1
|
Xiao J, Li H, Li X, Lei H, Li Z, Li C. Establishment of a nomogram-based prognostic model (LASSO-Cox regression) for predicting platelet storage lesions under different storage conditions. Front Mol Biosci 2025; 12:1561114. [PMID: 40230453 PMCID: PMC11994887 DOI: 10.3389/fmolb.2025.1561114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Accepted: 03/17/2025] [Indexed: 04/16/2025] Open
Abstract
Introduction Platelet concentrates (PCs) are critical blood products used for transfusion, but stored platelets often experience quality deterioration, resulting in reduced efficacy post-transfusion. Currently, the lack of effective prediction models hinders the assessment of platelet storage quality. To address this, we developed a miRNA-based prognosis prediction model that comprehensively evaluates platelet quality under diverse storage conditions, offering valuable insights into platelet shelf life. Methods We enrolled 249 eligible PC samples, divided into a training dataset and internal validation dataset (7:3). Through microRNA sequencing, we identified 13 differentially expressed miRNAs with platelets storage lesions (PSLs). Leveraging the LASSO-Cox regression model, we constructed a nomogram-based classifier based on the association between miRNA expression and the duration of PSLs-free survival. Performance evaluation using measures like concordance index, area under the curve, calibration curves, and decision curve analyses to confirm the model's robustness. Results The nomogram classifier, incorporating miRNAs (miR-4485-3p, miR-12136, miR-25-5p, miR-148b-5p) and storage method, effectively categorized PCs into high-risk and low-risk groups. Notably, significant differences in PSLs-free survival were observed across all datasets, underscoring the precision and accuracy of our nomogram-based model. Discussion This innovative classifier provides clinicians with a reliable tool to predict PSLs occurrence in PCs stored under different methods, facilitating improved clinical decision-making.
Collapse
Affiliation(s)
- Jun Xiao
- Department of Blood Transfusion, Air Force Medical Center, Air force medical University, Beijing, China
| | - Huimin Li
- Department of Blood Transfusion, Air Force Medical Center, Air force medical University, Beijing, China
| | - Xiaowei Li
- Department of Blood Transfusion, Air Force Medical Center, Air force medical University, Beijing, China
| | - Huifen Lei
- Department of Blood Transfusion, Air Force Medical Center, Air force medical University, Beijing, China
| | - Zhicai Li
- The Fifth School of Clinical Medicine, Anhui Medical University, Hefei, China
| | - Cuiying Li
- Department of Blood Transfusion, Air Force Medical Center, Air force medical University, Beijing, China
- The Fifth School of Clinical Medicine, Anhui Medical University, Hefei, 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
|
Pastrovic F, Novak R, Grgurevic I, Hrkac S, Salai G, Zarak M, Grgurevic L. Serum proteomic profiling of patients with compensated advanced chronic liver disease with and without clinically significant portal hypertension. PLoS One 2024; 19:e0301416. [PMID: 38603681 PMCID: PMC11008873 DOI: 10.1371/journal.pone.0301416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 03/16/2024] [Indexed: 04/13/2024] Open
Abstract
INTRODUCTION Portal hypertension (PH) drives the progression of liver cirrhosis to decompensation and death. Hepatic venous pressure gradient (HVPG) measurement is the standard of PH quantification, and HVPG≥10 mmHg defines clinically significant PH (CSPH). We performed proteomics-based serum profiling to search for a proteomic signature of CSPH in patients with compensated advanced chronic liver disease (cACLD). MATERIALS AND METHODS Consecutive patients with histologically confirmed cACLD and results of HVPG measurements were prospectively included. Serum samples were pooled according to the presence/absence of CSPH and analysed by liquid chromatography-mass spectrometry. Gene set enrichment analysis was performed, followed by comprehensive literature review for proteins identified with the most striking difference between the groups. RESULTS We included 48 patients (30 with, and 18 without CSPH). Protein CD44, involved in the inflammatory response, vascular endothelial growth factor C (VEGF-C) and lymphatic vessel endothelial hyaluronan receptor-1 (LYVE-1), both involved in lymphangiogenesis were found solely in the CSPH group. Although identified in both groups, proteins involved in neutrophil extracellular traps (NET) formation, as well as tenascin C, autotaxin and nephronectin which mediate vascular contractility and lymphangiogenesis were more abundant in CSPH. DISCUSSION AND CONCLUSION We propose that altered inflammatory response, including NET formation, vascular contractility and formation of new lymph vessels are key steps in PH development. Proteins such as CD44, VEGF-C, LYVE-1, tenascin C, Plasminogen activator inhibitor 1, Nephronectin, Bactericidal permeability-increasing protein, Autotaxin, Myeloperoxidase and a disintegrin and metalloproteinase with thrombospondin motifs-like protein 4 might be considered for further validation as potential therapeutic targets and candidate biomarkers of CSPH in cACLD.
Collapse
Affiliation(s)
- Frane Pastrovic
- Department of Gastroenterology, Hepatology and Clinical Nutrition, Laboratory for Liver Diseases and Portal Hypertension, University Hospital Dubrava, Zagreb, Croatia
- Faculty of Pharmacy and Biochemistry, University of Zagreb, Zagreb, Croatia
| | - Rudjer Novak
- Department of Proteomics, Center for Translational and Clinical Research, University of Zagreb School of Medicine, Zagreb, Croatia
- University of Zagreb, School of Medicine, Zagreb, Croatia
- Biomedical Research Center Salata, University of Zagreb, School of Medicine, Zagreb, Croatia
| | - Ivica Grgurevic
- Department of Gastroenterology, Hepatology and Clinical Nutrition, Laboratory for Liver Diseases and Portal Hypertension, University Hospital Dubrava, Zagreb, Croatia
- Faculty of Pharmacy and Biochemistry, University of Zagreb, Zagreb, Croatia
- University of Zagreb, School of Medicine, Zagreb, Croatia
| | - Stela Hrkac
- Department of Clinical Immunology, Allergology and Rheumatology, University Hospital Dubrava, Zagreb, Croatia
| | - Grgur Salai
- Department of Pulmonology, University Hospital Dubrava, Zagreb, Croatia
| | - Marko Zarak
- Faculty of Pharmacy and Biochemistry, University of Zagreb, Zagreb, Croatia
- Clinical Department of Laboratory Diagnostics, University Hospital Dubrava, Zagreb, Croatia
| | - Lovorka Grgurevic
- Department of Proteomics, Center for Translational and Clinical Research, University of Zagreb School of Medicine, Zagreb, Croatia
- Biomedical Research Center Salata, University of Zagreb, School of Medicine, Zagreb, Croatia
- Department of Anatomy, ˝Drago Perovic˝, School of Medicine, University of Zagreb, Zagreb, Croatia
| |
Collapse
|
4
|
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
|
5
|
Cox AA, Liu A, Ng CJ. Clusterin knockdown has effects on intracellular and secreted von Willebrand factor in human umbilical vein endothelial cells. PLoS One 2024; 19:e0298133. [PMID: 38363768 PMCID: PMC10871512 DOI: 10.1371/journal.pone.0298133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 01/17/2024] [Indexed: 02/18/2024] Open
Abstract
Alterations in von Willebrand factor (VWF) have an important role in human health and disease. Deficiency of VWF is associated with symptoms of bleeding and excesses of VWF are associated with thrombotic outcomes. Understanding the mechanisms that drive VWF regulation can lead to a better understanding of modulation of VWF levels in humans. We identified clusterin (CLU) as a potential candidate regulator of VWF based on a single cell RNA sequencing (scRNA-seq) analysis in control endothelial cells (ECs) and von Willebrand disease (VWD) endothelial colony-forming-cells (ECFCs). We found that patients with deficiencies of VWF (von Willebrand disease, VWD) had decreased CLU expression and ECs with low VWF expression also had low CLU expression. Based on these findings, we sought to evaluate the role of CLU in the regulation of VWF, specifically as it relates to VWD. As CLU is primarily thought to be a golgi protein involved in protein chaperoning, we hypothesized that knockdown of CLU would lead to decreases in VWF and alterations in Weibel-Palade bodies (WPBs). We used both siRNA- and CRISPR-Cas9-based approaches to modulate CLU in human umbilical vein endothelial cells (HUVECs) and evaluated VWF protein levels, VWF mRNA copy number, and WPB quantity and size. We demonstrated that siRNA-based knockdown of CLU resulted in decreases in VWF content in cellular lysates and supernatants, but no significant change in WPB quantity or size. A CRISPR-Cas9-based knockdown of CLU demonstrated similar findings of decreases in intracellular VWF content but no significant change in WPB quantity or size. Our data suggests that CLU knockdown is associated with decreases in cellular VWF content but does not affect VWF mRNA levels or WPB quantity or size.
Collapse
Affiliation(s)
- Allaura A. Cox
- Department of Pediatrics, University of Colorado–Anschutz Medical Campus, Aurora, CO, United States of America
| | - Alice Liu
- Department of Bioengineering, Washington University, St. Louis, MO, United States of America
| | - Christopher J. Ng
- Department of Pediatrics, University of Colorado–Anschutz Medical Campus, Aurora, CO, United States of America
| |
Collapse
|
6
|
Han J, Li‐Gao R, de Mutsert R, Rosendaal FR, van Hylckama Vlieg A. Association between venous thromboembolism-associated genetic variants, coagulation factor levels, and thrombin generation potential. EJHAEM 2024; 5:47-54. [PMID: 38406509 PMCID: PMC10887265 DOI: 10.1002/jha2.863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 01/23/2024] [Accepted: 01/26/2024] [Indexed: 02/27/2024]
Abstract
Recently three large meta-analyses of genome-wide association studies for venous thromboembolism (VTE) identified over 130 genetic variants. However, mechanisms by which newly identified and therefore underexplored VTE-associated genetic variants influence VTE remain unclear. To elucidate the mechanism, we investigated the association between 61 newly identified VTE-associated genetic variants and the levels of coagulation factor (F) VIII, FIX, FXI, and fibrinogen as well as thrombin generation parameters (lag time, peak, endogenous thrombin potential, time-to-peak, and velocity), which are well-known biological traits associated with VTE. This study was conducted on 5341 participants of the Netherlands Epidemiology of Obesity study. The associations between VTE-associated genetic variants and coagulation factor levels and thrombin generation parameters were examined using linear regression analyses, adjusted for age, sex, body mass index, oral contraceptive use, hormone replacement therapy, and menopausal status. Of 61 genetic variants, 33 were associated with one or more of the coagulation factor levels and thrombin generation parameters. Following multiple testing corrections, five genetic variants remained significant, of which MAP1A rs55707100 exhibited the most robust association with thrombin generation parameters and FXI levels (β = -5.33%, 95% confidence interval: -8.44, -2.22). Our findings shed light on the underlying mechanisms by which these genetic variants influence the risk of VTE.
Collapse
Affiliation(s)
- Jihee Han
- Department of Clinical EpidemiologyLeiden University Medical CenterLeidenthe Netherlands
| | - Ruifang Li‐Gao
- Department of Clinical EpidemiologyLeiden University Medical CenterLeidenthe Netherlands
| | - Renée de Mutsert
- Department of Clinical EpidemiologyLeiden University Medical CenterLeidenthe Netherlands
| | - Frits R. Rosendaal
- Department of Clinical EpidemiologyLeiden University Medical CenterLeidenthe Netherlands
| | | |
Collapse
|
7
|
LOWENSTEIN CHARLESJ. GENETIC DETERMINANTS OF THROMBOSIS. TRANSACTIONS OF THE AMERICAN CLINICAL AND CLIMATOLOGICAL ASSOCIATION 2024; 134:230-238. [PMID: 39135563 PMCID: PMC11316864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 08/15/2024]
Abstract
Venous thromboembolism (VTE) is a major cause of morbidity and mortality in the United States. VTE is caused by genetic and acquired conditions, but the genetic variants that increase the risk of VTE are not fully characterized. Recent genome-wide association studies (GWAS) have discovered novel genetic loci linked to VTE. Some of these loci have been characterized, uncovering new pathways that regulate VTE. Functional characterization of candidate genes discovered by GWAS may reveal new therapeutic targets to treat and prevent abnormal thrombosis or bleeding.
