (IBiS), Virgen del Rocio University Hospital/CSIC/University of Sevilla/CIBERONC, Molecular Pathology of Sarcomas, 41013 Seville, Spain
- Department of Normal and Pathological Cytology and Histology, School of Medicine, University of Seville, 41009 Seville, Spain
| | - Joaquín Olmedo-Pelayo
- Institute of Biomedicine of Sevilla (IBiS), Virgen del Rocio University Hospital/CSIC/University of Sevilla/CIBERONC, Molecular Pathology of Sarcomas, 41013 Seville, Spain
- Department of Normal and Pathological Cytology and Histology, School of Medicine, University of Seville, 41009 Seville, Spain
| | - Alfonso Bolado-Carrancio
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Carmen Salguero-Aranda
- Institute of Biomedicine of Sevilla (IBiS), Virgen del Rocio University Hospital/CSIC/University of Sevilla/CIBERONC, Molecular Pathology of Sarcomas, 41013 Seville, Spain
- Department of Normal and Pathological Cytology and Histology, School of Medicine, University of Seville, 41009 Seville, Spain
| | - Carmen Jordán-Pérez
- Institute of Biomedicine of Sevilla (IBiS), Virgen del Rocio University Hospital/CSIC/University of Sevilla/CIBERONC, Molecular Pathology of Sarcomas, 41013 Seville, Spain
| | - Marina Esteban-Medina
- Clinical Bioinformatics Area, Fundación Progreso y Salud (FPS), CDCA, Hospital Virgen del Rocío, 41013 Seville, Spain
| | - Inmaculada Álamo-Álvarez
- Clinical Bioinformatics Area, Fundación Progreso y Salud (FPS), CDCA, Hospital Virgen del Rocío, 41013 Seville, Spain
| | - Daniel Delgado-Bellido
- Institute of Biomedicine of Sevilla (IBiS), Virgen del Rocio University Hospital/CSIC/University of Sevilla/CIBERONC, Molecular Pathology of Sarcomas, 41013 Seville, Spain
| | - Laura Lobo-Selma
- Institute of Biomedicine of Sevilla (IBiS), Virgen del Rocio University Hospital/CSIC/University of Sevilla/CIBERONC, Molecular Pathology of Sarcomas, 41013 Seville, Spain
- Department of Normal and Pathological Cytology and Histology, School of Medicine, University of Seville, 41009 Seville, Spain
| | - Joaquín Dopazo
- Clinical Bioinformatics Area, Fundación Progreso y Salud (FPS), CDCA, Hospital Virgen del Rocío, 41013 Seville, Spain
| | - Ana Sastre
- Unidad Hemato-oncología Pediátrica, Hospital Infantil Universitario La Paz, 28046 Madrid, Spain
| | - Javier Alonso
- Unidad Hemato-oncología Pediátrica, Hospital Infantil Universitario La Paz, 28046 Madrid, Spain
- Unidad de Tumores Sólidos Infantiles, Instituto de Investigación de Enfermedades Raras, Instituto de Salud Carlos III (IIER-ISCIII), 28029 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III (CB06/07/1009; CIBERER-ISCIII), 28029 Madrid, Spain
| | - Thomas G. P. Grünewald
- Division of Translational Pediatric Sarcoma Research, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), 69120 Heidelberg, Germany
- Hopp-Children’s Cancer Center Heidelberg (KiTZ), 69120 Heidelberg, Germany
- Institute of Pathology, Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - Carmelo Bernabeu
- Division of Translational Pediatric Sarcoma Research, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), 69120 Heidelberg, Germany
- Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas (CSIC), 28040 Madrid, Spain
| | - Adam Byron
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XU, UK
- Division of Molecular and Cellular Function, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, UK
| | - Valerie G. Brunton
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Ana Teresa Amaral
- Institute of Biomedicine of Sevilla (IBiS), Virgen del Rocio University Hospital/CSIC/University of Sevilla/CIBERONC, Molecular Pathology of Sarcomas, 41013 Seville, Spain
- Department of Normal and Pathological Cytology and Histology, School of Medicine, University of Seville, 41009 Seville, Spain
- Correspondence: (A.T.A.); (E.D.Á.)
| | - Enrique De Álava
- Institute of Biomedicine of Sevilla (IBiS), Virgen del Rocio University Hospital/CSIC/University of Sevilla/CIBERONC, Molecular Pathology of Sarcomas, 41013 Seville, Spain
- Department of Normal and Pathological Cytology and Histology, School of Medicine, University of Seville, 41009 Seville, Spain
- Correspondence: (A.T.A.); (E.D.Á.)
| |
Collapse
|
5
|
Gao Y, Zhang J, Chen H, Wang Z, Hou J, Wang L. Dimethylamine enhances platelet hyperactivity in chronic kidney disease model. J Bioenerg Biomembr 2021; 53:585-595. [PMID: 34327565 DOI: 10.1007/s10863-021-09913-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 07/13/2021] [Indexed: 10/20/2022]
Abstract
Chronic kidney disease (CKD) remains a major health threat worldwide which is associated with elevated blood level of dimethylamine (DMA) and unbalanced platelet functions. Dimethylamine, a simple aliphatic amine, is abundantly found in human urine as well as other body fluids like plasma. However, the relation between dimethylamine and platelet activation is unclear. This study aims to unravel the mechanism of DMA and platelet function in chronic kidney disease. Through in vitro platelet characterization assay and in vivo CKD mouse model, the level of DMA, platelet activity and renal function were assessed by established methods. PKCδ and its downstream kinase MEK1/2 were examined by immunoblotting analysis of human platelet extract. Rescue experiments with PKCδ inhibitor or choline deficient diet were also conducted. DMA level in plasma of mouse CKD model was elevated along with enhanced platelet activation and comprised renal function. DMA can activate platelet in vitro and in vivo. Inhibition of PKCδ could antagonize the effect of DMA on platelet activation. When choline as the dietary source of DMA was deprived from CKD mouse, the level DMA was reduced and platelet activation was attenuated. Our results demonstrate that dimethylamine could enhance platelet activation in CKD model, potentially through activation of PKCδ.
Collapse
Affiliation(s)
- Yongning Gao
- Department of Hemodialysis, the Second Hospital of Hebei Medical University, No.215 Heping Western Road, Shijiazhuang, 053000, Hebei, China
| | - Jingyu Zhang
- Department of Hematology, the Second Hospital of Hebei Medical University, No.215 Heping Western Road, Shijiazhuang, 053000, Hebei, China.
| | - Hui Chen
- Department of Emergency, the Second Hospital of Hebei Medical University, No.215 Heping Western Road, Shijiazhuang, 053000, Hebei, China
| | - Zhu Wang
- Department of Gynaecology, the Second Hospital of Hebei Medical University, No.215 Heping Western Road, Shijiazhuang, 053000, Hebei, China
| | - Jingjing Hou
- Department of Hemodialysis, the Second Hospital of Hebei Medical University, No.215 Heping Western Road, Shijiazhuang, 053000, Hebei, China
| | - Lihua Wang
- Department of Hemodialysis, the Second Hospital of Hebei Medical University, No.215 Heping Western Road, Shijiazhuang, 053000, Hebei, China
| |
Collapse
|
6
|
Wang L, Liu G, Wu N, Dai B, Han S, Liu Q, Huang F, Chen Z, Xu W, Xia D, Gao C. mTOR regulates GPVI-mediated platelet activation. J Transl Med 2021; 19:201. [PMID: 33971888 PMCID: PMC8111939 DOI: 10.1186/s12967-021-02756-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 02/16/2021] [Indexed: 11/10/2022] Open
Abstract
Background Due to mTOR (mammalian/mechanistic target of rapamycin) gene-loss mice die during embryonic development, the role of mTOR in platelets has not been evaluated using gene knockout technology. Methods A mouse model with megakaryocyte/platelet-specific deletion of mTOR was established, and be used to evaluate the role of mTOR in platelet activation and thrombus formation. Results mTOR−/− platelets were deficient in thrombus formation when grown on low-concentration collagen-coated surfaces; however, no deficiency in thrombus formation was observed when mTOR−/− platelets were perfused on higher concentration collagen-coated surfaces. In FeCl3-induced mouse mesenteric arteriole thrombosis models, wild-type (WT) and mTOR−/− mice displayed significantly different responses to low-extent injury with respect to the ratio of occluded mice, especially within the first 40 min. Additionally, mTOR−/− platelets displayed reduced aggregation and dense granule secretion (ATP release) in response to low doses of the glycoprotein VI (GPVI) agonist collagen related peptide (CRP) and the protease-activated receptor-4 (PAR4) agonist GYPGKF-NH2; these deficiencies were overcame by stimulation with higher concentration agonists, suggesting dose dependence of the response. At low doses of GPVI or PAR agonist, the activation of αIIbβ3 in mTOR−/− platelets was reduced. Moreover, stimulation of mTOR−/− platelets with low-dose CRP attenuated the phosphorylation of S6K1, S6 and Akt Ser473, and increased the phosphorylation of PKCδ Thr505 and PKCε Ser729. Using isoform-specific inhibitors of PKCs (δ, ɛ, and α/β), we established that PKCδ/ɛ, and especially PKCδ but not PKCα/β or PKCθ, may be involved in low-dose GPVI-mediated/mTOR-dependent signaling. Conclusion These observations indicate that mTOR plays an important role in GPVI-dependent platelet activation and thrombus formation.
Collapse
Affiliation(s)
- Longsheng Wang
- Chronic Disease Research Institute, Department of Nutrition and Food Hygiene, Zhejiang University School of Public Health, 866 Yu-Hang-Tang Road, Hangzhou, 310058, China
| | - Gang Liu
- Chronic Disease Research Institute, Department of Nutrition and Food Hygiene, Zhejiang University School of Public Health, 866 Yu-Hang-Tang Road, Hangzhou, 310058, China.,Department of Pharmacology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou, China
| | - Nannan Wu
- Chronic Disease Research Institute, Department of Nutrition and Food Hygiene, Zhejiang University School of Public Health, 866 Yu-Hang-Tang Road, Hangzhou, 310058, China
| | - Baiyun Dai
- Chronic Disease Research Institute, Department of Nutrition and Food Hygiene, Zhejiang University School of Public Health, 866 Yu-Hang-Tang Road, Hangzhou, 310058, China
| | - Shuang Han
- Chronic Disease Research Institute, Department of Nutrition and Food Hygiene, Zhejiang University School of Public Health, 866 Yu-Hang-Tang Road, Hangzhou, 310058, China
| | - Qiaoyun Liu
- Department of Toxicology, Zhejiang University School of Public Health, 866 Yu-Hang-Tang Road, Hangzhou, 310058, China
| | - Fang Huang
- Department of Toxicology, Zhejiang University School of Public Health, 866 Yu-Hang-Tang Road, Hangzhou, 310058, China
| | - Zhihua Chen
- Department of Respiratory Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, China
| | - Weihong Xu
- Zhejiang Hospital, 12 Lingyin Road, Hangzhou, 310013, China
| | - Dajing Xia
- Department of Toxicology, Zhejiang University School of Public Health, 866 Yu-Hang-Tang Road, Hangzhou, 310058, China
| | - Cunji Gao
- Chronic Disease Research Institute, Department of Nutrition and Food Hygiene, Zhejiang University School of Public Health, 866 Yu-Hang-Tang Road, Hangzhou, 310058, China. .,Blood Research Institute, Blood Center of Wisconsin, Milwaukee, Milwaukee, WI, 53201, USA.
| |
Collapse
|
7
|
Tsai HJ, Cheng JC, Kao ML, Chiu HP, Chiang YH, Chen DP, Rau KM, Liao HR, Tseng CP. Integrin αIIbβ3 outside-in signaling activates human platelets through serine 24 phosphorylation of Disabled-2. Cell Biosci 2021; 11:32. [PMID: 33557943 PMCID: PMC7869483 DOI: 10.1186/s13578-021-00532-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 01/05/2021] [Indexed: 11/29/2022] Open
Abstract
Background Bidirectional integrin αIIbβ3 signaling is essential for platelet activation. The platelet adaptor protein Disabled-2 (Dab2) is a key regulator of integrin signaling and is phosphorylated at serine 24 in eukaryotic cells. However, the mechanistic insight and function of Dab2-serine 24 phosphorylation (Dab2-pSer24) in platelet biology are barely understood. This study aimed to define whether and how Dab2 is phosphorylated at Ser24 during platelet activation and to investigate the effect of Dab2-pSer24 on platelet function. Results An antibody with confirmed specificity for Dab2-pSer24 was generated. By using this antibody as a tool, we showed that protein kinase C (PKC)-mediated Dab2-pSer24 was a conservative signaling event when human platelets were activated by the platelet agonists such as thrombin, collagen, ADP, 12-O-tetradecanoylphorbol-13-acetate, and the thromboxane A2 activator U46619. The agonists-stimulated Dab2-pSer24 was attenuated by pretreatment of platelets with the RGDS peptide which inhibits integrin outside-in signaling by competitive binding of integrin αIIb with fibrinogen. Direct activation of platelet integrin outside-in signaling by combined treatment of platelets with manganese dichloride and fibrinogen or by spreading of platelets on fibrinogen also resulted in Dab2-pSer24. These findings implicate that Dab2-pSer24 was associated with the outside-in signaling of integrin. Further analysis revealed that Dab2-pSer24 was downstream of Src-PKC-axis and phospholipase D1 underlying the integrin αIIbβ3 outside-in signaling. A membrane penetrating peptide R11-Ser24 which contained 11 repeats of arginine linked to the Dab2-Ser24 phosphorylation site and its flanking sequences (RRRRRRRRRRR19APKAPSKKEKK29) and the R11-S24A peptide with Ser24Ala mutation were designed to elucidate the functions of Dab2-pSer24. R11-Ser24 but not R11-S24A inhibited agonists-stimulated Dab2-pSer24 and consequently suppressed platelet spreading on fibrinogen, with no effect on platelet aggregation and fibrinogen binding. Notably, Ser24 and the previously reported Ser723 phosphorylation (Dab2-pSer723) occurred exclusively in a single Dab2 molecule and resulted in distinctive subcellular distribution and function of Dab2. Dab2-pSer723 was mainly distributed in the cytosol of activated platelets and associated with integrin inside-out signaling, while Dab2-pSer24 was mainly distributed in the membrane fraction of activated platelets and associated with integrin outside-in signaling. Conclusions These findings demonstrate for the first time that Dab2-pSer24 is conservative in integrin αIIbβ3 outside-in signaling during platelet activation and plays a novel role in the control of cytoskeleton reorganization and platelet spreading on fibrinogen.
