1
|
Flint JP, Welstead M, Cox SR, Russ TC, Marshall A, Luciano M. Validation of a polygenic risk score for frailty in the Lothian Birth Cohort 1936 and English longitudinal study of ageing. Sci Rep 2024; 14:12586. [PMID: 38822050 PMCID: PMC11143351 DOI: 10.1038/s41598-024-63229-y] [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: 04/18/2023] [Accepted: 05/24/2024] [Indexed: 06/02/2024] Open
Abstract
Frailty is a complex trait. Twin studies and high-powered Genome Wide Association Studies conducted in the UK Biobank have demonstrated a strong genetic basis of frailty. The present study utilized summary statistics from a Genome Wide Association Study on the Frailty Index to create and test the predictive power of frailty polygenic risk scores (PRS) in two independent samples - the Lothian Birth Cohort 1936 (LBC1936) and the English Longitudinal Study of Ageing (ELSA) aged 67-84 years. Multiple regression models were built to test the predictive power of frailty PRS at five time points. Frailty PRS significantly predicted frailty, measured via the FI, at all-time points in LBC1936 and ELSA, explaining 2.1% (β = 0.15, 95%CI, 0.085-0.21) and 1.8% (β = 0.14, 95%CI, 0.10-0.17) of the variance, respectively, at age ~ 68/ ~ 70 years (p < 0.001). This work demonstrates that frailty PRS can predict frailty in two independent cohorts, particularly at early ages (~ 68/ ~ 70). PRS have the potential to be valuable instruments for identifying those at risk for frailty and could be important for controlling for genetic confounders in epidemiological studies.
Collapse
Affiliation(s)
- J P Flint
- Advanced Care Research Centre, School of Engineering, College of Science and Engineering, The University of Edinburgh, Edinburgh, UK.
- Lothian Birth Cohorts, Department of Psychology, University of Edinburgh, Edinburgh, UK.
- Department of Psychology, School of Philosophy, Psychology and Language Sciences, University of Edinburgh, Edinburgh, UK.
- Alzheimer Scotland Dementia Research Centre, University of Edinburgh, Edinburgh, UK.
| | - M Welstead
- Department of Psychology, School of Philosophy, Psychology and Language Sciences, University of Edinburgh, Edinburgh, UK
- Alzheimer Scotland Dementia Research Centre, University of Edinburgh, Edinburgh, UK
| | - S R Cox
- Lothian Birth Cohorts, Department of Psychology, University of Edinburgh, Edinburgh, UK
- Department of Psychology, School of Philosophy, Psychology and Language Sciences, University of Edinburgh, Edinburgh, UK
| | - T C Russ
- Alzheimer Scotland Dementia Research Centre, University of Edinburgh, Edinburgh, UK
- Division of Psychiatry, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - A Marshall
- Advanced Care Research Centre, School of Engineering, College of Science and Engineering, The University of Edinburgh, Edinburgh, UK
- School of Social and Political Science, University of Edinburgh, Edinburgh, UK
| | - M Luciano
- Lothian Birth Cohorts, Department of Psychology, University of Edinburgh, Edinburgh, UK
- Department of Psychology, School of Philosophy, Psychology and Language Sciences, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
2
|
Li C, Barroeta AB, Wong SS, Kim HJ, Pathak M, Dreveny I, Meijers JCM, Emsley J. Structures of factor XI and prekallikrein bound to domain 6 of high-molecular weight kininogen reveal alternate domain 6 conformations and exosites. J Thromb Haemost 2023; 21:2378-2389. [PMID: 37068593 DOI: 10.1016/j.jtha.2023.03.042] [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: 09/22/2022] [Revised: 03/15/2023] [Accepted: 03/24/2023] [Indexed: 04/19/2023]
Abstract
BACKGROUND High-molecular weight kininogen (HK) circulates in plasma as a complex with zymogen prekallikrein (PK). HK is both a substrate and a cofactor for activated plasma kallikrein, and the principal exosite interactions occur between PK N-terminal apple domains and the C-terminal D6 domain of HK. OBJECTIVES To determine the structure of the complex formed between PK apple domains and an HKD6 fragment and compare this with the coagulation factor XI (FXI)-HK complex. METHODS We produced recombinant FXI and PK heavy chains (HCs) spanning all 4 apple domains. We cocrystallized PKHC (and subsequently FXIHC) with a 31-amino acid synthetic peptide spanning HK residues Ser565-Lys595 and determined the crystal structure. We also analyzed the full-length FXI-HK complex in solution using hydrogen deuterium exchange mass spectrometry. RESULTS The 2.3Å PKHC-HK peptide crystal structure revealed that the HKD6 sequence WIPDIQ (Trp569-Gln574) binds to the apple 1 domain and HK FNPISDFPDT (Phe582-Thr591) binds to the apple 2 domain with a flexible intervening sequence resulting in a bent double conformation. A second 3.2Å FXIHC-HK peptide crystal structure revealed a similar interaction with the apple 2 domain but an alternate, straightened conformation of the HK peptide where residues LSFN (Leu579-Asn583) interacts with a unique pocket formed between the apple 2 and 3 domains. HDX-MS of full length FXI-HK complex in solution confirmed interactions with both apple 2 and apple 3. CONCLUSIONS The alternate conformations and exosite binding of the HKD6 peptide likely reflects the diverging relationship of HK to the functions of PK and FXI.