Collapse
|
8
|
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: 2] [Impact Index Per Article: 1.0] [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
|
9
|
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
|
10
|
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] [Grants] [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
| |
Collapse
|
11
|
Differential Expression of microRNAs in Serum of Patients with Chronic Painful Polyneuropathy and Healthy Age-Matched Controls. Biomedicines 2023; 11:biomedicines11030764. [PMID: 36979743 PMCID: PMC10045018 DOI: 10.3390/biomedicines11030764] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 02/24/2023] [Accepted: 02/28/2023] [Indexed: 03/06/2023] Open
Abstract
Polyneuropathies (PNP) are the most common type of disorder of the peripheral nervous system in adults. However, information on microRNA expression in PNP is lacking. Following microRNA sequencing, we compared the expression of microRNAs in the serum of patients experiencing chronic painful PNP with healthy age-matched controls. We have been able to identify four microRNAs (hsa-miR-3135b, hsa-miR-584-5p, hsa-miR-12136, and hsa-miR-550a-3p) that provide possible molecular links between degenerative processes, blood flow regulation, and signal transduction, that eventually lead to PNP. In addition, these microRNAs are discussed regarding the targeting of proteins that are involved in high blood flow/pressure and neural activity dysregulations/disbalances, presumably resulting in PNP-typical symptoms such as chronical numbness/pain. Within our study, we have identified four microRNAs that may serve as potential novel biomarkers of chronic painful PNP, and that may potentially bear therapeutic implications.
Collapse
|
12
|
Kat M, Margadant C, Voorberg J, Bierings R. Dispatch and delivery at the ER-Golgi interface: how endothelial cells tune their hemostatic response. FEBS J 2022; 289:6863-6870. [PMID: 35246944 PMCID: PMC9790534 DOI: 10.1111/febs.16421] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 03/01/2022] [Accepted: 03/03/2022] [Indexed: 01/13/2023]
Abstract
Von Willebrand factor (VWF) is a glycoprotein that is secreted into the circulation and controls bleeding by promoting adhesion and aggregation of blood platelets at sites of vascular injury. Substantial inter-individual variation in VWF plasma levels exists among the healthy population. Prior to secretion, VWF polymers are assembled and condensed into helical tubules, which are packaged into Weibel-Palade bodies (WPBs), a highly specialized post-Golgi storage compartment in vascular endothelial cells. In the inherited bleeding disorder Von Willebrand disease (VWD), mutations in the VWF gene can cause qualitative or quantitative defects, limiting protein function, secretion, or plasma survival. However, pathogenic VWF mutations cannot be found in all VWD cases. Although an increasing number of genetic modifiers have been identified, even more rare genetic variants that impact VWF plasma levels likely remain to be discovered. Here, we summarize recent evidence that modulation of the early secretory pathway has great impact on the biogenesis and release of WPBs. Based on these findings, we propose that rare, as yet unidentified quantitative trait loci influencing intracellular VWF transport contribute to highly variable VWF levels in the population. These may underlie the thrombotic complications linked to high VWF levels, as well as the bleeding tendency in individuals with low VWF levels.
Collapse
Affiliation(s)
- Marije Kat
- Molecular HematologySanquin Research and Landsteiner LaboratoryAmsterdam University Medical CenterUniversity of AmsterdamThe Netherlands
| | - Coert Margadant
- Angiogenesis laboratoryCancer Center AmsterdamAmsterdam University Medical Center location VUmcThe Netherlands
| | - Jan Voorberg
- Molecular HematologySanquin Research and Landsteiner LaboratoryAmsterdam University Medical CenterUniversity of AmsterdamThe Netherlands,Experimental Vascular MedicineAmsterdam University Medical CenterUniversity of AmsterdamThe Netherlands
| | - Ruben Bierings
- Hematology, Erasmus University Medical CenterRotterdamThe Netherlands
| |
Collapse
|
13
|
Delshad M, Sanaei MJ, Pourbagheri-Sigaroodi A, Bashash D. Host genetic diversity and genetic variations of SARS-CoV-2 in COVID-19 pathogenesis and the effectiveness of vaccination. Int Immunopharmacol 2022; 111:109128. [PMID: 35963158 PMCID: PMC9359488 DOI: 10.1016/j.intimp.2022.109128] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 07/15/2022] [Accepted: 08/03/2022] [Indexed: 12/14/2022]
Abstract
The severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), responsible for the outbreak of coronavirus disease 2019 (COVID-19), has shown a vast range of clinical manifestations from asymptomatic to life-threatening symptoms. To figure out the cause of this heterogeneity, studies demonstrated the trace of genetic diversities whether in the hosts or the virus itself. With this regard, this review provides a comprehensive overview of how host genetic such as those related to the entry of the virus, the immune-related genes, gender-related genes, disease-related genes, and also host epigenetic could influence the severity of COVID-19. Besides, the mutations in the genome of SARS-CoV-2 __leading to emerging of new variants__ per se affect the affinity of the virus to the host cells and enhance the immune escape capacity. The current review discusses these variants and also the latest data about vaccination effectiveness facing the most important variants.
Collapse
Affiliation(s)
- Mahda Delshad
- Department of Laboratory Sciences, School of Allied Medical Sciences, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mohammad-Javad Sanaei
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Atieh Pourbagheri-Sigaroodi
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Davood Bashash
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
14
|
Sadler B, Castaman G, O’Donnell JS. von Willebrand disease and von Willebrand factor. Haemophilia 2022; 28 Suppl 4:11-17. [PMID: 35521725 PMCID: PMC9094051 DOI: 10.1111/hae.14547] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 02/28/2022] [Accepted: 02/28/2022] [Indexed: 12/31/2022]
Abstract
Progress in both basic and translational research into the molecular mechanisms of VWD can be seen in multiple fields. GENETICS OF VWD In the past several decades, knowledge of the underlying pathogenesis of von Willebrand disease (VWD) has increased tremendously, thanks in no small part to detailed genetic mapping of the von Willebrand Factor (VWF) gene and advances in genetic and bioinformatic technology. However, these advances do not always easily translate into improved management for patients with VWD and low-VWF levels. VWD AND PREGNANCY For example, the treatment of pregnant women with VWD both pre- and postpartum can be complicated. While knowledge of the VWF genotype at some amino acid positions can aid in knowledge of who may be at increased risk of thrombocytopenia or insufficient increase in VWF levels during pregnancy, in many cases, VWF levels and bleeding severity is highly heterogeneous, making monitoring recommended during pregnancy to optimize treatment strategies. VWF AND COVID-19: New challenges related to the consequences of dysregulation of hemostasis continue to be discovered. The ongoing COVID-19 pandemic has highlighted that VWF has additional biological roles in the regulation of inflammatory disorders and angiogenesis, disruption of which may contribute to COVID-19 induced vasculopathy. Increased endothelial cell activation and Weibel-Palade body exocytosis in severe COVID-19 lead to markedly increased plasma VWF levels. Coupled with impairment of normal ADAMTS13 multimer regulation, these data suggest a role for VWF in the pathogenesis underlying pulmonary microvascular angiopathy in severe COVID-19. CONCLUSION With the increased affordability and availability of next-generation sequencing techniques, as well as a push towards a multi-omic approach and personalized medicine in human genetics, there is hope that translational research will improve VWD patient outcomes.
Collapse
Affiliation(s)
- Brooke Sadler
- Washington University School of Medicine, Department of Pediatrics, Division of Hematology/Oncology, St. Louis, MO USA
| | - Giancarlo Castaman
- Center for Bleeding Disorders and Coagulation, Department of Oncology, Careggi University Hospital, Florence, Italy
| | - James S. O’Donnell
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin 2, Ireland,National Coagulation Centre, St James’s Hospital, Dublin, Ireland
| |
Collapse
|
15
|
Ng CJ, Liu A, Venkataraman S, Ashworth KJ, Baker CD, O'Rourke R, Vibhakar R, Jones KL, Di Paola J. Single-cell transcriptional analysis of human endothelial colony-forming cells from patients with low VWF levels. Blood 2022; 139:2240-2251. [PMID: 35143643 PMCID: PMC8990376 DOI: 10.1182/blood.2021010683] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 07/21/2021] [Indexed: 11/20/2022] Open
Abstract
von Willebrand factor (VWF) plays a key role in normal hemostasis, and deficiencies of VWF lead to clinically significant bleeding. We sought to identify novel modifiers of VWF levels in endothelial colony-forming cells (ECFCs) using single-cell RNA sequencing (scRNA-seq). ECFCs were isolated from patients with low VWF levels (plasma VWF antigen levels between 30 and 50 IU/dL) and from healthy controls. Human umbilical vein endothelial cells were used as an additional control cell line. Cells were characterized for their Weibel Palade body (WPB) content and VWF release. scRNA-seq of all cell lines was performed to evaluate for gene expression heterogeneity and for candidate modifiers of VWF regulation. Candidate modifiers identified by scRNA-seq were further characterized with small-interfering RNA (siRNA) experiments to evaluate for effects on VWF. We observed that ECFCs derived from patients with low VWF demonstrated alterations in baseline WPB metrics and exhibit impaired VWF release. scRNA-seq analyses of these endothelial cells revealed overall decreased VWF transcription, mosaicism of VWF expression, and genes that are differentially expressed in low VWF ECFCs and control endothelial cells (control ECs). An siRNA screen of potential VWF modifiers provided further evidence of regulatory candidates, and 1 such candidate, FLI1, alters the transcriptional activity of VWF. In conclusion, ECFCs from individuals with low VWF demonstrate alterations in their baseline VWF packaging and release compared with control ECs. scRNA-seq revealed alterations in VWF transcription, and siRNA screening identified multiple candidate regulators of VWF.
Collapse
Affiliation(s)
- Christopher J Ng
- Department of Pediatrics, University of Colorado and Children's Hospital Colorado, Aurora, CO
- University of Colorado, Anschutz Medical Campus, Aurora, CO
| | - Alice Liu
- Department of Pediatrics, University of Colorado and Children's Hospital Colorado, Aurora, CO
- University of Colorado, Anschutz Medical Campus, Aurora, CO
| | - Sujatha Venkataraman
- Department of Pediatrics, University of Colorado and Children's Hospital Colorado, Aurora, CO
- University of Colorado, Anschutz Medical Campus, Aurora, CO
| | - Katrina J Ashworth
- Division of Hematology Oncology, Department of Pediatrics, Washington University School of Medicine in St. Louis, St. Louis, MO; and
| | - Christopher D Baker
- Department of Pediatrics, University of Colorado and Children's Hospital Colorado, Aurora, CO
- University of Colorado, Anschutz Medical Campus, Aurora, CO
| | - Rebecca O'Rourke
- Department of Pediatrics, University of Colorado and Children's Hospital Colorado, Aurora, CO
- University of Colorado, Anschutz Medical Campus, Aurora, CO
| | - Rajeev Vibhakar
- Department of Pediatrics, University of Colorado and Children's Hospital Colorado, Aurora, CO
- University of Colorado, Anschutz Medical Campus, Aurora, CO
| | - Kenneth L Jones
- Department of Cell Biology and
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Jorge Di Paola
- Division of Hematology Oncology, Department of Pediatrics, Washington University School of Medicine in St. Louis, St. Louis, MO; and
| |
Collapse
|
16
|
Finding the “switch” in platelet activation prediction of key mediators involved in reversal of platelet activation using a novel network biology approach. J Proteomics 2022; 261:104577. [DOI: 10.1016/j.jprot.2022.104577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 02/24/2022] [Accepted: 03/23/2022] [Indexed: 11/15/2022]
|
17
|
Salum GM, el Meguid MA, Abelhafez TH, Medhat E, Abdel Aziz AO, Dawood R. Evaluation of seven gene signature for predicting HCV recurrence post-liver transplantation. J Genet Eng Biotechnol 2021; 19:174. [PMID: 34757522 PMCID: PMC8581076 DOI: 10.1186/s43141-021-00266-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Accepted: 10/05/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND Orthotropic liver transplantation (OLT) offers a therapeutic choice for hepatocellular carcinoma (HCC) patients. The poor outcome of liver transplantation is HCV recurrence. Several genome-wide associated studies (GWAS) have reported many genetic variants to be associated with HCV recurrence. Seven gene polymorphisms formed a cirrhosis risk score (CRS) signature that could be used to distinguish chronic HCV patients at high risk from those at low risk for cirrhosis in non-transplant patients. This study aims to examine the association of CRS score and other clinical parameters with the probability for HCC emergence and/or the rate of HCV recurrence following liver transplantation. RESULTS Seven gene polymorphisms, forming the CRS, were genotyped by real-time PCR using allelic discrimination protocol in 199 end-stage liver disease patients (79 child A, 43 child B, and 77child C), comprising 106 patients who encountered liver transplantation. Recipient CRS scores were correlated with HCV recurrence (HCV-Rec) at the end of the third year after OLT. Around 81% (39) recipients with low steatosis (LS; < 3.5%) donor percentage revealed no HCV recurrence (non-Rec) (p<0.001). CRS score could distinguish between child A, child B, and child C only at the low-risk group. Among the HCV Rec group 27% (8/30), 40% (12/30), and 33% (10/30) fell into the high, moderate, and low CRS risk groups, respectively. Stepwise logistic regression evinced two features more likely to be seen in HCV-Rec patients: abnormal ALT [OR, 1.1; 95% CI, 1.02-1.2] and donor steatosis >3.5% [OR, 46.07; 95% CI, 1.5-1407.8]. CONCLUSIONS Accordingly, the CRS score seems to be less useful to predict HCV recurrence after OLT. ALT and donor steatosis (exceed 3.5%) can significantly promote the HCV recurrence post-OLT. Moreover, the combination of MMF and CNI positively heightens HCV recurrence.
