1
|
Zhang LJ, Hu YX, Huang RZ, Xu YY, Dong SH, Guo FH, Guo JJ, Qiu JJ, Cao ZY, Wei LJ, Mao JH, Lyu A, Liu JL, Zhao XX, Guo ZF, Jing Q. Intraplatelet miRNA-126 regulates thrombosis and its reduction contributes to platelet inhibition. Cardiovasc Res 2024; 120:1622-1635. [PMID: 38900927 DOI: 10.1093/cvr/cvae138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 07/17/2023] [Accepted: 01/08/2024] [Indexed: 06/22/2024] Open
Abstract
AIMS MicroRNA-126 (miR-126), one of the most abundant microRNAs in platelets, is involved in the regulation of platelet activity and the circulating miR-126 is reduced during antiplatelet therapy. However, whether intraplatelet miR-126 plays a role in thrombosis and platelet inhibition remains unclear. METHODS AND RESULTS Here, using tissue-specific knockout mice, we reported that the deficiency of miR-126 in platelets and vascular endothelial cells significantly prevented thrombosis and prolonged bleeding time. Using chimeric mice, we identified that the lack of intraplatelet miR-126 significantly prevented thrombosis. Ex vivo experiments further demonstrated that miR-126-deficient platelets displayed impaired platelet aggregation, spreading, and secretory functions. Next, miR-126 was confirmed to target phosphoinositol-3 kinase regulatory subunit 2 (PIK3R2) in platelet, which encodes a negative regulator of the phosphoinositide 3-kinase/protein kinase B pathway, enhancing platelet activation through activating the integrin αIIbβ3-mediated outside-in signalling. After undergoing myocardial infarction (MI), chimeric mice lacking intraplatelet miR-126 displayed reduced microvascular obstruction and prevented MI expansion in vivo. In contrast, overexpression of miR-126 by the administration of miR-126 agonist (agomiR-126) in wild-type mice aggravated microvascular obstruction and promoted MI expansion, which can be almost abolished by aspirin administration. In patients with cardiovascular diseases, antiplatelet therapies, either aspirin alone or combined with clopidogrel, decreased the level of intraplatelet miR-126. The reduction of intraplatelet miR-126 level was associated with the decrease in platelet activity. CONCLUSION Our murine and human data reveal that (i) intraplatelet miR-126 contributes to platelet activity and promotes thrombus formation, and (ii) the reduction of intraplatelet miR-126 contributes to platelet inhibition during antiplatelet therapy.
Collapse
Affiliation(s)
- Lu-Jun Zhang
- Department of Cardiology, Shanghai Changhai Hospital, 168 Changhai Road, Shanghai 200433, China
| | - Yang-Xi Hu
- Department of Cardiology, Shanghai Changzheng Hospital, Shanghai, China
| | - Rong-Zhong Huang
- Department of Geriatrics, Second Hospital Affiliated to Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Yan-Yan Xu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shao-Hua Dong
- Department of Cardiology, Shanghai Changhai Hospital, 168 Changhai Road, Shanghai 200433, China
| | - Fang-Hao Guo
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Innovation Center for Intervention of Chronic Disease and Promotion of Health, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, China
| | - Jun-Jun Guo
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Innovation Center for Intervention of Chronic Disease and Promotion of Health, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, China
| | - Jing-Jing Qiu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Innovation Center for Intervention of Chronic Disease and Promotion of Health, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, China
| | - Zi-Yun Cao
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Innovation Center for Intervention of Chronic Disease and Promotion of Health, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, China
| | - Li-Jiang Wei
- Department of Cardiology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jia-Hao Mao
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Innovation Center for Intervention of Chronic Disease and Promotion of Health, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, China
| | - Ankang Lyu
- Department of Cardiology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun-Ling Liu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xian-Xian Zhao
- Department of Cardiology, Shanghai Changhai Hospital, 168 Changhai Road, Shanghai 200433, China
| | - Zhi-Fu Guo
- Department of Cardiology, Shanghai Changhai Hospital, 168 Changhai Road, Shanghai 200433, China
| | - Qing Jing
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Innovation Center for Intervention of Chronic Disease and Promotion of Health, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, China
| |
Collapse
|
2
|
Qi H, Wu Y, Zhang W, Yu N, Lu X, Liu J. The syntaxin-binding protein STXBP5 regulates progerin expression. Sci Rep 2024; 14:23376. [PMID: 39379476 PMCID: PMC11461833 DOI: 10.1038/s41598-024-74621-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Accepted: 09/27/2024] [Indexed: 10/10/2024] Open
Abstract
Hutchinson-Gilfor progeria syndrome (HGPS) is caused by a mutation in Lamin A resulting in the production of a protein called progerin. The accumulation of progerin induces inflammation, cellular senescence and activation of the P53 pathway. In this study, through public dataset analysis, we identified Syntaxin Binding Protein 5 (STXBP5) as an influencing factor of progerin expression. STXBP5 overexpression accelerated the onset of senescence, while STXBP5 deletion suppressed progerin expression, delayed senility, and decreased the expression of senescence-related factors. STXBP5 and progerin have synergistic effects and a protein-protein interaction. Through bioinformatics analysis, we found that STXBP5 affects ageing-related signalling pathways such as the mitogen-activated protein kinase (MAPK) pathway, the hippo pathway and the interleukin 17 (IL17) signalling pathway in progerin-expressing cells. In addition, STXBP5 overexpression induced changes in transposable elements (TEs), such as the human endogenous retrovirus H internal coding sequence (HERVH-int) changes. Our protein coimmunoprecipitation (Co-IP) results indicated that STXBP5 bound directly to progerin. Therefore, decreasing STXBP5 expression is a potential new therapeutic strategy for treating ageing-related phenotypes in patients with HGPS.
Collapse
Affiliation(s)
- Hongqian Qi
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300350, China
- College of Pharmacy, Nankai University, Tianjin, 300350, China
| | - Yingying Wu
- College of Artificial Intelligence, Nankai University, Tianjin, 300350, China
- Engineering Research Center of Trusted Behavior Intelligence, Ministry of Education, Nankai University, Tianjin, 300350, China
| | - Weiyu Zhang
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, 14853-2703, USA
| | - Ningbo Yu
- College of Artificial Intelligence, Nankai University, Tianjin, 300350, China
- Engineering Research Center of Trusted Behavior Intelligence, Ministry of Education, Nankai University, Tianjin, 300350, China
| | - Xinyi Lu
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300350, China.
| | - Jinchao Liu
- College of Artificial Intelligence, Nankai University, Tianjin, 300350, China.
- Engineering Research Center of Trusted Behavior Intelligence, Ministry of Education, Nankai University, Tianjin, 300350, China.
| |
Collapse
|
3
|
Meijer M, Öttl M, Yang J, Subkhangulova A, Kumar A, Feng Z, van Voorst TW, Groffen AJ, van Weering JRT, Zhang Y, Verhage M. Tomosyns attenuate SNARE assembly and synaptic depression by binding to VAMP2-containing template complexes. Nat Commun 2024; 15:2652. [PMID: 38531902 PMCID: PMC10965968 DOI: 10.1038/s41467-024-46828-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 03/12/2024] [Indexed: 03/28/2024] Open
Abstract
Tomosyns are widely thought to attenuate membrane fusion by competing with synaptobrevin-2/VAMP2 for SNARE-complex assembly. Here, we present evidence against this scenario. In a novel mouse model, tomosyn-1/2 deficiency lowered the fusion barrier and enhanced the probability that synaptic vesicles fuse, resulting in stronger synapses with faster depression and slower recovery. While wild-type tomosyn-1m rescued these phenotypes, substitution of its SNARE motif with that of synaptobrevin-2/VAMP2 did not. Single-molecule force measurements indeed revealed that tomosyn's SNARE motif cannot substitute synaptobrevin-2/VAMP2 to form template complexes with Munc18-1 and syntaxin-1, an essential intermediate for SNARE assembly. Instead, tomosyns extensively bind synaptobrevin-2/VAMP2-containing template complexes and prevent SNAP-25 association. Structure-function analyses indicate that the C-terminal polybasic region contributes to tomosyn's inhibitory function. These results reveal that tomosyns regulate synaptic transmission by cooperating with synaptobrevin-2/VAMP2 to prevent SNAP-25 binding during SNARE assembly, thereby limiting initial synaptic strength and equalizing it during repetitive stimulation.
Collapse
Affiliation(s)
- Marieke Meijer
- Department of Human Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam University Medical Center, 1081HV, Amsterdam, The Netherlands.
| | - Miriam Öttl
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, 1081HV, Amsterdam, The Netherlands
| | - Jie Yang
- Department of Cell Biology, Yale School of Medicine, New Haven, CT, 06511, USA.
| | - Aygul Subkhangulova
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, 1081HV, Amsterdam, The Netherlands
| | - Avinash Kumar
- Department of Cell Biology, Yale School of Medicine, New Haven, CT, 06511, USA
| | - Zicheng Feng
- Department of Cell Biology, Yale School of Medicine, New Haven, CT, 06511, USA
| | - Torben W van Voorst
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, 1081HV, Amsterdam, The Netherlands
| | - Alexander J Groffen
- Department of Human Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam University Medical Center, 1081HV, Amsterdam, The Netherlands
| | - Jan R T van Weering
- Department of Human Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam University Medical Center, 1081HV, Amsterdam, The Netherlands
| | - Yongli Zhang
- Department of Cell Biology, Yale School of Medicine, New Haven, CT, 06511, USA.
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, 06511, USA.
| | - Matthijs Verhage
- Department of Human Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam University Medical Center, 1081HV, Amsterdam, The Netherlands.
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, 1081HV, Amsterdam, The Netherlands.
| |
Collapse
|
4
|
Vu LT, Ahmed F, Zhu H, Iu DSH, Fogarty EA, Kwak Y, Chen W, Franconi CJ, Munn PR, Tate AE, Levine SM, Stevens J, Mao X, Shungu DC, Moore GE, Keller BA, Hanson MR, Grenier JK, Grimson A. Single-cell transcriptomics of the immune system in ME/CFS at baseline and following symptom provocation. Cell Rep Med 2024; 5:101373. [PMID: 38232699 PMCID: PMC10829790 DOI: 10.1016/j.xcrm.2023.101373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 08/10/2023] [Accepted: 12/14/2023] [Indexed: 01/19/2024]
Abstract
Myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) is a serious and poorly understood disease. To understand immune dysregulation in ME/CFS, we use single-cell RNA sequencing (scRNA-seq) to examine immune cells in patient and control cohorts. Postexertional malaise (PEM), an exacerbation of symptoms following strenuous exercise, is a characteristic symptom of ME/CFS. To detect changes coincident with PEM, we applied scRNA-seq on the same cohorts following exercise. At baseline, ME/CFS patients display classical monocyte dysregulation suggestive of inappropriate differentiation and migration to tissue. We identify both diseased and more normal monocytes within patients, and the fraction of diseased cells correlates with disease severity. Comparing the transcriptome at baseline and postexercise challenge, we discover patterns indicative of improper platelet activation in patients, with minimal changes elsewhere in the immune system. Taken together, these data identify immunological defects present at baseline in patients and an additional layer of dysregulation in platelets.
Collapse
Affiliation(s)
- Luyen Tien Vu
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA
| | - Faraz Ahmed
- Genomics Innovation Hub and TREx Facility, Institute of Biotechnology, Cornell University, Ithaca, NY 14853, USA
| | - Hongya Zhu
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA
| | - David Shing Huk Iu
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA
| | - Elizabeth A Fogarty
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA
| | - Yeonui Kwak
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA
| | - Weizhong Chen
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA
| | - Carl J Franconi
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA
| | - Paul R Munn
- Genomics Innovation Hub and TREx Facility, Institute of Biotechnology, Cornell University, Ithaca, NY 14853, USA
| | - Ann E Tate
- Genomics Innovation Hub and TREx Facility, Institute of Biotechnology, Cornell University, Ithaca, NY 14853, USA
| | | | | | - Xiangling Mao
- Department of Radiology, Weill Cornell Medicine, New York, NY, USA
| | - Dikoma C Shungu
- Department of Radiology, Weill Cornell Medicine, New York, NY, USA
| | - Geoffrey E Moore
- Department of Exercise Science and Athletic Training, Ithaca College, Ithaca, NY, USA
| | - Betsy A Keller
- Department of Exercise Science and Athletic Training, Ithaca College, Ithaca, NY, USA
| | - Maureen R Hanson
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA
| | - Jennifer K Grenier
- Genomics Innovation Hub and TREx Facility, Institute of Biotechnology, Cornell University, Ithaca, NY 14853, USA.
| | - Andrew Grimson
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA.
| |
Collapse
|
5
|
Joshi S, Prakhya KS, Smith AN, Chanzu H, Zhang M, Whiteheart SW. The complementary roles of VAMP-2, -3, and -7 in platelet secretion and function. Platelets 2023; 34:2237114. [PMID: 37545110 PMCID: PMC10564522 DOI: 10.1080/09537104.2023.2237114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 06/21/2023] [Accepted: 07/10/2023] [Indexed: 08/08/2023]
Abstract
Platelet secretion requires Soluble N-ethylmaleimide Sensitive Attachment Protein Receptors (SNAREs). Vesicle SNAREs/Vesicle-Associated Membrane Proteins (v-SNAREs/VAMPs) on granules and t-SNAREs in plasma membranes mediate granule release. Platelet VAMP heterogeneity has complicated the assessment of how/if each is used and affects hemostasis. To address the importance of VAMP-7 (V7), we analyzed mice with global deletions of V3 and V7 together or platelet-specific deletions of V2, V3, and global deletion of V7. We measured the kinetics of cargo release, and its effects on three injury models to define the context-specific roles of these VAMPs. Loss of V7 minimally affected dense and α granule release but did affect lysosomal release. V3-/-7-/- and V2Δ3Δ7-/- platelets showed partial defects in α and lysosomal release; dense granule secretion was unaffected. In vivo assays showed that loss of V2, V3, and V7 caused no bleeding or occlusive thrombosis. These data indicate a role for V7 in lysosome release that is partially compensated by V3. V7 and V3, together, contribute to α granule release, however none of these deletions affected hemostasis/thrombosis. Our results confirm the dominance of V8. When it is present, deletion of V2, V3, or V7 alone or in combination minimally affects platelet secretion and hemostasis.