Collapse
Affiliation(s)
- Chan Li
- Biodiscovery Institute, School of Pharmacy, University of Nottingham, Nottingham, UK
| | - Awital Bar Barroeta
- Department of Molecular Hematology, Sanquin Research, Amsterdam, The Netherlands
| | - Szu Shen Wong
- School of Pharmacy and Bioengineering, Keele University, Staffordshire, UK
| | - Hyo Jung Kim
- Biodiscovery Institute, School of Pharmacy, University of Nottingham, Nottingham, UK
| | - Monika Pathak
- Biodiscovery Institute, School of Pharmacy, University of Nottingham, Nottingham, UK
| | - Ingrid Dreveny
- Biodiscovery Institute, School of Pharmacy, University of Nottingham, Nottingham, UK
| | - Joost C M Meijers
- Department of Molecular Hematology, Sanquin Research, Amsterdam, The Netherlands; Amsterdam UMC, University of Amsterdam, department of Experimental Vascular Medicine, Amsterdam, The Netherlands; Amsterdam Cardiovascular Sciences, Pulmonary Hypertension and Thrombosis, Amsterdam, The Netherlands
| | - Jonas Emsley
- Biodiscovery Institute, School of Pharmacy, University of Nottingham, Nottingham, UK.
| |
Collapse
|
3
|
Besin V, Yulianti T, Notopuro PB, Humardani FM. Genetic Polymorphisms of Ischemic Stroke in Asians. Clin Chim Acta 2023; 549:117527. [PMID: 37666385 DOI: 10.1016/j.cca.2023.117527] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 08/25/2023] [Accepted: 08/25/2023] [Indexed: 09/06/2023]
Abstract
The increasing incidence of ischemic stroke emphasizes the necessity for early detection and preventive strategies. Diagnostic biomarkers currently available for ischemic stroke only become detectable shortly before the manifestation of stroke symptoms. Genetic variants associated with ischemic stroke offer a potential solution to address this diagnostic limitation. However, it is crucial to acknowledge that genetic variants cannot be modified in the same way as epigenetic changes. Nevertheless, individuals carrying risk or protective variants can modify their lifestyle to potentially influence the associated epigenetic factors. This study aims to summarize specific variants relevant to Asian populations that may aid in the early detection of ischemic stroke and explore their impact on the disease's pathophysiology. These variants give us important information about the genes that play a role in ischemic stroke by affecting things like atherosclerosis pathway, blood coagulation pathway, homocysteine metabolism, transporter function, transcription, and the activity of neurons regulation. It is important to recognize the variations in genetic variants among different ethnicities and avoid generalizing the pathogenesis of ischemic stroke.
Collapse
Affiliation(s)
- Valentinus Besin
- Faculty of Medicine, University of Surabaya, Surabaya 60292, Indonesia
| | - Trilis Yulianti
- Faculty of Medicine, Universitas Sumatera Utara, Medan 20155, Indonesia
| | - Paulus Budiono Notopuro
- Department of Clinical Pathology, Faculty of Medicine, Universitas Airlangga, Surabaya 60132, Indonesia
| | - Farizky Martriano Humardani
- Faculty of Medicine, University of Surabaya, Surabaya 60292, Indonesia; Magister in Biomedical Science Program, Faculty of Medicine Universitas Brawijaya, Malang 65112, Indonesia.