Collapse
Affiliation(s)
- Hui-Ju Tsai
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan, Republic of China
| | - Ju-Chien Cheng
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung, 404, Taiwan, Republic of China
| | - Man-Leng Kao
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan, Republic of China
| | - Hung-Pin Chiu
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan, Republic of China
| | - Yi-Hsuan Chiang
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan, Republic of China
| | - Ding-Ping Chen
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan, Republic of China.,Department of Laboratory Medicine, Chang Gung Memorial Hospital, Taoyuan, 333, Taiwan, Republic of China
| | - Kun-Ming Rau
- Department of Hematology-Oncology, E-Da Cancer Hospital, Kaohsiung, 824, Taiwan, Republic of China.,School of Medicine, College of Medicine, I-Shou University, Kaohsiung, 824, Taiwan, Republic of China
| | - Hsiang-Ruei Liao
- Graduate institute of Natural Products, College of Medicine, Chang-Gung University, Taoyuan, 333, Taiwan, Republic of China.,Graduate institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan, Republic of China.,Department of Anesthesiology, Chang Gung Memorial Hospital, Taoyuan, 333, Taiwan, Republic of China
| | - Ching-Ping Tseng
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan, Republic of China. .,Department of Laboratory Medicine, Chang Gung Memorial Hospital, Taoyuan, 333, Taiwan, Republic of China. .,Graduate institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan, Republic of China. .,Molecular Medicine Research Center, Chang Gung University, Taoyuan, 333, Taiwan, Republic of China.
| |
Collapse
|
8
|
Chaudhary PK, Kim S, Jee Y, Lee SH, Kim S. Characterization of Integrin αIIbβ3-Mediated Outside-in Signaling by Protein Kinase Cδ in Platelets. Int J Mol Sci 2020; 21:ijms21186563. [PMID: 32911704 PMCID: PMC7555476 DOI: 10.3390/ijms21186563] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 09/01/2020] [Accepted: 09/07/2020] [Indexed: 11/16/2022] Open
Abstract
Engagement of integrin αIIbβ3 promotes platelet-platelet interaction and stimulates outside-in signaling that amplifies activation. Protein kinase Cδ (PKCδ) is known to play an important role in platelet activation, but its role in outside-in signaling has not been established. In the present study, we determined the role of PKCδ and its signaling pathways in integrin αIIbβ3-mediated outside-in signaling in platelets using PKCδ-deficient platelets. Platelet spreading to immobilized fibrinogen resulted in PKCδ phosphorylation, suggesting that αIIbβ3 activation caused PKCδ activation. αIIbβ3-mediated phosphorylation of Akt was significantly inhibited in PKCδ -/- platelets, indicating a role of PKCδ in outside-in signaling. αIIbβ3-mediated PKCδ phosphorylation was inhibited by proline-rich tyrosine kinase 2 (Pyk2) selective inhibitor, suggesting that Pyk2 contributes to the regulation of PKCδ phosphorylation in outside-in signaling. Additionally, Src-family kinase inhibitor PP2 inhibited integrin-mediated Pyk2 and PKCδ phosphorylation. Lastly, platelet spreading was inhibited in PKCδ -/- platelets compared to the wild-type (WT) platelets, and clot retraction from PKCδ -/- platelets was markedly delayed, indicating that PKCδ is involved in the regulation of αIIbβ3-dependent interactivities with cytoskeleton elements. Together, these results provide evidence that PKCδ plays an important role in outside-in signaling, which is regulated by Pyk2 in platelets.
Collapse
Affiliation(s)
- Preeti Kumari Chaudhary
- College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Korea; (P.K.C.); (S.K.); (S.-H.L.)
| | - Sanggu Kim
- College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Korea; (P.K.C.); (S.K.); (S.-H.L.)
| | - Youngheun Jee
- College of Veterinary Medicine and Veterinary Medical Research Institute, Jeju National University, Jeju 63243, Korea;
| | - Seung-Hun Lee
- College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Korea; (P.K.C.); (S.K.); (S.-H.L.)
| | - Soochong Kim
- College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Korea; (P.K.C.); (S.K.); (S.-H.L.)
- Correspondence: ; Tel.: +82-43-249-1846
| |
Collapse
|
9
|
Schmoker AM, Perez Pearson LM, Cruz C, Colon Flores LG, Branfeild S, Pagán Torres FD, Fonseca K, Cantres YM, Salgado Ramirez CA, Melendez LM, Ballif BA, Washington AV. Defining the TLT-1 interactome from resting and activated human platelets. J Proteomics 2020; 215:103638. [PMID: 31923473 PMCID: PMC7044047 DOI: 10.1016/j.jprot.2020.103638] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 01/03/2020] [Accepted: 01/05/2020] [Indexed: 12/14/2022]
Abstract
The triggering receptor expressed on myeloid cells (TREM) protein family forms a class of type I transmembrane proteins expressed in immune cells that play important roles in innate and adaptive immune responses. The TREM family member TREM-like transcript 1 (TLT-1, also TREML1) is expressed in megakaryocytes and packaged into platelet granules. TLT-1 binds fibrinogen and plays a role in bleeding initiated by inflammatory insults. Here, we describe a proteomics screen that maps the TLT-1 interactome in resting and activated human platelets. Several identified TLT-1 interactors are involved in cell adhesion and migration, as well as platelet activation. Select interactors, including β3-integrin, RACK1, GRB2, and Rabs 5A, 7, and 11A, were additionally characterized in co-immunoprecipitation/immunoblotting experiments. Finally, several phosphorylation sites were found on immunoprecipitated TLT-1, including Thr280, a novel, regulated site on a conserved residue near the TLT-1 ITIM regulatory sequence. SIGNIFICANCE: Platelet function relies on the secretion of active molecules from intracellular vesicles, or granules, which contain soluble and membrane-bound proteins that are essential for platelet aggregation, coagulation reactions, and pathogen defense mechanisms. TLT-1 is sequestered in α-granules and transported to the plasma membrane, where it plays a unique role in hemostasis after inflammatory insults. Despite the known importance of TLT-1 in platelet biology, our knowledge of TLT-1 mechanistic signaling is limited. This study defines the TLT-1 interactome in resting and active human platelets, identifying several novel TLT-1 interactors, as well as TLT-1 phosphorylation sites, all with likely signaling implications in platelet aggregation dynamics.
Collapse
Affiliation(s)
- Anna M Schmoker
- Department of Biology, University of Vermont, 109 Carrigan Drive, 120A Marsh Life Sciences, Burlington, VT 05405, USA.
| | - Leishla M Perez Pearson
- Department of Biology, University of Vermont, 109 Carrigan Drive, 120A Marsh Life Sciences, Burlington, VT 05405, USA; Department of Biology, University of Puerto Rico-Río Piedras, Department of Biology, San Juan, PR, USA
| | - Claudia Cruz
- Department of Biology, University of Vermont, 109 Carrigan Drive, 120A Marsh Life Sciences, Burlington, VT 05405, USA; Department of Biology, University of Puerto Rico-Río Piedras, Department of Biology, San Juan, PR, USA
| | - Luis G Colon Flores
- Department of Biology, University of Puerto Rico-Río Piedras, Department of Biology, San Juan, PR, USA
| | - Siobhan Branfeild
- Department of Biology, University of Puerto Rico-Río Piedras, Department of Biology, San Juan, PR, USA
| | - Fabiola D Pagán Torres
- Department of Biology, University of Vermont, 109 Carrigan Drive, 120A Marsh Life Sciences, Burlington, VT 05405, USA
| | - Karmen Fonseca
- Department of Biology, University of Vermont, 109 Carrigan Drive, 120A Marsh Life Sciences, Burlington, VT 05405, USA
| | - Yadira M Cantres
- Translational Proteomics Center, Comprehensive Cancer Center, University of Puerto Rico, Medical Sciences Campus, San Juan, PR, USA
| | - Carla A Salgado Ramirez
- Translational Proteomics Center, Comprehensive Cancer Center, University of Puerto Rico, Medical Sciences Campus, San Juan, PR, USA
| | - Loyda M Melendez
- Department of Microbiology and Medical Zoology, University of Puerto Rico, Medical Sciences Campus, San Juan, PR, USA; Translational Proteomics Center, Comprehensive Cancer Center, University of Puerto Rico, Medical Sciences Campus, San Juan, PR, USA
| | - Bryan A Ballif
- Department of Biology, University of Vermont, 109 Carrigan Drive, 120A Marsh Life Sciences, Burlington, VT 05405, USA.
| | - A Valance Washington
- Department of Biology, University of Puerto Rico-Río Piedras, Department of Biology, San Juan, PR, USA.
| |
Collapse
|
10
|
Bialkowska K, Sossey-Alaoui K, Pluskota E, Izem L, Qin J, Plow EF. Site-specific phosphorylation regulates the functions of kindlin-3 in a variety of cells. Life Sci Alliance 2020; 3:3/3/e201900594. [PMID: 32024667 PMCID: PMC7010036 DOI: 10.26508/lsa.201900594] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 01/22/2020] [Accepted: 01/23/2020] [Indexed: 12/12/2022] Open
Abstract
Studies of isolated cells, mice, and humans have demonstrated the vital role of the FERM domain protein kindlin-3 in integrin activation in certain hematopoietic and non-hematopoietic cells, consequent to binding to integrin β-subunits. To explore regulatory mechanisms, we developed a monoclonal antibody that selectively recognizes the phosphorylated form of Ser484 (pS484) in kindlin-3. Activation of platelets, HEL megakaryocytic-like cells and BT549 breast cancer cells led to enhanced expression of pS484 as assessed by immunofluorescence or Western blotting. In platelets, pS484 rose rapidly and transiently upon stimulation. When a mutant form of kindlin-3, T482S484/AA kindlin-3, was transduced into mouse megakaryocytes, it failed to support activation of integrin αIIbβ3, whereas wild-type kindlin-3 did. In MDA-MB231 breast cancer cells, expression of T482S484/AA kindlin-3 suppressed cell spreading, migration, invasion, and VEGF production. Wild-type kindlin-3 expressing cells markedly increased tumor growth in vivo, whereas T482S484/AA kindlin-3 significantly blunted tumor progression. Thus, our data establish that a unique phosphorylation event in kindlin-3 regulates its cellular functions.
Collapse
Affiliation(s)
- Katarzyna Bialkowska
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute Cleveland Clinic, Cleveland, OH, USA
| | - Khalid Sossey-Alaoui
- Department of Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Elzbieta Pluskota
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute Cleveland Clinic, Cleveland, OH, USA
| | - Lahoucine Izem
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute Cleveland Clinic, Cleveland, OH, USA
| | - Jun Qin
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute Cleveland Clinic, Cleveland, OH, USA
| | - Edward F Plow
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
11
|
Huang J, Li X, Shi X, Zhu M, Wang J, Huang S, Huang X, Wang H, Li L, Deng H, Zhou Y, Mao J, Long Z, Ma Z, Ye W, Pan J, Xi X, Jin J. Platelet integrin αIIbβ3: signal transduction, regulation, and its therapeutic targeting. J Hematol Oncol 2019; 12:26. [PMID: 30845955 PMCID: PMC6407232 DOI: 10.1186/s13045-019-0709-6] [Citation(s) in RCA: 212] [Impact Index Per Article: 42.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Accepted: 02/21/2019] [Indexed: 12/18/2022] Open
Abstract
Integrins are a family of transmembrane glycoprotein signaling receptors that can transmit bioinformation bidirectionally across the plasma membrane. Integrin αIIbβ3 is expressed at a high level in platelets and their progenitors, where it plays a central role in platelet functions, hemostasis, and arterial thrombosis. Integrin αIIbβ3 also participates in cancer progression, such as tumor cell proliferation and metastasis. In resting platelets, integrin αIIbβ3 adopts an inactive conformation. Upon agonist stimulation, the transduction of inside-out signals leads integrin αIIbβ3 to switch from a low- to high-affinity state for fibrinogen and other ligands. Ligand binding causes integrin clustering and subsequently promotes outside-in signaling, which initiates and amplifies a range of cellular events to drive essential platelet functions such as spreading, aggregation, clot retraction, and thrombus consolidation. Regulation of the bidirectional signaling of integrin αIIbβ3 requires the involvement of numerous interacting proteins, which associate with the cytoplasmic tails of αIIbβ3 in particular. Integrin αIIbβ3 and its signaling pathways are considered promising targets for antithrombotic therapy. This review describes the bidirectional signal transduction of integrin αIIbβ3 in platelets, as well as the proteins responsible for its regulation and therapeutic agents that target integrin αIIbβ3 and its signaling pathways.