Collapse
Affiliation(s)
- Ghada M. Salum
- Department of Microbial Biotechnology, Genetic Engineering Division, National Research Centre, Dokki, P.O. 12622, Giza, Egypt
| | - Mai Abd el Meguid
- Department of Microbial Biotechnology, Genetic Engineering Division, National Research Centre, Dokki, P.O. 12622, Giza, Egypt
| | - Tawfeek H. Abelhafez
- Department of Microbial Biotechnology, Genetic Engineering Division, National Research Centre, Dokki, P.O. 12622, Giza, Egypt
| | - Eman Medhat
- Department of Endemic Medicine and Hepato-gastroenterology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Ashraf O. Abdel Aziz
- Department of Endemic Medicine and Hepato-gastroenterology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Reham Dawood
- Department of Microbial Biotechnology, Genetic Engineering Division, National Research Centre, Dokki, P.O. 12622, Giza, Egypt
| |
Collapse
|
18
|
Ranjan J, Ravindra A, Mishra B. Gender and genetic factors impacting COVID-19 severity. J Family Med Prim Care 2021; 10:3956-3963. [PMID: 35136752 PMCID: PMC8797126 DOI: 10.4103/jfmpc.jfmpc_769_21] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 07/08/2021] [Accepted: 07/10/2021] [Indexed: 11/04/2022] Open
Abstract
COVID-19 pandemic is a cause of global concern and is impacting lives and economy globally. Infection due to SARS-CoV-2 leads to varied clinical manifestations, which can vary from asymptomatic to severe acute respiratory syndrome and death. The clinical features are proposed to depend upon various host factors, namely, gender and genetic factors. The significantly high mortality among males has revealed the role of gender, androgens, age, genetics, and risk factors in determining the severity of COVID-19 among the population. The interplay of various host factors and their association with clinically severe infections is crucial for our understanding of COVID-19 pathogenesis. A PubMed and Google scholar search was made using keywords such as "COVID-19 + sex differences," "COVID-19 + androgens," "COVID-19 + ACE2 receptor," and "COVID-19 + smoking alcoholism pregnancy." The articles which highlight the association of gender and genetic factors to COVID-19 were selected and included in our study. It is mainly the primary care or family physicians who act as the first contact of COVID-19 patients. With the recent increase in SARS-CoV-2 infections in the Indian subcontinent and probability of upcoming surges, it has become imperative to understand its interaction with the various gender and genetic factors to devise effective triage and management protocols. Our review highlights the possible mechanisms by which these factors impact the severity of COVID-19. A better understanding of these factors will be of immense help to primary care physicians.
Collapse
Affiliation(s)
- Jai Ranjan
- Department of Microbiology, All India Institute of Medical Sciences, Bhubaneswar, Odisha, India
| | - Akshatha Ravindra
- Department of Microbiology, All India Institute of Medical Sciences, Bhubaneswar, Odisha, India
| | - Baijayantimala Mishra
- Department of Microbiology, All India Institute of Medical Sciences, Bhubaneswar, Odisha, India
- Address for correspondence: Dr. Baijayantimala Mishra, Department of Microbiology, All India Institute of Medical Sciences, Bhubaneswar - 751 019, Odisha, India. E-mail:
| |
Collapse
|
19
|
Secretome and Tunneling Nanotubes: A Multilevel Network for Long Range Intercellular Communication between Endothelial Cells and Distant Cells. Int J Mol Sci 2021; 22:ijms22157971. [PMID: 34360735 PMCID: PMC8347715 DOI: 10.3390/ijms22157971] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 07/14/2021] [Accepted: 07/22/2021] [Indexed: 02/07/2023] Open
Abstract
As a cellular interface between the blood and tissues, the endothelial cell (EC) monolayer is involved in the control of key functions including vascular tone, permeability and homeostasis, leucocyte trafficking and hemostasis. EC regulatory functions require long-distance communications between ECs, circulating hematopoietic cells and other vascular cells for efficient adjusting thrombosis, angiogenesis, inflammation, infection and immunity. This intercellular crosstalk operates through the extracellular space and is orchestrated in part by the secretory pathway and the exocytosis of Weibel Palade Bodies (WPBs), secretory granules and extracellular vesicles (EVs). WPBs and secretory granules allow both immediate release and regulated exocytosis of messengers such as cytokines, chemokines, extracellular membrane proteins, coagulation or growth factors. The ectodomain shedding of transmembrane protein further provide the release of both receptor and ligands with key regulatory activities on target cells. Thin tubular membranous channels termed tunneling nanotubes (TNTs) may also connect EC with distant cells. EVs, in particular exosomes, and TNTs may contain and transfer different biomolecules (e.g., signaling mediators, proteins, lipids, and microRNAs) or pathogens and have emerged as a major triggers of horizontal intercellular transfer of information.
Collapse
|
20
|
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
|
21
|
Hu J, Li C, Wang S, Li T, Zhang H. Genetic variants are identified to increase risk of COVID-19 related mortality from UK Biobank data. Hum Genomics 2021; 15:10. [PMID: 33536081 PMCID: PMC7856608 DOI: 10.1186/s40246-021-00306-7] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 01/15/2021] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND The severity of coronavirus disease 2019 (COVID-19) caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is highly heterogeneous. Studies have reported that males and some ethnic groups are at increased risk of death from COVID-19, which implies that individual risk of death might be influenced by host genetic factors. METHODS In this project, we consider the mortality as the trait of interest and perform a genome-wide association study (GWAS) of data for 1778 infected cases (445 deaths, 25.03%) distributed by the UK Biobank. Traditional GWAS fails to identify any genome-wide significant genetic variants from this dataset. To enhance the power of GWAS and account for possible multi-loci interactions, we adopt the concept of super variant for the detection of genetic factors. A discovery-validation procedure is used for verifying the potential associations. RESULTS We find 8 super variants that are consistently identified across multiple replications as susceptibility loci for COVID-19 mortality. The identified risk factors on chromosomes 2, 6, 7, 8, 10, 16, and 17 contain genetic variants and genes related to cilia dysfunctions (DNAH7 and CLUAP1), cardiovascular diseases (DES and SPEG), thromboembolic disease (STXBP5), mitochondrial dysfunctions (TOMM7), and innate immune system (WSB1). It is noteworthy that DNAH7 has been reported recently as the most downregulated gene after infecting human bronchial epithelial cells with SARS-CoV-2. CONCLUSIONS Eight genetic variants are identified to significantly increase the risk of COVID-19 mortality among the patients with white British ancestry. These findings may provide timely clues and potential directions for better understanding the molecular pathogenesis of COVID-19 and the genetic basis of heterogeneous susceptibility, with potential impact on new therapeutic options.
Collapse
Affiliation(s)
- Jianchang Hu
- Department of Biostatistics, Yale University, 300 George Street, Ste 523, New Haven, CT, 06511, USA
| | - Cai Li
- Department of Biostatistics, Yale University, 300 George Street, Ste 523, New Haven, CT, 06511, USA
| | - Shiying Wang
- Department of Biostatistics, Yale University, 300 George Street, Ste 523, New Haven, CT, 06511, USA
| | - Ting Li
- Department of Biostatistics, Yale University, 300 George Street, Ste 523, New Haven, CT, 06511, USA
| | - Heping Zhang
- Department of Biostatistics, Yale University, 300 George Street, Ste 523, New Haven, CT, 06511, USA.
| |
Collapse
|
22
|
Hu J, Li C, Wang S, Li T, Zhang H. Genetic variants are identified to increase risk of COVID-19 related mortality from UK Biobank data. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2020:2020.11.05.20226761. [PMID: 33200144 PMCID: PMC7668757 DOI: 10.1101/2020.11.05.20226761] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Background The severity of coronavirus disease 2019 (COVID-19) caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is highly heterogenous. Studies have reported that males and some ethnic groups are at increased risk of death from COVID-19, which implies that individual risk of death might be influenced by host genetic factors. Methods In this project, we consider the mortality as the trait of interest and perform a genome-wide association study (GWAS) of data for 1,778 infected cases (445 deaths, 25.03%) distributed by the UK Biobank. Traditional GWAS failed to identify any genome-wide significant genetic variants from this dataset. To enhance the power of GWAS and account for possible multi-loci interactions, we adopt the concept of super-variant for the detection of genetic factors. A discovery-validation procedure is used for verifying the potential associations. Results We find 8 super-variants that are consistently identified across multiple replications as susceptibility loci for COVID-19 mortality. The identified risk factors on Chromosomes 2, 6, 7, 8, 10, 16, and 17 contain genetic variants and genes related to cilia dysfunctions (DNAH7 and CLUAP1), cardiovascular diseases (DES and SPEG), thromboembolic disease (STXBP5), mitochondrial dysfunctions (TOMM7), and innate immune system (WSB1). It is noteworthy that DNAH7 has been reported recently as the most downregulated gene after infecting human bronchial epithelial cells with SARS-CoV2. Conclusions Eight genetic variants are identified to significantly increase risk of COVID-19 mortality among the patients with white British ancestry. These findings may provide timely evidence and clues for better understanding the molecular pathogenesis of COVID-19 and genetic basis of heterogeneous susceptibility, with potential impact on new therapeutic options.
Collapse
Affiliation(s)
| | | | | | - Ting Li
- Department of Biostatistics, Yale University
| | | |
Collapse
|
23
|
Zhang YH, Pan X, Zeng T, Chen L, Huang T, Cai YD. Identifying the RNA signatures of coronary artery disease from combined lncRNA and mRNA expression profiles. Genomics 2020; 112:4945-4958. [PMID: 32919019 DOI: 10.1016/j.ygeno.2020.09.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 07/28/2020] [Accepted: 09/05/2020] [Indexed: 12/23/2022]
Abstract
Coronary artery disease (CAD) is the most common cardiovascular disease. CAD research has greatly progressed during the past decade. mRNA is a traditional and popular pipeline to investigate various disease, including CAD. Compared with mRNA, lncRNA has better stability and thus may serve as a better disease indicator in blood. Investigating potential CAD-related lncRNAs and mRNAs will greatly contribute to the diagnosis and treatment of CAD. In this study, a computational analysis was conducted on patients with CAD by using a comprehensive transcription dataset with combined mRNA and lncRNA expression data. Several machine learning algorithms, including feature selection methods and classification algorithms, were applied to screen for the most CAD-related RNA molecules. Decision rules were also reported to provide a quantitative description about the effect of these RNA molecules on CAD progression. These new findings (CAD-related RNA molecules and rules) can help understand mRNA and lncRNA expression levels in CAD.