| |
Collapse
|
4
|
Flint JP, Welstead M, Cox SR, Russ TC, Marshall A, Luciano M. Validation of a polygenic risk score for Frailty in the Lothian Birth Cohort and English Longitudinal Study of Ageing. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.04.03.23288064. [PMID: 37066324 PMCID: PMC10104224 DOI: 10.1101/2023.04.03.23288064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/29/2023]
Abstract
Frailty is a complex trait. Twin studies and a high-powered Genome Wide Association Study (GWAS) conducted in the UK Biobank have demonstrated a strong genetic basis of frailty. The present study utilized summary statistics from this GWAS to create and test the predictive power of frailty polygenic risk scores (PRS) in two independent samples - the Lothian Birth Cohort 1936 (LBC1936) and the English Longitudinal Study of Ageing (ELSA) aged 67-84 years. Multiple regression models were built to test the predictive power of frailty PRS at five time points. Frailty PRS significantly predicted frailty at all-time points in LBC1936 and ELSA, explaining 2.1% (β = 0.15, 95%CI, 0.085-0.21) and 1.6% (β = 0.14, 95%CI, 0.10-0.17) of the variance, respectively, at age ~68/~70 years (p < 0.001). This work demonstrates that frailty PRS can predict frailty in two independent cohorts, particularly at early ages (~68/~70). PRS have the potential to be valuable instruments for identifying those at risk for frailty and could be important for controlling for genetic confounders in epidemiological studies.
Collapse
Affiliation(s)
- J P Flint
- Advanced Care Research Centre School of Engineering, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, UK
- Lothian Birth Cohorts, Department of Psychology, University of Edinburgh, Edinburgh, UK
- Department of Psychology, School of Philosophy, Psychology and Language Sciences, University of Edinburgh, Edinburgh, UK
- Alzheimer Scotland Dementia Research Centre, University of Edinburgh, Edinburgh, UK
| | - M Welstead
- Department of Psychology, School of Philosophy, Psychology and Language Sciences, University of Edinburgh, Edinburgh, UK
- Alzheimer Scotland Dementia Research Centre, University of Edinburgh, Edinburgh, UK
| | - S R Cox
- Lothian Birth Cohorts, Department of Psychology, University of Edinburgh, Edinburgh, UK
- Department of Psychology, School of Philosophy, Psychology and Language Sciences, University of Edinburgh, Edinburgh, UK
| | - T C Russ
- Alzheimer Scotland Dementia Research Centre, University of Edinburgh, Edinburgh, UK
- Division of Psychiatry, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - A Marshall
- Advanced Care Research Centre School of Engineering, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, UK
- School of Social and Political Science, University of Edinburgh, Edinburgh, UK
| | - M Luciano
- Lothian Birth Cohorts, Department of Psychology, University of Edinburgh, Edinburgh, UK
- Department of Psychology, School of Philosophy, Psychology and Language Sciences, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
5
|
Muko R, Sunouchi T, Urayama S, Toishi Y, Kusano K, Sato H, Muranaka M, Shin T, Oikawa MA, Ojima Y, Ali M, Nomura Y, Matsuda H, Tanaka A. Unique insertion/deletion polymorphisms within histidine-rich region of histidine-rich glycoprotein in Thoroughbred horses. Sci Rep 2023; 13:300. [PMID: 36609619 PMCID: PMC9822902 DOI: 10.1038/s41598-023-27374-0] [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: 08/17/2022] [Accepted: 01/02/2023] [Indexed: 01/07/2023] Open
Abstract
Histidine-rich glycoprotein (HRG) is abundant plasma protein with various effects on angiogenesis, coagulation, and immune responses. Previously, we identified the base and amino acid sequences of equine HRG (eHRG) and revealed that eHRG regulates neutrophil functions. In this study, we first conducted a large-scale gene analysis with DNA samples extracted from 1700 Thoroughbred horses and identified unique insertion/deletion polymorphisms in the histidine-rich region (HRR) of eHRG. Here we report two types of polymorphisms (deletion type 1 [D1] and deletion type 2 [D2]) containing either a 45 bp or 90 bp deletion in the HRR of eHRG, and five genotypes of eHRG (insertion/insertion [II], ID1, ID2, D1D1, and D1D2) in Thoroughbred horses. Allele frequency of I, D1, and D2, was 0.483, 0.480, and 0.037 and the incidence of each genotype was II: 23.4%, ID1: 46.2%, ID2: 3.6%, D1D1: 23.1%, and D1D2: 3.7%, respectively. The molecular weights of each plasma eHRG protein collected from horses with each genotype was detected as bands of different molecular size, which corresponded to the estimated amino acid sequence. The nickel-binding affinity of the D1 or D2 deletion eHRG was reduced, indicating a loss of function at the site. eHRG proteins show a variety of biological and immunological activities in vivo, and HRR is its active center, suggesting that genetic polymorphisms in eHRG may be involved in the performance in athletic ability, productivity, and susceptibility to infectious diseases in Thoroughbred horses.