Collapse
Affiliation(s)
- Smita Joshi
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY 40536, USA
| | | | - Alexis N. Smith
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY 40536, USA
| | - Harry Chanzu
- GenScript USA Inc., 860 Centennial Ave. Piscataway, NJ 08854, USA
| | - Ming Zhang
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY 40536, USA
| | - Sidney W. Whiteheart
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY 40536, USA
| |
Collapse
|
6
|
Subkhangulova A, Gonzalez-Lozano MA, Groffen AJA, van Weering JRT, Smit AB, Toonen RF, Verhage M. Tomosyn affects dense core vesicle composition but not exocytosis in mammalian neurons. eLife 2023; 12:e85561. [PMID: 37695731 PMCID: PMC10495110 DOI: 10.7554/elife.85561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 08/28/2023] [Indexed: 09/13/2023] Open
Abstract
Tomosyn is a large, non-canonical SNARE protein proposed to act as an inhibitor of SNARE complex formation in the exocytosis of secretory vesicles. In the brain, tomosyn inhibits the fusion of synaptic vesicles (SVs), whereas its role in the fusion of neuropeptide-containing dense core vesicles (DCVs) is unknown. Here, we addressed this question using a new mouse model with a conditional deletion of tomosyn (Stxbp5) and its paralogue tomosyn-2 (Stxbp5l). We monitored DCV exocytosis at single vesicle resolution in tomosyn-deficient primary neurons using a validated pHluorin-based assay. Surprisingly, loss of tomosyns did not affect the number of DCV fusion events but resulted in a strong reduction of intracellular levels of DCV cargos, such as neuropeptide Y (NPY) and brain-derived neurotrophic factor (BDNF). BDNF levels were largely restored by re-expression of tomosyn but not by inhibition of lysosomal proteolysis. Tomosyn's SNARE domain was dispensable for the rescue. The size of the trans-Golgi network and DCVs was decreased, and the speed of DCV cargo flux through Golgi was increased in tomosyn-deficient neurons, suggesting a role for tomosyns in DCV biogenesis. Additionally, tomosyn-deficient neurons showed impaired mRNA expression of some DCV cargos, which was not restored by re-expression of tomosyn and was also observed in Cre-expressing wild-type neurons not carrying loxP sites, suggesting a direct effect of Cre recombinase on neuronal transcription. Taken together, our findings argue against an inhibitory role of tomosyns in neuronal DCV exocytosis and suggests an evolutionary conserved function of tomosyns in the packaging of secretory cargo at the Golgi.
Collapse
Affiliation(s)
- Aygul Subkhangulova
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research (CNCR), Vrije Universiteit (VU) AmsterdamAmsterdamNetherlands
| | - Miguel A Gonzalez-Lozano
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research (CNCR), Vrije Universiteit (VU) AmsterdamAmsterdamNetherlands
| | - Alexander JA Groffen
- Department of Human Genetics, Center for Neurogenomics and Cognitive Research (CNCR), Amsterdam University Medical Center (UMC)AmsterdamNetherlands
| | - Jan RT van Weering
- Department of Human Genetics, Center for Neurogenomics and Cognitive Research (CNCR), Amsterdam University Medical Center (UMC)AmsterdamNetherlands
| | - August B Smit
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research (CNCR), Vrije Universiteit (VU) AmsterdamAmsterdamNetherlands
| | - Ruud F Toonen
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research (CNCR), Vrije Universiteit (VU) AmsterdamAmsterdamNetherlands
| | - Matthijs Verhage
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research (CNCR), Vrije Universiteit (VU) AmsterdamAmsterdamNetherlands
- Department of Human Genetics, Center for Neurogenomics and Cognitive Research (CNCR), Amsterdam University Medical Center (UMC)AmsterdamNetherlands
| |
Collapse
|
7
|
Swystun LL, Michels A, Lillicrap D. The contribution of the sinusoidal endothelial cell receptors CLEC4M, stabilin-2, and SCARA5 to VWF-FVIII clearance in thrombosis and hemostasis. J Thromb Haemost 2023; 21:2007-2019. [PMID: 37085036 PMCID: PMC11539076 DOI: 10.1016/j.jtha.2023.04.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 04/11/2023] [Accepted: 04/12/2023] [Indexed: 04/23/2023]
Abstract
Quantitative abnormalities in factor VIII (FVIII) and its binding partner, von Willebrand factor (VWF), are associated with an increased risk of bleeding or thrombosis, and pathways that regulate the clearance of VWF-FVIII can strongly influence their plasma levels. In 2010, the Cohorts for Heart and Aging Research in Genome Epidemiology (CHARGE) on genome-wide association study meta-analysis identified variants in the genes for the sinusoidal endothelial receptors C-type lectin domain family 4 member M (CLEC4M), stabilin-2, and scavenger receptor class A member 5 (SCARA5) as being associated with plasma levels of VWF and/or FVIII in normal individuals. The ability of these receptors to bind, internalize, and clear the VWF-FVIII complex from the circulation has now been reported in a series of studies using in vitro and in vivo models. The receptor stabilin-2 has also been shown to modulate the immune response to infused VWF-FVIII concentrates in a murine model. In addition, the influence of genetic variants in CLEC4M, STAB2, and SCARA5 on type 1 von Willebrand disease/low VWF phenotype, FVIII pharmacokinetics, and the risk of venous thromboembolism has been described in a number of patient-based studies. Understanding the role of these receptors in the regulation of VWF-FVIII clearance has led to significant insights into the genomic architecture that modulates plasma VWF and FVIII levels, improving the understanding of pathways that regulate VWF-FVIII clearance and the mechanistic basis of quantitative VWF-FVIII pathologies.
Collapse
Affiliation(s)
- Laura L Swystun
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada
| | - Alison Michels
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada. https://twitter.com/michels_alison
| | - David Lillicrap
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada.
| |
Collapse
|
8
|
Swystun LL, Lillicrap D. Current Understanding of Inherited Modifiers of FVIII Pharmacokinetic Variation. Pharmgenomics Pers Med 2023; 16:239-252. [PMID: 36998673 PMCID: PMC10046206 DOI: 10.2147/pgpm.s383221] [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: 12/20/2022] [Accepted: 03/06/2023] [Indexed: 04/01/2023] Open
Abstract
The inherited bleeding disorder hemophilia A involves the quantitative deficiency of the coagulation cofactor factor VIII (FVIII). Prophylactic treatment of severe hemophilia A patients with FVIII concentrates aims to reduce the frequency of spontaneous joint bleeding and requires personalized tailoring of dosing regimens to account for the substantial inter-individual variability of FVIII pharmacokinetics. The strong reproducibility of FVIII pharmacokinetic (PK) metrics between repeat analyses in the same individual suggests this trait is genetically regulated. While the influence of plasma von Willebrand factor antigen (VWF:Ag) levels, ABO blood group, and patient age on FVIII PK is well established, estimates suggest these factors account for less than 35% of the overall variability in FVIII PK. More recent studies have identified genetic determinants that modify FVIII clearance or half-life including VWF gene variants that impair VWF-FVIII binding resulting in the accelerated clearance of VWF-free FVIII. Additionally, variants in receptors that regulate the clearance of FVIII or the VWF-FVIII complex have been associated with FVIII PK. The characterization of genetic modifiers of FVIII PK will provide mechanistic insight into a subject of clinical significance and support the development of personalized treatment plans for patients with hemophilia A.
Collapse
Affiliation(s)
- Laura L Swystun
- Department of Pathology and Molecular Medicine, Queen’s University, Kingston, ON, Canada
| | - David Lillicrap
- Department of Pathology and Molecular Medicine, Queen’s University, Kingston, ON, Canada
- Correspondence: David Lillicrap, Richardson Laboratory, Queen’s University, 88 Stuart Street, Kingston, Ontario, K7L 3N6, Canada, Tel +1 613 548-1304, Fax +1 613 548-1356, Email
| |
Collapse
|
9
|
Harper MT. Platelet-Derived Extracellular Vesicles in Arterial Thrombosis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1418:259-275. [PMID: 37603285 DOI: 10.1007/978-981-99-1443-2_17] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/22/2023]
Abstract
Blood platelets are necessary for normal haemostasis but also form life-threatening arterial thrombi when atherosclerotic plaques rupture. Activated platelets release many extracellular vesicles during thrombosis. Phosphatidylserine-exposing microparticles promote coagulation. Small exosomes released during granule secretion deliver cargoes including microRNAs to cells throughout the cardiovascular system. Here, we discuss the mechanisms by which platelets release these extracellular vesicles, together with the possibility of inhibiting this release as an antithrombotic strategy.
Collapse
Affiliation(s)
- Matthew T Harper
- Department of Pharmacology, University of Cambridge, Cambridge, UK.
| |
Collapse
|
10
|
Campbell RA, Manne BK, Banerjee M, Middleton EA, Ajanel A, Schwertz H, Denorme F, Stubben C, Montenont E, Saperstein S, Page L, Tolley ND, Lim DL, Brown SM, Grissom CK, Sborov DW, Krishnan A, Rondina MT. IFITM3 regulates fibrinogen endocytosis and platelet reactivity in nonviral sepsis. J Clin Invest 2022; 132:e153014. [PMID: 36194487 PMCID: PMC9711880 DOI: 10.1172/jci153014] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 09/29/2022] [Indexed: 01/13/2023] Open
Abstract
Platelets and megakaryocytes are critical players in immune responses. Recent reports suggest infection and inflammation alter the megakaryocyte and platelet transcriptome to induce altered platelet reactivity. We determined whether nonviral sepsis induces differential platelet gene expression and reactivity. Nonviral sepsis upregulated IFN-induced transmembrane protein 3 (IFITM3), an IFN-responsive gene that restricts viral replication. As IFITM3 has been linked to clathrin-mediated endocytosis, we determined whether IFITM3 promoted endocytosis of α-granule proteins. IFN stimulation enhanced fibrinogen endocytosis in megakaryocytes and platelets from Ifitm+/+ mice, but not Ifitm-/- mice. IFITM3 overexpression or deletion in megakaryocytes demonstrated IFITM3 was necessary and sufficient to regulate fibrinogen endocytosis. Mechanistically, IFITM3 interacted with clathrin and αIIb and altered their plasma membrane localization into lipid rafts. In vivo IFN administration increased fibrinogen endocytosis, platelet reactivity, and thrombosis in an IFITM-dependent manner. In contrast, Ifitm-/- mice were completely rescued from IFN-induced platelet hyperreactivity and thrombosis. During murine sepsis, platelets from Ifitm+/+ mice demonstrated increased fibrinogen content and platelet reactivity, which was dependent on IFN-α and IFITMs. Platelets from patients with nonviral sepsis had increases in platelet IFITM3 expression, fibrinogen content, and hyperreactivity. These data identify IFITM3 as a regulator of platelet endocytosis, hyperreactivity, and thrombosis during inflammatory stress.