Collapse
Affiliation(s)
- Yu-Hang Zhang
- School of Life Sciences, Shanghai University, Shanghai 200444, China; Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Xiaoyong Pan
- Institute of Image Processing and Pattern Recognition, Shanghai Jiao Tong University, Key Laboratory of System Control and Information Processing, Ministry of Education of China, 200240 Shanghai, China.
| | - Tao Zeng
- Shanghai Research Center for Brain Science and Brain-Inspired Intelligence, Shanghai 201210, China.
| | - Lei Chen
- College of Information Engineering, Shanghai Maritime University, Shanghai 201306, China.
| | - Tao Huang
- Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China.
| | - Yu-Dong Cai
- School of Life Sciences, Shanghai University, Shanghai 200444, China.
| |
Collapse
|
24
|
Li Y, Zhang X, Zheng Q, Zhang Y, Ma Y, Zhu C, Yang L, Peng X, Wang Q, Wang B, Meng X, Li H, Liu J. YAP1 Inhibition in HUVECs Is Associated with Released Exosomes and Increased Hepatocarcinoma Invasion and Metastasis. MOLECULAR THERAPY-NUCLEIC ACIDS 2020; 21:86-97. [PMID: 32516736 PMCID: PMC7281784 DOI: 10.1016/j.omtn.2020.05.021] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 05/04/2020] [Accepted: 05/18/2020] [Indexed: 12/21/2022]
Abstract
Hepatocellular carcinoma is one of the most common gastrointestinal malignancies. Anti-angiogenesis therapies have recently demonstrated promise in the treatment of malignancies, although early treatment benefits may be accompanied by metastasis over time. Additional and more effective anti-angiogenic treatment modalities are therefore needed. We previously found that Yes-associated protein 1 (YAP1) expression is increased in hepatocellular carcinoma (HCC), particularly around tumor-associated blood vessels, suggesting a role in angiogenesis. The YAP1 inhibitor verteporfin is presently in anti-angiogenic clinical trials for the treatment of various cancers. Depleted YAP1 from vascular endothelial cells effectively reduced proliferation and tube formation, validating its utility as an anti-angiogenesis target. We also showed that YAP1 depletion or inhibition in vascular endothelial cells leads to increased release of exosomes containing the long non-coding RNA (lncRNA) MALAT1 into the tumor microenvironment. Direct exosomal transfer of MALAT1 to hepatic cells leads to increased hepatic cell invasion and migration via activation of extracellular signal-regulated kinase 1/2 (ERK1/2) signaling. These observations may explain the occurrence of distant tumor metastasis with YAP1-associated anti-angiogenic therapy over time. It provides insight into new pathways and treatment paradigms that may be targeted to increase the long-term success of anti-angiogenic therapies.
Collapse
Affiliation(s)
- Yan Li
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China; Department of Radiation Oncology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Xiaodong Zhang
- General Surgery Department, Beijing Friendship Hospital, Capital Medical University, Beijing, China; National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Qianqian Zheng
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Yijun Zhang
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yingbo Ma
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| | - Chen Zhu
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, China
| | - Liang Yang
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| | - Xueqiang Peng
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| | - Qi Wang
- Department of Geriatrics, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Biao Wang
- Department of Biochemistry and Molecular Biology, Academy of life sciences of China Medical University, Shenyang, China
| | - Xin Meng
- Department of Biochemistry and Molecular Biology, Academy of life sciences of China Medical University, Shenyang, China
| | - Hangyu Li
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China.
| | - Jingang Liu
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China.
| |
Collapse
|
25
|
Yu M, Du H, Wang B, Chen J, Lu F, Peng S, Sun Y, Liu N, Sun X, Shiyun D, Zhao Y, Wang Y, Zhao D, Lu F, Zhang W. Exogenous H 2S Induces Hrd1 S-sulfhydration and Prevents CD36 Translocation via VAMP3 Ubiquitylation in Diabetic Hearts. Aging Dis 2020; 11:286-300. [PMID: 32257542 PMCID: PMC7069459 DOI: 10.14336/ad.2019.0530] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 05/30/2019] [Indexed: 12/21/2022] Open
Abstract
Hydrogen sulfide (H2S) plays physiological roles in vascular tone regulation, cytoprotection, and ATP synthesis. HMG-CoA reductase degradation protein (Hrd1), an E3 ubiquitin ligase, is involved in protein trafficking. H2S may play a role in controlling fatty acid uptake in diabetic cardiomyopathy (DCM) in a manner correlated with modulation of Hrd1 S-sulfhydration; however, this role remains to be elucidated. The aim of the present study was to examine whether H2S can attenuate lipid accumulation and to explain the possible mechanisms involved in the regulation of the H2S-Hrd1/VAMP3 pathway. Db/db mice and neonatal rat cardiomyocytes treated with high glucose, palmitate and oleate were used as animal and cellular models of type 2 diabetes, respectively. The expression of cystathionine-γ-lyase (CSE), Hrd1, CD36 and VAMP3 was detected by Western blot analysis. In addition, Hrd1 was mutated at Cys115, and Hrd1 S-sulfhydration was examined using an S-sulfhydration assay. VAMP3 ubiquitylation was investigated by immunoprecipitation. Lipid droplet formation was tested by TEM, BODIPY 493/503 staining and oil red O staining. The expression of CSE and Hrd1 was decreased in db/db mice compared to control mice, whereas CD36 and VAMP3 expression was increased. NaHS administration reduced droplet formation, and exogenous H2S restored Hrd1 expression, modified S-sulfhydration, and decreased VAMP3 expression in the plasma membrane. Using LC-MS/MS analysis, we identified 85 proteins with decreased ubiquitylation, including 3 vesicle-associated membrane proteins, in the cardiac tissues of model db/db mice compared with NaHS-treated db/db mice. Overexpression of Hrd1 mutated at Cys115 diminished VAMP3 ubiquitylation, whereas it increased CD36 and VAMP3 expression and droplet formation. siRNA-mediated Hrd1 deletion increased the expression of CD36 in the cell membrane. These findings suggested that H2S regulates VAMP3 ubiquitylation via Hrd1 S-sulfhydration at Cys115 to prevent CD36 translocation in diabetes.
Collapse
Affiliation(s)
- Miao Yu
- 1Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Haining Du
- 1Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Bingzhu Wang
- 1Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Jian Chen
- 1Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Fangping Lu
- 1Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Shuo Peng
- 1Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Yu Sun
- 1Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Ning Liu
- 1Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Xiaojiao Sun
- 1Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Dong Shiyun
- 1Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Yajun Zhao
- 1Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Yan Wang
- 2Department of Urologic Surgery, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Dechao Zhao
- 3Department of Cardiology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Fanghao Lu
- 1Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Weihua Zhang
- 1Department of Pathophysiology, Harbin Medical University, Harbin, China.,4Key Laboratory of Cardiovascular Medicine Research (Harbin Medical University), Ministry of Education, Harbin, 150086, China
| |
Collapse
|
26
|
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: 12] [Impact Index Per Article: 2.4] [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
|
27
|
Lacroix-Desmazes S, Voorberg J, Lillicrap D, Scott DW, Pratt KP. Tolerating Factor VIII: Recent Progress. Front Immunol 2020; 10:2991. [PMID: 31998296 PMCID: PMC6965068 DOI: 10.3389/fimmu.2019.02991] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Accepted: 12/05/2019] [Indexed: 02/02/2023] Open
Abstract
Development of neutralizing antibodies against biotherapeutic agents administered to prevent or treat various clinical conditions is a longstanding and growing problem faced by patients, medical providers and pharmaceutical companies. The hemophilia A community has deep experience with attempting to manage such deleterious immune responses, as the lifesaving protein drug factor VIII (FVIII) has been in use for decades. Hemophilia A is a bleeding disorder caused by genetic mutations that result in absent or dysfunctional FVIII. Prophylactic treatment consists of regular intravenous FVIII infusions. Unfortunately, 1/4 to 1/3 of patients develop neutralizing anti-FVIII antibodies, referred to clinically as “inhibitors,” which result in a serious bleeding diathesis. Until recently, the only therapeutic option for these patients was “Immune Tolerance Induction,” consisting of intensive FVIII administration, which is extraordinarily expensive and fails in ~30% of cases. There has been tremendous recent progress in developing novel potential clinical alternatives for the treatment of hemophilia A, ranging from encouraging results of gene therapy trials, to use of other hemostatic agents (either promoting coagulation or slowing down anti-coagulant or fibrinolytic pathways) to “bypass” the need for FVIII or supplement FVIII replacement therapy. Although these approaches are promising, there is widespread agreement that preventing or reversing inhibitors remains a high priority. Risk profiles of novel therapies are still unknown or incomplete, and FVIII will likely continue to be considered the optimal hemostatic agent to support surgery and manage trauma, or to combine with other therapies. We describe here recent exciting studies, most still pre-clinical, that address FVIII immunogenicity and suggest novel interventions to prevent or reverse inhibitor development. Studies of FVIII uptake, processing and presentation on antigen-presenting cells, epitope mapping, and the roles of complement, heme, von Willebrand factor, glycans, and the microbiome in FVIII immunogenicity are elucidating mechanisms of primary and secondary immune responses and suggesting additional novel targets. Promising tolerogenic therapies include development of FVIII-Fc fusion proteins, nanoparticle-based therapies, oral tolerance, and engineering of regulatory or cytotoxic T cells to render them FVIII-specific. Importantly, these studies are highly applicable to other scenarios where establishing immune tolerance to a defined antigen is a clinical priority.
Collapse
Affiliation(s)
| | - Jan Voorberg
- Sanquin Research and Landsteiner Laboratory, Department of Molecular and Cellular Hemostasis, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - David Lillicrap
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON, Canada
| | - David W Scott
- Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Kathleen P Pratt
- Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| |
Collapse
|
28
|
Sabater-Lleal M, Huffman JE, de Vries PS, Marten J, Mastrangelo MA, Song C, Pankratz N, Ward-Caviness CK, Yanek LR, Trompet S, Delgado GE, Guo X, Bartz TM, Martinez-Perez A, Germain M, de Haan HG, Ozel AB, Polasek O, Smith AV, Eicher JD, Reiner AP, Tang W, Davies NM, Stott DJ, Rotter JI, Tofler GH, Boerwinkle E, de Maat MPM, Kleber ME, Welsh P, Brody JA, Chen MH, Vaidya D, Soria JM, Suchon P, van Hylckama Vlieg A, Desch KC, Kolcic I, Joshi PK, Launer LJ, Harris TB, Campbell H, Rudan I, Becker DM, Li JZ, Rivadeneira F, Uitterlinden AG, Hofman A, Franco OH, Cushman M, Psaty BM, Morange PE, McKnight B, Chong MR, Fernandez-Cadenas I, Rosand J, Lindgren A, Gudnason V, Wilson JF, Hayward C, Ginsburg D, Fornage M, Rosendaal FR, Souto JC, Becker LC, Jenny NS, März W, Jukema JW, Dehghan A, Trégouët DA, Morrison AC, Johnson AD, O'Donnell CJ, Strachan DP, Lowenstein CJ, Smith NL. Genome-Wide Association Transethnic Meta-Analyses Identifies Novel Associations Regulating Coagulation Factor VIII and von Willebrand Factor Plasma Levels. Circulation 2019; 139:620-635. [PMID: 30586737 DOI: 10.1161/circulationaha.118.034532] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Factor VIII (FVIII) and its carrier protein von Willebrand factor (VWF) are associated with risk of arterial and venous thrombosis and with hemorrhagic disorders. We aimed to identify and functionally test novel genetic associations regulating plasma FVIII and VWF. METHODS We meta-analyzed genome-wide association results from 46 354 individuals of European, African, East Asian, and Hispanic ancestry. All studies performed linear regression analysis using an additive genetic model and associated ≈35 million imputed variants with natural log-transformed phenotype levels. In vitro gene silencing in cultured endothelial cells was performed for candidate genes to provide additional evidence on association and function. Two-sample Mendelian randomization analyses were applied to test the causal role of FVIII and VWF plasma levels on the risk of arterial and venous thrombotic events. RESULTS We identified 13 novel genome-wide significant ( P≤2.5×10-8) associations, 7 with FVIII levels ( FCHO2/TMEM171/TNPO1, HLA, SOX17/RP1, LINC00583/NFIB, RAB5C-KAT2A, RPL3/TAB1/SYNGR1, and ARSA) and 11 with VWF levels ( PDHB/PXK/KCTD6, SLC39A8, FCHO2/TMEM171/TNPO1, HLA, GIMAP7/GIMAP4, OR13C5/NIPSNAP, DAB2IP, C2CD4B, RAB5C-KAT2A, TAB1/SYNGR1, and ARSA), beyond 10 previously reported associations with these phenotypes. Functional validation provided further evidence of association for all loci on VWF except ARSA and DAB2IP. Mendelian randomization suggested causal effects of plasma FVIII activity levels on venous thrombosis and coronary artery disease risk and plasma VWF levels on ischemic stroke risk. CONCLUSIONS The meta-analysis identified 13 novel genetic loci regulating FVIII and VWF plasma levels, 10 of which we validated functionally. We provide some evidence for a causal role of these proteins in thrombotic events.