Collapse
Affiliation(s)
- Ryo Muko
- grid.136594.c0000 0001 0689 5974Institute of Global Innovation Research, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Tomoya Sunouchi
- grid.136594.c0000 0001 0689 5974Laboratory of Comparative Animal Medicine, Division of Animal Life Science, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo, 183-8509 Japan
| | - Shuntaro Urayama
- grid.482817.00000 0001 0710 998XRace Horse Clinic, Ritto Training Center, Japan Racing Association, Shiga, Japan
| | - Yuko Toishi
- Shadai Stallion Station, Shadai Corporation, Hokkaido, Japan
| | - Kanichi Kusano
- grid.482817.00000 0001 0710 998XRace Horse Clinic, Ritto Training Center, Japan Racing Association, Shiga, Japan
| | - Hiroaki Sato
- grid.482817.00000 0001 0710 998XRace Integrity Section, Stewards Department, Japan Racing Association, Tokyo, Japan
| | - Masanori Muranaka
- grid.482817.00000 0001 0710 998XRace Horse Clinic, Ritto Training Center, Japan Racing Association, Shiga, Japan
| | - Taekyun Shin
- grid.411277.60000 0001 0725 5207Department of Veterinary Anatomy, College of Veterinary Medicine and Veterinary Medical Research Institute, Jeju National University, Jeju, South Korea
| | - Masa-aki Oikawa
- grid.507451.20000 0004 7662 6210Diagnostic Laboratory, Equine Veterinary Medical Center, Education City, Doha, Qatar
| | - Yoshinobu Ojima
- grid.136594.c0000 0001 0689 5974Institute of Global Innovation Research, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Mohammad Ali
- grid.507451.20000 0004 7662 6210Diagnostic Laboratory, Equine Veterinary Medical Center, Education City, Doha, Qatar
| | - Yoshihiro Nomura
- grid.136594.c0000 0001 0689 5974Scleroprotein and Leather Research Institute, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Hiroshi Matsuda
- grid.136594.c0000 0001 0689 5974Laboratory of Comparative Animal Medicine, Division of Animal Life Science, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo, 183-8509 Japan
| | - Akane Tanaka
- Institute of Global Innovation Research, Tokyo University of Agriculture and Technology, Tokyo, Japan. .,Laboratory of Comparative Animal Medicine, Division of Animal Life Science, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo, 183-8509, Japan. .,Cooperative Major in Advanced Health Science, Graduate School of Bio-Applications and System Engineering, Tokyo University of Agriculture and Technology, Tokyo, Japan.
| |
Collapse
|
6
|
Gholami M, Zoughi M, Hasanzad M, Larijani B, Amoli MM. Haplotypic variants of COVID-19 related genes are associated with blood pressure and metabolites levels. J Med Virol 2023; 95:e28355. [PMID: 36443248 PMCID: PMC9877746 DOI: 10.1002/jmv.28355] [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: 12/25/2021] [Revised: 07/27/2022] [Accepted: 11/22/2022] [Indexed: 12/03/2022]
Abstract
The genetic association of coronavirus disease 2019 (COVID-19) with its complications has not been fully understood. This study aimed to identify variants and haplotypes of candidate genes implicated in COVID-19 related traits by combining the literature review and pathway analysis. To explore such genes, the protein-protein interactions and relevant pathways of COVID-19-associated genes were assessed. A number of variants on candidate genes were identified from Genome-wide association studies (GWASs) which were associated with COVID-19 related traits (p ˂ 10-6 ). Haplotypic blocks were assessed using haplotypic structures among the 1000 Genomes Project (r2 ≥ 0.8, D' ≥ 0.8). Further functional analyses were performed on the selected variants. The results demonstrated that a group of variants in ACE and AGT genes were significantly correlated with COVID-19 related traits. Three haplotypes were identified to be involved in the blood metabolites levels and the development of blood pressure. Functional analyses revealed that most GWAS index variants were expression quantitative trait loci and had transcription factor binding sites, exonic splicing enhancers or silencer activities. Furthermore, the proxy haplotype variants, rs4316, rs4353, rs4359, and three variants, namely rs2493133, rs2478543, and rs5051, were associated with blood metabolite and systolic blood pressure, respectively. These variants exerted more regulatory effects compared with other GWAS variants. The present study indicates that the genetic variants and candidate haplotypes of COVID-19 related genes are associated with blood pressure and blood metabolites. However, further observational studies are warranted to confirm these results.