Collapse
Affiliation(s)
- Jiansong Huang
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Key Laboratory of Hematologic Malignancies, Diagnosis and Treatment, Hangzhou, Zhejiang, China.,Institute of Hematology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xia Li
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Key Laboratory of Hematologic Malignancies, Diagnosis and Treatment, Hangzhou, Zhejiang, China.,Institute of Hematology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xiaofeng Shi
- Department of Hematology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Mark Zhu
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jinghan Wang
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Key Laboratory of Hematologic Malignancies, Diagnosis and Treatment, Hangzhou, Zhejiang, China.,Institute of Hematology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Shujuan Huang
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Key Laboratory of Hematologic Malignancies, Diagnosis and Treatment, Hangzhou, Zhejiang, China.,Institute of Hematology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xin Huang
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Key Laboratory of Hematologic Malignancies, Diagnosis and Treatment, Hangzhou, Zhejiang, China.,Institute of Hematology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Huafeng Wang
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Key Laboratory of Hematologic Malignancies, Diagnosis and Treatment, Hangzhou, Zhejiang, China.,Institute of Hematology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Department of Hematological Malignancies Translational Science, Gehr Family Center for Leukemia Research, Hematologic Malignancies and Stem Cell Transplantation Institute, Beckman Research Institute, City of Hope Medical Center, Duarte, CA, 91010, USA
| | - Ling Li
- Department of Hematological Malignancies Translational Science, Gehr Family Center for Leukemia Research, Hematologic Malignancies and Stem Cell Transplantation Institute, Beckman Research Institute, City of Hope Medical Center, Duarte, CA, 91010, USA
| | - Huan Deng
- Department of Pathology, The Fourth Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Yulan Zhou
- Department of Hematology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Jianhua Mao
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Collaborative Innovation Center of Hematology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Sino-French Research Centre for Life Sciences and Genomics, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhangbiao Long
- Department of Hematology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zhixin Ma
- Clinical Prenatal Diagnosis Center, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Wenle Ye
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Key Laboratory of Hematologic Malignancies, Diagnosis and Treatment, Hangzhou, Zhejiang, China.,Institute of Hematology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jiajia Pan
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Key Laboratory of Hematologic Malignancies, Diagnosis and Treatment, Hangzhou, Zhejiang, China.,Institute of Hematology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xiaodong Xi
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Collaborative Innovation Center of Hematology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China. .,Sino-French Research Centre for Life Sciences and Genomics, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Jie Jin
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China. .,Key Laboratory of Hematologic Malignancies, Diagnosis and Treatment, Hangzhou, Zhejiang, China. .,Institute of Hematology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| |
Collapse
|
12
|
Carubbi C, Masselli E, Pozzi G, Mattioli M, Martini S, Goldoni M, Aloe R, Cervellin G, Vitale M, Gobbi G. Combination of Platelet expression of PKCepsilon and cardiac troponin-I for early diagnosis of chest pain patients in the emergency department. Sci Rep 2019; 9:2125. [PMID: 30765820 PMCID: PMC6375996 DOI: 10.1038/s41598-019-38624-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 12/28/2018] [Indexed: 01/02/2023] Open
Abstract
A rapid differential diagnosis of the clinical conditions underlying chest pain is a relevant clinical issue. Specifically, a fast rule-in or -out of acute myocardial infarction (AMI) is mandatory to improve diagnostic outcome and cost-effectiveness of patient management. We demonstrated that Protein Kinase C (PKC) epsilon is selectively expressed by platelets from AMI patients, accounting for increased platelet activation. Thus, we hypothesized that PKCepsilon-expressing platelets may represent a pathophysiological marker of AMI that could be utilized in combination with troponin-I, the conventional marker of cardiac injury, to add diagnostic information in chest pain workup. In 94 chest pain patients consecutively admitted to Parma University Hospital, we tested the diagnostic performance of flow-cytometric detection of PKCepsilon expressing platelets in discriminating AMI vs. non-AMI conditions. We demonstrated that PKCepsilon-expressing platelets were significantly higher in patients with AMI. Flow cytometry detection of PKCepsilon-expressing platelets showed high sensitivity and specificity (87.5% and 84.4%, respectively) and good diagnostic accuracy (AUC: 0.875). The combination of PKCepsilon expressing platelets and cardiac troponin clearly discriminates patients with 100% and 0% of probability to be affected by AMI. Overall, we highlighted a dual marker strategy potentially useful for a rapid rule-in or -out of myocardial infarction in chest pain patients.
Collapse
Affiliation(s)
- Cecilia Carubbi
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Elena Masselli
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Giulia Pozzi
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Maria Mattioli
- Division of Cardiology, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| | - Silvia Martini
- Protein Phosphorylation Laboratory, Francis Crick Institute, London, United Kingdom
| | - Matteo Goldoni
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Rosalia Aloe
- Dipartimento di Biochimica ad Elevata Automazione, Dipartimento Diagnostico, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| | | | - Marco Vitale
- Department of Medicine and Surgery, University of Parma, Parma, Italy.
- Sport and exercise medicine center (SEM), University of Parma, Parma, Italy.
- CoreLab, Azienda Ospedaliero-Universitaria di Parma, University of Parma, Parma, Italy.
| | - Giuliana Gobbi
- Department of Medicine and Surgery, University of Parma, Parma, Italy
- Sport and exercise medicine center (SEM), University of Parma, Parma, Italy
| |
Collapse
|
13
|
|
14
|
Abstract
B cells are essential to the adaptive immune system for providing the humoral immunity against cohorts of pathogens. The presentation of antigen to the B cell receptor (BCR) leads to the initiation of B cell activation, which is a process sensitive to the stiffness features of the substrates presenting the antigens. Mechanosensing of the B cells, potentiated through BCR signaling and the adhesion molecules, efficiently regulates B cell activation, proliferation and subsequent antibody responses. Defects in sensing of the antigen-presenting substrates can lead to the activation of autoreactive B cells in autoimmune diseases. The use of high-resolution, high-speed live-cell imaging along with the sophisticated biophysical materials, has uncovered the mechanisms underlying the initiation of B cell activation within seconds of its engagement with the antigen presenting substrates. In this chapter, we reviewed studies that have contributed to uncover the molecular mechanisms of B cell mechanosensing during the initiation of B cell activation.
Collapse
Affiliation(s)
- Samina Shaheen
- Center for life sciences, MOE Key Laboratory of Protein Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Beijing Key Lab for Immunological Research on Chronic Diseases, School of Life Sciences, Institute for Immunology, Tsinghua University, Beijing, China
| | - Zhengpeng Wan
- Center for life sciences, MOE Key Laboratory of Protein Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Beijing Key Lab for Immunological Research on Chronic Diseases, School of Life Sciences, Institute for Immunology, Tsinghua University, Beijing, China
| | - Kabeer Haneef
- Center for life sciences, MOE Key Laboratory of Protein Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Beijing Key Lab for Immunological Research on Chronic Diseases, School of Life Sciences, Institute for Immunology, Tsinghua University, Beijing, China
| | - Yingyue Zeng
- Center for life sciences, MOE Key Laboratory of Protein Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Beijing Key Lab for Immunological Research on Chronic Diseases, School of Life Sciences, Institute for Immunology, Tsinghua University, Beijing, China
| | - Wang Jing
- Center for life sciences, MOE Key Laboratory of Protein Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Beijing Key Lab for Immunological Research on Chronic Diseases, School of Life Sciences, Institute for Immunology, Tsinghua University, Beijing, China
| | - Wanli Liu
- Center for life sciences, MOE Key Laboratory of Protein Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Beijing Key Lab for Immunological Research on Chronic Diseases, School of Life Sciences, Institute for Immunology, Tsinghua University, Beijing, China.
| |
Collapse
|
15
|
Abstract
The WD40 domain is one of the most abundant and interacting domains in the eukaryotic genome. In proteins the WD domain folds into a β-propeller structure, providing a platform for the interaction and assembly of several proteins into a signalosome. WD40 repeats containing proteins, in lower eukaryotes, are mainly involved in growth, cell cycle, development and virulence, while in higher organisms, they play an important role in diverse cellular functions like signal transduction, cell cycle control, intracellular transport, chromatin remodelling, cytoskeletal organization, apoptosis, development, transcriptional regulation, immune responses. To play the regulatory role in various processes, they act as a scaffold for protein-protein or protein-DNA interaction. So far, no WD40 domain has been identified with intrinsic enzymatic activity. Several WD40 domain-containing proteins have been recently characterized in prokaryotes as well. The review summarizes the vast array of functions performed by different WD40 domain containing proteins, their domain organization and functional conservation during the course of evolution.
Collapse
Affiliation(s)
- Buddhi Prakash Jain
- Department of Zoology, School of Life Sciences, Mahatma Gandhi Central University, Motihari, Bihar, 845401, India.
| | - Shweta Pandey
- APSGMNS Govt P G College, Kawardha, Chhattisgarh, 491995, India
| |
Collapse
|
16
|
Liu D, Cao Y, Zhang X, Peng C, Tian X, Yan C, Liu Y, Liu M, Han Y. Chemokine CC-motif ligand 2 participates in platelet function and arterial thrombosis by regulating PKCα-P38MAPK-HSP27 pathway. Biochim Biophys Acta Mol Basis Dis 2018; 1864:2901-2912. [PMID: 29864522 DOI: 10.1016/j.bbadis.2018.05.025] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 05/10/2018] [Accepted: 05/30/2018] [Indexed: 12/22/2022]
Abstract
BACKGROUND Studies indicate that chemokine CC-motif ligand 2 (CCL2) is involved in inflammation and atherosclerosis. However, the roles and mechanisms of CCL2 on platelet function and arterial thrombosis are unknown. METHODS The expressions of CCL2 or CCR2 in the plasma, platelets and coronary thrombus of ST-elevated myocardial infarction (STEMI) patients were examined by ELISA, Western blot, immunohistochemistry and immunofluorescence. The roles of CCL2 on platelet aggregation, activation and secretion were examined by light transmission aggregometry, flow cytometry and ELISA. RESULTS The expressions of CCL2 or CCR2 in the plasma or platelets of STEMI patients with platelet high response were higher than those with platelet normal response; In vitro, exogenous recombinant human CCL2 markedly increased platelet aggregation, activation and granule secretion, which were abolished by CCL2 neutralizing antibody or CCR2 inhibiter. CCL2 increased the phosphorylation levels of PKCα (Thr638), P38MAPK (Thr180/Tyr182) and HSP27 (S78/S82) in human platelets, which were abrogated by PKCα inhibitor (RO 318220) or P38MAPK inhibitor (SB 203580). RO 318220 or SB 203580 diminished CCL2-induced platelet function. In CCL2-/- mice, platelet aggregation and secretion were attenuated; the phosphorylation of PKCα, P38MAPK and HSP27 were decreased. In a carotid arterial thrombus mouse model, CCL2-/- mice displayed a significantly extended carotid artery occlusion time compared with wild type. CONCLUSIONS CCL2 played important roles in regulating platelet function and arterial thrombosis through the PKCα-P38MAPK-HSP27 pathway, which might provide theoretical basis for searching new antiplatelet drugs and the treatment for cardiovascular diseases.
Collapse
Affiliation(s)
- Dan Liu
- Cardiovascular Research Institute and Department of Cardiology, General Hospital of Shenyang Military Region, Shenyang, China
| | - Yu Cao
- Cardiovascular Research Institute and Department of Cardiology, General Hospital of Shenyang Military Region, Shenyang, China
| | - Xiaolin Zhang
- Cardiovascular Research Institute and Department of Cardiology, General Hospital of Shenyang Military Region, Shenyang, China
| | - Chengfei Peng
- Cardiovascular Research Institute and Department of Cardiology, General Hospital of Shenyang Military Region, Shenyang, China
| | - Xiaoxiang Tian
- Cardiovascular Research Institute and Department of Cardiology, General Hospital of Shenyang Military Region, Shenyang, China
| | - Chenghui Yan
- Cardiovascular Research Institute and Department of Cardiology, General Hospital of Shenyang Military Region, Shenyang, China
| | - Yanxia Liu
- Cardiovascular Research Institute and Department of Cardiology, General Hospital of Shenyang Military Region, Shenyang, China
| | - Meili Liu
- Cardiovascular Research Institute and Department of Cardiology, General Hospital of Shenyang Military Region, Shenyang, China
| | - Yaling Han
- Cardiovascular Research Institute and Department of Cardiology, General Hospital of Shenyang Military Region, Shenyang, China.
| |
Collapse
|
17
|
Mugami S, Dobkin-Bekman M, Rahamim-Ben Navi L, Naor Z. Differential roles of PKC isoforms (PKCs) in GnRH stimulation of MAPK phosphorylation in gonadotrope derived cells. Mol Cell Endocrinol 2018; 463:97-105. [PMID: 28392410 DOI: 10.1016/j.mce.2017.04.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 04/04/2017] [Accepted: 04/05/2017] [Indexed: 12/30/2022]
Abstract
The role of protein kinase C (PKC) isoforms (PKCs) in GnRH-stimulated MAPK [ERK1/2, JNK1/2 and p38) phosphorylation was examined in gonadotrope derived cells. GnRH induced a protracted activation of ERK1/2 and a slower and more transient activation of JNK1/2 and p38MAPK. Gonadotropes express conventional PKCα and PKCβII, novel PKCδ, PKCε and PKCθ, and atypical PKC-ι/λ. The use of green fluorescent protein (GFP)-PKCs constructs revealed that GnRH induced rapid translocation of PKCα and PKCβII to the plasma membrane, followed by their redistribution to the cytosol. PKCδ and PKCε localized to the cytoplasm and Golgi, followed by the rapid redistribution by GnRH of PKCδ to the perinuclear zone and of PKCε to the plasma membrane. The use of dominant negatives for PKCs and peptide inhibitors for the receptors for activated C kinase (RACKs) has revealed differential role for PKCα, PKCβII, PKCδ and PKCε in ERK1/2, JNK1/2 and p38MAPK phosphorylation in a ligand-and cell context-dependent manner. The paradoxical findings that PKCs activated by GnRH and PMA play a differential role in MAPKs phosphorylation may be explained by persistent vs. transient redistribution of selected PKCs or redistribution of a given PKC to the perinuclear zone vs. the plasma membrane. Thus, we have identified the PKCs involved in GnRH stimulated MAPKs phosphorylation in gonadotrope derived cells. Once activated, the MAPKs will mediate the transcription of the gonadotropin subunits and GnRH receptor genes.