Collapse
Affiliation(s)
- Maria Sabater-Lleal
- Cardiovascular Medicine Unit, Department of Medicine, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden (M.S.-L.).,Unit of Genomics of Complex Diseases, Institut d'Investigació Biomèdica Sant Pau, IIB-Sant Pau, Barcelona, Spain (M.S.-L., A.M.-P., J.M.S.)
| | - Jennifer E Huffman
- Population Sciences Branch, National Heart, Lung, and Blood Institute, Framingham, MA (J.E.H., C.S., J.D.E., M.-H.C., A.D.J., C.J.O.).,Framingham Heart Study, MA (J.E.H., C.S., J.D.E., M.-H.C., A.D.J., C.J.O.)
| | - Paul S de Vries
- Human Genetics Center, Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health (P.S.d.V., E.B., M.F., A.C.M.), University of Texas Health Science Center at Houston.,Department of Epidemiology (P.S.d.V., A.H., O.H.F., A.D.), Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Jonathan Marten
- Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine (J.M., J.F.W., C.H.), University of Edinburgh, Scotland
| | - Michael A Mastrangelo
- Aab Cardiovascular Research Institute, University of Rochester Medical Center, NY (M.A.M., C.J.L.)
| | - Ci Song
- Population Sciences Branch, National Heart, Lung, and Blood Institute, Framingham, MA (J.E.H., C.S., J.D.E., M.-H.C., A.D.J., C.J.O.).,Framingham Heart Study, MA (J.E.H., C.S., J.D.E., M.-H.C., A.D.J., C.J.O.)
| | - Nathan Pankratz
- Department of Laboratory Medicine and Pathology, University of Minnesota School of Medicine, Minneapolis (N.P.)
| | - Cavin K Ward-Caviness
- Environmental Public Health Division, National Health and Environmental Effects Research Laboratory, US Environmental Protection Agency, Chapel Hill, NC (C.K.W.-C.)
| | - Lisa R Yanek
- GeneSTAR Research Program, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD (L.R.Y., D.V., D.M.B., L.C.B.)
| | - Stella Trompet
- Department of Geriatrics and Gerontology (S.T.), Leiden University Medical Center, the Netherlands.,Department of Cardiology (S.T., J.W.J.), Leiden University Medical Center, the Netherlands
| | - Graciela E Delgado
- Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany (G.E.D., M.E.K., W.M.)
| | - Xiuqing Guo
- Institute for Translational Genomics and Population Sciences, Department of Pediatrics and Medicine, LABioMed at Harbor-UCLA Medical Center, Torrance, CA (X.G., J.I.R.)
| | - Traci M Bartz
- Department of Biostatistics (T.M.B., B.M.), University of Washington, Seattle
| | - Angel Martinez-Perez
- Unit of Genomics of Complex Diseases, Institut d'Investigació Biomèdica Sant Pau, IIB-Sant Pau, Barcelona, Spain (M.S.-L., A.M.-P., J.M.S.)
| | - Marine Germain
- Institut national de la santé et de la recherche médicale (INSERM), UMR_S 1166, Team Genomics and Pathophysiology of Cardiovascular Diseases, Sorbonne Universités, Université Pierre-et-Marie-Curie, Paris, France (M.G., D.-A.T.).,ICAN Institute for Cardiometabolism and Nutrition, Paris, France (M.G., D.-A.T.)
| | - Hugoline G de Haan
- Department of Clinical Epidemiology (H.G.d.H., A.v.H.V., F.R.R.), Leiden University Medical Center, the Netherlands
| | - Ayse B Ozel
- Department of Human Genetics (A.B.O., J.Z.L., D.G.), University of Michigan, Ann Arbor
| | - Ozren Polasek
- Faculty of Medicine, University of Split, Croatia (O.P., I.K.)
| | - Albert V Smith
- School of Public Health, Department of Biostatistics (A.V.S.), University of Michigan, Ann Arbor
| | - John D Eicher
- Framingham Heart Study, MA (J.E.H., C.S., J.D.E., M.-H.C., A.D.J., C.J.O.)
| | - Alex P Reiner
- Department of Epidemiology, (A.P.R., B.M.P., N.L.S.), University of Washington, Seattle.,Fred Hutchinson Cancer Research Center, Seattle, WA (A.P.R.)
| | - Weihong Tang
- Division of Epidemiology and Community Health, University of Minnesota School of Public Health, Minneapolis (W.T.)
| | - Neil M Davies
- Medical Research Council Integrative Epidemiology Unit and Bristol Medical School (N.M.D.), University of Bristol, UK
| | - David J Stott
- Academic Section of Geriatrics, Faculty of Medicine (J.D.S.), University of Glasgow, UK
| | - Jerome I Rotter
- Institute for Translational Genomics and Population Sciences, Department of Pediatrics and Medicine, LABioMed at Harbor-UCLA Medical Center, Torrance, CA (X.G., J.I.R.)
| | - Geoffrey H Tofler
- Royal North Shore Hospital, University of Sydney, Australia (G.H.T.)
| | - Eric Boerwinkle
- Human Genetics Center, Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health (P.S.d.V., E.B., M.F., A.C.M.), University of Texas Health Science Center at Houston.,Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX (E.B.)
| | - Moniek P M de Maat
- Department of Hematology (M.P.M.d.M.), Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Marcus E Kleber
- Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany (G.E.D., M.E.K., W.M.).,Institute of Nutrition, Friedrich-Schiller-University Jena, Mannheim, Germany (M.E.K.)
| | - Paul Welsh
- Institute of Cardiovascular and Medical Sciences (P.W.), University of Glasgow, UK
| | - Jennifer A Brody
- Department of Medicine (J.A.B., B.M.P.), University of Washington, Seattle
| | - Ming-Huei Chen
- Population Sciences Branch, National Heart, Lung, and Blood Institute, Framingham, MA (J.E.H., C.S., J.D.E., M.-H.C., A.D.J., C.J.O.).,Framingham Heart Study, MA (J.E.H., C.S., J.D.E., M.-H.C., A.D.J., C.J.O.)
| | - Dhananjay Vaidya
- GeneSTAR Research Program, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD (L.R.Y., D.V., D.M.B., L.C.B.)
| | - José Manuel Soria
- Unit of Genomics of Complex Diseases, Institut d'Investigació Biomèdica Sant Pau, IIB-Sant Pau, Barcelona, Spain (M.S.-L., A.M.-P., J.M.S.)
| | - Pierre Suchon
- Laboratory of Haematology, La Timone Hospital, Marseille, France (P.S., P.-E.M.).,Institut national de la santé et de la recherche médicale (INSERM), UMR_S 1062, Nutrition Obesity and Risk of Thrombosis, Marseille, France (P.S., P.-E.M.)
| | - Astrid van Hylckama Vlieg
- Department of Clinical Epidemiology (H.G.d.H., A.v.H.V., F.R.R.), Leiden University Medical Center, the Netherlands
| | - Karl C Desch
- Department of Pediatrics and Communicable Disease (K.D.C.), University of Michigan, Ann Arbor
| | - Ivana Kolcic
- Faculty of Medicine, University of Split, Croatia (O.P., I.K.)
| | - Peter K Joshi
- Centre for Global Health Research, Usher Institute for Population Health Sciences and Informatics (P.K.J., H.C., I.R., J.F.W.), University of Edinburgh, Scotland
| | - Lenore J Launer
- Laboratory of Epidemiology and Population Sciences National Institute on Aging, Bethesda, MD (L.J.L., T.B.H.)
| | - Tamara B Harris
- Laboratory of Epidemiology and Population Sciences National Institute on Aging, Bethesda, MD (L.J.L., T.B.H.)
| | - Harry Campbell
- Centre for Global Health Research, Usher Institute for Population Health Sciences and Informatics (P.K.J., H.C., I.R., J.F.W.), University of Edinburgh, Scotland
| | - Igor Rudan
- Centre for Global Health Research, Usher Institute for Population Health Sciences and Informatics (P.K.J., H.C., I.R., J.F.W.), University of Edinburgh, Scotland
| | - Diane M Becker
- GeneSTAR Research Program, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD (L.R.Y., D.V., D.M.B., L.C.B.)
| | - Jun Z Li
- Department of Human Genetics (A.B.O., J.Z.L., D.G.), University of Michigan, Ann Arbor
| | - Fernando Rivadeneira
- Department of Internal Medicine (F.R., A.G.U.), Erasmus University Medical Center, Rotterdam, the Netherlands
| | - André G Uitterlinden
- Department of Internal Medicine (F.R., A.G.U.), Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Albert Hofman
- Department of Epidemiology (P.S.d.V., A.H., O.H.F., A.D.), Erasmus University Medical Center, Rotterdam, the Netherlands.,Department of Epidemiology, Harvard H.T. Chan School of Public Health, Boston, MA (A.H.)
| | - Oscar H Franco
- Department of Epidemiology (P.S.d.V., A.H., O.H.F., A.D.), Erasmus University Medical Center, Rotterdam, the Netherlands.,Institute of Social and Preventive Medicine, University of Bern, Switzerland (O.H.F.)
| | - Mary Cushman
- Larner College of Medicine, University of Vermont, Colchester (M.C.)
| | - Bruce M Psaty
- Department of Epidemiology, (A.P.R., B.M.P., N.L.S.), University of Washington, Seattle.,Department of Medicine (J.A.B., B.M.P.), University of Washington, Seattle.,Department of Health Services (B.M.P.), University of Washington, Seattle.,Kaiser Permanente Washington Research Institute, Kaiser Permanente Washington, Seattle (B.M.P., N.L.S.)
| | - Pierre-Emmanuel Morange
- Laboratory of Haematology, La Timone Hospital, Marseille, France (P.S., P.-E.M.).,Institut national de la santé et de la recherche médicale (INSERM), UMR_S 1062, Nutrition Obesity and Risk of Thrombosis, Marseille, France (P.S., P.-E.M.)
| | - Barbara McKnight
- Department of Biostatistics (T.M.B., B.M.), University of Washington, Seattle.,Cardiovascular Health Research Unit (B.M.), University of Washington, Seattle
| | - Michael R Chong
- McMaster University, Population Health Research Institute, Population Health Research Institute, Biochemistry and Biomedical Sciences, Hamilton, Canada (M.R.C.)
| | - Israel Fernandez-Cadenas
- Stroke Pharmacogenomics and genetics, Department of Neurology, Institut d'Investigació Biomedica Sant Pau, IIB-Sant Pau, Barcelona, Spain (I.F.-C.)
| | - Jonathan Rosand
- Massachusetts General Hospital, Broad Institute, Harvard Medical School, Boston (J.R.)
| | - Arne Lindgren
- Department of Clinical Sciences Lund, Neurology, Lund University, Sweden (A.L.).,Department of Neurology and Rehabilitation Medicine, Neurology, Skåne University Hospital, Lund, Sweden (A.L.)
| | | | - Vilmundur Gudnason
- Icelandic Heart Association, Kopavogur (V.G.).,Faculty of Medicine, University of Iceland, Reykjavik (V.G.)
| | - James F Wilson
- Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine (J.M., J.F.W., C.H.), University of Edinburgh, Scotland.,Centre for Global Health Research, Usher Institute for Population Health Sciences and Informatics (P.K.J., H.C., I.R., J.F.W.), University of Edinburgh, Scotland
| | - Caroline Hayward
- Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine (J.M., J.F.W., C.H.), University of Edinburgh, Scotland
| | - David Ginsburg
- Department of Human Genetics (A.B.O., J.Z.L., D.G.), University of Michigan, Ann Arbor
| | - Myriam Fornage
- Human Genetics Center, Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health (P.S.d.V., E.B., M.F., A.C.M.), University of Texas Health Science Center at Houston.,Brown Foundation Institute of Molecular Medicine (M.F.), University of Texas Health Science Center at Houston
| | - Frits R Rosendaal
- Department of Clinical Epidemiology (H.G.d.H., A.v.H.V., F.R.R.), Leiden University Medical Center, the Netherlands.,Einthoven Laboratory of Experimental Vascular Medicine (F.R.R., J.W.J.), Leiden University Medical Center, the Netherlands
| | - Juan Carlos Souto
- Unit of Hemostasis and Thrombosis, Hospital de la Sant Creu i Sant Pau, Barcelona, Spain (J.C.S.)
| | - Lewis C Becker
- GeneSTAR Research Program, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD (L.R.Y., D.V., D.M.B., L.C.B.)
| | - Nancy S Jenny
- Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Colchester (N.S.J.)
| | - Winfried März
- Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany (G.E.D., M.E.K., W.M.).,SYNLAB Academy, SYNLAB Holding Deutschland GmbH, Mannheim, Germany (W.M.).,Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University Graz, Mannheim, Germany (W.M.)
| | - J Wouter Jukema
- Department of Cardiology (S.T., J.W.J.), Leiden University Medical Center, the Netherlands.,Einthoven Laboratory of Experimental Vascular Medicine (F.R.R., J.W.J.), Leiden University Medical Center, the Netherlands.,Interuniversity Cardiology Institute of the Netherlands, Utrecht (J.W.J.)