Collapse
Affiliation(s)
- Morteza Gholami
- Metabolic Disorders Research Center, Endocrinology and Metabolism Molecular‐Cellular Sciences InstituteTehran University of Medical SciencesTehranIran,Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences InstituteTehran University of Medical SciencesTehranIran
| | - Marziyeh Zoughi
- Metabolomics and genomics research center endocrinology and metabolism molecular‐cellular sciences instituteTehran University of medical sciencesTehranIran
| | - Mandana Hasanzad
- Personalized Medicine Research Center, Endocrinology and Metabolism Clinical Sciences InstituteTehran University of Medical SciencesTehranIran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences InstituteTehran University of Medical SciencesTehranIran
| | - Mahsa M. Amoli
- Metabolic Disorders Research Center, Endocrinology and Metabolism Molecular‐Cellular Sciences InstituteTehran University of Medical SciencesTehranIran
| |
Collapse
|
7
|
Genetic Polymorphisms Associated with Prothrombin Time and Activated Partial Thromboplastin Time in Chinese Healthy Population. Genes (Basel) 2022; 13:genes13101867. [PMID: 36292752 PMCID: PMC9602091 DOI: 10.3390/genes13101867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 09/30/2022] [Accepted: 10/12/2022] [Indexed: 11/04/2022] Open
Abstract
(1) Background: The purpose of this study was to evaluate the effect of gene polymorphisms on prothrombin time (PT) and activated partial thromboplastin time (APTT) in a healthy Chinese population. (2) Methods: A total of 403 healthy volunteers from a series of novel oral anticoagulants (NOACs) bioequivalence trials in China were included. Coagulation tests for PT and APTT were performed in the central lab at Peking University First Hospital. Whole-exome sequencing (WES) and genome-wide association analysis were performed. (3) Results: In the correlation analysis of PT, 105 SNPs from 84 genes reached the genome-wide significance threshold (p < 1 × 10−5). Zinc Finger Protein 594 (ZNF594) rs184838268 (p = 4.50 × 10−19) was most significantly related to PT, and Actinin Alpha 1 (ACTN1) was found to interact most with other candidate genes. Significant associations with previously reported candidate genes Aurora Kinase B (AURKB), Complement C5(C5), Clock Circadian Regulator (CLOCK), and Histone Deacetylase 9(HDAC9) were detected in our dataset (p < 1 × 10−5). PiggyBac Transposable Element Derived 2(PGBD2) rs75935520 (p = 4.49 × 10−6), Bromodomain Adjacent To Zinc Finger Domain 2A(BAZ2A) rs199970765 (p = 5.69 × 10−6) and Protogenin (PRTG) rs80064850 (p = 8.69 × 10−6) were significantly correlated with APTT (p < 1 × 10−5). The heritability values of PT and APTT were 0.83 and 0.64, respectively; (4) Conclusion: The PT and APTT of healthy populations are affected by genetic polymorphisms. ZNF594 and ACTN1 variants could be novel genetic markers of PT, while PRTG polymorphisms might be associated with APTT levels. The findings could be attributed to ethnic differences, and need further investigation.
Collapse
|
8
|
Singh PK, Chen Z, Horn K, Norris EH. Blocking domain 6 of high molecular weight kininogen to understand intrinsic clotting mechanisms. Res Pract Thromb Haemost 2022; 6:e12815. [PMID: 36254255 PMCID: PMC9561425 DOI: 10.1002/rth2.12815] [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: 09/01/2022] [Accepted: 09/05/2022] [Indexed: 11/05/2022] Open
Abstract
Background The contact system is initiated by factor (F) XII activation and the assembly of high molecular weight kininogen (HK) with either FXI or prekallikrein (PK) on a negatively charged surface. Overactivation of this system contributes to thrombosis and inflammation in numerous diseases. To develop effective therapeutics for contact system disorders, a detailed understanding of this pathway is needed. Methods We performed coagulation assays in normal human plasma and various factor-deficient plasmas. To evaluate how HK-mediated PK and FXI activation contributes to coagulation, we used an anti-HK antibody to block access to domain 6 of HK, the region required for efficient activation of PK and FXI. Results FXI's binding to HK and its subsequent activation by activated FXII contributes to coagulation. We found that the 3E8 anti-HK antibody can inhibit the binding of FXI or PK to HK, delaying clot formation in human plasma. Our data show that in the absence of FXI, however, PK can substitute for FXI in this process. Addition of activated FXI (FXIa) or activated PK (PKa) abolished the inhibitory effect of 3E8. Moreover, the requirement of HK in intrinsic coagulation can be largely bypassed by adding FXIa. Like FXIa, exogenous PKa shortened the clotting time in HK-deficient plasma, which was not due to feedback activation of FXII. Conclusions This study improves our understanding of HK-mediated coagulation and provides an explanation for the absence of bleeding in HK-deficient individuals. 3E8 specifically prevented HK-mediated FXI activation; therefore, it could be used to prevent contact activation-mediated thrombosis without altering hemostasis.