Collapse
Affiliation(s)
- Shany Mugami
- Department of Biochemistry and Molecular Biology, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Ramat Aviv 69978, Israel
| | - Masha Dobkin-Bekman
- Department of Biochemistry and Molecular Biology, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Ramat Aviv 69978, Israel
| | - Liat Rahamim-Ben Navi
- Department of Biochemistry and Molecular Biology, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Ramat Aviv 69978, Israel
| | - Zvi Naor
- Department of Biochemistry and Molecular Biology, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Ramat Aviv 69978, Israel.
| |
Collapse
|
18
|
Abstract
Integrin αIIbβ3 is a highly abundant heterodimeric platelet receptor that can transmit information bidirectionally across the plasma membrane, and plays a critical role in hemostasis and thrombosis. Upon platelet activation, inside-out signaling pathways increase the affinity of αIIbβ3 for fibrinogen and other ligands. Ligand binding and integrin clustering subsequently stimulate outside-in signaling, which initiates and amplifies a range of cellular events driving essential platelet processes such as spreading, thrombus consolidation, and clot retraction. Integrin αIIbβ3 has served as an excellent model for the study of integrin biology, and it has become clear that integrin outside-in signaling is highly complex and involves a vast array of enzymes, signaling adaptors, and cytoskeletal components. In this review, we provide a concise but comprehensive overview of αIIbβ3 outside-in signaling, focusing on the key players involved, and how they cooperate to orchestrate this critical aspect of platelet biology. We also discuss gaps in the current understanding of αIIbβ3 outside-in signaling and highlight avenues for future investigation.
Collapse
|
19
|
Shaheen S, Wan Z, Li Z, Chau A, Li X, Zhang S, Liu Y, Yi J, Zeng Y, Wang J, Chen X, Xu L, Chen W, Wang F, Lu Y, Zheng W, Shi Y, Sun X, Li Z, Xiong C, Liu W. Substrate stiffness governs the initiation of B cell activation by the concerted signaling of PKCβ and focal adhesion kinase. eLife 2017; 6. [PMID: 28755662 PMCID: PMC5536945 DOI: 10.7554/elife.23060] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 07/03/2017] [Indexed: 12/17/2022] Open
Abstract
The mechanosensing ability of lymphocytes regulates their activation in response to antigen stimulation, but the underlying mechanism remains unexplored. Here, we report that B cell mechanosensing-governed activation requires BCR signaling molecules. PMA-induced activation of PKCβ can bypass the Btk and PLC-γ2 signaling molecules that are usually required for B cells to discriminate substrate stiffness. Instead, PKCβ-dependent activation of FAK is required, leading to FAK-mediated potentiation of B cell spreading and adhesion responses. FAK inactivation or deficiency impaired B cell discrimination of substrate stiffness. Conversely, adhesion molecules greatly enhanced this capability of B cells. Lastly, B cells derived from rheumatoid arthritis (RA) patients exhibited an altered BCR response to substrate stiffness in comparison with healthy controls. These results provide a molecular explanation of how initiation of B cell activation discriminates substrate stiffness through a PKCβ-mediated FAK activation dependent manner.
Collapse
Affiliation(s)
- Samina Shaheen
- MOE Key Laboratory of Protein Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Life Sciences, Institute for Immunology, Tsinghua University, Beijing, China
| | - Zhengpeng Wan
- MOE Key Laboratory of Protein Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Life Sciences, Institute for Immunology, Tsinghua University, Beijing, China
| | - Zongyu Li
- MOE Key Laboratory of Protein Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Life Sciences, Institute for Immunology, Tsinghua University, Beijing, China
| | - Alicia Chau
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Xinxin Li
- MOE Key Laboratory of Protein Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Life Sciences, Institute for Immunology, Tsinghua University, Beijing, China
| | - Shaosen Zhang
- MOE Key Laboratory of Protein Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Life Sciences, Institute for Immunology, Tsinghua University, Beijing, China
| | - Yang Liu
- MOE Key Laboratory of Protein Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Life Sciences, Institute for Immunology, Tsinghua University, Beijing, China
| | - Junyang Yi
- MOE Key Laboratory of Protein Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Life Sciences, Institute for Immunology, Tsinghua University, Beijing, China
| | - Yingyue Zeng
- MOE Key Laboratory of Protein Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Life Sciences, Institute for Immunology, Tsinghua University, Beijing, China
| | - Jing Wang
- MOE Key Laboratory of Protein Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Life Sciences, Institute for Immunology, Tsinghua University, Beijing, China
| | - Xiangjun Chen
- MOE Key Laboratory of Protein Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Life Sciences, Institute for Immunology, Tsinghua University, Beijing, China
| | - Liling Xu
- MOE Key Laboratory of Protein Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Life Sciences, Institute for Immunology, Tsinghua University, Beijing, China
| | - Wei Chen
- School of Medicine, Zhejiang University, Hangzhou, China
| | - Fei Wang
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, China
| | - Yun Lu
- State Key Joint Laboratory of Environment Simulation and Pollution Control, School of Environment, Tsinghua University, Beijing, China
| | - Wenjie Zheng
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Yan Shi
- Center for Life Sciences, Department of Basic Medical Sciences, Institute of Immunology, Tsinghua University, Beijing, China
| | - Xiaolin Sun
- Department of Rheumatology and Immunology, Clinical Immunology Center, Peking University People's Hospital, Beijing, China
| | - Zhanguo Li
- Department of Rheumatology and Immunology, Clinical Immunology Center, Peking University People's Hospital, Beijing, China
| | - Chunyang Xiong
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China.,College of Engineering, Peking University, Beijing, China
| | - Wanli Liu
- MOE Key Laboratory of Protein Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Life Sciences, Institute for Immunology, Tsinghua University, Beijing, China
| |
Collapse
|
20
|
Beck S, Leitges M, Stegner D. Protein kinase Cι/λ is dispensable for platelet function in thrombosis and hemostasis in mice. Cell Signal 2017; 38:223-229. [PMID: 28739484 DOI: 10.1016/j.cellsig.2017.07.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 07/05/2017] [Accepted: 07/20/2017] [Indexed: 11/17/2022]
Abstract
Platelet activation at sites of vascular injury is crucial for hemostasis, but it may also cause myocardial infarction or ischemic stroke. Upon platelet activation, cytoskeletal reorganization is essential for platelet secretion and thrombus formation. Members of the protein kinase C family, which includes 12 isoforms, are involved in most platelet responses required for thrombus formation. The atypical protein kinase Cι/λ (PKCι/λ) has been implicated as an important mediator of cell polarity, carcinogenesis and immune cell responses. PKCι/λ is known to be associated with the small GTPase Cdc42, an important mediator of multiple platelet functions; however, its exact function in platelets is not known. To study the role of PKCι/λ, we generated platelet- and megakaryocyte-specific PKCι/λ knockout mice (Prkcifl/fl, Pf4-Cre) and used them to investigate the function of PKCι/λ in platelet activation and aggregation in vitro and in vivo. Surprisingly, lack of PKCι/λ had no detectable effect on platelet spreading and function in vitro and in vivo under all tested conditions. These results indicate that PKCι/λ is dispensable for Cdc42-triggered processes and for thrombosis and hemostasis in mice.
Collapse
Affiliation(s)
- Sarah Beck
- Institute of Experimental Biomedicine, University Hospital and Rudolf Virchow Center, University of Würzburg, Würzburg, Germany
| | | | - David Stegner
- Institute of Experimental Biomedicine, University Hospital and Rudolf Virchow Center, University of Würzburg, Würzburg, Germany.
| |
Collapse
|
21
|
Masselli E, Carubbi C, Pozzi G, Martini S, Aversa F, Galli D, Gobbi G, Mirandola P, Vitale M. Platelet expression of PKCepsilon oncoprotein in myelofibrosis is associated with disease severity and thrombotic risk. ANNALS OF TRANSLATIONAL MEDICINE 2017; 5:273. [PMID: 28758099 DOI: 10.21037/atm.2017.06.22] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND Myelofibrosis (MF) is the most aggressive Philadelphia-negative chronic myeloproliferative neoplasm (MPN) with high morbidity and mortality due to thrombo-hemorrhagic complications and leukemic transformation. MF is characterized by profound alterations of megakaryocytopoiesis, with consequent abnormalities in platelet number and function. We recently showed that the overexpression of the oncoprotein PKCepsilon plays a key role in the aberrant differentiation of MF megakaryocyte clone and that its levels correlate with disease burden. Moreover, our group previously demonstrated that PKCepsilon is over-expressed in platelets from patients with acute myocardial infarction (MI) and accounts for their increased reactivity. On these bases, we investigated here the activation state and PKCepsilon expression of MF platelets, testing potential correlations with thrombotic risk and disease aggressiveness. METHODS Platelets were isolated from peripheral blood samples of MF patients and healthy donors (HDs). Patients were stratified according to the IPSS/DIPSS risk category and history of cardiovascular events. Platelet activation was assessed by flow cytometry. PKCepsilon mRNA and protein levels were determined by real time-PCR and western blot. RESULTS MF platelets circulate in an activated status and display significantly higher levels of PKCepsilon compared to HDs. In MF patients, PKCepsilon platelet levels were associated with high-risk disease as well as with a positive history of major cardiovascular events. CONCLUSIONS PKCepsilon is configuring as the common denominator of neoplastic transformation and thrombus formation in MF. Overall, our data pinpoint PKCepsilon as a potential novel biomarker of disease aggressiveness and thrombotic risk in this hematologic neoplasm.
Collapse
Affiliation(s)
- Elena Masselli
- Department of Medicine and Surgery, University of Parma, Ospedale Maggiore, Parma 43126, Italy
| | - Cecilia Carubbi
- Department of Medicine and Surgery, University of Parma, Ospedale Maggiore, Parma 43126, Italy
| | - Giulia Pozzi
- Department of Medicine and Surgery, University of Parma, Ospedale Maggiore, Parma 43126, Italy
| | - Silvia Martini
- Department of Medicine and Surgery, University of Parma, Ospedale Maggiore, Parma 43126, Italy
| | - Franco Aversa
- Hematology and BMT Center, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| | - Daniela Galli
- Department of Medicine and Surgery, University of Parma, Ospedale Maggiore, Parma 43126, Italy
| | - Giuliana Gobbi
- Department of Medicine and Surgery, University of Parma, Ospedale Maggiore, Parma 43126, Italy
| | - Prisco Mirandola
- Department of Medicine and Surgery, University of Parma, Ospedale Maggiore, Parma 43126, Italy
| | - Marco Vitale
- Department of Medicine and Surgery, University of Parma, Ospedale Maggiore, Parma 43126, Italy.,CoreLab, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| |
Collapse
|
22
|
Unsworth AJ, Bye AP, Gibbins JM. Platelet-Derived Inhibitors of Platelet Activation. PLATELETS IN THROMBOTIC AND NON-THROMBOTIC DISORDERS 2017. [PMCID: PMC7123044 DOI: 10.1007/978-3-319-47462-5_37] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
23
|
Affiliation(s)
- Amnon Altman
- Division of Cell Biology, La Jolla Institute for Allergy and Immunology, La Jolla, California 92037; ,
| | - Kok-Fai Kong
- Division of Cell Biology, La Jolla Institute for Allergy and Immunology, La Jolla, California 92037; ,
| |
Collapse
|
24
|
Zaid Y, Senhaji N, Naya A, Fadainia C, Kojok K. PKCs in thrombus formation. ACTA ACUST UNITED AC 2015; 63:268-71. [PMID: 26476932 DOI: 10.1016/j.patbio.2015.09.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2015] [Accepted: 09/01/2015] [Indexed: 10/22/2022]
Abstract
The protein kinase C (PKC) family has been implicated in several physiological processes regulating platelet activation. Each isoform of PKC expressed on platelets, may have a positive and/or negative role depending on the nature and concentration of the agonist. Mice lacking PKCα show much reduced thrombus formation in vivo, while PKCθ(-/-) showed inhibition of aggregation in response to PAR4. On the other hand, PKCδ by associating with Fyn, inhibits platelet aggregation. In addition, PKCβ by interacting with its receptor RACK1 has been implicated in the primary phases of signaling via the αIIbβ3 and finally PKCɛ appears to be involved in platelet function downstream GPVI. The present review discusses the latest observations relevant to the role of individual PKC isoforms in platelet activation and thrombus formation.
Collapse
Affiliation(s)
- Y Zaid
- Laboratory of Thrombosis and Hemostasis, Montreal Heart Institute, 5000 Belanger, Montreal, H1T 1C8 Quebec, Canada.
| | - N Senhaji
- Laboratory of Genetic and Molecular Pathology (LGPM), Medical School, Hassan II University, Casablanca, Morocco
| | - A Naya
- Laboratory of Physiology and Molecular Genetic, Faculty of Sciences, Hassan II University, Casablanca, Morocco
| | - C Fadainia
- Laboratory of Thrombosis and Hemostasis, Montreal Heart Institute, 5000 Belanger, Montreal, H1T 1C8 Quebec, Canada
| | - K Kojok
- Laboratory of Thrombosis and Hemostasis, Montreal Heart Institute, 5000 Belanger, Montreal, H1T 1C8 Quebec, Canada
| |
Collapse
|
25
|
Xiang B, Zhang G, Ye S, Zhang R, Huang C, Liu J, Tao M, Ruan C, Smyth SS, Whiteheart SW, Li Z. Characterization of a Novel Integrin Binding Protein, VPS33B, Which Is Important for Platelet Activation and In Vivo Thrombosis and Hemostasis. Circulation 2015; 132:2334-44. [PMID: 26399659 DOI: 10.1161/circulationaha.115.018361] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2015] [Accepted: 09/08/2015] [Indexed: 11/16/2022]
Abstract
BACKGROUND Integrins are heterodimeric (α/β) membrane proteins that play fundamental roles in many biological processes, for example, cell adhesion and spreading, which are important for platelet function and hemostasis. The molecular mechanism that regulates integrin activation is not completely understood. METHODS AND RESULTS Here, we show that VPS33B, a member of the Sec1/Munc18 family, binds directly to the integrin β subunit. Overexpression of VPS33B in Chinese hamster ovary cells potentiated αIIbβ3 outside-in signaling but not inside-out signaling. Platelets, from megakaryocyte- and platelet-specific VPS33B conditional knockout mice, had normal morphology, yet their spreading on fibrinogen was impaired and they failed to support clot retraction. Platelet aggregation and ATP secretion in response to low-dose agonists were reduced in the VPS33B knockout mice. αIIbβ3-mediated endocytosis of fibrinogen was also defective. Tail bleeding times and times to occlusion in an FeCl3-induced thrombosis model were prolonged in the VPS33B knockout mice. Furthermore, VPS33B acted upstream of the RhoA-ROCK-MLC and Rac1-dependent pathways that lead to clot retraction and cell spreading, respectively. CONCLUSIONS Our work demonstrates that vesicular trafficking complexes, containing VPS33B, are a novel class of modifiers of integrin function. Our data also provide insights into the molecular mechanism and treatment of arthrogryposis, renal dysfunction, and cholestasis syndrome.