| | - Abbas Dehghan
- Department of Epidemiology (P.S.d.V., A.H., O.H.F., A.D.), Erasmus University Medical Center, Rotterdam, the Netherlands.,Department of Epidemiology and Biostatistics, Imperial College London, UK (A.D.)
| | - David-Alexandre Trégouët
- Institut national de la santé et de la recherche médicale (INSERM), UMR_S 1166, Team Genomics and Pathophysiology of Cardiovascular Diseases, Sorbonne Universités, Université Pierre-et-Marie-Curie, Paris, France (M.G., D.-A.T.).,ICAN Institute for Cardiometabolism and Nutrition, Paris, France (M.G., D.-A.T.)
| | - Alanna C Morrison
- Human Genetics Center, Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health (P.S.d.V., E.B., M.F., A.C.M.), University of Texas Health Science Center at Houston
| | - Andrew D Johnson
- Population Sciences Branch, National Heart, Lung, and Blood Institute, Framingham, MA (J.E.H., C.S., J.D.E., M.-H.C., A.D.J., C.J.O.).,Framingham Heart Study, MA (J.E.H., C.S., J.D.E., M.-H.C., A.D.J., C.J.O.)
| | - Christopher J O'Donnell
- Population Sciences Branch, National Heart, Lung, and Blood Institute, Framingham, MA (J.E.H., C.S., J.D.E., M.-H.C., A.D.J., C.J.O.).,Framingham Heart Study, MA (J.E.H., C.S., J.D.E., M.-H.C., A.D.J., C.J.O.).,Cardiology Section Administration, Boston VA Healthcare System, West Roxbury, MA (C.J.O.)
| | - David P Strachan
- Population Health Research Institute, St George's, University of London, UK (D.P.S.)
| | - Charles J Lowenstein
- Aab Cardiovascular Research Institute, University of Rochester Medical Center, NY (M.A.M., C.J.L.)
| | - Nicholas L Smith
- Department of Epidemiology, (A.P.R., B.M.P., N.L.S.), University of Washington, Seattle.,Kaiser Permanente Washington Research Institute, Kaiser Permanente Washington, Seattle (B.M.P., N.L.S.).,Seattle Epidemiologic Research and Information Center, Department of Veterans Affairs Office of Research and Development, WA (N.L.S.)
| |
Collapse
|
29
|
Flood VH, Garcia J, Haberichter SL. The role of genetics in the pathogenesis and diagnosis of type 1 Von Willebrand disease. Curr Opin Hematol 2019; 26:331-335. [PMID: 31261173 PMCID: PMC6727843 DOI: 10.1097/moh.0000000000000524] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW Von Willebrand disease (VWD) is a common bleeding disorder, but diagnosis of VWD is challenging, particularly with type 1 VWD. Although most clinicians use specific tests of von Willebrand factor (VWF) activity to classify patients with VWD, genetic testing for VWF defects is another potential method of diagnosis. RECENT FINDINGS Studies of patients with type 1 VWD report consistently that many, but not all, study participants have VWF gene defects. Certain populations, including those with VWF levels less than 30 IU/dl and those with clearance defects, are more likely to have a VWF sequence variant. In addition, a number of loci outside the VWF gene have been shown to affect VWF levels, including ABO, CLEC4M, STXBP5, and STAB2. SUMMARY Genetic defects in VWF are common, but not all defects lead to disease. Type 1 VWD in particular does not always have an associated VWF sequence variant. New data stemming from genome-wide association studies on modifier genes suggest that the etiology of type 1 VWD is multifactorial.
Collapse
Affiliation(s)
- Veronica H Flood
- Department of Pediatrics, Division of Hematology/Oncology, Medical College of Wisconsin
- Children's Research Institute, Children's Hospital of Wisconsin
- Versiti Blood Research Institute, Milwaukee, Wisconsin
| | - Jessica Garcia
- Department of Pediatrics, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Sandra L Haberichter
- Department of Pediatrics, Division of Hematology/Oncology, Medical College of Wisconsin
- Children's Research Institute, Children's Hospital of Wisconsin
- Versiti Blood Research Institute, Milwaukee, Wisconsin
| |
Collapse
|
30
|
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
|
31
|
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: 97] [Impact Index Per Article: 16.2] [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
|
32
|
Bennett JA, Ture SK, Schmidt RA, Mastrangelo MA, Cameron SJ, Terry LE, Yule DI, Morrell CN, Lowenstein CJ. Acetylcholine Inhibits Platelet Activation. J Pharmacol Exp Ther 2019; 369:182-187. [PMID: 30765424 PMCID: PMC6439456 DOI: 10.1124/jpet.118.253583] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 02/12/2019] [Indexed: 12/18/2022] Open
Abstract
Platelets are key mediators of thrombosis. Many agonists of platelet activation are known, but fewer endogenous inhibitors of platelets, such as prostacyclin and nitric oxide (NO), have been identified. Acetylcholinesterase inhibitors, such as donepezil, can cause bleeding in patients, but the underlying mechanisms are not well understood. We hypothesized that acetylcholine is an endogenous inhibitor of platelets. We measured the effect of acetylcholine or analogs of acetylcholine on human platelet activation ex vivo. Acetylcholine and analogs of acetylcholine inhibited platelet activation, as measured by P-selectin translocation and glycoprotein IIb IIIa conformational changes. Conversely, we found that antagonists of the acetylcholine receptor, such as pancuronium, enhance platelet activation. Furthermore, drugs inhibiting acetylcholinesterase, such as donepezil, also inhibit platelet activation, suggesting that platelets release acetylcholine. We found that NO mediates acetylcholine inhibition of platelets. Our data suggest that acetylcholine is an endogenous inhibitor of platelet activation. The cholinergic system may be a novel target for antithrombotic therapies.
Collapse
Affiliation(s)
- John A Bennett
- Aab Cardiovascular Research Institute, Department of Medicine (J.A.B., S.K.T., R.A.S., M.A.M., S.J.C., C.N.M., C.J.L.) and Department of Pharmacology and Physiology (L.E.T., D.I.Y.), University of Rochester Medical Center, Rochester, New York
| | - Sara K Ture
- Aab Cardiovascular Research Institute, Department of Medicine (J.A.B., S.K.T., R.A.S., M.A.M., S.J.C., C.N.M., C.J.L.) and Department of Pharmacology and Physiology (L.E.T., D.I.Y.), University of Rochester Medical Center, Rochester, New York
| | - Rachel A Schmidt
- Aab Cardiovascular Research Institute, Department of Medicine (J.A.B., S.K.T., R.A.S., M.A.M., S.J.C., C.N.M., C.J.L.) and Department of Pharmacology and Physiology (L.E.T., D.I.Y.), University of Rochester Medical Center, Rochester, New York
| | - Michael A Mastrangelo
- Aab Cardiovascular Research Institute, Department of Medicine (J.A.B., S.K.T., R.A.S., M.A.M., S.J.C., C.N.M., C.J.L.) and Department of Pharmacology and Physiology (L.E.T., D.I.Y.), University of Rochester Medical Center, Rochester, New York
| | - Scott J Cameron
- Aab Cardiovascular Research Institute, Department of Medicine (J.A.B., S.K.T., R.A.S., M.A.M., S.J.C., C.N.M., C.J.L.) and Department of Pharmacology and Physiology (L.E.T., D.I.Y.), University of Rochester Medical Center, Rochester, New York
| | - Lara E Terry
- Aab Cardiovascular Research Institute, Department of Medicine (J.A.B., S.K.T., R.A.S., M.A.M., S.J.C., C.N.M., C.J.L.) and Department of Pharmacology and Physiology (L.E.T., D.I.Y.), University of Rochester Medical Center, Rochester, New York
| | - David I Yule
- Aab Cardiovascular Research Institute, Department of Medicine (J.A.B., S.K.T., R.A.S., M.A.M., S.J.C., C.N.M., C.J.L.) and Department of Pharmacology and Physiology (L.E.T., D.I.Y.), University of Rochester Medical Center, Rochester, New York
| | - Craig N Morrell
- Aab Cardiovascular Research Institute, Department of Medicine (J.A.B., S.K.T., R.A.S., M.A.M., S.J.C., C.N.M., C.J.L.) and Department of Pharmacology and Physiology (L.E.T., D.I.Y.), University of Rochester Medical Center, Rochester, New York
| | - Charles J Lowenstein
- Aab Cardiovascular Research Institute, Department of Medicine (J.A.B., S.K.T., R.A.S., M.A.M., S.J.C., C.N.M., C.J.L.) and Department of Pharmacology and Physiology (L.E.T., D.I.Y.), University of Rochester Medical Center, Rochester, New York
| |
Collapse
|
33
|
Shimojo M, Madara J, Pankow S, Liu X, Yates J, Südhof TC, Maximov A. Synaptotagmin-11 mediates a vesicle trafficking pathway that is essential for development and synaptic plasticity. Genes Dev 2019; 33:365-376. [PMID: 30808661 PMCID: PMC6411015 DOI: 10.1101/gad.320077.118] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 12/21/2018] [Indexed: 11/25/2022]
Abstract
Synaptotagmin-11 (Syt11) is a Synaptotagmin isoform that lacks an apparent ability to bind calcium, phospholipids, or SNARE proteins. While human genetic studies have linked mutations in the Syt11 gene to schizophrenia and Parkinson's disease, the localization or physiological role of Syt11 remain unclear. We found that in neurons, Syt11 resides on abundant vesicles that differ from synaptic vesicles and resemble trafficking endosomes. These vesicles recycle via the plasma membrane in an activity-dependent manner, but their exocytosis is slow and desynchronized. Constitutive knockout mice lacking Syt11 died shortly after birth, suggesting Syt11-mediated membrane transport is required for survival. In contrast, selective ablation of Syt11 in excitatory forebrain neurons using a conditional knockout did not affect life span but impaired synaptic plasticity and memory. Syt11-deficient neurons displayed normal secretion of fast neurotransmitters and peptides but exhibited a reduction of long-term synaptic potentiation. Hence, Syt11 is an essential component of a neuronal vesicular trafficking pathway that differs from the well-characterized synaptic vesicle trafficking pathway but is also essential for life.
Collapse
Affiliation(s)
- Masafumi Shimojo
- Department of Neuroscience, Scripps Research, La Jolla, California 92037, USA
- The Dorris Neuroscience, Scripps Research, La Jolla, California 92037, USA
| | - Joseph Madara
- Department of Neuroscience, Scripps Research, La Jolla, California 92037, USA
- The Dorris Neuroscience, Scripps Research, La Jolla, California 92037, USA
| | - Sandra Pankow
- Department of Molecular Medicine, Scripps Research, La Jolla, California 92037, USA
| | - Xinran Liu
- Department of Neuroscience, University of Texas Southwestern Medical Center at Dallas, Dallas 75235, Texas, USA
| | - John Yates
- Department of Molecular Medicine, Scripps Research, La Jolla, California 92037, USA
| | - Thomas C Südhof
- Department of Neuroscience, University of Texas Southwestern Medical Center at Dallas, Dallas 75235, Texas, USA
- Department of Molecular and Cellular Physiology, Stanford University, Palo Alto, California 94035, USA
| | - Anton Maximov
- Department of Neuroscience, Scripps Research, La Jolla, California 92037, USA
- The Dorris Neuroscience, Scripps Research, La Jolla, California 92037, USA
- Department of Neuroscience, University of Texas Southwestern Medical Center at Dallas, Dallas 75235, Texas, USA
| |
Collapse
|
34
|
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
|
35
|
Lenzi C, Stevens J, Osborn D, Hannah MJ, Bierings R, Carter T. Synaptotagmin 5 regulates Ca 2+-dependent Weibel-Palade body exocytosis in human endothelial cells. J Cell Sci 2019; 132:jcs.221952. [PMID: 30659119 DOI: 10.1242/jcs.221952] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 01/09/2019] [Indexed: 12/11/2022] Open
Abstract
Elevations of intracellular free Ca2+ concentration ([Ca2+]i) are a potent trigger for Weibel-Palade body (WPB) exocytosis and secretion of von Willebrand factor (VWF) from endothelial cells; however, the identity of WPB-associated Ca2+-sensors involved in transducing acute increases in [Ca2+]i into granule exocytosis remains unknown. Here, we show that synaptotagmin 5 (SYT5) is expressed in human umbilical vein endothelial cells (HUVECs) and is recruited to WPBs to regulate Ca2+-driven WPB exocytosis. Western blot analysis of HUVECs identified SYT5 protein, and exogenously expressed SYT5-mEGFP localised almost exclusively to WPBs. shRNA-mediated knockdown of endogenous SYT5 (shSYT5) reduced the rate and extent of histamine-evoked WPB exocytosis and reduced secretion of the WPB cargo VWF-propeptide (VWFpp). The shSYT5-mediated reduction in histamine-evoked WPB exocytosis was prevented by expression of shRNA-resistant SYT5-mCherry. Overexpression of SYT5-EGFP increased the rate and extent of histamine-evoked WPB exocytosis, and increased secretion of VWFpp. Expression of a Ca2+-binding defective SYT5 mutant (SYT5-Asp197Ser-EGFP) mimicked depletion of endogenous SYT5. We identify SYT5 as a WPB-associated Ca2+ sensor regulating Ca2+-dependent secretion of stored mediators from vascular endothelial cells.