Collapse
Affiliation(s)
- Pradeep K. Singh
- Patricia and John Rosenwald Laboratory of Neurobiology and GeneticsThe Rockefeller UniversityNew YorkNew YorkUSA
| | - Zu‐Lin Chen
- Patricia and John Rosenwald Laboratory of Neurobiology and GeneticsThe Rockefeller UniversityNew YorkNew YorkUSA
| | - Katharina Horn
- Patricia and John Rosenwald Laboratory of Neurobiology and GeneticsThe Rockefeller UniversityNew YorkNew YorkUSA
| | - Erin H. Norris
- Patricia and John Rosenwald Laboratory of Neurobiology and GeneticsThe Rockefeller UniversityNew YorkNew YorkUSA
| |
Collapse
|
9
|
Temprano‐Sagrera G, Sitlani CM, Bone WP, Martin‐Bornez M, Voight BF, Morrison AC, Damrauer SM, de Vries PS, Smith NL, Sabater‐Lleal M. Multi-phenotype analyses of hemostatic traits with cardiovascular events reveal novel genetic associations. J Thromb Haemost 2022; 20:1331-1349. [PMID: 35285134 PMCID: PMC9314075 DOI: 10.1111/jth.15698] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 02/15/2022] [Accepted: 03/08/2022] [Indexed: 12/01/2022]
Abstract
BACKGROUND Multi-phenotype analysis of genetically correlated phenotypes can increase the statistical power to detect loci associated with multiple traits, leading to the discovery of novel loci. This is the first study to date to comprehensively analyze the shared genetic effects within different hemostatic traits, and between these and their associated disease outcomes. OBJECTIVES To discover novel genetic associations by combining summary data of correlated hemostatic traits and disease events. METHODS Summary statistics from genome wide-association studies (GWAS) from seven hemostatic traits (factor VII [FVII], factor VIII [FVIII], von Willebrand factor [VWF] factor XI [FXI], fibrinogen, tissue plasminogen activator [tPA], plasminogen activator inhibitor 1 [PAI-1]) and three major cardiovascular (CV) events (venous thromboembolism [VTE], coronary artery disease [CAD], ischemic stroke [IS]), were combined in 27 multi-trait combinations using metaUSAT. Genetic correlations between phenotypes were calculated using Linkage Disequilibrium Score Regression (LDSC). Newly associated loci were investigated for colocalization. We considered a significance threshold of 1.85 × 10-9 obtained after applying Bonferroni correction for the number of multi-trait combinations performed (n = 27). RESULTS Across the 27 multi-trait analyses, we found 4 novel pleiotropic loci (XXYLT1, KNG1, SUGP1/MAU2, TBL2/MLXIPL) that were not significant in the original individual datasets, were not described in previous GWAS for the individual traits, and that presented a common associated variant between the studied phenotypes. CONCLUSIONS The discovery of four novel loci contributes to the understanding of the relationship between hemostasis and CV events and elucidate common genetic factors between these traits.
Collapse
Affiliation(s)
- Gerard Temprano‐Sagrera
- Genomics of Complex Disease UnitSant Pau Biomedical Research Institute. IIB‐Sant PauBarcelonaSpain
| | - Colleen M. Sitlani
- Cardiovascular Health Research UnitDepartment of MedicineUniversity of WashingtonSeattleWashingtonUSA
| | - William P. Bone
- Genomics and Computational Biology Graduate GroupPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Miguel Martin‐Bornez
- Genomics of Complex Disease UnitSant Pau Biomedical Research Institute. IIB‐Sant PauBarcelonaSpain
| | - Benjamin F. Voight
- Department of Systems Pharmacology and Translational Therapeutics and Department of GeneticsUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvaniaUSA
- Institute of Translational Medicine and TherapeuticsUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvaniaUSA
| | - Alanna C. Morrison
- Human Genetics CenterDepartment of EpidemiologyHuman Genetics, and Environmental SciencesSchool of Public HealthThe University of Texas Health Science Center at HoustonHoustonTexasUSA
| | - Scott M. Damrauer
- Department of Surgery and Department of GeneticsPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Corporal Michael Crescenz VA Medical CenterPhiladelphiaPennsylvaniaUSA
| | - Paul S. de Vries
- Human Genetics CenterDepartment of EpidemiologyHuman Genetics, and Environmental SciencesSchool of Public HealthThe University of Texas Health Science Center at HoustonHoustonTexasUSA
| | - Nicholas L. Smith
- Department of EpidemiologyUniversity of WashingtonSeattleWashingtonUSA
- Kaiser Permanente Washington Health Research InstituteKaiser PermanenteSeattleWashingtonUSA
- Seattle Epidemiologic Research and Information CenterDepartment of Veterans Affairs Office of Research and DevelopmentSeattleWashingtonUSA
| | - Maria Sabater‐Lleal
- Genomics of Complex Disease UnitSant Pau Biomedical Research Institute. IIB‐Sant PauBarcelonaSpain
- Cardiovascular Medicine UnitDepartment of MedicineCenter for Molecular MedicineKarolinska InstitutetStockholmSweden
| |
Collapse
|
10
|