Collapse
Affiliation(s)
- Binggang Xiang
- From Division of Cardiovascular Medicine, Saha Cardiovascular Research Center (B.X., G.Z., S.Y., R.Z., S.S.S., Z.L.), Department of Molecular and Cellular Biochemistry (S.Y., S.W.W.), and Markey Cancer Center (C.H.), University of Kentucky, Lexington; Department of Biochemistry and Molecular Biology, Mayo Clinic in Arizona, Scottsdale (J.L.); Department of Oncology (M.T.) and Jiangsu Institute of Hematology, Department of Hematology (C.R.), First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China; and Lexington Veterans Affairs Medical Center, Lexington, KY (S.S.S.)
| | - Guoying Zhang
- From Division of Cardiovascular Medicine, Saha Cardiovascular Research Center (B.X., G.Z., S.Y., R.Z., S.S.S., Z.L.), Department of Molecular and Cellular Biochemistry (S.Y., S.W.W.), and Markey Cancer Center (C.H.), University of Kentucky, Lexington; Department of Biochemistry and Molecular Biology, Mayo Clinic in Arizona, Scottsdale (J.L.); Department of Oncology (M.T.) and Jiangsu Institute of Hematology, Department of Hematology (C.R.), First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China; and Lexington Veterans Affairs Medical Center, Lexington, KY (S.S.S.)
| | - Shaojing Ye
- From Division of Cardiovascular Medicine, Saha Cardiovascular Research Center (B.X., G.Z., S.Y., R.Z., S.S.S., Z.L.), Department of Molecular and Cellular Biochemistry (S.Y., S.W.W.), and Markey Cancer Center (C.H.), University of Kentucky, Lexington; Department of Biochemistry and Molecular Biology, Mayo Clinic in Arizona, Scottsdale (J.L.); Department of Oncology (M.T.) and Jiangsu Institute of Hematology, Department of Hematology (C.R.), First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China; and Lexington Veterans Affairs Medical Center, Lexington, KY (S.S.S.)
| | - Rui Zhang
- From Division of Cardiovascular Medicine, Saha Cardiovascular Research Center (B.X., G.Z., S.Y., R.Z., S.S.S., Z.L.), Department of Molecular and Cellular Biochemistry (S.Y., S.W.W.), and Markey Cancer Center (C.H.), University of Kentucky, Lexington; Department of Biochemistry and Molecular Biology, Mayo Clinic in Arizona, Scottsdale (J.L.); Department of Oncology (M.T.) and Jiangsu Institute of Hematology, Department of Hematology (C.R.), First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China; and Lexington Veterans Affairs Medical Center, Lexington, KY (S.S.S.)
| | - Cai Huang
- From Division of Cardiovascular Medicine, Saha Cardiovascular Research Center (B.X., G.Z., S.Y., R.Z., S.S.S., Z.L.), Department of Molecular and Cellular Biochemistry (S.Y., S.W.W.), and Markey Cancer Center (C.H.), University of Kentucky, Lexington; Department of Biochemistry and Molecular Biology, Mayo Clinic in Arizona, Scottsdale (J.L.); Department of Oncology (M.T.) and Jiangsu Institute of Hematology, Department of Hematology (C.R.), First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China; and Lexington Veterans Affairs Medical Center, Lexington, KY (S.S.S.)
| | - Jun Liu
- From Division of Cardiovascular Medicine, Saha Cardiovascular Research Center (B.X., G.Z., S.Y., R.Z., S.S.S., Z.L.), Department of Molecular and Cellular Biochemistry (S.Y., S.W.W.), and Markey Cancer Center (C.H.), University of Kentucky, Lexington; Department of Biochemistry and Molecular Biology, Mayo Clinic in Arizona, Scottsdale (J.L.); Department of Oncology (M.T.) and Jiangsu Institute of Hematology, Department of Hematology (C.R.), First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China; and Lexington Veterans Affairs Medical Center, Lexington, KY (S.S.S.)
| | - Min Tao
- From Division of Cardiovascular Medicine, Saha Cardiovascular Research Center (B.X., G.Z., S.Y., R.Z., S.S.S., Z.L.), Department of Molecular and Cellular Biochemistry (S.Y., S.W.W.), and Markey Cancer Center (C.H.), University of Kentucky, Lexington; Department of Biochemistry and Molecular Biology, Mayo Clinic in Arizona, Scottsdale (J.L.); Department of Oncology (M.T.) and Jiangsu Institute of Hematology, Department of Hematology (C.R.), First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China; and Lexington Veterans Affairs Medical Center, Lexington, KY (S.S.S.)
| | - Changgeng Ruan
- From Division of Cardiovascular Medicine, Saha Cardiovascular Research Center (B.X., G.Z., S.Y., R.Z., S.S.S., Z.L.), Department of Molecular and Cellular Biochemistry (S.Y., S.W.W.), and Markey Cancer Center (C.H.), University of Kentucky, Lexington; Department of Biochemistry and Molecular Biology, Mayo Clinic in Arizona, Scottsdale (J.L.); Department of Oncology (M.T.) and Jiangsu Institute of Hematology, Department of Hematology (C.R.), First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China; and Lexington Veterans Affairs Medical Center, Lexington, KY (S.S.S.)
| | - Susan S Smyth
- From Division of Cardiovascular Medicine, Saha Cardiovascular Research Center (B.X., G.Z., S.Y., R.Z., S.S.S., Z.L.), Department of Molecular and Cellular Biochemistry (S.Y., S.W.W.), and Markey Cancer Center (C.H.), University of Kentucky, Lexington; Department of Biochemistry and Molecular Biology, Mayo Clinic in Arizona, Scottsdale (J.L.); Department of Oncology (M.T.) and Jiangsu Institute of Hematology, Department of Hematology (C.R.), First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China; and Lexington Veterans Affairs Medical Center, Lexington, KY (S.S.S.)
| | - Sidney W Whiteheart
- From Division of Cardiovascular Medicine, Saha Cardiovascular Research Center (B.X., G.Z., S.Y., R.Z., S.S.S., Z.L.), Department of Molecular and Cellular Biochemistry (S.Y., S.W.W.), and Markey Cancer Center (C.H.), University of Kentucky, Lexington; Department of Biochemistry and Molecular Biology, Mayo Clinic in Arizona, Scottsdale (J.L.); Department of Oncology (M.T.) and Jiangsu Institute of Hematology, Department of Hematology (C.R.), First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China; and Lexington Veterans Affairs Medical Center, Lexington, KY (S.S.S.)
| | - Zhenyu Li
- From Division of Cardiovascular Medicine, Saha Cardiovascular Research Center (B.X., G.Z., S.Y., R.Z., S.S.S., Z.L.), Department of Molecular and Cellular Biochemistry (S.Y., S.W.W.), and Markey Cancer Center (C.H.), University of Kentucky, Lexington; Department of Biochemistry and Molecular Biology, Mayo Clinic in Arizona, Scottsdale (J.L.); Department of Oncology (M.T.) and Jiangsu Institute of Hematology, Department of Hematology (C.R.), First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China; and Lexington Veterans Affairs Medical Center, Lexington, KY (S.S.S.)
| |
Collapse
|
26
|
Lesyk G, Fong T, Ruvolo PP, Jurasz P. The potential of enzastaurin to enhance platelet aggregation and growth factor secretion: implications for cancer cell survival. J Thromb Haemost 2015; 13:1514-20. [PMID: 25990653 DOI: 10.1111/jth.13010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Accepted: 05/13/2015] [Indexed: 11/30/2022]
Abstract
BACKGROUND Enzastaurin is a protein kinase C (PKC)β inhibitor with antiproliferative and proapoptotic effects that was in clinical development for the treatment of a variety of cancers. However, the primary endpoints in several clinical trials of enzastaurin were not met, and thrombosis was reported as an adverse effect in some trials. While investigating the role of PKC in regulating growth factor release from platelets, we found that, unlike other PKC inhibitors, enzastaurin may potentiate platelet aggregation. OBJECTIVE To investigate the effects of enzastaurin on platelet aggregation, growth factor secretion from α-granules and cancer cell apoptosis in the presence of platelets. METHODS Prostacyclin-washed platelets and platelet-rich plasma were isolated from the blood of healthy human volunteers. Platelet light-aggregometry was performed in the presence and absence of enzastaurin and acetylsalicylic acid (ASA). P-selectin was measured by flow cytometry, and vascular endothelial growth factor (VEGF) release was measured by ELISA. A549 lung carcinoma cells were treated with releasates from enzastaurin-titrated platelets. A cell death ELISA was performed to measure A549 apoptosis. RESULTS AND CONCLUSIONS Enzastaurin (10(-8) -10(-6) m) potentiated aggregation of prostacyclin-washed platelets and caused an increase in VEGF release from α-granules that, in turn, promoted cancer cell survival. In platelet-rich plasma, 10(-6) m enzastaurin inhibited platelet aggregation, but not 10(-7) m enzastaurin, which also failed to suppress VEGF secretion. ASA abrogated enzastaurin-potentiated washed-platelet aggregation and VEGF release. These findings indicate that, at high plasma protein-free drug concentrations, enzastaurin potentiates platelet aggregation and growth factor secretion, an effect that may counteract its anticancer activity. ASA nullifies this effect.
Collapse
Affiliation(s)
- G Lesyk
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
- Cardiovascular Research Centre, University of Alberta, Edmonton, AB, Canada
| | - T Fong
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - P P Ruvolo
- Department of Leukemia Research, MD Anderson Cancer Center, University of Texas, Houston, TX, USA
| | - P Jurasz
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
- Cardiovascular Research Centre, University of Alberta, Edmonton, AB, Canada
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
27
|
Bialkowska K, Byzova TV, Plow EF. Site-specific phosphorylation of kindlin-3 protein regulates its capacity to control cellular responses mediated by integrin αIIbβ3. J Biol Chem 2015; 290:6226-42. [PMID: 25609252 DOI: 10.1074/jbc.m114.634436] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The contributions of integrins to cellular responses depend upon their activation, which is regulated by binding of proteins to their cytoplasmic tails. Kindlins are integrin cytoplasmic tail binding partners and are essential for optimal integrin activation, and kindlin-3 fulfills this role in hematopoietic cells. Here, we used human platelets and human erythroleukemia (HEL) cells, which express integrin αIIbβ3, to investigate whether phosphorylation of kindlin-3 regulates integrin activation. When HEL cells were stimulated with thrombopoietin or phorbol 12-myristate 13-acetate (PMA), αIIbβ3 became activated as evidenced by binding of an activation-specific monoclonal antibody and soluble fibrinogen, adherence and spreading on fibrinogen, colocalization of β3 integrin and kindlin-3 in focal adhesions, and enhanced β3 integrin-kindlin-3 association in immunoprecipitates. Kindlin-3 knockdown impaired adhesion and spreading on fibrinogen. Stimulation of HEL cells with agonists significantly increased kindlin-3 phosphorylation as detected by mass spectrometric sequencing. Thr(482) or Ser(484) was identified as a phosphorylation site, which resides in a sequence not conserved in kindlin-1 or kindlin-2. These same residues were phosphorylated in kindlin-3 when platelets were stimulated with thrombin. When expressed in HEL cells, T482A/S484A kindlin-3 decreased soluble ligand binding and cell spreading on fibrinogen compared with wild-type kindlin-3. A membrane-permeable peptide containing residues 476-485 of kindlin-3 was introduced into HEL cells and platelets; adhesion and spreading of both cell types were blunted compared with a scrambled control peptide. These data identify a role of kindlin-3 phosphorylation in integrin β3 activation and provide a basis for functional differences between kindlin-3 and the two other kindlin paralogs.