Collapse
Affiliation(s)
- Robert A. Campbell
- University of Utah Molecular Medicine Program, Salt Lake City, Utah, USA
- Department of Internal Medicine
- Department of Pathology, and
| | - Bhanu Kanth Manne
- University of Utah Molecular Medicine Program, Salt Lake City, Utah, USA
| | - Meenakshi Banerjee
- University of Utah Molecular Medicine Program, Salt Lake City, Utah, USA
| | - Elizabeth A. Middleton
- University of Utah Molecular Medicine Program, Salt Lake City, Utah, USA
- Department of Internal Medicine
| | | | - Hansjorg Schwertz
- University of Utah Molecular Medicine Program, Salt Lake City, Utah, USA
- Rocky Mountain Center for Occupational and Environmental Health, University of Utah, Salt Lake City, Utah, USA
- Occupational Medicine, Billings Clinic Bozeman, Bozeman, Montana, USA
| | - Frederik Denorme
- University of Utah Molecular Medicine Program, Salt Lake City, Utah, USA
| | - Chris Stubben
- Bioinformatics Shared Resource, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
| | - Emilie Montenont
- University of Utah Molecular Medicine Program, Salt Lake City, Utah, USA
| | | | - Lauren Page
- University of Utah Molecular Medicine Program, Salt Lake City, Utah, USA
| | - Neal D. Tolley
- University of Utah Molecular Medicine Program, Salt Lake City, Utah, USA
| | - Diana L. Lim
- University of Utah Molecular Medicine Program, Salt Lake City, Utah, USA
| | - Samuel M. Brown
- Division of Pulmonary and Critical Medicine, Department of Medicine, Intermountain Medical Center, Murray, Utah, USA
| | - Colin K. Grissom
- Division of Pulmonary and Critical Medicine, Department of Medicine, Intermountain Medical Center, Murray, Utah, USA
| | - Douglas W. Sborov
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
| | - Anandi Krishnan
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California, USA
- Department of Pathology, Stanford University, Stanford, California, USA
| | - Matthew T. Rondina
- University of Utah Molecular Medicine Program, Salt Lake City, Utah, USA
- Department of Internal Medicine
- Department of Pathology, and
- George E. Wahlen Department of Veterans Affairs Medical Center, Department of Internal Medicine, and Geriatric Research, Education, and Clinical Center, Salt Lake City, Utah, USA
| |
Collapse
|
11
|
Chandrakanthan M, Nguyen TQ, Hasan Z, Muralidharan S, Vu TM, Li AWL, Le UTN, Thi Thuy Ha H, Baik SH, Tan SH, Foo JC, Wenk MR, Cazenave-Gassiot A, Torta F, Ong WY, Chan MYY, Nguyen LN. Deletion of Mfsd2b impairs thrombotic functions of platelets. Nat Commun 2021; 12:2286. [PMID: 33863882 PMCID: PMC8052357 DOI: 10.1038/s41467-021-22642-x] [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: 02/10/2020] [Accepted: 03/23/2021] [Indexed: 11/09/2022] Open
Abstract
We recently discovered that Mfsd2b, which is the S1P exporter found in blood cells. Here, we report that Mfsd2b is critical for the release of all S1P species in both resting and activated platelets. We show that resting platelets store S1P in the cytoplasm. After activation, this S1P pool is delivered to the plasma membrane, where Mfsd2b is predominantly localized for export. Employing knockout mice of Mfsd2b, we reveal that platelets contribute a minor amount of plasma S1P. Nevertheless, Mfsd2b deletion in whole body or platelets impairs platelet morphology and functions. In particular, Mfsd2b knockout mice show significantly reduced thrombus formation. We show that loss of Mfsd2b affects intrinsic platelet functions as part of remarkable sphingolipid accumulation. These findings indicate that accumulation of sphingolipids including S1P by deletion of Mfsd2b strongly impairs platelet functions, which suggests that the transporter may be a target for the prevention of thrombotic disorders.
Collapse
Affiliation(s)
- Madhuvanthi Chandrakanthan
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Toan Quoc Nguyen
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Zafrul Hasan
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Sneha Muralidharan
- Department of Medicine, Yong Loo-Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Thiet Minh Vu
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,NTT Hi-Tech Institute, Nguyen Tat Thanh University, Ho Chi Minh City, Vietnam
| | - Aaron Wei Liang Li
- Department of Medicine, Yong Loo-Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Uyen Thanh Nha Le
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Hoa Thi Thuy Ha
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Sang-Ha Baik
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Sock Hwee Tan
- Department of Medicine, Yong Loo-Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Juat Chin Foo
- Singapore Lipidomics Incubator (SLING), Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Markus R Wenk
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Singapore Lipidomics Incubator (SLING), Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Amaury Cazenave-Gassiot
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Singapore Lipidomics Incubator (SLING), Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Federico Torta
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Singapore Lipidomics Incubator (SLING), Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Wei Yi Ong
- Department of Anatomy, Yong Loo-Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Mark Yan Yee Chan
- Department of Medicine, Yong Loo-Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Long N Nguyen
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore. .,Singapore Lipidomics Incubator (SLING), Life Sciences Institute, National University of Singapore, Singapore, Singapore. .,Cardiovascular Disease Research (CVD) Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore. .,Immunology Program Research Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
12
|
Chanzu H, Lykins J, Wigna-Kumar S, Joshi S, Pokrovskaya I, Storrie B, Pejler G, Wood JP, Whiteheart SW. Platelet α-granule cargo packaging and release are affected by the luminal proteoglycan, serglycin. J Thromb Haemost 2021; 19:1082-1095. [PMID: 33448622 DOI: 10.1111/jth.15243] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 12/16/2020] [Accepted: 01/04/2021] [Indexed: 01/27/2023]
Abstract
BACKGROUND Serglycin (SRGN) is an intragranular, sulfated proteoglycan in hematopoietic cells that affects granule composition and function. OBJECTIVE To understand how SRGN affects platelet granule packaging, cargo release, and extra-platelet microenvironments. METHODS Platelets and megakaryocytes from SRGN-/- mice were assayed for secretion kinetics, cargo levels, granule morphology upon activation, and receptor shedding. RESULTS Metabolic, 35 SO4 labeling identified SRGN as a major sulfated macromolecule in megakaryocytes. SRGN colocalized with α-granule markers (platelet factor 4 [PF4], von Willebrand factor [VWF], and P-selectin), but its deletion did not affect α-granule morphology or number. Platelet α-granule composition was altered, with a reduction in basic proteins (pI ≥8; e.g., PF4, SDF-1, angiogenin) and constitutive release of PF4 from SRGN-/- megakaryocytes. P-Selectin, VWF, and fibrinogen were unaffected. Serotonin (5-HT) uptake and β-hexosaminidase (HEXB) were slightly elevated. Thrombin-induced exocytosis of PF4 from platelets was defective; however, release of RANTES/CCL5 was normal and osteopontin secretion was more rapid. Release of 5-HT and HEXB (from dense granules and lysosomes, respectively) were unaffected. Ultrastructural studies showed distinct morphologies in activated platelets. The α-granule lumen of SRGN-/- platelet had a grainy staining pattern, whereas that of wild-type granules had only fibrous material remaining. α-Granule swelling and decondensation were reduced in SRGN-/- platelets. Upon stimulation of platelets, a SRGN/PF4 complex was released in a time- and agonist-dependent manner. Shedding of GPVI from SRGN-/- platelets was modestly enhanced. Shedding of GP1b was unaffected. CONCLUSION The polyanionic proteoglycan SRGN influences α-granule packaging, cargo release, and shedding of platelet membrane proteins.
Collapse
Affiliation(s)
- Harry Chanzu
- Department of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Joshua Lykins
- Department of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Subershan Wigna-Kumar
- Department of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Smita Joshi
- Department of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, KY, USA
- Lexington VA Medical Center, Lexington, KY, USA
| | - Irina Pokrovskaya
- Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Brian Storrie
- Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Gunnar Pejler
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Jeremy P Wood
- Department of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, KY, USA
- Division of Cardiovascular Medicine, Gill Heart and Vascular Institute, University of Kentucky, Lexington, KY, USA
| | - Sidney W Whiteheart
- Department of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, KY, USA
- Lexington VA Medical Center, Lexington, KY, USA
| |
Collapse
|
13
|
Ménasché G, Longé C, Bratti M, Blank U. Cytoskeletal Transport, Reorganization, and Fusion Regulation in Mast Cell-Stimulus Secretion Coupling. Front Cell Dev Biol 2021; 9:652077. [PMID: 33796537 PMCID: PMC8007931 DOI: 10.3389/fcell.2021.652077] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 02/03/2021] [Indexed: 01/16/2023] Open
Abstract
Mast cells are well known for their role in allergies and many chronic inflammatory diseases. They release upon stimulation, e.g., via the IgE receptor, numerous bioactive compounds from cytoplasmic secretory granules. The regulation of granule secretion and its interaction with the cytoskeleton and transport mechanisms has only recently begun to be understood. These studies have provided new insight into the interaction between the secretory machinery and cytoskeletal elements in the regulation of the degranulation process. They suggest a tight coupling of these two systems, implying a series of specific signaling effectors and adaptor molecules. Here we review recent knowledge describing the signaling events regulating cytoskeletal reorganization and secretory granule transport machinery in conjunction with the membrane fusion machinery that occur during mast cell degranulation. The new insight into MC biology offers novel strategies to treat human allergic and inflammatory diseases targeting the late steps that affect harmful release from granular stores leaving regulatory cytokine secretion intact.
Collapse
Affiliation(s)
- Gaël Ménasché
- Laboratory of Molecular Basis of Altered Immune Homeostasis, Imagine Institute, INSERM UMR 1163, Université de Paris, Paris, France
| | - Cyril Longé
- Laboratory of Molecular Basis of Altered Immune Homeostasis, Imagine Institute, INSERM UMR 1163, Université de Paris, Paris, France
| | - Manuela Bratti
- Centre de Recherche sur l'Inflammation, INSERM UMR 1149, CNRS ERL8252, Faculté de Médecine site Bichat, Université de Paris, Paris, France.,Laboratoire d'Excellence Inflamex, Université de Paris, Paris, France
| | - Ulrich Blank
- Centre de Recherche sur l'Inflammation, INSERM UMR 1149, CNRS ERL8252, Faculté de Médecine site Bichat, Université de Paris, Paris, France.,Laboratoire d'Excellence Inflamex, Université de Paris, Paris, France
| |
Collapse
|
14
|
Fan C, Yang X, Wang WW, Wang J, Li W, Guo M, Huang S, Wang Z, Liu K. Role of Kv1.3 Channels in Platelet Functions and Thrombus Formation. Arterioscler Thromb Vasc Biol 2020; 40:2360-2375. [PMID: 32787516 DOI: 10.1161/atvbaha.120.314278] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Objective:
Platelet activation by stimulatory factors leads to an increase in intracellular calcium concentration ([Ca
2+
]
i
), which is essential for almost all platelet functions. Modulation of Ca
2+
influx and [Ca
2+
]
i
in platelets has been emerging as a possible strategy for preventing and treating platelet-dependent thrombosis. Voltage-gated potassium 1.3 channels (Kv1.3) are highly expressed in platelets and able to regulate agonist-evoked [Ca
2+
]
i
increase. However, the role of Kv1.3 channels in regulating platelet functions and thrombosis has not yet been elucidated. In addition, it is difficult to obtain a specific blocker for this channel, since Kv1.3 shares identical drug-binding sites with other K
+
channels. Here, we investigate whether specific blockade of Kv1.3 channels by monoclonal antibodies affects platelet functions and thrombosis.
Approach and Results:
In this study, we produced the anti-Kv1.3 monoclonal antibody 6E12#15, which could specifically recognize both human and mouse Kv1.3 proteins and sufficiently block Kv1.3 channel currents. We found Kv1.3 blockade by 6E12#15 inhibited platelet aggregation, adhesion, and activation upon agonist stimulation. In vivo treatment with 6E12#15 alleviated thrombus formation in a mesenteric arteriole thrombosis mouse model and protected mice from collagen/epinephrine-induced pulmonary thromboembolism. Furthermore, we observed Kv1.3 regulated platelet functions by modulating Ca
2+
influx and [Ca
2+
]
i
elevation, and that this is mediated in part by P2X
1
. Interestingly,
Kv1.3
−/−
mice showed impaired platelet aggregation while displayed no abnormalities in in vivo thrombus formation. This phenomenon was related to more megakaryocytes and platelets produced in
Kv1.3
−/−
mice compared with wild-type mice.
Conclusions:
We showed specific inhibition of Kv1.3 by the novel monoclonal antibody 6E12#15 suppressed platelet functions and platelet-dependent thrombosis through modulating platelet [Ca
2+
]
i
elevation. These results indicate that Kv1.3 could act as a promising therapeutic target for antiplatelet therapies.