Collapse
Affiliation(s)
- Camille Lenzi
- Molecular and Clinical Sciences Research Institute, St George's, University of London, London SW18 ORE, UK
| | | | - Daniel Osborn
- Molecular and Clinical Sciences Research Institute, St George's, University of London, London SW18 ORE, UK
| | - Matthew J Hannah
- Microbiology Services Colindale, Public Health England, London, NW9 5EQ, UK
| | - Ruben Bierings
- Plasma Proteins, Sanquin Research and Landsteiner Laboratory, Academic Medical Centre, University of Amsterdam, 1006 AD Amsterdam, PO Box 9190, The Netherlands
| | - Tom Carter
- Molecular and Clinical Sciences Research Institute, St George's, University of London, London SW18 ORE, UK
| |
Collapse
|
36
|
Schillemans M, Karampini E, Kat M, Bierings R. Exocytosis of Weibel-Palade bodies: how to unpack a vascular emergency kit. J Thromb Haemost 2019; 17:6-18. [PMID: 30375718 PMCID: PMC7379738 DOI: 10.1111/jth.14322] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Indexed: 01/17/2023]
Abstract
The blood vessel wall has a number of self-healing properties, enabling it to minimize blood loss and prevent or overcome infections in the event of vascular trauma. Endothelial cells prepackage a cocktail of hemostatic, inflammatory and angiogenic mediators in their unique secretory organelles, the Weibel-Palade bodies (WPBs), which can be immediately released on demand. Secretion of their contents into the vascular lumen through a process called exocytosis enables the endothelium to actively participate in the arrest of bleeding and to slow down and direct leukocytes to areas of inflammation. Owing to their remarkable elongated morphology and their secretory contents, which span the entire size spectrum of small chemokines all the way up to ultralarge von Willebrand factor multimers, WPBs constitute an ideal model system for studying the molecular mechanisms of secretory organelle biogenesis, exocytosis, and content expulsion. Recent studies have now shown that, during exocytosis, WPBs can undergo several distinct modes of fusion, and can utilize fundamentally different mechanisms to expel their contents. In this article, we discuss recent advances in our understanding of the composition of the WPB exocytotic machinery and how, because of its configuration, it is able to support WPB release in its various forms.
Collapse
Affiliation(s)
- M. Schillemans
- Molecular and Cellular HemostasisSanquin Research and Landsteiner LaboratoryAmsterdam UMCUniversity of AmsterdamAmsterdamthe Netherlands
| | - E. Karampini
- Molecular and Cellular HemostasisSanquin Research and Landsteiner LaboratoryAmsterdam UMCUniversity of AmsterdamAmsterdamthe Netherlands
| | - M. Kat
- Molecular and Cellular HemostasisSanquin Research and Landsteiner LaboratoryAmsterdam UMCUniversity of AmsterdamAmsterdamthe Netherlands
| | - R. Bierings
- Molecular and Cellular HemostasisSanquin Research and Landsteiner LaboratoryAmsterdam UMCUniversity of AmsterdamAmsterdamthe Netherlands
- HematologyErasmus University Medical CenterRotterdamthe Netherlands
| |
Collapse
|
37
|
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: 48] [Impact Index Per Article: 6.9] [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
|
38
|
Xiao C, Stahel P, Carreiro AL, Hung YH, Dash S, Bookman I, Buhman KK, Lewis GF. Oral Glucose Mobilizes Triglyceride Stores From the Human Intestine. Cell Mol Gastroenterol Hepatol 2018; 7:313-337. [PMID: 30704982 PMCID: PMC6357697 DOI: 10.1016/j.jcmgh.2018.10.002] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 10/05/2018] [Accepted: 10/05/2018] [Indexed: 12/22/2022]
Abstract
BACKGROUND & AIMS The small intestine regulates plasma triglyceride (TG) concentration. Within enterocytes, dietary TGs are packaged into chylomicrons (CMs) for secretion or stored temporarily in cytoplasmic lipid droplets (CLDs) until further mobilization. We and others have shown that oral and intravenous glucose enhances CM particle secretion in human beings, however, the mechanisms through which this occurs are incompletely understood. METHODS Two separate cohorts of participants ingested a high-fat liquid meal and, 5 hours later, were assigned randomly to ingest either a glucose solution or an equivalent volume of water. In 1 group (N = 6), plasma and lipoprotein TG responses were assessed in a randomized cross-over study. In a separate group (N = 24), duodenal biopsy specimens were obtained 1 hour after ingestion of glucose or water. Ultrastructural and proteomic analyses were performed on duodenal biopsy specimens. RESULTS Compared with water, glucose ingestion increased circulating TGs within 30 minutes, mainly in the CM fraction. It decreased the total number of CLDs and the proportion of large-sized CLDs within enterocytes. We identified 2919 proteins in human duodenal tissue, 270 of which are related to lipid metabolism and 134 of which were differentially present in response to glucose compared with water ingestion. CONCLUSIONS Oral glucose mobilizes TGs stored within enterocyte CLDs to provide substrate for CM synthesis and secretion. Future studies elucidating the underlying signaling pathways may provide mechanistic insights that lead to the development of novel therapeutics for the treatment of hypertriglyceridemia.
Collapse
Affiliation(s)
- Changting Xiao
- Division of Endocrinology and Metabolism, Departments of Medicine and Physiology, Banting and Best Diabetes Centre, University of Toronto, Toronto, Ontario, Canada
| | - Priska Stahel
- Division of Endocrinology and Metabolism, Departments of Medicine and Physiology, Banting and Best Diabetes Centre, University of Toronto, Toronto, Ontario, Canada
| | - Alicia L. Carreiro
- Department of Nutrition Science, Purdue University, West Lafayette, Indiana
| | - Yu-Han Hung
- Department of Nutrition Science, Purdue University, West Lafayette, Indiana
| | - Satya Dash
- Division of Endocrinology and Metabolism, Departments of Medicine and Physiology, Banting and Best Diabetes Centre, University of Toronto, Toronto, Ontario, Canada
| | - Ian Bookman
- Kensington Screening Clinic, Toronto, Ontario, Canada
| | - Kimberly K. Buhman
- Department of Nutrition Science, Purdue University, West Lafayette, Indiana
| | - Gary F. Lewis
- Division of Endocrinology and Metabolism, Departments of Medicine and Physiology, Banting and Best Diabetes Centre, University of Toronto, Toronto, Ontario, Canada,Correspondence Address correspondence to: Gary F. Lewis, MD, FRCPC, Toronto General Hospital, 200 Elizabeth Street, EN12-218, Toronto, Ontario, M5G 2C4 Canada. fax: (416) 340-3314.
| |
Collapse
|
39
|
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]
|
40
|
Schillemans M, Karampini E, van den Eshof BL, Gangaev A, Hofman M, van Breevoort D, Meems H, Janssen H, Mulder AA, Jost CR, Escher JC, Adam R, Carter T, Koster AJ, van den Biggelaar M, Voorberg J, Bierings R. Weibel-Palade Body Localized Syntaxin-3 Modulates Von Willebrand Factor Secretion From Endothelial Cells. Arterioscler Thromb Vasc Biol 2018; 38:1549-1561. [PMID: 29880488 PMCID: PMC6039413 DOI: 10.1161/atvbaha.117.310701] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Accepted: 05/17/2018] [Indexed: 01/08/2023]
Abstract
Supplemental Digital Content is available in the text. Objective— Endothelial cells store VWF (von Willebrand factor) in rod-shaped secretory organelles, called Weibel-Palade bodies (WPBs). WPB exocytosis is coordinated by a complex network of Rab GTPases, Rab effectors, and SNARE (soluble NSF attachment protein receptor) proteins. We have previously identified STXBP1 as the link between the Rab27A-Slp4-a complex on WPBs and the SNARE proteins syntaxin-2 and -3. In this study, we investigate the function of syntaxin-3 in VWF secretion. Approach and Results— In human umbilical vein endothelial cells and in blood outgrowth endothelial cells (BOECs) from healthy controls, endogenous syntaxin-3 immunolocalized to WPBs. A detailed analysis of BOECs isolated from a patient with variant microvillus inclusion disease, carrying a homozygous mutation in STX3(STX3−/−), showed a loss of syntaxin-3 protein and absence of WPB-associated syntaxin-3 immunoreactivity. Ultrastructural analysis revealed no detectable differences in morphology or prevalence of immature or mature WPBs in control versus STX3−/− BOECs. VWF multimer analysis showed normal patterns in plasma of the microvillus inclusion disease patient, and media from STX3−/− BOECs, together indicating WPB formation and maturation are unaffected by absence of syntaxin-3. However, a defect in basal as well as Ca2+- and cAMP-mediated VWF secretion was found in the STX3−/− BOECs. We also show that syntaxin-3 interacts with the WPB-associated SNARE protein VAMP8 (vesicle-associated membrane protein-8). Conclusions— Our data reveal syntaxin-3 as a novel WPB-associated SNARE protein that controls WPB exocytosis.
Collapse
Affiliation(s)
- Maaike Schillemans
- From the Plasma Proteins, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, The Netherlands (M.S., E.K., B.L.v.d.E., A.G., M.H., D.v.B., H.M., M.v.d.B., J.V., R.B.)
| | - Ellie Karampini
- From the Plasma Proteins, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, The Netherlands (M.S., E.K., B.L.v.d.E., A.G., M.H., D.v.B., H.M., M.v.d.B., J.V., R.B.)
| | - Bart L van den Eshof
- From the Plasma Proteins, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, The Netherlands (M.S., E.K., B.L.v.d.E., A.G., M.H., D.v.B., H.M., M.v.d.B., J.V., R.B.)
| | - Anastasia Gangaev
- From the Plasma Proteins, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, The Netherlands (M.S., E.K., B.L.v.d.E., A.G., M.H., D.v.B., H.M., M.v.d.B., J.V., R.B.)
| | - Menno Hofman
- From the Plasma Proteins, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, The Netherlands (M.S., E.K., B.L.v.d.E., A.G., M.H., D.v.B., H.M., M.v.d.B., J.V., R.B.)
| | - Dorothee van Breevoort
- From the Plasma Proteins, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, The Netherlands (M.S., E.K., B.L.v.d.E., A.G., M.H., D.v.B., H.M., M.v.d.B., J.V., R.B.)
| | - Henriët Meems
- From the Plasma Proteins, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, The Netherlands (M.S., E.K., B.L.v.d.E., A.G., M.H., D.v.B., H.M., M.v.d.B., J.V., R.B.)
| | - Hans Janssen
- Cell Biology, The Netherlands Cancer Institute, Amsterdam (H.J.)
| | - Aat A Mulder
- Molecular Cell Biology, Section Electron Microscopy, Leiden University Medical Center, The Netherlands (A.A.M., C.R.J., A.J.K.)
| | - Carolina R Jost
- Molecular Cell Biology, Section Electron Microscopy, Leiden University Medical Center, The Netherlands (A.A.M., C.R.J., A.J.K.)
| | - Johanna C Escher
- Pediatric Gastroenterology, Sophia Children's Hospital, Erasmus MC, Rotterdam, The Netherlands (J.C.E.)
| | - Rüdiger Adam
- Pediatric Gastroenterology, University Medical Centre, Mannheim, Germany (R.A.)
| | - Tom Carter
- St George's, University of London, United Kingdom (T.C.)
| | - Abraham J Koster
- Molecular Cell Biology, Section Electron Microscopy, Leiden University Medical Center, The Netherlands (A.A.M., C.R.J., A.J.K.)
| | - Maartje van den Biggelaar
- From the Plasma Proteins, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, The Netherlands (M.S., E.K., B.L.v.d.E., A.G., M.H., D.v.B., H.M., M.v.d.B., J.V., R.B.)
| | - Jan Voorberg
- From the Plasma Proteins, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, The Netherlands (M.S., E.K., B.L.v.d.E., A.G., M.H., D.v.B., H.M., M.v.d.B., J.V., R.B.).,Department of Vascular Medicine, Academic Medical Center, University of Amsterdam, The Netherlands (J.V.)
| | - Ruben Bierings
- From the Plasma Proteins, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, The Netherlands (M.S., E.K., B.L.v.d.E., A.G., M.H., D.v.B., H.M., M.v.d.B., J.V., R.B.)
| |
Collapse
|
41
|
Trégouët DA, Morange PE. What is currently known about the genetics of venous thromboembolism at the dawn of next generation sequencing technologies. Br J Haematol 2018; 180:335-345. [PMID: 29082522 DOI: 10.1111/bjh.15004] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Venous thromboembolism (VTE) has a strong genetic component. This review summarizes what is known at the seventeen genes that are now well established to harbour VTE-associated genetic variants. In addition, it discusses additional candidate genes that deserve further validation before being claimed as VTE associated genes. Finally, several research strategies are briefly described to identify other molecular determinants of the disease.