Collapse
Affiliation(s)
- Katarzyna Bialkowska
- From the Joseph J. Jacobs Center for Thrombosis and Vascular Biology, Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195
| | - Tatiana V Byzova
- From the Joseph J. Jacobs Center for Thrombosis and Vascular Biology, Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195
| | - Edward F Plow
- From the Joseph J. Jacobs Center for Thrombosis and Vascular Biology, Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195
| |
Collapse
|
28
|
Khatlani T, Pradhan S, Da Q, Gushiken FC, Bergeron AL, Langlois KW, Molkentin JD, Rumbaut RE, Vijayan KV. The β isoform of the catalytic subunit of protein phosphatase 2B restrains platelet function by suppressing outside-in αII b β3 integrin signaling. J Thromb Haemost 2014; 12:2089-101. [PMID: 25330904 PMCID: PMC4268338 DOI: 10.1111/jth.12761] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Accepted: 10/07/2014] [Indexed: 01/25/2023]
Abstract
BACKGROUND Calcium-dependent signaling mechanisms play a critical role in platelet activation. Unlike calcium-activated protease and kinase, the contribution of calcium-activated protein serine/threonine phosphatase in platelet activation is poorly understood. OBJECTIVE To assess the role of catalytic subunit of protein phosphatase 2B (PP2B) or calcineurin in platelet function. RESULTS Here, we showed that an increase in PP2B activity was associated with agonist-induced activation of human and murine platelets. Pharmacological inhibitors of the catalytic subunit of protein phosphatase 2B (PP2B-A) such as cyclosporine A or tacrolimus (FK506) potentiated aggregation of human platelets. Murine platelets lacking the β isoform of PP2B-A (PP2B-Aβ(-/-) ) displayed increased aggregation with low doses of agonist concentrations. Loss of PP2B-Aβ did not affect agonist-induced integrin αII b β3 inside-out signaling, but increased basal Src activation and outside-in αII b β3 signaling to p38 mitogen-activated protein kinase (MAPK), with a concomitant enhancement in platelet spreading on immobilized fibrinogen and greater fibrin clot retraction. Fibrinogen-induced increased p38 activation in PP2B-Aβ(-/-) platelets were blocked by Src inhibitor. Both PP2B-Aβ(-/-) platelets and PP2B-Aβ-depleted human embryonal kidney 293 αII b β3 cells displayed increased adhesion to immobilized fibrinogen. Filamin A, an actin crosslinking phosphoprotein that is known to associate with β3 , was dephosphorylated on Ser(2152) in fibrinogen-adhered wild-type but not in PP2B-Aβ(-/-) platelets. In a FeCl3 injury thrombosis model, PP2B-Aβ(-/-) mice showed decreased time to occlusion in the carotid artery. CONCLUSION These observations indicate that PP2B-Aβ by suppressing outside-in αII b β3 integrin signaling limits platelet response to vascular injury.
Collapse
Affiliation(s)
- Tanvir Khatlani
- Department of Medicine, Michael E. DeBakey VA Medical Center, Houston, TX 77030, USA
| | - Subhashree Pradhan
- Department of Medicine, Michael E. DeBakey VA Medical Center, Houston, TX 77030, USA
| | - Qi Da
- Department of Medicine, Michael E. DeBakey VA Medical Center, Houston, TX 77030, USA
| | - Francisca C. Gushiken
- Department of Medicine, Michael E. DeBakey VA Medical Center, Houston, TX 77030, USA
| | - Angela L. Bergeron
- Department of Medicine, Michael E. DeBakey VA Medical Center, Houston, TX 77030, USA
| | - Kimberly W. Langlois
- Department of Pediatrics, Molecular Physiology, Michael E. DeBakey VA Medical Center, Houston, TX 77030, USA
| | - Jeffery D. Molkentin
- Molecular Cardiovascular Biology Program, Howard Hughes Medical Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Rolando E. Rumbaut
- Department of Medicine, Michael E. DeBakey VA Medical Center, Houston, TX 77030, USA
- Department of Pediatrics, Molecular Physiology, Michael E. DeBakey VA Medical Center, Houston, TX 77030, USA
- Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey VA Medical Center, Houston, TX 77030, USA
| | - K. Vinod Vijayan
- Department of Medicine, Michael E. DeBakey VA Medical Center, Houston, TX 77030, USA
- Department of Pediatrics, Molecular Physiology, Michael E. DeBakey VA Medical Center, Houston, TX 77030, USA
- Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey VA Medical Center, Houston, TX 77030, USA
- Department of Biophysics, Baylor College of Medicine, Michael E. DeBakey VA Medical Center, Houston, TX 77030, USA
| |
Collapse
|
29
|
Omosigho NN, Swaminathan K, Plomann M, Müller-Taubenberger A, Noegel AA, Riyahi TY. The Dictyostelium discoideum RACK1 orthologue has roles in growth and development. Cell Commun Signal 2014; 12:37. [PMID: 24930026 PMCID: PMC4094278 DOI: 10.1186/1478-811x-12-37] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Accepted: 06/06/2014] [Indexed: 12/19/2022] Open
Abstract
Background The receptor for activated C-kinase 1 (RACK1) is a conserved protein belonging to the WD40 repeat family of proteins. It folds into a beta propeller with seven blades which allow interactions with many proteins. Thus it can serve as a scaffolding protein and have roles in several cellular processes. Results We identified the product of the Dictyostelium discoideum gpbB gene as the Dictyostelium RACK1 homolog. The protein is mainly cytosolic but can also associate with cellular membranes. DdRACK1 binds to phosphoinositides (PIPs) in protein-lipid overlay and liposome-binding assays. The basis of this activity resides in a basic region located in the extended loop between blades 6 and 7 as revealed by mutational analysis. Similar to RACK1 proteins from other organisms DdRACK1 interacts with G protein subunits alpha, beta and gamma as shown by yeast two-hybrid, pulldown, and immunoprecipitation assays. Unlike the Saccharomyces cerevisiae and Cryptococcus neoformans RACK1 proteins it does not appear to take over Gβ function in D. discoideum as developmental and other defects were not rescued in Gβ null mutants overexpressing GFP-DdRACK1. Overexpression of GFP-tagged DdRACK1 and a mutant version (DdRACK1mut) which carried a charge-reversal mutation in the basic region in wild type cells led to changes during growth and development. Conclusion DdRACK1 interacts with heterotrimeric G proteins and can through these interactions impact on processes specifically regulated by these proteins.
Collapse
Affiliation(s)
| | | | | | | | - Angelika A Noegel
- Institute of Biochemistry I, Medical Faculty, Center for Molecular Medicine Cologne (CMMC) and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Köln, Germany.
| | | |
Collapse
|
30
|
RACK1, a versatile hub in cancer. Oncogene 2014; 34:1890-8. [PMID: 24882575 DOI: 10.1038/onc.2014.127] [Citation(s) in RCA: 130] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Revised: 04/07/2014] [Accepted: 04/10/2014] [Indexed: 01/15/2023]
Abstract
RACK1 is a highly conserved intracellular adaptor protein with significant homology to Gβ and was originally identified as the anchoring protein for activated protein kinase C. In the past 20 years, the number of binding partners and validated cellular functions for RACK1 has increased, which facilitates clarification of its involvement in different biological events. In this review, we will focus on its role in cancer, summarizing its aberrant expression, pro- or anti-oncogenic effects and the underlying mechanisms in various cancers.
Collapse
|
31
|
Mruk DD, Xiao X, Lydka M, Li MWM, Bilinska B, Cheng CY. Intercellular adhesion molecule 1: recent findings and new concepts involved in mammalian spermatogenesis. Semin Cell Dev Biol 2013; 29:43-54. [PMID: 23942142 DOI: 10.1016/j.semcdb.2013.07.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Revised: 07/26/2013] [Accepted: 07/29/2013] [Indexed: 01/05/2023]
Abstract
Spermatogenesis, the process of spermatozoa production, is regulated by several endocrine factors, including testosterone, follicle stimulating hormone, luteinizing hormone and estradiol 17β. For spermatogenesis to reach completion, developing germ cells must traverse the seminiferous epithelium while remaining transiently attached to Sertoli cells. If germ cell adhesion were to be compromised for a period of time longer than usual, germ cells would slough from the seminiferous epithelium and infertility would result. Presently, Sertoli-germ cell adhesion is known to be mediated largely by classical and desmosomal cadherins. More recent studies, however, have begun to expand long-standing concepts and to examine the roles of other proteins such as intercellular adhesion molecules. In this review, we focus on the biology of intercellular adhesion molecules in the mammalian testis, hoping that this information is useful in the design of future studies.
Collapse
Affiliation(s)
- Dolores D Mruk
- Center for Biomedical Research, Population Council, 1230 York Avenue, New York, NY 10065, United States.
| | - Xiang Xiao
- Center for Biomedical Research, Population Council, 1230 York Avenue, New York, NY 10065, United States
| | - Marta Lydka
- Center for Biomedical Research, Population Council, 1230 York Avenue, New York, NY 10065, United States
| | - Michelle W M Li
- Center for Biomedical Research, Population Council, 1230 York Avenue, New York, NY 10065, United States
| | - Barbara Bilinska
- Institute of Zoology, Department of Endocrinology, The Jagiellonian University, Gronostajowa 9, 30-387 Krakow, Poland
| | - C Yan Cheng
- Center for Biomedical Research, Population Council, 1230 York Avenue, New York, NY 10065, United States
| |
Collapse
|
32
|
RACK1 to the future--a historical perspective. Cell Commun Signal 2013; 11:53. [PMID: 23915285 PMCID: PMC3750812 DOI: 10.1186/1478-811x-11-53] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Accepted: 07/26/2013] [Indexed: 12/18/2022] Open
Abstract
This perspective summarises the first and long overdue RACK1 meeting held at the University of Limerick, Ireland, May 2013, in which RACK1's role in the immune system, the heart and the brain were discussed and its contribution to disease states such as cancer, cardiac hypertrophy and addiction were described. RACK1 is a scaffolding protein and a member of the WD repeat family of proteins. These proteins have a unique architectural assembly that facilitates protein anchoring and the stabilisation of protein activity. A large body of evidence is accumulating which is helping to define the versatile role of RACK1 in assembling and dismantling complex signaling pathways from the cell membrane to the nucleus in health and disease. In this commentary, we first provide a historical perspective on RACK1. We also address many of the pertinent and topical questions about this protein such as its role in transcription, epigenetics and translation, its cytoskeletal contribution and the merits of targeting RACK1 in disease.
Collapse
|
33
|
di Giacomo V, Pacella S, Rapino M, Di Giulio M, Zara S, Pasquantonio G, Cellini L, Cataldi A. pPKC α regulates through integrin β 1 human gingival fibroblasts/Streptococcus mitisadhesion in response to HEMA. Int Endod J 2013; 46:1164-72. [DOI: 10.1111/iej.12113] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2012] [Accepted: 03/27/2013] [Indexed: 01/17/2023]
Affiliation(s)
- V. di Giacomo
- Dipartimento di Farmacia; Università G. d'Annunzio; Chieti-Pescara Italy
| | - S. Pacella
- Dipartimento di Medicina e Scienze dell'Invecchiamento; Università G. d'Annunzio; Chieti-Pescara Italy
| | - M. Rapino
- Istituto di Genetica Molecolare del CNR; Unità di Chieti; Chieti; Italy
| | - M. Di Giulio
- Dipartimento di Farmacia; Università G. d'Annunzio; Chieti-Pescara Italy
| | - S. Zara
- Dipartimento di Farmacia; Università G. d'Annunzio; Chieti-Pescara Italy
| | - G. Pasquantonio
- Dipartimento di Materiali Dentali e Tecnologie Prostodontiche; Università di Roma Tor Vergata; Roma Italy
| | - L. Cellini
- Dipartimento di Farmacia; Università G. d'Annunzio; Chieti-Pescara Italy
| | - A. Cataldi
- Dipartimento di Farmacia; Università G. d'Annunzio; Chieti-Pescara Italy
| |
Collapse
|
34
|
Moncada de la Rosa C, Radziwon-Balicka A, El-Sikhry H, Seubert J, Ruvolo PP, Radomski MW, Jurasz P. Pharmacologic Protein Kinase CαInhibition Uncouples Human Platelet-Stimulated Angiogenesis from Collagen-Induced Aggregation. J Pharmacol Exp Ther 2013; 345:15-24. [DOI: 10.1124/jpet.112.200881] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
|
35
|
Allison GL, Lowe GM, Rahman K. Aged garlic extract inhibits platelet activation by increasing intracellular cAMP and reducing the interaction of GPIIb/IIIa receptor with fibrinogen. Life Sci 2012; 91:1275-80. [PMID: 23069586 DOI: 10.1016/j.lfs.2012.09.019] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2012] [Revised: 08/15/2012] [Accepted: 09/26/2012] [Indexed: 11/16/2022]
Abstract
AIMS Increased platelet aggregation plays an important role in the etiology of cardiovascular disease. Garlic inhibits platelet aggregation; however, the mechanisms involved have not clearly been defined. This study was undertaken to investigate the mechanisms by which an aged garlic extract (AGE) inhibits both the activation and aggregation of human platelets. MAIN METHODS Isolated human platelets were stimulated with ADP and their adhesion to fibrinogen was assessed using Rose Bengal or (51)Cr uptake. Activation of platelets was assessed using fluorescence activated cell sorting (FACS) analysis along with measurement of intracellular cAMP. KEY FINDINGS AGE at concentrations in the range of 3.12 to 12.5% (v/v) inhibited the binding of platelets to fibrinogen by approximately 40% when compared to control values in the Rose Bengal assay (P<0.05). In the (51)Cr experiments AGE significantly inhibited the binding of ADP-activated platelets to immobilized fibrinogen by 61.5% at 1.56% and 6.25% (v/v) of AGE respectively. At a concentration of 12.5% (v/v) the inhibition was 70.4% and at 25% (v/v) it was 64.5% respectively (P<0.05). In the fluorescence activated cell sorting (FACS) analysis, AGE significantly decreased the amount of PAC-1 binding to GPIIb/IIIa by approximately 72% compared with PBS control. In conjunction to these observations, AGE also increased platelet cAMP (P<0.01) levels. SIGNIFICANCE These findings suggest that AGE inhibits platelet aggregation via inhibition of the GPIIb/IIIa receptor and an increase in cAMP.