Collapse
Affiliation(s)
- Cheng Fan
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (C.F., M.G., S.H., Z.W.)
| | - Xiaofang Yang
- Center for Cardiac Intensive Care, Beijing Anzhen Hospital, Capital Medical University, China (X.Y.)
| | | | - Jue Wang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (J.W.)
| | - Wenzhu Li
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston (W.L.)
| | - Mengyuan Guo
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (C.F., M.G., S.H., Z.W.)
| | - Shiyuan Huang
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (C.F., M.G., S.H., Z.W.)
| | - Zhaohui Wang
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (C.F., M.G., S.H., Z.W.)
| | - Kun Liu
- Institute of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (K.L.)
| |
Collapse
|
15
|
Shen W, Kilander MBC, Bridi MS, Frei JA, Niescier RF, Huang S, Lin YC. Tomosyn regulates the small RhoA GTPase to control the dendritic stability of neurons and the surface expression of AMPA receptors. J Neurosci Res 2020; 98:1213-1231. [PMID: 32133675 PMCID: PMC7216846 DOI: 10.1002/jnr.24608] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 01/22/2020] [Accepted: 02/20/2020] [Indexed: 12/20/2022]
Abstract
Tomosyn, a protein encoded by syntaxin‐1‐binding protein 5 (STXBP5) gene, has a well‐established presynaptic role in the inhibition of neurotransmitter release and the reduction of synaptic transmission by its canonical interaction with the soluble N‐ethylmaleimide‐sensitive factor attachment protein receptor machinery. However, the postsynaptic role of tomosyn in dendritic arborization, spine stability, and trafficking of ionotropic glutamate receptors remains to be elucidated. We used short hairpin RNA to knock down tomosyn in mouse primary neurons to evaluate the postsynaptic cellular function and molecular signaling regulated by tomosyn. Knockdown of tomosyn led to an increase of RhoA GTPase activity accompanied by compromised dendritic arborization, loss of dendritic spines, decreased surface expression of AMPA receptors, and reduced miniature excitatory postsynaptic current frequency. Inhibiting RhoA signaling was sufficient to rescue the abnormal dendritic morphology and the surface expression of AMPA receptors. The function of tomosyn regulating RhoA is mediated through the N‐terminal WD40 motif, where two variants each carrying a single nucleotide mutation in this region were found in individuals with autism spectrum disorder (ASD). We demonstrated that these variants displayed loss‐of‐function phenotypes. Unlike the wild‐type tomosyn, these two variants failed to restore the reduced dendritic complexity, spine density, as well as decreased surface expression of AMPA receptors in tomosyn knockdown neurons. This study uncovers a novel role of tomosyn in maintaining neuronal function by inhibiting RhoA activity. Further analysis of tomosyn variants also provides a potential mechanism for explaining cellular pathology in ASD.
Collapse
Affiliation(s)
- Wenjuan Shen
- Program in Neuroscience, Hussman Institute for Autism, Baltimore, MD, USA
| | | | - Morgan S Bridi
- Program in Neuroscience, Hussman Institute for Autism, Baltimore, MD, USA
| | - Jeannine A Frei
- Program in Neuroscience, Hussman Institute for Autism, Baltimore, MD, USA
| | - Robert F Niescier
- Program in Neuroscience, Hussman Institute for Autism, Baltimore, MD, USA
| | - Shiyong Huang
- Program in Neuroscience, Hussman Institute for Autism, Baltimore, MD, USA
| | - Yu-Chih Lin
- Program in Neuroscience, Hussman Institute for Autism, Baltimore, MD, USA
| |
Collapse
|
16
|
Interaction networks of Weibel-Palade body regulators syntaxin-3 and syntaxin binding protein 5 in endothelial cells. J Proteomics 2019; 205:103417. [PMID: 31201948 DOI: 10.1016/j.jprot.2019.103417] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 05/17/2019] [Accepted: 06/11/2019] [Indexed: 12/27/2022]
Abstract
The endothelium stores the hemostatic protein Von Willebrand factor (VWF) in endothelial storage organelles called Weibel-Palade bodies (WPBs). During maturation, WPBs recruit a complex of Rab GTPases and effectors that associate with components of the SNARE machinery that control WPB exocytosis. Recent genome wide association studies have found links between genetic variations in the SNAREs syntaxin-2 (STX2) and syntaxin binding protein 5 (STXBP5) and VWF plasma levels, suggesting a role for SNARE proteins in regulating VWF release. Moreover, we have previously identified the SNARE proteins syntaxin-3 and STXBP1 as regulators of WPB release. In this study we used an unbiased iterative interactomic approach to identify new components of the WPB exocytotic machinery. An interactome screen of syntaxin-3 identifies a number of SNAREs and SNARE associated proteins (STXBP2, STXBP5, SNAP23, NAPA and NSF). We show that the VAMP-like domain (VLD) of STXBP5 is indispensable for the interaction with SNARE proteins and this capacity of the VLD could be exploited to identify an extended set of novel endothelial SNARE interactors of STXBP5. In addition, an STXBP5 variant with an N436S substitution, which is linked to lower VWF plasma levels, does not show a difference in interactome when compared with WT STXBP5. SIGNIFICANCE: The hemostatic protein Von Willebrand factor plays a pivotal role in vascular health: quantitative or qualitative deficiencies of VWF can lead to bleeding, while elevated levels of VWF are associated with increased risk of thrombosis. Tight regulation of VWF secretion from WPBs is therefore essential to maintain vascular homeostasis. We used an unbiased proteomic screen to identify new components of the regulatory machinery that controls WPB exocytosis. Our data expand the endothelial SNARE protein network and provide a set of novel candidate WPB regulators that may contribute to regulation of VWF plasma levels and vascular health.
Collapse
|
17
|
Delevoye C, Marks MS, Raposo G. Lysosome-related organelles as functional adaptations of the endolysosomal system. Curr Opin Cell Biol 2019; 59:147-158. [PMID: 31234051 PMCID: PMC6726539 DOI: 10.1016/j.ceb.2019.05.003] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 05/10/2019] [Accepted: 05/13/2019] [Indexed: 12/19/2022]
Abstract
Unique functions of specialised cells such as those of the immune and haemostasis systems, skin, blood vessels, lung, and bone require specialised compartments, collectively referred to as lysosome-related organelles (LROs), that share features of endosomes and lysosomes. LROs harbour unique morphological features and cell type-specific contents, and most if not all undergo regulated secretion for diverse functions. Ongoing research, largely driven by analyses of inherited diseases and their model systems, is unravelling the mechanisms involved in LRO generation, maturation, transport and secretion. A molecular understanding of these features will provide targets and markers that can be exploited for diagnosis and therapy of a myriad of diseases.
Collapse
Affiliation(s)
- Cédric Delevoye
- Structure and Membrane Compartments, Institut Curie, Paris Sciences and Lettres Research University, Centre National de la Recherche Scientifique, UMR144, Paris, France
| | - Michael S Marks
- Dept. of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia Research Institute, Philadelphia, PA 19104, USA; Dept. of Pathology and Laboratory Medicine and Dept. of Physiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Graça Raposo
- Structure and Membrane Compartments, Institut Curie, Paris Sciences and Lettres Research University, Centre National de la Recherche Scientifique, UMR144, Paris, France.
| |
Collapse
|
18
|
Huang J, Xie N, Huang H, Yao J, Hu W. Long noncoding RNA STXBP5-AS1 inhibits cell proliferation, migration, and invasion via preventing the PI3K/AKT against STXBP5 expression in non-small-cell lung carcinoma. J Cell Biochem 2019; 120:7489-7498. [PMID: 30450569 DOI: 10.1002/jcb.28023] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 10/15/2018] [Indexed: 01/24/2023]
Abstract
Long noncoding RNAs participate in carcinogenesis and tumor progression in non-small-cell lung carcinoma (NSCLC), but the mechanisms underlying NSCLC tumorigenesis remain to be fully elucidated. Here, we reported the functional role and potential mechanism of long noncoding RNA syntaxin-binding protein 5-antisense RNA 1 (STXBP5-AS1) in NSCLC. First, our data revealed that the expression levels of STXBP5-AS1 in 31 NSCLC tissues were lower than in adjacent tissues using quantitative polymerase chain reaction (qPCR) and its expression was significantly associated with tumor metastasis of NSCLC patients. Moreover, CCK-8, scratch wound healing and transwell assay suggested that upregulation of STXBP5-AS1 repressed the proliferation, migration, and invasion in A549, NCI-H292, and NCI-H460 cells. To explore the potential mechanism of STXBP5-AS1 in NSCLC, we first investigated the relationship among STXBP5-AS1, STXBP5, and AKT1 in A549 cells. Results indicated that STXBP5-AS1 was negatively related with STXBP5 and AKT1 at messenger RNA expression level using qPCR. In addition, we observed that STXBP5-AS1 had reverse effects on the protein levels of STXBP5 and phosphorylated AKT1 (p-AKT1) in A549 cells via Western blot assay, despite no significant effects on AKT1. Subsequently, LY294002, as the phosphatidylinositol 3 kinase/protein kinase B (PI3K/AKT) pathway inhibitor, was used to further confirm the regulatory mechanism of STXBP5-AS1, which showed that knockdown of STXBP5-AS1 could rescue the expression of STXBP5 and p-AKT1 protein expression levels in A549 cells. Taken together, our results suggested that STXBP5-AS1, as a tumor suppressor, inhibits cell proliferation, migration, and invasion by preventing the PI3K/AKT against STXBP5 expression in NSCLC.
Collapse
Affiliation(s)
- Jingyu Huang
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Nianlin Xie
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Hu Huang
- Department of Oncology, The 161th Hospital of Chinese People's Liberation Army, Wuhan, China
| | - Jie Yao
- Department of Urological Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Weidong Hu
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
19
|
Gorski MM, Lecchi A, Femia EA, La Marca S, Cairo A, Pappalardo E, Lotta LA, Artoni A, Peyvandi F. Complications of whole-exome sequencing for causal gene discovery in primary platelet secretion defects. Haematologica 2019; 104:2084-2090. [PMID: 30819905 PMCID: PMC6886420 DOI: 10.3324/haematol.2018.204990] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 02/22/2019] [Indexed: 01/24/2023] Open
Abstract
Primary platelet secretion defects constitute a heterogeneous group of functional defects characterized by reduced platelet granule secretion upon stimulation by different agonists. The clinical and laboratory heterogeneity of primary platelet secretion defects warrants a tailored approach. We performed a pilot study in order to develop DNA sequence analysis pipelines for gene discovery and to create a list of candidate causal genes for platelet secretion defects. Whole-exome sequencing analysis of 14 unrelated Italian patients with primary secretion defects and 16 controls was performed on Illumina HiSeq. Variant prioritization was carried out using two filtering approaches: identification of rare, potentially damaging variants in platelet candidate genes or by selecting singletons. To corroborate the results, exome sequencing was applied in a family in which platelet secretion defects and a bleeding diathesis were present. Platelet candidate gene analysis revealed gene defects in 10/14 patients, which included ADRA2A, ARHGAP1, DIAPH1, EXOC1, FCGR2A, ITPR1, LTBP1, PTPN7, PTPN12, PRKACG, PRKCD, RAP1GAP, STXBP5L, and VWF. The analysis of singletons identified additional gene defects in PLG and PHACTR2 in two other patients. The family analysis confirmed a missense variant p.D1144N in the STXBP5L gene and p.P83H in the KCNMB3 gene as potentially causal. In summary, exome sequencing revealed potential causal variants in 12 of 14 patients with primary platelet secretion defects, highlighting the limitations of the genomic approaches for causal gene identification in this heterogeneous clinical and laboratory phenotype.
Collapse
Affiliation(s)
- Marcin M Gorski
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Angelo Bianchi Bonomi Hemophilia and Thrombosis Center, Milan.,Università degli Studi di Milano, Department of Pathophysiology and Transplantation and Fondazione Luigi Villa, Milan, Italy
| | - Anna Lecchi
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Angelo Bianchi Bonomi Hemophilia and Thrombosis Center, Milan
| | - Eti A Femia
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Angelo Bianchi Bonomi Hemophilia and Thrombosis Center, Milan
| | - Silvia La Marca
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Angelo Bianchi Bonomi Hemophilia and Thrombosis Center, Milan
| | - Andrea Cairo
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Angelo Bianchi Bonomi Hemophilia and Thrombosis Center, Milan
| | - Emanuela Pappalardo
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Angelo Bianchi Bonomi Hemophilia and Thrombosis Center, Milan.,Università degli Studi di Milano, Department of Pathophysiology and Transplantation and Fondazione Luigi Villa, Milan, Italy
| | - Luca A Lotta
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Angelo Bianchi Bonomi Hemophilia and Thrombosis Center, Milan
| | - Andrea Artoni
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Angelo Bianchi Bonomi Hemophilia and Thrombosis Center, Milan
| | - Flora Peyvandi
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Angelo Bianchi Bonomi Hemophilia and Thrombosis Center, Milan .,Università degli Studi di Milano, Department of Pathophysiology and Transplantation and Fondazione Luigi Villa, Milan, Italy
| |
Collapse
|
20
|
|
21
|
Swystun LL, Lillicrap D. Genetic regulation of plasma von Willebrand factor levels in health and disease. J Thromb Haemost 2018; 16:2375-2390. [PMID: 30246494 PMCID: PMC7147242 DOI: 10.1111/jth.14304] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Indexed: 02/06/2023]
Abstract
Plasma levels of the multimeric glycoprotein von Willebrand factor (VWF) constitute a complex quantitative trait with a continuous distribution and wide range in the normal population (50-200%). Quantitative deficiencies of VWF (< 50%) are associated with an increased risk of bleeding, whereas high plasma levels of VWF (> 150%) influence the risk of arterial and venous thromboembolism. Although environmental factors can strongly influence plasma VWF levels, it is estimated that approximately 65% of this variability is heritable. Interestingly, although variability in VWF can account for ~ 5% of the genetic influence on plasma VWF levels, other genetic loci also strongly modify plasma VWF levels. The identification of the additional sources of VWF heritability has been the focus of recent observational trait-mapping studies, including genome-wide association studies or linkage analyses, as well as hypothesis-driven research studies. Quantitative trait loci influencing VWF glycosylation, secretion and clearance have been associated with plasma VWF antigen levels in normal individuals, and may contribute to quantitative VWF abnormalities in patients with a thrombotic tendency or type 1 von Willebrand disease (VWD). The identification of genetic modifiers of plasma VWF levels may allow for better molecular diagnosis of type 1 VWD, and enable the identification of individuals at increased risk for thrombosis. Validation of trait-mapping studies with in vitro and in vivo methodologies has led to novel insights into the life cycle of VWF and the pathogenesis of quantitative VWF abnormalities.