Salomi BSB, Solomon R, Turaka VP, Aaron S, Christudass CS. Cryptogenic Stroke in the Young: Role of Candidate Gene Polymorphisms in Indian Patients with Ischemic Etiology. Neurol India 2021; 69:1655-1662. [PMID: 34979665 DOI: 10.4103/0028-3886.333441] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
CONTEXT Strokes that remain without a definite cause even after an extensive workup, termed cryptogenic strokes, constitute up to 30-40% of ischemic strokes (ISs) in the young. Some of them can have a genetic basis. However, the well-established genetic causes account for only a small percentage of these cases. AIM To evaluate the association of cryptogenic young IS with 16 candidate gene polymorphisms. SETTINGS AND DESIGN A case-control study with cryptogenic young IS patients (South and North Indians; n = 105) and age, sex, and ethnicity-matched controls (n = 215). SUBJECTS AND METHODS Genotyping was carried out by PCR-RFLP method using DNA extracted from the blood. STATISTICAL ANALYSIS USED Association of the genotypes with the disease was studied using Chi-square test. RESULTS MTHFR rs1801133 and KNG1 rs710446 showed significant statistical association with cryptogenic young IS (P = 0.0261 and 0.0157, respectively) in the Indian population. Significant association of KNG1 rs710446 (P 0.0036) and FXII rs1801020 (P 0.0376) with cryptogenic young stroke in South Indian males, SERPINC1 rs2227589 in South Indian female patients (P = 0.0374), and CYP4V2 rs13146272 in North Indian males (P = 0.0293) was observed. CONCLUSIONS Our study indicates that in the Indian population MTHFR rs1801133, KNG rs710446, FXII rs1801020, SERPINC1 rs2227589, CYP4V2 rs13146272, and FXIII V34L may be significant risk factors for cryptogenic IS in the young. In addition, ethnicity and gender play a significant role. Further studies with larger sample size are required to completely establish these polymorphisms as risk factors for cryptogenic IS in young Indians.
Collapse
Affiliation(s)
- Bodda S B Salomi
- Department of Neurological Sciences, Christian Medical College - Vellore, Vellore, Tamil Nadu, India
| | - Raja Solomon
- Department of Neurological Sciences, Christian Medical College - Vellore, Vellore, Tamil Nadu, India
| | - Vijay Prakash Turaka
- Department of General Medicine, Christian Medical College - Vellore, Vellore, Tamil Nadu, India
| | - Sanjith Aaron
- Department of Neurological Sciences, Christian Medical College - Vellore, Vellore, Tamil Nadu, India
| | | |
Collapse
|
11
|
Moksnes MR, Røsjø H, Richmond A, Lyngbakken MN, Graham SE, Hansen AF, Wolford BN, Gagliano Taliun SA, LeFaive J, Rasheed H, Thomas LF, Zhou W, Aung N, Surakka I, Douville NJ, Campbell A, Porteous DJ, Petersen SE, Munroe PB, Welsh P, Sattar N, Smith GD, Fritsche LG, Nielsen JB, Åsvold BO, Hveem K, Hayward C, Willer CJ, Brumpton BM, Omland T. Genome-wide association study of cardiac troponin I in the general population. Hum Mol Genet 2021; 30:2027-2039. [PMID: 33961016 PMCID: PMC8522636 DOI: 10.1093/hmg/ddab124] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 04/23/2021] [Accepted: 04/27/2021] [Indexed: 11/16/2022] Open
Abstract
Circulating cardiac troponin proteins are associated with structural heart disease and predict incident cardiovascular disease in the general population. However, the genetic contribution to cardiac troponin I (cTnI) concentrations and its causal effect on cardiovascular phenotypes are unclear. We combine data from two large population-based studies, the Trøndelag Health Study and the Generation Scotland Scottish Family Health Study, and perform a genome-wide association study of high-sensitivity cTnI concentrations with 48 115 individuals. We further use two-sample Mendelian randomization to investigate the causal effects of circulating cTnI on acute myocardial infarction (AMI) and heart failure (HF). We identified 12 genetic loci (8 novel) associated with cTnI concentrations. Associated protein-altering variants highlighted putative functional genes: CAND2, HABP2, ANO5, APOH, FHOD3, TNFAIP2, KLKB1 and LMAN1. Phenome-wide association tests in 1688 phecodes and 83 continuous traits in UK Biobank showed associations between a genetic risk score for cTnI and cardiac arrhythmias, metabolic and anthropometric measures. Using two-sample Mendelian randomization, we confirmed the non-causal role of cTnI in AMI (5948 cases, 355 246 controls). We found indications for a causal role of cTnI in HF (47 309 cases and 930 014 controls), but this was not supported by secondary analyses using left ventricular mass as outcome (18 257 individuals). Our findings clarify the biology underlying the heritable contribution to circulating cTnI and support cTnI as a non-causal biomarker for AMI in the general population. Using genetically informed methods for causal inference helps inform the role and value of measuring cTnI in the general population.