Collapse
Affiliation(s)
- Gillian L Allison
- School of Pharmacy, Faculty of Medical Sciences, The University of the West Indies, Eric Williams Medical Sciences Complex, Champ Fleurs, Trinidad and Tobago
| | | | | |
Collapse
|
36
|
Carubbi C, Mirandola P, Mattioli M, Galli D, Marziliano N, Merlini PA, Lina D, Notarangelo F, Cozzi MR, Gesi M, Ardissino D, De Marco L, Vitale M, Gobbi G. Protein kinase C ε expression in platelets from patients with acute myocardial infarction. PLoS One 2012; 7:e46409. [PMID: 23071564 PMCID: PMC3465320 DOI: 10.1371/journal.pone.0046409] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2012] [Accepted: 08/29/2012] [Indexed: 01/16/2023] Open
Abstract
Objective Platelets play crucial roles in the pathophysiology of thrombosis and myocardial infarction. Protein kinase C ε (PKCε) is virtually absent in human platelets and its expression is precisely regulated during human megakaryocytic differentiation. On the basis of what is known on the role of platelet PKCε in other species, we hypothesized that platelets from myocardial infarction patients might ectopically express PKCε with a pathophysiological role in the disease. Methods and Results We therefore studied platelet PKCε expression from 24 patients with myocardial infarction, 24 patients with stable coronary artery disease and 24 healthy subjects. Indeed, platelets from myocardial infarction patients expressed PKCε with a significant frequency as compared to both stable coronary artery disease and healthy subjects. PKCε returned negative during patient follow-up. The forced expression of PKCε in normal donor platelets significantly increased their response to adenosine diphosphate-induced activation and adhesion to subendothelial collagen. Conclusions Our data suggest that platelet generations produced before the acute event retain PKCε-mRNA that is not down-regulated during terminal megakaryocyte differentiation. Results are discussed in the perspective of peri-infarctual megakaryocytopoiesis as a critical component of myocardial infarction pathophysiology.
Collapse
Affiliation(s)
- Cecilia Carubbi
- Department of Biomedical, Biotechnological and Translational Sciences, University of Parma, Parma, Italy
| | - Prisco Mirandola
- Department of Biomedical, Biotechnological and Translational Sciences, University of Parma, Parma, Italy
| | - Maria Mattioli
- Department of Biomedical, Biotechnological and Translational Sciences, University of Parma, Parma, Italy
| | - Daniela Galli
- Department of Biomedical, Biotechnological and Translational Sciences, University of Parma, Parma, Italy
| | | | | | - Daniela Lina
- Division of Cardiology, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| | | | - Maria Rita Cozzi
- Department of Laboratory Medicine, CRO National Cancer Institute, Aviano, Italy
| | - Marco Gesi
- Department of Human Morphology and Applied Biology, University of Pisa, Pisa, Italy
| | - Diego Ardissino
- Division of Cardiology, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| | - Luigi De Marco
- Department of Laboratory Medicine, CRO National Cancer Institute, Aviano, Italy
| | - Marco Vitale
- Department of Biomedical, Biotechnological and Translational Sciences, University of Parma, Parma, Italy
- * E-mail:
| | - Giuliana Gobbi
- Department of Biomedical, Biotechnological and Translational Sciences, University of Parma, Parma, Italy
| |
Collapse
|
37
|
UNSWORTH AJ, FINNEY BA, NAVARRO-NUNEZ L, SEVERIN S, WATSON SP, PEARS CJ. Protein kinase Cε and protein kinase Cθ double-deficient mice have a bleeding diathesis. J Thromb Haemost 2012; 10:1887-94. [PMID: 22812584 PMCID: PMC3532618 DOI: 10.1111/j.1538-7836.2012.04857.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2012] [Accepted: 07/10/2012] [Indexed: 01/24/2023]
Abstract
BACKGROUND In comparison to the classical isoforms of protein kinase C (PKC), the novel isoforms are thought to play minor or inhibitory roles in the regulation of platelet activation and thrombosis. OBJECTIVES To measure the levels of PKCθ and PKCε and to investigate the phenotype of mice deficient in both novel PKC isoforms. METHODS Tail bleeding and platelet activation assays were monitored in mice and platelets from mice deficient in both PKCθ and PKCε. RESULTS PKCε plays a minor role in supporting aggregation and secretion following stimulation of the collagen receptor GPVI in mouse platelets but has no apparent role in spreading on fibrinogen. PKCθ, in contrast, plays a minor role in supporting adhesion and filopodial generation on fibrinogen but has no apparent role in aggregation and secretion induced by GPVI despite being expressed at over 10 times the level of PKCε. Platelets deficient in both novel isoforms have a similar pattern of aggregation downstream of GPVI and spreading on fibrinogen as the single null mutants. Strikingly, a marked reduction in aggregation on collagen under arteriolar shear conditions is observed in blood from the double but not single-deficient mice along with a significant increase in tail bleeding. CONCLUSIONS These results reveal a greater than additive role for PKCθ and PKCε in supporting platelet activation under shear conditions and demonstrate that, in combination, the two novel PKCs support platelet activation.
Collapse
Affiliation(s)
- A J UNSWORTH
- Department of Biochemistry, University of OxfordOxford
| | - B A FINNEY
- Centre for Cardiovascular Sciences, Institute of Biomedical Research, School of Clinical and Experimental Medicine, College of Medical and Dental Sciences, University of BirminghamBirmingham, UK
| | - L NAVARRO-NUNEZ
- Centre for Cardiovascular Sciences, Institute of Biomedical Research, School of Clinical and Experimental Medicine, College of Medical and Dental Sciences, University of BirminghamBirmingham, UK
| | - S SEVERIN
- Centre for Cardiovascular Sciences, Institute of Biomedical Research, School of Clinical and Experimental Medicine, College of Medical and Dental Sciences, University of BirminghamBirmingham, UK
| | - S P WATSON
- Centre for Cardiovascular Sciences, Institute of Biomedical Research, School of Clinical and Experimental Medicine, College of Medical and Dental Sciences, University of BirminghamBirmingham, UK
| | - C J PEARS
- Department of Biochemistry, University of OxfordOxford
| |
Collapse
|
38
|
The leucocyte β2 (CD18) integrins: the structure, functional regulation and signalling properties. Biosci Rep 2012; 32:241-69. [PMID: 22458844 DOI: 10.1042/bsr20110101] [Citation(s) in RCA: 126] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Leucocytes are highly motile cells. Their ability to migrate into tissues and organs is dependent on cell adhesion molecules. The integrins are a family of heterodimeric transmembrane cell adhesion molecules that are also signalling receptors. They are involved in many biological processes, including the development of metazoans, immunity, haemostasis, wound healing and cell survival, proliferation and differentiation. The leucocyte-restricted β2 integrins comprise four members, namely αLβ2, αMβ2, αXβ2 and αDβ2, which are required for a functional immune system. In this paper, the structure, functional regulation and signalling properties of these integrins are reviewed.
Collapse
|
39
|
Horn M, Bertling A, Brodde MF, Müller A, Roth J, Van Aken H, Jurk K, Heilmann C, Peters G, Kehrel BE. Human neutrophil alpha-defensins induce formation of fibrinogen and thrombospondin-1 amyloid-like structures and activate platelets via glycoprotein IIb/IIIa. J Thromb Haemost 2012; 10:647-61. [PMID: 22268819 DOI: 10.1111/j.1538-7836.2012.04640.x] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND Human neutrophil α-defensins (HNPs) are important constituents of the innate immune system. Beyond their antimicrobial properties, HNPs also have pro-inflammatory features. While HNPs in plasma from healthy individuals are barely detectable, their level is strongly elevated in septic plasma and plasma from patients with acute coronary syndromes. OBJECTIVES As thrombosis and inflammation are intertwined processes and activation of human polymorphonuclear leukocytes (PMNL) and subsequent degranulation is associated with full activation of surrounding platelets, we studied the effect of HNPs on platelet function. METHODS The effect of HNPs on platelet activation parameters and apoptosis was investigated via aggregometry, flow cytometry, confocal microscopy and the ELISA technique. RESULTS It was found that HNPs activate platelets in pathophysiologically relevant doses, inducing fibrinogen and thrombospondin-1 binding, aggregation, granule secretion, sCD40L shedding, and procoagulant activity. HNPs bound directly to the platelet membrane, induced membrane pore formation, microparticle formation, mitochondrial membrane depolarization and caspase-3-activity. Confocal microscopy revealed the HNP-induced formation of polymeric fibrinogen and thrombospondin-1 amyloid-like structures, which bound microorganisms. Platelets adhered to these structures and formed aggregates. Blocking of glycoprotein IIb/IIIa (GPIIb/IIIa) markedly inhibited HNP-induced platelet activation. In addition, heparin, heparinoid, serpins and α(2)-macroglobulin, which all bind to HNPs, blocked HNP-1-induced platelet activation in contrast to direct thrombin inhibitors such as hirudin. CONCLUSIONS HNPs activate platelets and induce platelet apoptosis by formation of amyloid-like proteins. As these structures entrapped bacteria and fungi, they might reflect an additional function of HNPs in host defense. The described mechanism links again thrombosis and infection.
Collapse
Affiliation(s)
- M Horn
- Department of Anaesthesiology and Intensive Care, Experimental and Clinical Haemostasis, University of Muenster, Muenster, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Bertram A, Zhang H, von Vietinghoff S, de Pablo C, Haller H, Shushakova N, Ley K. Protein kinase C-θ is required for murine neutrophil recruitment and adhesion strengthening under flow. THE JOURNAL OF IMMUNOLOGY 2012; 188:4043-51. [PMID: 22403440 DOI: 10.4049/jimmunol.1101651] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Protein kinase C (PKC)-θ is involved in T cell activation via regulating the avidity of the β(2) integrin LFA-1 in the immunological synapse. LFA-1 also mediates leukocyte adhesion. To investigate the role of PKC-θ in neutrophil adhesion, we performed intravital microscopy in cremaster venules of mice reconstituted with bone marrow from LysM-GFP(+) (wild-type [WT]) and PKC-θ gene-deficient (Prkcq(-/-)) mice. Following stimulation with CXCL1, both WT and Prkcq(-/-) cells became adherent. Although most WT neutrophils remained adherent for at least 180 s, 50% of Prkcq(-/-) neutrophils were detached after 105 s and most by 180 s. Upon CXCL1 injection, rolling of all WT neutrophils stopped for 90 s, but rolling of Prkcq(-/-) neutrophils started 30 s after CXCL1 stimulation. A similar neutrophil adhesion defect was seen in vitro, and spreading of Prkcq(-/-) neutrophils was delayed. Prkcq(-/-) neutrophil recruitment was impaired in fMLP-induced transmigration into the cremaster muscle, thioglycollate-induced peritonitis, and LPS-induced lung injury. We conclude that PKC-θ mediates integrin-dependent neutrophil functions and is required to sustain neutrophil adhesion in postcapillary venules in vivo. These findings suggest that the role of PKC-θ in outside-in signaling following engagement of neutrophil integrins is relevant for inflammation in vivo.
Collapse
Affiliation(s)
- Anna Bertram
- Department of Nephrology and Hypertensiology, Hannover Medical School, 30625 Hannover, Germany.
| | | | | | | | | | | | | |
Collapse
|
41
|
Bynagari-Settipalli YS, Lakhani P, Jin J, Bhavaraju K, Rico MC, Kim S, Woulfe D, Kunapuli SP. Protein kinase C isoform ε negatively regulates ADP-induced calcium mobilization and thromboxane generation in platelets. Arterioscler Thromb Vasc Biol 2012; 32:1211-9. [PMID: 22362759 DOI: 10.1161/atvbaha.111.242388] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
OBJECTIVE Members of the protein kinase C (PKC) family are shown to positively and negatively regulate platelet activation. Although positive regulatory roles are extensively studied, negative regulatory roles of PKCs are poorly understood. We investigated the mechanism and specific isoforms involved in PKC-mediated negative regulation of ADP-induced functional responses. METHODS AND RESULTS A pan-PKC inhibitor, GF109203X, potentiated ADP-induced cPLA(2) phosphorylation and thromboxane generation as well as ERK activation and intracellular calcium (Ca(2+)(i)) mobilization, 2 signaling molecules, upstream of cPLA(2) activation. Thus, PKCs inhibit cPLA(2) activation by inhibiting ERK and Ca(2+)(i) mobilization. Because the inhibitor of classic PKC isoforms, GO-6976, did not affect ADP-mediated thromboxane generation, we investigated the role of novel class of PKC isoforms. ADP-induced thromboxane generation, calcium mobilization, and ERK phosphorylation were potentiated in PKCε null murine platelets compared with platelets from wild-type littermates. Interestingly, when thromboxane release is blocked, ADP-induced aggregation in PKCε knockout and wild-type was similar, suggesting that PKCε does not affect ADP-induced aggregation directly. PKCε knockout mice exhibited shorter times to occlusion in an FeCl(3)-induced arterial injury model and shorter bleeding times in tail-bleeding experiments. CONCLUSIONS We conclude that PKCε negatively regulates ADP-induced thromboxane generation in platelets and offers protection against thrombosis.
Collapse
|
42
|
Feng C, Li YF, Yau YH, Lee HS, Tang XY, Xue ZH, Zhou YC, Lim WM, Cornvik TC, Ruedl C, Shochat SG, Tan SM. Kindlin-3 mediates integrin αLβ2 outside-in signaling, and it interacts with scaffold protein receptor for activated-C kinase 1 (RACK1). J Biol Chem 2012; 287:10714-26. [PMID: 22334666 DOI: 10.1074/jbc.m111.299594] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Integrins are heterodimeric type I membrane cell adhesion molecules that are involved in many biological processes. Integrins are bidirectional signal transducers because their cytoplasmic tails are docking sites for cytoskeletal and signaling molecules. Kindlins are cytoplasmic molecules that mediate inside-out signaling and activation of the integrins. The three kindlin paralogs in humans are kindlin-1, -2, and -3. Each of these contains a 4.1-ezrin-radixin-moesin (FERM) domain and a pleckstrin homology domain. Kindlin-3 is expressed in platelets, hematopoietic cells, and endothelial cells. Here we show that kindlin-3 is involved in integrin αLβ2 outside-in signaling. It also promotes micro-clustering of integrin αLβ2. We provide evidence that kindlin-3 interacts with the receptor for activated-C kinase 1 (RACK1), a scaffold protein that folds into a seven-blade propeller. This interaction involves the pleckstrin homology domain of kindlin-3 and blades 5-7 of RACK1. Using the SKW3 human T lymphoma cells, we show that integrin αLβ2 engagement by its ligand ICAM-1 promotes the association of kindlin-3 with RACK1. We also show that kindlin-3 co-localizes with RACK1 in polarized SKW3 cells and human T lymphoblasts. Our findings suggest that kindlin-3 plays an important role in integrin αLβ2 outside-in signaling.