Collapse
Affiliation(s)
- L L Swystun
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada
| | - D Lillicrap
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada
| |
Collapse
|
22
|
Madera-Salcedo IK, Danelli L, Tiwari N, Dema B, Pacreau E, Vibhushan S, Birnbaum J, Agabriel C, Liabeuf V, Klingebiel C, Menasche G, Macias-Silva M, Benhamou M, Charles N, González-Espinosa C, Vitte J, Blank U. Tomosyn functions as a PKCδ-regulated fusion clamp in mast cell degranulation. Sci Signal 2018; 11:11/537/eaan4350. [DOI: 10.1126/scisignal.aan4350] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
23
|
Adam F, Kauskot A, Kurowska M, Goudin N, Munoz I, Bordet JC, Huang JD, Bryckaert M, Fischer A, Borgel D, de Saint Basile G, Christophe OD, Ménasché G. Kinesin-1 Is a New Actor Involved in Platelet Secretion and Thrombus Stability. Arterioscler Thromb Vasc Biol 2018. [PMID: 29519941 DOI: 10.1161/atvbaha.117.310373] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
OBJECTIVE Platelet secretion is crucial for many physiological platelet responses. Even though several regulators of the fusion machinery for secretory granule exocytosis have been identified in platelets, the underlying mechanisms are not yet fully characterized. APPROACH AND RESULTS By studying a mouse model (cKO [conditional knockout]Kif5b) lacking Kif5b (kinesin-1 heavy chain) in its megakaryocytes and platelets, we evidenced unstable hemostasis characterized by an increase of blood loss associated to a marked tendency to rebleed in a tail-clip assay and thrombus instability in an in vivo thrombosis model. This instability was confirmed in vitro in a whole-blood perfusion assay under blood flow conditions. Aggregations induced by thrombin and collagen were also impaired in cKOKif5b platelets. Furthermore, P-selectin exposure, PF4 (platelet factor 4) secretion, and ATP release after thrombin stimulation were impaired in cKOKif5b platelets, highlighting the role of kinesin-1 in α-granule and dense granule secretion. Importantly, exogenous ADP rescued normal thrombin induced-aggregation in cKOKif5b platelets, which indicates that impaired aggregation was because of defective release of ADP and dense granules. Last, we demonstrated that kinesin-1 interacts with the molecular machinery comprising the granule-associated Rab27 (Ras-related protein Rab-27) protein and the Slp4 (synaptotagmin-like protein 4/SYTL4) adaptor protein. CONCLUSIONS Our results indicate that a kinesin-1-dependent process plays a role for platelet function by acting into the mechanism underlying α-granule and dense granule secretion.
Collapse
Affiliation(s)
- Frédéric Adam
- From the INSERM, UMR_S 1176, Paris-Sud University, Université Paris-Saclay, Le Kremlin-Bicêtre, France (F.A., A.K., M.B., D.B., O.D.C.)
| | - Alexandre Kauskot
- From the INSERM, UMR_S 1176, Paris-Sud University, Université Paris-Saclay, Le Kremlin-Bicêtre, France (F.A., A.K., M.B., D.B., O.D.C.)
| | - Mathieu Kurowska
- INSERM, UMR_S 1163, Laboratory of Normal and Pathological Homeostasis of the Immune System, Paris, France (M.K., I.M., A.F., G.d.S.B., G.M.).,Imagine Institute (M.K., I.M., A.F., G.d.S.B., G.M.)
| | - Nicolas Goudin
- Cell Imaging Facility, Imagine Institute (N.G.), Paris Descartes University, Sorbonne Paris Cité, France
| | - Isabelle Munoz
- INSERM, UMR_S 1163, Laboratory of Normal and Pathological Homeostasis of the Immune System, Paris, France (M.K., I.M., A.F., G.d.S.B., G.M.).,Imagine Institute (M.K., I.M., A.F., G.d.S.B., G.M.)
| | - Jean-Claude Bordet
- Laboratoire d'Hémostase, Centre de Biologie Est, Hospices Civils de Lyon, Bron, France (J.-C.B.).,Laboratoire de Recherche sur l'Hémophilie, UCBL1, Lyon, France (J.-C.B.)
| | - Jian-Dong Huang
- School of Biomedical Sciences, The University of Hong Kong, China (J.-D.H.)
| | - Marijke Bryckaert
- From the INSERM, UMR_S 1176, Paris-Sud University, Université Paris-Saclay, Le Kremlin-Bicêtre, France (F.A., A.K., M.B., D.B., O.D.C.)
| | - Alain Fischer
- INSERM, UMR_S 1163, Laboratory of Normal and Pathological Homeostasis of the Immune System, Paris, France (M.K., I.M., A.F., G.d.S.B., G.M.).,Imagine Institute (M.K., I.M., A.F., G.d.S.B., G.M.).,Department of Immunology and Pediatric Hematology (A.F.)
| | - Delphine Borgel
- From the INSERM, UMR_S 1176, Paris-Sud University, Université Paris-Saclay, Le Kremlin-Bicêtre, France (F.A., A.K., M.B., D.B., O.D.C.).,Biological Hematology Service (D.B.), Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris, France; and Collège de France, Paris (A.F.)
| | - Geneviève de Saint Basile
- INSERM, UMR_S 1163, Laboratory of Normal and Pathological Homeostasis of the Immune System, Paris, France (M.K., I.M., A.F., G.d.S.B., G.M.).,Imagine Institute (M.K., I.M., A.F., G.d.S.B., G.M.)
| | - Olivier D Christophe
- From the INSERM, UMR_S 1176, Paris-Sud University, Université Paris-Saclay, Le Kremlin-Bicêtre, France (F.A., A.K., M.B., D.B., O.D.C.)
| | - Gaël Ménasché
- INSERM, UMR_S 1163, Laboratory of Normal and Pathological Homeostasis of the Immune System, Paris, France (M.K., I.M., A.F., G.d.S.B., G.M.).,Imagine Institute (M.K., I.M., A.F., G.d.S.B., G.M.)
| |
Collapse
|
24
|
Abstract
Platelet granules are unique among secretory vesicles in both their content and their life cycle. Platelets contain three major granule types—dense granules, α-granules, and lysosomes—although other granule types have been reported. Dense granules and α-granules are the most well-studied and the most physiologically important. Platelet granules are formed in large, multilobulated cells, termed megakaryocytes, prior to transport into platelets. The biogenesis of dense granules and α-granules involves common but also distinct pathways. Both are formed from the
trans-Golgi network and early endosomes and mature in multivesicular bodies, but the formation of dense granules requires trafficking machinery different from that of α-granules. Following formation in the megakaryocyte body, both granule types are transported through and mature in long proplatelet extensions prior to the release of nascent platelets into the bloodstream. Granules remain stored in circulating platelets until platelet activation triggers the exocytosis of their contents. Soluble
N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) proteins, located on both the granules and target membranes, provide the mechanical energy that enables membrane fusion during both granulogenesis and exocytosis. The function of these core fusion engines is controlled by SNARE regulators, which direct the site, timing, and extent to which these SNAREs interact and consequently the resulting membrane fusion. In this review, we assess new developments in the study of platelet granules, from their generation to their exocytosis.
Collapse
Affiliation(s)
- Anish Sharda
- Division of Hemostasis and Thrombosis, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, USA
| | - Robert Flaumenhaft
- Division of Hemostasis and Thrombosis, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, USA
| |
Collapse
|
25
|
Xu S, Zhou Z, Li H, Liu Z, Pan X, Wang F, Huang Y, Li X, Xiao Y, Pan J, Wang C, Li D. BMSCs ameliorate septic coagulopathy by suppressing inflammation in cecal ligation and puncture-induced sepsis. J Cell Sci 2018; 131:jcs.211151. [PMID: 29242228 DOI: 10.1242/jcs.211151] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 12/07/2017] [Indexed: 01/08/2023] Open
Abstract
Sepsis is an aggressive and life-threatening systemic inflammatory response with a high mortality. Inflammation and coagulation play crucial roles in the pathogenesis of sepsis in a mutually promoting manner. Unlike other single-target molecular therapies that have no obvious effects on clinical sepsis, bone marrow stromal cell (BMSC) therapy offers a broader spectrum of activities ranging from immune and inflammation suppression to tissue regeneration. In this report, we demonstrate that BMSC injection attenuates septic coagulopathy. It decreased the mortality, mitigated lung injury and reduced the surge of proinflammatory factors in mice with sepsis induced by cecal ligation and puncture (CLP). An in vitro cell model also revealed that co-culture with BMSCs reduced secretion of proinflammatory factors and injury of endothelial cells in response to lipopolysaccharide (LPS), an endotoxin of gram-negative bacteria. Together, our results demonstrate that BMSCs suppress sepsis-induced inflammation, endothelial dysfunction and defective coagulation.
Collapse
Affiliation(s)
- Shunyao Xu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China.,Department of Intensive Care Unit, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Zhen Zhou
- Department of Intensive Care Unit, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Hao Li
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Ziying Liu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Xiaojun Pan
- Department of Intensive Care Unit, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Fen Wang
- Department of Intensive Care Unit, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Yueyue Huang
- Department of Intensive Care Unit, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Xiaogang Li
- Ningbo Fourth Hospital, Xiangshan, Zhejiang 315000, P.R. China
| | - Yunbei Xiao
- Department of Urology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Jingye Pan
- Department of Intensive Care Unit, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Cong Wang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China .,Ningbo Fourth Hospital, Xiangshan, Zhejiang 315000, P.R. China
| | - Dequan Li
- Department of Traumatology Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| |
Collapse
|
26
|
TRAF3 negatively regulates platelet activation and thrombosis. Sci Rep 2017; 7:17112. [PMID: 29215030 PMCID: PMC5719392 DOI: 10.1038/s41598-017-17189-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 11/22/2017] [Indexed: 11/21/2022] Open
Abstract
CD40 ligand (CD40L), a member of the tumor necrosis factor (TNF) superfamily, binds to CD40, leading to many effects depending on target cell type. Platelets express CD40L and are a major source of soluble CD40L. CD40L has been shown to potentiate platelet activation and thrombus formation, involving both CD40-dependent and -independent mechanisms. A family of proteins called TNF receptor associated factors (TRAFs) plays key roles in mediating CD40L-CD40 signaling. Platelets express several TRAFs. It has been shown that TRAF2 plays a role in CD40L-mediated platelet activation. Here we show that platelet also express TRAF3, which plays a negative role in regulating platelet activation. Thrombin- or collagen-induced platelet aggregation and secretion are increased in TRAF3 knockout mice. The expression levels of collagen receptor GPVI and integrin αIIbβ3 in platelets were not affected by deletion of TRAF3, suggesting that increased platelet activation in the TRAF3 knockout mice was not due to increased expression platelet receptors. Time to formation of thrombi in a FeCl3-induced thrombosis model was significantly shortened in the TRAF3 knockout mice. However, mouse tail-bleeding times were not affected by deletion of TRAF3. Thus, TRAF3 plays a negative role in platelet activation and in thrombus formation in vivo.
Collapse
|
27
|
Suchon P, Trégouët DA, Morange PE. Genetics of Venous Thrombosis: update in 2015. Thromb Haemost 2017; 114:910-9. [DOI: 10.1160/th15-05-0410] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 07/14/2015] [Indexed: 11/05/2022]
Abstract
SummaryVenous thrombosis (VT) is a common multifactorial disease with a genetic component that was first suspected nearly 60 years ago. In this review, we document the genetic determinants of the disease, and update recent findings delivered by the application of high-throughput genotyping and sequencing technologies. To date, 17 genes have been robustly demonstrated to harbour genetic variations associated with VT risk: ABO, F2, F5, F9, F11, FGG, GP6, KNG1, PROC, PROCR, PROS1, SERPINC1, SLC44A2, STXBP5, THBD, TSPAN15 and VWF. The common polymorphisms are estimated to account only for a modest part (~5 %) of the VT heritability. Much remains to be done to fully disentangle the exact genetic (and epigenetic) architecture of the disease. A large suite of powerful tools and research strategies can be deployed on the large collections of patients that have already been assembled (and additional are ongoing).