Collapse
Affiliation(s)
- David-Alexandre Trégouët
- Department of Genomics & Pathophysiology of Cardiovascular Diseases, Sorbonne Universités, UPMC Univ. Paris 06, Institut National pour la Santé et la Recherche Médicale (INSERM), Unité Mixte de Recherche en Santé (UMR_S) 1166, Paris, France
- ICAN Institute for Cardiometabolism and Nutrition, Paris, France
| | - Pierre-Emmanuel Morange
- Laboratory of Haematology, La Timone Hospital, Marseille, France
- INSERM UMR_S 1062, Nutrition Obesity and Risk of Thrombosis, Aix-Marseille University, Marseille, France
| |
Collapse
|
42
|
Abstract
Von Willebrand factor (VWF) is a multimeric plasma glycoprotein that plays a central role in the initiation of blood coagulation. Through interactions between its specific functional domains, the vascular wall, coagulation factor VIII, and platelet receptors, VWF maintains hemostasis by binding to platelets and delivering factor VIII to the sites of vascular injury. In the healthy human population, plasma VWF levels vary widely. The important role of VWF is illustrated by individuals at the extremes of the normal distribution of plasma VWF concentrations where individuals with low VWF levels are more likely to present with mucocutaneous bleeding. Conversely, people with high VWF levels are at higher risk for venous thromboembolic disease, stroke, and coronary artery disease. This report will summarize recent advances in our understanding of environmental influences and the genetic control of VWF plasma variation in healthy and symptomatic populations and will also highlight the unanswered questions that are currently driving this field of study.
Collapse
Affiliation(s)
- Karl C Desch
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
43
|
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
|
44
|
McCormack JJ, Lopes da Silva M, Ferraro F, Patella F, Cutler DF. Weibel-Palade bodies at a glance. J Cell Sci 2017; 130:3611-3617. [PMID: 29093059 DOI: 10.1242/jcs.208033] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
The vascular environment can rapidly alter, and the speed with which responses to both physiological and pathological changes are required necessitates the existence of a highly responsive system. The endothelium can quickly deliver bioactive molecules by regulated exocytosis of its secretory granules, the Weibel-Palade bodies (WPBs). WPBs include proteins that initiate both haemostasis and inflammation, as well those that modulate blood pressure and angiogenesis. WPB formation is driven by von Willebrand factor, their most abundant protein, which controls both shape and size of WPBs. WPB are generated in a range of sizes, with the largest granules over ten times the size of the smallest. In this Cell Science at a Glance and the accompanying poster, we discuss the emerging mechanisms by which WPB size is controlled and how this affects the ability of this organelle to modulate haemostasis. We will also outline the different modes of exocytosis and their polarity that are currently being explored, and illustrate that these large secretory organelles provide a model for how elements of secretory granule biogenesis and exocytosis cooperate to support a complex and diverse set of functions.
Collapse
Affiliation(s)
- Jessica J McCormack
- MRC Laboratory of Molecular Cell Biology, University College London, Gower Street, London, WC1E6BT, UK
| | - Mafalda Lopes da Silva
- MRC Laboratory of Molecular Cell Biology, University College London, Gower Street, London, WC1E6BT, UK
| | - Francesco Ferraro
- MRC Laboratory of Molecular Cell Biology, University College London, Gower Street, London, WC1E6BT, UK
| | - Francesca Patella
- MRC Laboratory of Molecular Cell Biology, University College London, Gower Street, London, WC1E6BT, UK
| | - Daniel F Cutler
- MRC Laboratory of Molecular Cell Biology, University College London, Gower Street, London, WC1E6BT, UK
| |
Collapse
|
45
|
Abstract
PURPOSE OF REVIEW In the last nine decades, large advances have been made toward the characterization of the pathogenic basis and clinical management of von Willebrand disease (VWD), the most prevalent inherited bleeding disorder. Pathological variations at the von Willebrand factor (VWF) locus present as a range of both quantitative and qualitative abnormalities that make up the complex clinical spectrum of VWD. This review describes the current understanding of the pathobiological basis of VWD. RECENT FINDINGS The molecular basis of type 2 (qualitative abnormalities) and type 3 VWD (total quantitative deficiency) have been well characterized in recent decades. However, knowledge of type 1 VWD (partial quantitative deficiency) remains incomplete because of the allelic and locus heterogeneity of this trait, and is complicated by genetic variability at the VWF gene, interactions between the VWF gene and the environment, and the involvement of external modifying loci. Recent genome wide association studies and linkage analyses have sought to identify additional genes that modify the type 1 VWD phenotype. SUMMARY Understanding the pathogenic basis of VWD will facilitate the development of novel treatment regimens for this disorder, and improve the ability to provide complementary molecular diagnostics for type 1 VWD.
Collapse
|
46
|
VAMP3 and SNAP23 mediate the disturbed flow-induced endothelial microRNA secretion and smooth muscle hyperplasia. Proc Natl Acad Sci U S A 2017; 114:8271-8276. [PMID: 28716920 DOI: 10.1073/pnas.1700561114] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Vascular endothelial cells (ECs) at arterial branches and curvatures experience disturbed blood flow and induce a quiescent-to-activated phenotypic transition of the adjacent smooth muscle cells (SMCs) and a subsequent smooth muscle hyperplasia. However, the mechanism underlying the flow pattern-specific initiation of EC-to-SMC signaling remains elusive. Our previous study demonstrated that endothelial microRNA-126-3p (miR-126-3p) acts as a key intercellular molecule to increase turnover of the recipient SMCs, and that its release is reduced by atheroprotective laminar shear (12 dynes/cm2) to ECs. Here we provide evidence that atherogenic oscillatory shear (0.5 ± 4 dynes/cm2), but not atheroprotective pulsatile shear (12 ± 4 dynes/cm2), increases the endothelial secretion of nonmembrane-bound miR-126-3p and other microRNAs (miRNAs) via the activation of SNAREs, vesicle-associated membrane protein 3 (VAMP3) and synaptosomal-associated protein 23 (SNAP23). Knockdown of VAMP3 and SNAP23 reduces endothelial secretion of miR-126-3p and miR-200a-3p, as well as the proliferation, migration, and suppression of contractile markers in SMCs caused by EC-coculture. Pharmacological intervention of mammalian target of rapamycin complex 1 in ECs blocks endothelial secretion and EC-to-SMC transfer of miR-126-3p through transcriptional inhibition of VAMP3 and SNAP23. Systemic inhibition of VAMP3 and SNAP23 by rapamycin or periadventitial application of the endocytosis inhibitor dynasore ameliorates the disturbed flow-induced neointimal formation, whereas intraluminal overexpression of SNAP23 aggravates it. Our findings demonstrate the flow-pattern-specificity of SNARE activation and its contribution to the miRNA-mediated EC-SMC communication.
Collapse
|
47
|
Da Q, Shaw T, Pradhan S, Roche PA, Cruz MA, Vijayan KV. Disruption of protein complexes containing protein phosphatase 2B and Munc18c reduces the secretion of von Willebrand factor from endothelial cells. J Thromb Haemost 2017; 15:1032-1039. [PMID: 28294518 PMCID: PMC5413388 DOI: 10.1111/jth.13671] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Indexed: 12/01/2022]
Abstract
Essentials Endothelial secretion of von Willebrand factor (VWF) promotes inflammation and thrombosis. We studied the role of protein phosphatase 2B (PP2B) and Munc18c protein complex in VWF secretion. Disruption of PP2B-Munc18c complex in endothelial cells reduced agonist-induced VWF secretion. PP2B-Munc18c complex represents a potential target for thrombotic and inflammatory conditions. SUMMARY Background Aberrant secretion of von Willebrand factor (VWF) from endothelial cells contributes to inflammation and vascular thrombosis. Agonist-induced VWF secretion is facilitated by protein kinase and phosphatase-mediated signaling. Although the catalytic subunit of protein phosphatase 2B (PP2B-Aα) is targeted to the secretory machinery via an interaction with the vesicle trafficking protein Munc18c in endothelial cells, the functional relevance of this phosphatase complex is unclear. Objective To assess the contribution of the PP2B-Aα-Munc18c complex to endothelial VWF secretion. Results Here, we show that amino acids 120-130 of PP2B-Aα are important to support an interaction with Munc18c. A synthetic myristylated cell-permeable peptide, which is derived from amino acids 121-130 of PP2B-Aα, disrupted endogenous PP2B-Aα-Munc18c complexes in human umbilical vein endothelial cells, and decreased low-dose histamine-stimulated and thrombin-stimulated VWF secretion. Conclusion These studies indicate that PP2B-Aα-Munc18c complex supports agonist-induced VWF secretion, and suggest the potential of targeting this phosphatase complex in thrombotic and inflammatory conditions.
Collapse
Affiliation(s)
- Qi Da
- Department of Medicine, Michael E. DeBakey Veterans Affairs Medical Center (MEDVAMC), Houston, TX 77030, U.S.A
- Baylor College of Medicine and Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey Veterans Affairs Medical Center (MEDVAMC), Houston, TX 77030, U.S.A
| | - Tanner Shaw
- Department of Medicine, Michael E. DeBakey Veterans Affairs Medical Center (MEDVAMC), Houston, TX 77030, U.S.A
- Baylor College of Medicine and Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey Veterans Affairs Medical Center (MEDVAMC), Houston, TX 77030, U.S.A
| | - Subhashree Pradhan
- Department of Medicine, Michael E. DeBakey Veterans Affairs Medical Center (MEDVAMC), Houston, TX 77030, U.S.A
- Baylor College of Medicine and Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey Veterans Affairs Medical Center (MEDVAMC), Houston, TX 77030, U.S.A
| | - Paul A. Roche
- National Cancer Institute, Bethesda, MD 20892, U.S.A
| | - Miguel A. Cruz
- Department of Medicine, Michael E. DeBakey Veterans Affairs Medical Center (MEDVAMC), Houston, TX 77030, U.S.A
- Baylor College of Medicine and Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey Veterans Affairs Medical Center (MEDVAMC), Houston, TX 77030, U.S.A
- Department of Pediatrics, Michael E. DeBakey Veterans Affairs Medical Center (MEDVAMC), Houston, TX 77030, U.S.A
- Department of Molecular Physiology and Biophysics, Michael E. DeBakey Veterans Affairs Medical Center (MEDVAMC), Houston, TX 77030, U.S.A
| | - K. Vinod Vijayan
- Department of Medicine, Michael E. DeBakey Veterans Affairs Medical Center (MEDVAMC), Houston, TX 77030, U.S.A
- Baylor College of Medicine and Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey Veterans Affairs Medical Center (MEDVAMC), Houston, TX 77030, U.S.A
- Department of Pediatrics, Michael E. DeBakey Veterans Affairs Medical Center (MEDVAMC), Houston, TX 77030, U.S.A
- Department of Molecular Physiology and Biophysics, Michael E. DeBakey Veterans Affairs Medical Center (MEDVAMC), Houston, TX 77030, U.S.A
| |
Collapse
|
48
|
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
|
49
|
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
|
50
|
Ganesana M, Lee ST, Wang Y, Venton BJ. Analytical Techniques in Neuroscience: Recent Advances in Imaging, Separation, and Electrochemical Methods. Anal Chem 2017; 89:314-341. [PMID: 28105819 PMCID: PMC5260807 DOI: 10.1021/acs.analchem.6b04278] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
| | | | | | - B. Jill Venton
- Department of Chemistry, PO Box 400319, University of Virginia, Charlottesville, VA 22904
| |
Collapse
|