Collapse
Affiliation(s)
- Chen Feng
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Goggs R, Poole AW. Platelet signaling-a primer. J Vet Emerg Crit Care (San Antonio) 2012; 22:5-29. [PMID: 22316389 DOI: 10.1111/j.1476-4431.2011.00704.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2011] [Accepted: 11/25/2011] [Indexed: 12/28/2022]
Abstract
OBJECTIVE To review the receptors and signal transduction pathways involved in platelet plug formation and to highlight links between platelets, leukocytes, endothelium, and the coagulation system. DATA SOURCES Original studies, review articles, and book chapters in the human and veterinary medical fields. DATA SYNTHESIS Platelets express numerous surface receptors. Critical among these are glycoprotein VI, the glycoprotein Ib-IX-V complex, integrin α(IIb) β(3) , and the G-protein-coupled receptors for thrombin, ADP, and thromboxane. Activation of these receptors leads to various important functional events, in particular activation of the principal adhesion receptor α(IIb) β(3) . Integrin activation allows binding of ligands such as fibrinogen, mediating platelet-platelet interaction in the process of aggregation. Signals activated by these receptors also couple to 3 other important functional events, secretion of granule contents, change in cell shape through cytoskeletal rearrangement, and procoagulant membrane expression. These processes generate a stable thrombus to limit blood loss and promote restoration of endothelial integrity. CONCLUSIONS Improvements in our understanding of how platelets operate through their signaling networks are critical for diagnosis of unusual primary hemostatic disorders and for rational antithrombotic drug design.
Collapse
Affiliation(s)
- Robert Goggs
- School of Physiology and Pharmacology, Faculty of Medical and Veterinary Sciences, University of Bristol, UK.
| | | |
Collapse
|
44
|
Abstract
Three classes of inhibitors of platelet aggregation have demonstrated substantial clinical benfits. Aspirin acts by irreversibly inhibiting COX-1 and therefore blocking the synthesis of proaggregatory thromboxane A (2) (TxA(2)). The indirect acting (ticlopidine, clopidogrel, prasugrel) and the direct acting (ticagrelor) antagonists of P2Y(12) block the thrombus stabilizing activity of ADP. Parenteral GP IIb-IIIa inhibitors directly block platelet-platelet interactions. Despite well-established benefits, all antiplatelet agents have important limitations: increased bleeding and gastrointestinal toxicities (aspirin), high incidence of thrombotic thrombocytopenic purpura (ticlopidine), potentially nonresponders (clopidogrel), severe bleeding (prasugrel, GP IIb-IIIa antagonists) and "complicated" relationships with aspirin ticagrelor). In this chapter, we present the genetic and pharmacological evidence that supports the development and expectations associated with novel antiplatelet strategies directed at intrasignaling pathways.
Collapse
Affiliation(s)
- Patrick Andre
- Portola Pharmaceuticals Inc, 270 E. Grand Avenue, Suite 22 South, San Francisco, CA 94080, USA.
| |
Collapse
|
45
|
Williams CM, Feng Y, Martin P, Poole AW. Protein kinase C alpha and beta are positive regulators of thrombus formation in vivo in a zebrafish (Danio rerio) model of thrombosis. J Thromb Haemost 2011; 9:2457-65. [PMID: 21951302 DOI: 10.1111/j.1538-7836.2011.04520.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
BACKGROUND The zebrafish (Danio rerio) is becoming an attractive model organism for the assessment of gene function in thrombosis in vivo. Zebrafish, as a thrombosis model, have several advantages, with the capacity to follow thrombus formation at high resolution in real time using intravital microscopy, without the need for complex surgical techniques, and the capability to rapidly knockdown gene expression using morpholino antisense approaches. OBJECTIVES We have recently shown, in mouse models, that protein kinase C alpha (PKCα) plays a critical role in regulating thrombus formation in vivo. PKC beta (β) plays a non-redundant role also in platelet function in vitro, but the function of this gene had not yet been assessed in vivo. METHODS In the present study, we analyzed the function of both PKCα and PKCβ in the zebrafish model in vivo, by live imaging using a laser-induced injury of the main caudal artery in 3-day-old larvae. RESULTS We showed that D. rerio express orthologs of both the PKCα and PKCβ genes, with high sequence identity. Translation blocking and splice-blocking morpholinos effectively and specifically knockdown expression of these genes and knockdown with either morpholino leads to attenuated thrombus formation, as assessed by several quantitative parameters including time to initial adhesion and peak thrombus surface area. CONCLUSIONS Our data indicate that these two highly related genes play non-redundant roles in regulating thrombosis, an observation that supports our previous in vitro murine data, and suggests unique roles, and possibly unique regulation, for PKCα and PKCβ in controlling platelet function in vivo.
Collapse
Affiliation(s)
- C M Williams
- School of Physiology & Pharmacology, University Walk, Bristol, UK
| | | | | | | |
Collapse
|
46
|
O’TOOLE TIMOTHYE, BIALKOWSKA KATARZYNA, LI XIAOHONG, FOX JOANE. Tiam1 is recruited to β1-integrin complexes by 14-3-3ζ where it mediates integrin-induced Rac1 activation and motility. J Cell Physiol 2011; 226:2965-78. [PMID: 21302295 PMCID: PMC6385608 DOI: 10.1002/jcp.22644] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
14-3-3 is an adaptor protein that localizes to the leading edge of spreading cells, returning to the cytoplasm as spreading ceases. Previously, we showed that integrin-induced Rac1 activation and spreading were inhibited by sequestration of 14-3-3ζ and restored by its overexpression. Here, we determined whether 14-3-3 mediates integrin signaling by localizing a guanine nucleotide exchange factor (GEF) to Rac1-activating integrin complexes. We showed that GST-14-3-3ζ recruited the Rac1-GEF, Tiam1, from cell lysates through Tiam1 residues 1-182 (N(1-182) Tiam1). The physiological relevance of this interaction was examined in serum-starved Hela cells plated on fibronectin. Both Tiam1 and N(1-182) Tiam1 were recruited to 14-3-3-containing β1-integrin complexes, as shown by co-localization and co-immunoprecipitation. Integrin-induced Rac1 activation was inhibited when Tiam1 was depleted with siRNA or by overexpression of catalytically inactive N(1-182) Tiam1, which was incorporated into 14-3-3/β1-integrin complexes and inhibited spreading in a manner that was overcome by constitutively active Rac1. Integrin-induced Rac1 activation, spreading, and migration were also inhibited by overexpression of 14-3-3ζ S58D, which was unable to recruit Tiam1 from lysates, co-immunoprecipitate with Tiam1, or mediate its incorporation into β1-integrin complexes. Taken together, these findings suggest a previously unrecognized mechanism of integrin-induced Rac1 activation in which 14-3-3 dimers localize Tiam1 to integrin complexes, where it mediates integrin-dependent Rac1 activation, thus initiating motility-inducing pathways. Moreover, since Tiam1 is recruited to other sites of localized Rac1 activation through its PH-CC-EX domain, the present findings show that a mechanism involving its N-terminal 182 residues is utilized to recruit Tiam1 to motility-inducing integrin complexes.
Collapse
Affiliation(s)
- TIMOTHY E. O’TOOLE
- Department of Molecular Cardiology, Joseph J. Jacobs Center for Thrombosis and Vascular Biology, The Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - KATARZYNA BIALKOWSKA
- Department of Molecular Cardiology, Joseph J. Jacobs Center for Thrombosis and Vascular Biology, The Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - XIAOHONG LI
- Department of Molecular Cardiology, Joseph J. Jacobs Center for Thrombosis and Vascular Biology, The Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - JOAN E.B. FOX
- Department of Molecular Cardiology, Joseph J. Jacobs Center for Thrombosis and Vascular Biology, The Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
- Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, Ohio
| |
Collapse
|
47
|
Adams DR, Ron D, Kiely PA. RACK1, A multifaceted scaffolding protein: Structure and function. Cell Commun Signal 2011; 9:22. [PMID: 21978545 PMCID: PMC3195729 DOI: 10.1186/1478-811x-9-22] [Citation(s) in RCA: 333] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2011] [Accepted: 10/06/2011] [Indexed: 12/17/2022] Open
Abstract
The Receptor for Activated C Kinase 1 (RACK1) is a member of the tryptophan-aspartate repeat (WD-repeat) family of proteins and shares significant homology to the β subunit of G-proteins (Gβ). RACK1 adopts a seven-bladed β-propeller structure which facilitates protein binding. RACK1 has a significant role to play in shuttling proteins around the cell, anchoring proteins at particular locations and in stabilising protein activity. It interacts with the ribosomal machinery, with several cell surface receptors and with proteins in the nucleus. As a result, RACK1 is a key mediator of various pathways and contributes to numerous aspects of cellular function. Here, we discuss RACK1 gene and structure and its role in specific signaling pathways, and address how posttranslational modifications facilitate subcellular location and translocation of RACK1. This review condenses several recent studies suggesting a role for RACK1 in physiological processes such as development, cell migration, central nervous system (CN) function and circadian rhythm as well as reviewing the role of RACK1 in disease.
Collapse
Affiliation(s)
- David R Adams
- Department of Life Sciences, and Materials and Surface Science Institute, University of Limerick, Limerick, Ireland.
| | | | | |
Collapse
|
48
|
Zhang G, Xiang B, Ye S, Chrzanowska-Wodnicka M, Morris AJ, Gartner TK, Whiteheart SW, White GC, Smyth SS, Li Z. Distinct roles for Rap1b protein in platelet secretion and integrin αIIbβ3 outside-in signaling. J Biol Chem 2011; 286:39466-77. [PMID: 21940635 DOI: 10.1074/jbc.m111.239608] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Rap1b is activated by platelet agonists and plays a critical role in integrin α(IIb)β(3) inside-out signaling and platelet aggregation. Here we show that agonist-induced Rap1b activation plays an important role in stimulating secretion of platelet granules. We also show that α(IIb)β(3) outside-in signaling can activate Rap1b, and integrin outside-in signaling-mediated Rap1b activation is important in facilitating platelet spreading on fibrinogen and clot retraction. Rap1b-deficient platelets had diminished ATP secretion and P-selectin expression induced by thrombin or collagen. Importantly, addition of low doses of ADP and/or fibrinogen restored aggregation of Rap1b-deficient platelets. Furthermore, we found that Rap1b was activated by platelet spreading on immobilized fibrinogen, a process that was not affected by P2Y(12) or TXA(2) receptor deficiency, but was inhibited by the selective Src inhibitor PP2, the PKC inhibitor Ro-31-8220, or the calcium chelator demethyl-1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid tetrakis. Clot retraction was abolished, and platelet spreading on fibrinogen was diminished in Rap1b-deficient platelets compared with wild-type controls. The defects in clot retraction and spreading on fibrinogen of Rap1b-deficient platelets were not rescued by addition of MnCl(2), which elicits α(IIb)β(3) outside-in signaling in the absence of inside-out signaling. Thus, our results reveal two different activation mechanisms of Rap1b as well as novel functions of Rap1b in platelet secretion and in integrin α(IIb)β(3) outside-in signaling.
Collapse
Affiliation(s)
- Guoying Zhang
- Division of Cardiovascular Medicine, The Gill Heart Institute, University of Kentucky, Lexington, Kentucky 40536, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Protein kinase C-theta in platelet activation. FEBS Lett 2011; 585:3208-15. [DOI: 10.1016/j.febslet.2011.09.014] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2011] [Revised: 08/20/2011] [Accepted: 09/12/2011] [Indexed: 02/05/2023]
|
50
|
Protease-activated receptor 1 (PAR1) signalling desensitization is counteracted via PAR4 signalling in human platelets. Biochem J 2011; 436:469-80. [PMID: 21391917 DOI: 10.1042/bj20101360] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
PARs (protease-activated receptors) 1 and 4 belong to the family of G-protein-coupled receptors which induce both G(α12/13) and G(αq) signalling. By applying the specific PAR1- and PAR4-activating hexapeptides, SFLLRN and AYPGKF respectively, we found that aggregation of isolated human platelets mediated via PAR1, but not via PAR4, is abolished upon homologous receptor activation in a concentration- and time-dependent fashion. This effect was not due to receptor internalization, but to a decrease in Ca²⁺ mobilization, PKC (protein kinase C) signalling and α-granule secretion, as well as to a complete lack of dense granule secretion. Interestingly, subthreshold PAR4 activation rapidly abrogated PAR1 signalling desensitization by differentially reconstituting these affected signalling events and functional responses, which was sufficient to re-establish aggregation. The lack of ADP release and P2Y₁₂ receptor-induced G(αi) signalling accounted for the loss of the aggregation response, as mimicking G(αi/z) signalling with 2-MeS-ADP (2-methylthioadenosine-5'-O-diphosphate) or epinephrine (adrenaline) could substitute for intermediate PAR4 activation. Finally, we found that the re-sensitization of PAR1 signalling-induced aggregation via PAR4 relied on PKC-mediated release of both ADP from dense granules and fibrinogen from α-granules. The present study elucidates further differences in human platelet PAR signalling regulation and provides evidence for a cross-talk in which PAR4 signalling counteracts mechanisms involved in PAR1 signalling down-regulation.
Collapse
|