Collapse
|
28
|
Monitoring storage induced changes in the platelet proteome employing label free quantitative mass spectrometry. Sci Rep 2017; 7:11045. [PMID: 28887518 PMCID: PMC5591311 DOI: 10.1038/s41598-017-11643-w] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 08/29/2017] [Indexed: 01/10/2023] Open
Abstract
Shelf life of platelet concentrates is limited to 5-7 days due to loss of platelet function during storage, commonly referred to as the platelet storage lesion (PSL). To get more insight into the development of the PSL, we used label free quantitative mass spectrometry to identify changes in the platelet proteome during storage. In total 2501 proteins were accurately quantified in 3 biological replicates on at least 1 of the 7 different time-points analyzed. Significant changes in levels of 21 proteins were observed over time. Gene ontology enrichment analysis of these proteins revealed that the majority of this set was involved in platelet degranulation, secretion and regulated exocytosis. Twelve of these proteins have been shown to reside in α-granules. Upon prolonged storage (13-16 days) elevated levels of α-2-macroglobulin, glycogenin and Ig μ chain C region were identified. Taken together this study identifies novel markers for monitoring of the PSL that may potentially also be used for the detection of "young" and "old" platelets in the circulation.
Collapse
|
29
|
Batten SR, Matveeva EA, Whiteheart SW, Vanaman TC, Gerhardt GA, Slevin JT. Linking kindling to increased glutamate release in the dentate gyrus of the hippocampus through the STXBP5/tomosyn-1 gene. Brain Behav 2017; 7:e00795. [PMID: 28948088 PMCID: PMC5607557 DOI: 10.1002/brb3.795] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 06/29/2017] [Accepted: 07/02/2017] [Indexed: 12/19/2022] Open
Abstract
INTRODUCTION In kindling, repeated electrical stimulation of certain brain areas causes progressive and permanent intensification of epileptiform activity resulting in generalized seizures. We focused on the role(s) of glutamate and a negative regulator of glutamate release, STXBP5/tomosyn-1, in kindling. METHODS Stimulating electrodes were implanted in the amygdala and progression to two successive Racine stage 5 seizures was measured in wild-type and STXBP5/tomosyn-1-/- (Tom-/-) animals. Glutamate release measurements were performed in distinct brain regions using a glutamate-selective microelectrode array (MEA). RESULTS Naïve Tom-/- mice had significant increases in KCl-evoked glutamate release compared to naïve wild type as measured by MEA of presynaptic release in the hippocampal dentate gyrus (DG). Kindling progression was considerably accelerated in Tom-/- mice, requiring fewer stimuli to reach a fully kindled state. Following full kindling, MEA measurements of both kindled Tom+/+ and Tom-/- mice showed significant increases in KCl-evoked and spontaneous glutamate release in the DG, indicating a correlation with the fully kindled state independent of genotype. Resting glutamate levels in all hippocampal subregions were significantly lower in the kindled Tom-/- mice, suggesting possible changes in basal control of glutamate circuitry in the kindled Tom-/- mice. CONCLUSIONS Our studies demonstrate that increased glutamate release in the hippocampal DG correlates with acceleration of the kindling process. Although STXBP5/tomosyn-1 loss increased evoked glutamate release in naïve animals contributing to their prokindling phenotype, the kindling process can override any attenuating effect of STXBP5/tomosyn-1. Loss of this "braking" effect of STXBP5/tomosyn-1 on kindling progression may set in motion an alternative but ultimately equally ineffective compensatory response, detected here as reduced basal glutamate release.
Collapse
Affiliation(s)
- Seth R. Batten
- Department of PsychologyUniversity of KentuckyCollege of Arts and SciencesLexingtonKYUSA
| | - Elena A. Matveeva
- Department of Molecular & Cellular BiochemistryUniversity of Kentucky Medical CenterLexingtonKYUSA
| | - Sidney W. Whiteheart
- Department of Molecular & Cellular BiochemistryUniversity of Kentucky Medical CenterLexingtonKYUSA
| | - Thomas C. Vanaman
- Department of Molecular & Cellular BiochemistryUniversity of Kentucky Medical CenterLexingtonKYUSA
| | - Greg A. Gerhardt
- Department of NeuroscienceUniversity of Kentucky Medical CenterLexingtonKYUSA
- Department of NeurologyUniversity of Kentucky Medical CenterLexingtonKYUSA
| | - John T. Slevin
- Neurology ServiceVeterans Affairs Medical CenterLexingtonKYUSA
- Department of NeurologyUniversity of Kentucky Medical CenterLexingtonKYUSA
- Department of Pharmacology and Nutritional SciencesUniversity of Kentucky Medical CenterLexingtonKYUSA
| |
Collapse
|
30
|
Zhu QM, Ko KA, Ture S, Mastrangelo MA, Chen MH, Johnson AD, O'Donnell CJ, Morrell CN, Miano JM, Lowenstein CJ. Novel Thrombotic Function of a Human SNP in STXBP5 Revealed by CRISPR/Cas9 Gene Editing in Mice. Arterioscler Thromb Vasc Biol 2017; 37:264-270. [PMID: 28062498 PMCID: PMC5269475 DOI: 10.1161/atvbaha.116.308614] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 12/13/2016] [Indexed: 11/16/2022]
Abstract
OBJECTIVE To identify and characterize the effect of a SNP (single-nucleotide polymorphism) in the STXBP5 locus that is associated with altered thrombosis in humans. GWAS (genome-wide association studies) have identified numerous SNPs associated with human thrombotic phenotypes, but determining the functional significance of an individual candidate SNP can be challenging, particularly when in vivo modeling is required. Recent GWAS led to the discovery of STXBP5 as a regulator of platelet secretion in humans. Further clinical studies have identified genetic variants of STXBP5 that are linked to altered plasma von Willebrand factor levels and thrombosis in humans, but the functional significance of these variants in STXBP5 is not understood. APPROACH AND RESULTS We used CRISPR/Cas9 (clustered regularly interspaced short palindromic repeats/CRISPR-associated 9) techniques to produce a precise mouse model carrying a human coding SNP rs1039084 (encoding human p. N436S) in the STXBP5 locus associated with decreased thrombosis. Mice carrying the orthologous human mutation (encoding p. N437S in mouse STXBP5) have lower plasma von Willebrand factor levels, decreased thrombosis, and decreased platelet secretion compared with wild-type mice. This thrombosis phenotype recapitulates the phenotype of humans carrying the minor allele of rs1039084. Decreased plasma von Willebrand factor and platelet activation may partially explain the decreased thrombotic phenotype in mutant mice. CONCLUSIONS Using precise mammalian genome editing, we have identified a human nonsynonymous SNP rs1039084 in the STXBP5 locus as a causal variant for a decreased thrombotic phenotype. CRISPR/Cas9 genetic editing facilitates the rapid and efficient generation of animals to study the function of human genetic variation in vascular diseases.
Collapse
Affiliation(s)
- Qiuyu Martin Zhu
- From the Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester Medical Center, NY (Q.M.Z., K.A.K., S.T., M.A.M., C.N.M., J.M.M., C.J.L.); Division of Intramural Research, National Heart, Lung and Blood Institute (NHLBI), National Institutes of Health, and NHLBI Framingham Heart Study, Framingham, MA (M.-H.C., A.D.J., C.J.O.D.); and Cardiovascular Division, Massachusetts General Hospital, Harvard Medical School, Boston (C.J.O.D.)
| | - Kyung Ae Ko
- From the Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester Medical Center, NY (Q.M.Z., K.A.K., S.T., M.A.M., C.N.M., J.M.M., C.J.L.); Division of Intramural Research, National Heart, Lung and Blood Institute (NHLBI), National Institutes of Health, and NHLBI Framingham Heart Study, Framingham, MA (M.-H.C., A.D.J., C.J.O.D.); and Cardiovascular Division, Massachusetts General Hospital, Harvard Medical School, Boston (C.J.O.D.)
| | - Sara Ture
- From the Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester Medical Center, NY (Q.M.Z., K.A.K., S.T., M.A.M., C.N.M., J.M.M., C.J.L.); Division of Intramural Research, National Heart, Lung and Blood Institute (NHLBI), National Institutes of Health, and NHLBI Framingham Heart Study, Framingham, MA (M.-H.C., A.D.J., C.J.O.D.); and Cardiovascular Division, Massachusetts General Hospital, Harvard Medical School, Boston (C.J.O.D.)
| | - Michael A Mastrangelo
- From the Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester Medical Center, NY (Q.M.Z., K.A.K., S.T., M.A.M., C.N.M., J.M.M., C.J.L.); Division of Intramural Research, National Heart, Lung and Blood Institute (NHLBI), National Institutes of Health, and NHLBI Framingham Heart Study, Framingham, MA (M.-H.C., A.D.J., C.J.O.D.); and Cardiovascular Division, Massachusetts General Hospital, Harvard Medical School, Boston (C.J.O.D.)
| | - Ming-Huei Chen
- From the Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester Medical Center, NY (Q.M.Z., K.A.K., S.T., M.A.M., C.N.M., J.M.M., C.J.L.); Division of Intramural Research, National Heart, Lung and Blood Institute (NHLBI), National Institutes of Health, and NHLBI Framingham Heart Study, Framingham, MA (M.-H.C., A.D.J., C.J.O.D.); and Cardiovascular Division, Massachusetts General Hospital, Harvard Medical School, Boston (C.J.O.D.)
| | - Andrew D Johnson
- From the Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester Medical Center, NY (Q.M.Z., K.A.K., S.T., M.A.M., C.N.M., J.M.M., C.J.L.); Division of Intramural Research, National Heart, Lung and Blood Institute (NHLBI), National Institutes of Health, and NHLBI Framingham Heart Study, Framingham, MA (M.-H.C., A.D.J., C.J.O.D.); and Cardiovascular Division, Massachusetts General Hospital, Harvard Medical School, Boston (C.J.O.D.)
| | - Christopher J O'Donnell
- From the Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester Medical Center, NY (Q.M.Z., K.A.K., S.T., M.A.M., C.N.M., J.M.M., C.J.L.); Division of Intramural Research, National Heart, Lung and Blood Institute (NHLBI), National Institutes of Health, and NHLBI Framingham Heart Study, Framingham, MA (M.-H.C., A.D.J., C.J.O.D.); and Cardiovascular Division, Massachusetts General Hospital, Harvard Medical School, Boston (C.J.O.D.)
| | - Craig N Morrell
- From the Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester Medical Center, NY (Q.M.Z., K.A.K., S.T., M.A.M., C.N.M., J.M.M., C.J.L.); Division of Intramural Research, National Heart, Lung and Blood Institute (NHLBI), National Institutes of Health, and NHLBI Framingham Heart Study, Framingham, MA (M.-H.C., A.D.J., C.J.O.D.); and Cardiovascular Division, Massachusetts General Hospital, Harvard Medical School, Boston (C.J.O.D.)
| | - Joseph M Miano
- From the Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester Medical Center, NY (Q.M.Z., K.A.K., S.T., M.A.M., C.N.M., J.M.M., C.J.L.); Division of Intramural Research, National Heart, Lung and Blood Institute (NHLBI), National Institutes of Health, and NHLBI Framingham Heart Study, Framingham, MA (M.-H.C., A.D.J., C.J.O.D.); and Cardiovascular Division, Massachusetts General Hospital, Harvard Medical School, Boston (C.J.O.D.)
| | - Charles J Lowenstein
- From the Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester Medical Center, NY (Q.M.Z., K.A.K., S.T., M.A.M., C.N.M., J.M.M., C.J.L.); Division of Intramural Research, National Heart, Lung and Blood Institute (NHLBI), National Institutes of Health, and NHLBI Framingham Heart Study, Framingham, MA (M.-H.C., A.D.J., C.J.O.D.); and Cardiovascular Division, Massachusetts General Hospital, Harvard Medical School, Boston (C.J.O.D.).
| |
Collapse
|
31
|
McLeish KR, Merchant ML, Creed TM, Tandon S, Barati MT, Uriarte SM, Ward RA. Frontline Science: Tumor necrosis factor-α stimulation and priming of human neutrophil granule exocytosis. J Leukoc Biol 2017; 102:19-29. [PMID: 28096297 DOI: 10.1189/jlb.3hi0716-293rr] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2016] [Revised: 12/19/2016] [Accepted: 12/20/2016] [Indexed: 12/24/2022] Open
Abstract
Neutrophil granule exocytosis plays an important role in innate and adaptive immune responses. The present study examined TNF-α stimulation or priming of exocytosis of the 4 neutrophil granule subsets. TNF-α stimulated exocytosis of secretory vesicles and gelatinase granules and primed specific and azurophilic granule exocytosis to fMLF stimulation. Both stimulation and priming of exocytosis by TNF-α were dependent on p38 MAPK activity. Bioinformatic analysis of 1115 neutrophil proteins identified by mass spectrometry as being phosphorylated by TNF-α exposure found that actin cytoskeleton regulation was a major biologic function. A role for p38 MAPK regulation of the actin cytoskeleton was confirmed experimentally. Thirteen phosphoproteins regulated secretory vesicle quantity, formation, or release, 4 of which-Raf1, myristoylated alanine-rich protein kinase C (PKC) substrate (MARCKS), Abelson murine leukemia interactor 1 (ABI1), and myosin VI-were targets of the p38 MAPK pathway. Pharmacologic inhibition of Raf1 reduced stimulated exocytosis of gelatinase granules and priming of specific granule exocytosis. We conclude that differential regulation of exocytosis by TNF-α involves the actin cytoskeleton and is a necessary component for priming of the 2 major neutrophil antimicrobial defense mechanisms: oxygen radical generation and release of toxic granule contents.
Collapse
Affiliation(s)
- Kenneth R McLeish
- Department of Medicine, University of Louisville School of Medicine, Louisville, Kentucky, USA; and .,Robley Rex Veterans Affairs Medical Center, Louisville, Kentucky, USA
| | - Michael L Merchant
- Department of Medicine, University of Louisville School of Medicine, Louisville, Kentucky, USA; and
| | - T Michael Creed
- Department of Medicine, University of Louisville School of Medicine, Louisville, Kentucky, USA; and
| | - Shweta Tandon
- Department of Medicine, University of Louisville School of Medicine, Louisville, Kentucky, USA; and
| | - Michelle T Barati
- Department of Medicine, University of Louisville School of Medicine, Louisville, Kentucky, USA; and
| | - Silvia M Uriarte
- Department of Medicine, University of Louisville School of Medicine, Louisville, Kentucky, USA; and
| | - Richard A Ward
- Department of Medicine, University of Louisville School of Medicine, Louisville, Kentucky, USA; and
| |
Collapse
|
32
|
Abstract
PURPOSE OF REVIEW Von Willebrand factor (VWF) is a large multidomain, multimeric glycoprotein that plays an essential role in regulating the balance between blood clotting and bleeding. Aberrant VWF regulation can lead to a spectrum of diseases extending from bleeding disorders [Von Willebrand disease (VWD)] to aberrant thrombotic thrombocytopenic purpura (TTP). Understanding the biology of VWF expression and secretion is essential for developing novel targeted therapies for VWF-related hemostasis disorders. RECENT FINDINGS A number of recent elegant in-vitro and in-vivo studies will be highlighted, including the discovery of intronic splicing in the VWF gene, microRNA-regulated VWF gene expression, and syntaxin binding protein and autophagy mediated VWF secretion. Compared with the already established critical role of VWF in VWD and TTP pathophysiology, additional clinical studies have clarified and reinforced the association of elevated plasma levels of VWF with an increased risk of stroke, myocardial infarction, venous thrombosis, and diabetic thrombotic complications. Moreover, experimental mouse models of ischemic stroke and myocardial infarction have further supported VWF as a potential therapeutic target. SUMMARY VWF biosynthesis, maturation, and secretion is a complex process, which mandates tight regulation. Significant progress has been made in our understandings of VWF expression and secretion and its association with thrombotic diseases, contributing to the development of novel targeting VWF drugs for prevention and treatment of deficient and enhanced hemostasis.
Collapse
Affiliation(s)
- Yaozu Xiang
- aYale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, New Haven, Connecticut, USA bSchool of Life Sciences and Technology, Advanced Institute of Translational Medicine, Tongji University, Shanghai, China
| | | |
Collapse
|
33
|
Abstract
Secretion is essential to many of the roles that platelets play in the vasculature, e.g., thrombosis, angiogenesis, and inflammation, enabling platelets to modulate the microenvironment at sites of vascular lesions with a myriad of bioactive molecules stored in their granules. Past studies demonstrate that granule cargo release is mediated by Soluble NSF Attachment Protein Receptor (SNARE) proteins, which are required for granule-plasma membrane fusion. Several SNARE regulators, which control when, where, and how the SNAREs interact, have been identified in platelets. Additionally, platelet SNAREs are controlled by post-translational modifications, e.g., phosphorylation and acylation. Although there have been many recent insights into the mechanisms of platelet secretion, many questions remain: have we identified all the important regulators, does calcium directly control the process, and is platelet secretion polarized. In this review, we focus on the mechanics of platelet secretion and discuss how the secretory machinery functions in the pathway leading to membrane fusion and cargo release.
Collapse
Affiliation(s)
- Smita Joshi
- a Department of Molecular and Cellular Biochemistry , University of Kentucky , Lexington , KY , USA
| | - Sidney W Whiteheart
- a Department of Molecular and Cellular Biochemistry , University of Kentucky , Lexington , KY , USA
| |
Collapse
|
34
|
Temporal quantitative phosphoproteomics of ADP stimulation reveals novel central nodes in platelet activation and inhibition. Blood 2016; 129:e1-e12. [PMID: 28060719 DOI: 10.1182/blood-2016-05-714048] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 11/03/2016] [Indexed: 01/01/2023] Open
Abstract
Adenosine diphosphate (ADP) enhances platelet activation by virtually any other stimulant to complete aggregation. It binds specifically to the G-protein-coupled membrane receptors P2Y1 and P2Y12, stimulating intracellular signaling cascades, leading to integrin αIIbβ3 activation, a process antagonized by endothelial prostacyclin. P2Y12 inhibitors are among the most successful antiplatelet drugs, however, show remarkable variability in efficacy. We reasoned whether a more detailed molecular understanding of ADP-induced protein phosphorylation could identify (1) critical hubs in platelet signaling toward aggregation and (2) novel molecular targets for antiplatelet treatment strategies. We applied quantitative temporal phosphoproteomics to study ADP-mediated signaling at unprecedented molecular resolution. Furthermore, to mimic the antagonistic efficacy of endothelial-derived prostacyclin, we determined how Iloprost reverses ADP-mediated signaling events. We provide temporal profiles of 4797 phosphopeptides, 608 of which showed significant regulation. Regulated proteins are implicated in well-known activating functions such as degranulation and cytoskeletal reorganization, but also in less well-understood pathways, involving ubiquitin ligases and GTPase exchange factors/GTPase-activating proteins (GEF/GAP). Our data demonstrate that ADP-triggered phosphorylation occurs predominantly within the first 10 seconds, with many short rather than sustained changes. For a set of phosphorylation sites (eg, PDE3ASer312, CALDAG-GEFISer587, ENSASer109), we demonstrate an inverse regulation by ADP and Iloprost, suggesting that these are central modulators of platelet homeostasis. This study demonstrates an extensive spectrum of human platelet protein phosphorylation in response to ADP and Iloprost, which inversely overlap and represent major activating and inhibitory pathways.
Collapse
|
35
|
Abstract
Cytotoxic T lymphocytes (CTLs) and natural killer (NK) cells target infected or transformed cells with perforin-containing cytotoxic granules through immune synapses, while platelets secrete several types of granules which contents are essential for thrombosis and hemostasis. Recent work has culminated in the notion that an exocytic SNARE complex, based on a very similar set of components, is primarily responsible for exocytosis of the diverse granules in these different cell types. Granule exocytosis is, in particular, uniquely dependent on the atypical Q-SNARE syntaxin 11, its interacting partners of the Sec/Munc (SM) family, and is regulated by Rab27a. Mutations in these exocytic components underlie disease manifestations of familial hemophagocytic lymphohistiocytosis (FHL) subtypes, characterized by hyperactivation of the immune system, as well as platelet granule secretion defects. Here we discuss the key discoveries that led to the converging notion of the syntaxin 11-based exocytosis machinery for cytotoxic granules and platelet-derived granules.
Collapse
Affiliation(s)
- Bor Luen Tang
- a Department of Biochemistry , Yong Loo Lin School of Medicine, National University of Singapore , Singapore and.,b NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore , Singapore
| |
Collapse
|
36
|
Abstract
IgG carrying terminal α2,6-linked sialic acids added to conserved N-glycans within the Fc domain by the sialyltransferase ST6Gal1 accounts for the anti-inflammatory effects of large-dose i.v. Ig (IVIg) in autoimmunity. Here, B-cell-specific ablation of ST6Gal1 in mice revealed that IgG sialylation can occur in the extracellular environment of the bloodstream independently of the B-cell secretory pathway. We also discovered that secreted ST6Gal1 is produced by cells lining central veins in the liver and that IgG sialylation is powered by serum-localized nucleotide sugar donor CMP-sialic acid that is at least partially derived from degranulating platelets. Thus, antibody-secreting cells do not exclusively control the sialylation-dependent anti-inflammatory function of IgG. Rather, IgG sialylation can be regulated by the liver and platelets through the corresponding release of enzyme and sugar donor into the cardiovascular circulation.
Collapse
|
37
|
Arf6 controls platelet spreading and clot retraction via integrin αIIbβ3 trafficking. Blood 2016; 127:1459-67. [PMID: 26738539 DOI: 10.1182/blood-2015-05-648550] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 01/01/2016] [Indexed: 12/18/2022] Open
Abstract
Platelet and megakaryocyte endocytosis is important for loading certain granule cargo (ie, fibrinogen [Fg] and vascular endothelial growth factor); however, the mechanisms of platelet endocytosis and its functional acute effects are understudied. Adenosine 5'-diphosphate-ribosylation factor 6 (Arf6) is a small guanosine triphosphate-binding protein that regulates endocytic trafficking, especially of integrins. To study platelet endocytosis, we generated platelet-specific Arf6 knockout (KO) mice. Arf6 KO platelets had less associated Fg suggesting that Arf6 affects αIIbβ3-mediated Fg uptake and/or storage. Other cargo was unaffected. To measure Fg uptake, mice were injected with biotinylated- or fluorescein isothiocyanate (FITC)-labeled Fg. Platelets from the injected Arf6 KO mice showed lower accumulation of tagged Fg, suggesting an uptake defect. Ex vivo, Arf6 KO platelets were also defective in FITC-Fg uptake and storage. Immunofluorescence analysis showed initial trafficking of FITC-Fg to a Rab4-positive compartment followed by colocalization with Rab11-positive structures, suggesting that platelets contain and use both early and recycling endosomes. Resting and activated αIIbβ3 levels, as measured by flow cytometry, were unchanged; yet, Arf6 KO platelets exhibited enhanced spreading on Fg and faster clot retraction. This was not the result of alterations in αIIbβ3 signaling, because myosin light-chain phosphorylation and Rac1/RhoA activation were unaffected. Consistent with the enhanced clot retraction and spreading, Arf6 KO mice showed no deficits in tail bleeding or FeCl3-induced carotid injury assays. Our studies present the first mouse model for defining the functions of platelet endocytosis and suggest that altered integrin trafficking may affect the efficacy of platelet function.
Collapse
|
38
|
Role of Munc13-4 as a Ca2+-dependent tether during platelet secretion. Biochem J 2015; 473:627-39. [PMID: 26637270 DOI: 10.1042/bj20151150] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 12/04/2015] [Indexed: 11/17/2022]
Abstract
The Munc13 family of exocytosis regulators has multiple Ca(2+)-binding, C2 domains. Here, we probed the mechanism by which Munc13-4 regulates in vitro membrane fusion and platelet exocytosis. We show that Munc13-4 enhances in vitro soluble NSF attachment protein receptor (SNARE)-dependent, proteoliposome fusion in a Ca(2+)- and phosphatidylserine (PS)-dependent manner that was independent of SNARE concentrations. Munc13-4-SNARE interactions, under the conditions used, were minimal in the absence or presence of Ca(2+). However, Munc13-4 was able to bind and cluster liposomes harbouring PS in response to Ca(2+). Interestingly, Ca(2+)-dependent liposome binding/clustering and enhancement of proteoliposome fusion required both Munc13-4 C2 domains, but only the Ca(2+)-liganding aspartate residues of the C2B domain. Analytical ultracentrifugation (AUC) measurements indicated that, in solution, Munc13-4 was a monomeric prolate ellipsoid with dimensions consistent with a molecule that could bridge two fusing membranes. To address the potential role of Munc13-4 as a tethering protein in platelets, we examined mepacrine-stained, dense granule mobility and secretion in platelets from wild-type and Munc13-4 null (Unc13d(Jinx)) mice. In the absence of Munc13-4, dense granules were highly mobile in both resting and stimulated platelets, and stimulation-dependent granule release was absent. These observations suggest that dense granules are stably docked in resting platelets awaiting stimulation and that Munc13-4 plays a vesicle-stabilizing or tethering role in resting platelets and also in activated platelets in response to Ca(2+). In summary, we show that Munc13-4 conveys Ca(2+) sensitivity to platelet SNARE-mediated membrane fusion and reveal a potential mechanism by which Munc13-4 bridges and stabilizes apposing membranes destined for fusion.
Collapse
|
39
|
Heijnen H, van der Sluijs P. Platelet secretory behaviour: as diverse as the granules … or not? J Thromb Haemost 2015; 13:2141-51. [PMID: 26391322 DOI: 10.1111/jth.13147] [Citation(s) in RCA: 146] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 09/05/2015] [Indexed: 12/19/2022]
Abstract
Platelets play a central role in the arrest of bleeding after damage to a blood vessel and in the development of thrombosis. Platelets rapidly respond after interaction with sub-endothelial components and release cargo from their storage granules. The three principal granule types of platelets are α-granules, dense granules and lysosomes. Timed release of granule contents and regulated expression of critical receptors are essential for maintenance of the platelet thrombus, yet also have important functions beyond hemostasis (i.e. inflammatory reactions and immune responses). α-granules store adhesive molecules such as von Willebrand factor and fibrinogen, growth factors and inflammatory and angiogenic mediators, which play crucial roles in inflammatory responses and tumor genesis. The α-granules comprise a group of subcellular compartments with a unique composition and ultrastructure. Recent studies have suggested that differential secretory kinetics of α-granule subtypes is responsible for a thematic release of adhesive and inflammatory mediators. In addition, new results indicate that activation-dependent synthesis and release of cytokines also contribute to the inflammatory role of platelets. We will discuss the various methods that platelets use to regulate secretory processes and how these relate to potential differential secretion patterns, thereby promoting adhesiveness and/or inflammatory functions. We will focus on the heterogenic granule population, open canalicular system (OCS) plasticity, the role of contractile and mechanobiological forces, and the fusogenic machinery.
Collapse
Affiliation(s)
- H Heijnen
- Department of Clinical Chemistry and Hematology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - P van der Sluijs
- Department of Cell Biology, University Medical Center Utrecht, Utrecht, the Netherlands
| |
Collapse
|
40
|
Xu X, Sun B. Platelet granule secretion mechanisms: Are they modified in sepsis? Thromb Res 2015; 136:845-50. [DOI: 10.1016/j.thromres.2015.09.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Revised: 08/19/2015] [Accepted: 09/06/2015] [Indexed: 12/31/2022]
|
41
|
Abstract
In this issue of Blood, Sakurai et al examine the response of single platelets to fibrinogen- and collagen-coated microdots and show that platelets can orient their release of α-granule cargo to promote spreading beyond the dot's boundary.
Collapse
|
42
|
Abstract
Autophagy is important for maintaining cellular homeostasis, and thus its deficiency is implicated in a broad spectrum of human diseases. Its role in platelet function has only recently been examined. Our biochemical and imaging studies demonstrate that the core autophagy machinery exists in platelets, and that autophagy is constitutively active in resting platelets. Moreover, autophagy is induced upon platelet activation, as indicated by agonist-induced loss of the autophagy marker LC3II. Additional experiments, using inhibitors of platelet activation, proteases, and lysosomal acidification, as well as platelets from knockout mouse strains, show that agonist-induced LC3II loss is a consequence of platelet signaling cascades and requires proteases, acidic compartments, and membrane fusion. To assess the physiological role of platelet autophagy, we generated a mouse strain with a megakaryocyte- and platelet-specific deletion of Atg7, an enzyme required for LC3II production. Ex vivo analysis of platelets from these mice shows modest defects in aggregation and granule cargo packaging. Although these mice have normal platelet numbers and size distributions, they exhibit a robust bleeding diathesis in the tail-bleeding assay and a prolonged occlusion time in the FeCl3-induced carotid injury model. Our results demonstrate that autophagy occurs in platelets and is important for hemostasis and thrombosis.
Collapse
|
43
|
Walker B, Schmid E, Russo A, Schmidt EM, Burk O, Münzer P, Velic A, Macek B, Schaller M, Schwab M, Seabra MC, Gawaz M, Lang F, Borst O. Impact of the serum- and glucocorticoid-inducible kinase 1 on platelet dense granule biogenesis and secretion. J Thromb Haemost 2015; 13:1325-34. [PMID: 25944668 DOI: 10.1111/jth.12998] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2014] [Indexed: 02/04/2023]
Abstract
BACKGROUND Platelet secretion is critical to development of acute thrombotic occlusion. Platelet dense granules contain a variety of important hemostatically active substances. Nevertheless, biogenesis of platelet granules is poorly understood. OBJECTIVES Serum- and glucocorticoid-inducible kinase 1 (SGK1) has been shown to be highly expressed in platelets and megakaryocytes, but its role in the regulation of platelet granule biogenesis and its impact on thrombosis has not been investigated so far. METHODS AND RESULTS Electron microscopy analysis of the platelet ultrastructure revealed a significant reduction in the number and packing of dense granules in platelets lacking SGK1 (sgk1(-/-) ). In sgk1(-/-) platelets serotonin content was significantly reduced and activation-dependent secretion of ATP, serotonin and CD63 significantly impaired. In vivo adhesion after carotis ligation was significantly decreased in platelets lacking SGK1 and occlusive thrombus formation after FeCl3 -induced vascular injury was significantly diminished in sgk1(-/-) mice. Transcript levels and protein abundance of dense granule biogenesis regulating GTPase Rab27b were significantly reduced in sgk1(-/-) platelets without affecting Rab27b mRNA stability. In MEG-01 cells transfection with constitutively active (S422) (D) SGK1 but not with inactive (K127) (N) SGK1 significantly enhanced Rab27b mRNA levels. Sgk1(-/-) megakaryocytes show significantly reduced expression of Rab27b and serotonin/CD63 levels compared with sgk1(+/+) megakaryocytes. Proteome analysis identified nine further vesicular transport proteins regulated by SGK1, which may have an impact on impaired platelet granule biogenesis in sgk1(-/-) platelets independent of Rab27b. CONCLUSIONS The present observations identify SGK1 as a novel powerful regulator of platelet dense granule biogenesis, platelet secretion and thrombus formation. SGK1 is at least partially effective because it regulates transcription of Rab27b in megakaryocytes.
Collapse
Affiliation(s)
- B Walker
- Department of Cardiology and Cardiovascular Medicine, University of Tübingen, Tübingen, Germany
- Department of Physiology, University of Tübingen, Tübingen, Germany
| | - E Schmid
- Department of Pediatric Surgery and Pediatric Urology, University Children's Hospital Tübingen, Tübingen, Germany
| | - A Russo
- Department of Physiology, University of Tübingen, Tübingen, Germany
| | - E-M Schmidt
- Department of Physiology, University of Tübingen, Tübingen, Germany
| | - O Burk
- Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany
| | - P Münzer
- Department of Cardiology and Cardiovascular Medicine, University of Tübingen, Tübingen, Germany
- Department of Physiology, University of Tübingen, Tübingen, Germany
| | - A Velic
- Proteom Center Tübingen, University of Tübingen, Tübingen, Germany
| | - B Macek
- Proteom Center Tübingen, University of Tübingen, Tübingen, Germany
| | - M Schaller
- Department of Dermatology, University Hospital Tübingen, Tübingen, Germany
| | - M Schwab
- Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany
- Department of Clinical Pharmacology, University Hospital, Tübingen, Germany
| | - M C Seabra
- Molecular Medicine, National Heart and Lung Institute, Imperial College London, London, UK
| | - M Gawaz
- Department of Cardiology and Cardiovascular Medicine, University of Tübingen, Tübingen, Germany
| | - F Lang
- Department of Physiology, University of Tübingen, Tübingen, Germany
| | - O Borst
- Department of Cardiology and Cardiovascular Medicine, University of Tübingen, Tübingen, Germany
- Department of Physiology, University of Tübingen, Tübingen, Germany
| |
Collapse
|
44
|
VAMP-7 links granule exocytosis to actin reorganization during platelet activation. Blood 2015; 126:651-60. [PMID: 25999457 DOI: 10.1182/blood-2014-12-618744] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 05/09/2015] [Indexed: 01/30/2023] Open
Abstract
Platelet activation results in profound morphologic changes accompanied by release of granule contents. Recent evidence indicates that fusion of granules with the plasma membrane during activation provides auxiliary membrane to cover growing actin structures. Yet little is known about how membrane fusion is coupled with actin reorganization. Vesicle-associated membrane protein (VAMP)-7 is found on platelet vesicles and possesses an N-terminal longin domain capable of linking exocytosis to cytoskeletal remodeling. We have evaluated platelets from VAMP-7(-/-) mice to determine whether this VAMP isoform contributes to granule release and platelet spreading. VAMP-7(-/-) platelets demonstrated a partial defect in dense granule exocytosis and impaired aggregation. α Granule exocytosis from VAMP-7(-/-) platelets was diminished both in vitro and in vivo during thrombus formation. Consistent with a role of VAMP-7 in cytoskeletal remodeling, spreading on matrices was decreased in VAMP-7(-/-) platelets compared to wild-type controls. Immunoprecipitation of VAMP-7 revealed an association with VPS9-domain ankyrin repeat protein (VARP), an adaptor protein that interacts with both membrane-bound and cytoskeleton proteins and with Arp2/3. VAMP-7, VARP, and Arp2/3 localized to the platelet periphery during spreading. These studies demonstrate that VAMP-7 participates in both platelet granule secretion and spreading and suggest a mechanism whereby VAMP-7 links granule exocytosis with actin reorganization.
Collapse
|
45
|
Södergren AL, Svensson Holm ACB, Ramström S, Lindström EG, Grenegård M, Öllinger K. Thrombin-induced lysosomal exocytosis in human platelets is dependent on secondary activation by ADP and regulated by endothelial-derived substances. Platelets 2015; 27:86-92. [PMID: 25970449 DOI: 10.3109/09537104.2015.1042446] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Exocytosis of lysosomal contents from platelets has been speculated to participate in clearance of thrombi and vessel wall remodelling. The mechanisms that regulate lysosomal exocytosis in platelets are, however, still unclear. The aim of this study was to identify the pathways underlying platelet lysosomal secretion and elucidate how this process is controlled by platelet inhibitors. We found that high concentrations of thrombin induced partial lysosomal exocytosis as assessed by analysis of the activity of released N-acetyl-β-glucosaminidase (NAG) and by identifying the fraction of platelets exposing the lysosomal-associated membrane protein (LAMP)-1 on the cell surface by flow cytometry. Stimulation of thrombin receptors PAR1 or PAR4 with specific peptides was equally effective in inducing LAMP-1 surface expression. Notably, lysosomal exocytosis in response to thrombin was significantly reduced if the secondary activation by ADP was inhibited by the P2Y12 antagonist cangrelor, while inhibition of thromboxane A2 formation by treatment with acetylsalicylic acid was of minor importance in this regard. Moreover, the NO-releasing drug S-nitroso-N-acetyl penicillamine (SNAP) or the cyclic AMP-elevating eicosanoid prostaglandin I2 (PGI2) significantly suppressed lysosomal exocytosis. We conclude that platelet inhibitors that mimic functional endothelium such as PGI2 or NO efficiently counteract lysosomal exocytosis. Furthermore, we suggest that secondary release of ADP and concomitant signaling via PAR1/4- and P2Y12 receptors is important for efficient platelet lysosomal exocytosis by thrombin.
Collapse
Affiliation(s)
- Anna L Södergren
- a Department of Clinical and Experimental Medicine, Faculty of Health Sciences , Linköping University , Linköping , Sweden
| | - Ann-Charlotte B Svensson Holm
- a Department of Clinical and Experimental Medicine, Faculty of Health Sciences , Linköping University , Linköping , Sweden
| | - Sofia Ramström
- a Department of Clinical and Experimental Medicine, Faculty of Health Sciences , Linköping University , Linköping , Sweden
| | - Eva G Lindström
- b Department of Medical and Health Sciences, Faculty of Health Sciences , Linköping University , Linköping , Sweden , and
| | - Magnus Grenegård
- b Department of Medical and Health Sciences, Faculty of Health Sciences , Linköping University , Linköping , Sweden , and
- c Department of Clinical Medicine , School of Health Sciences, Örebro University , Örebro , Sweden
| | - Karin Öllinger
- a Department of Clinical and Experimental Medicine, Faculty of Health Sciences , Linköping University , Linköping , Sweden
| |
Collapse
|
46
|
Lillicrap D. Syntaxin-binding protein 5 exocytosis regulation: differential role in endothelial cells and platelets. J Clin Invest 2014; 124:4231-3. [PMID: 25244090 DOI: 10.1172/jci77511] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Details of the pathophysiologic mechanisms that underlie complex disorders, such as the thrombo-occlusive events associated with myocardial infarction, stroke, and venous thromboembolism, are challenging to address. Recent advances have been made through the application of genome-wide association studies (GWAS) to identify genetic loci associated with plasma levels of procoagulant proteins and risk of thrombotic disease. GWAS have consistently identified the gene encoding syntaxin-binding protein 5 (STXBP5) in this context. STXBP5 is expressed in both endothelium and platelets, and SNPs within the STXBP5 locus have been associated with plasma levels of vWF and increased venous thrombosis risk. In this issue of the JCI, two complementary reports from the laboratories of Charles Lowenstein and Sidney Whiteheart describe studies that highlight the complexity of the function of STXBP5 in control of storage granule development and exocytosis in platelets and endothelium. Together, these studies demonstrate that STXBP5 differentially regulates exocytosis in these two cell types. While STXBP5 facilitates granule release from platelets, it inhibits secretion from the Weibel-Palade bodies (WPBs) of endothelial cells.
Collapse
|