Collapse
Affiliation(s)
- Marta R Moksnes
- Department of Public Health and Nursing, K.G. Jebsen Center for Genetic Epidemiology, NTNU - Norwegian University of Science and Technology, 7491 Trondheim, Norway
| | - Helge Røsjø
- Division of Research and Innovation, Akershus University Hospital, 1478 Lørenskog, Norway
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, 0316 Oslo, Norway
| | - Anne Richmond
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Magnus N Lyngbakken
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, 0316 Oslo, Norway
- Division of Medicine, Department of Cardiology, Akershus University Hospital, 1478 Lørenskog, Norway
| | - Sarah E Graham
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Ailin Falkmo Hansen
- Department of Public Health and Nursing, K.G. Jebsen Center for Genetic Epidemiology, NTNU - Norwegian University of Science and Technology, 7491 Trondheim, Norway
| | - Brooke N Wolford
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Sarah A Gagliano Taliun
- Faculty of Medicine, Université de Montréal, Montréal, QC H3T 1J4, Canada
- Montréal Heart Institute, Montréal, QC H1T 1C8, Canada
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
- Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
| | - Jonathon LeFaive
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
- Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
| | - Humaira Rasheed
- Department of Public Health and Nursing, K.G. Jebsen Center for Genetic Epidemiology, NTNU - Norwegian University of Science and Technology, 7491 Trondheim, Norway
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol BS8 2BN, UK
| | - Laurent F Thomas
- Department of Public Health and Nursing, K.G. Jebsen Center for Genetic Epidemiology, NTNU - Norwegian University of Science and Technology, 7491 Trondheim, Norway
- Department of Clinical and Molecular Medicine, NTNU - Norwegian University of Science and Technology, 7491 Trondheim, Norway
- BioCore - Bioinformatics Core Facility, NTNU - Norwegian University of Science and Technology, 7491 Trondheim. Norway
- Clinic of Laboratory Medicine, St. Olavs Hospital, Trondheim University Hospital, 7491 Trondheim, Norway
| | - Wei Zhou
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA 02114, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Nay Aung
- William Harvey Research Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
- National Institute for Health Research Barts Cardiovascular Biomedical Research Centre, Queen Mary University of London, London E1 4NS, UK
- Barts Heart Centre, St. Bartholomew’s Hospital, Barts Health NHS Trust, London EC1A 7BE, UK
| | - Ida Surakka
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Nicholas J Douville
- Department of Anesthesiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Archie Campbell
- Medical Genetics Section, CGEM, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - David J Porteous
- Medical Genetics Section, CGEM, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Steffen E Petersen
- William Harvey Research Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
- National Institute for Health Research Barts Cardiovascular Biomedical Research Centre, Queen Mary University of London, London E1 4NS, UK
- Barts Heart Centre, St. Bartholomew’s Hospital, Barts Health NHS Trust, London EC1A 7BE, UK
| | - Patricia B Munroe
- William Harvey Research Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
- National Institute for Health Research Barts Cardiovascular Biomedical Research Centre, Queen Mary University of London, London E1 4NS, UK
| | - Paul Welsh
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow G12 8TA, UK
| | - Naveed Sattar
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow G12 8TA, UK
| | - George Davey Smith
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol BS8 2BN, UK
| | - Lars G Fritsche
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
- Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
| | - Jonas B Nielsen
- Department of Public Health and Nursing, K.G. Jebsen Center for Genetic Epidemiology, NTNU - Norwegian University of Science and Technology, 7491 Trondheim, Norway
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Epidemiology Research, Statens Serum Institute, 2300 Copenhagen, Denmark
- Department of Cardiology, Copenhagen University Hospital, 2100 Copenhagen, Denmark
| | - Bjørn Olav Åsvold
- Department of Public Health and Nursing, K.G. Jebsen Center for Genetic Epidemiology, NTNU - Norwegian University of Science and Technology, 7491 Trondheim, Norway
- Department of Public Health and Nursing, HUNT Research Centre, NTNU - Norwegian University of Science and Technology, 7600 Levanger, Norway
- Department of Endocrinology, St. Olavs Hospital, Trondheim University Hospital, 7006 Trondheim, Norway
| | - Kristian Hveem
- Department of Public Health and Nursing, K.G. Jebsen Center for Genetic Epidemiology, NTNU - Norwegian University of Science and Technology, 7491 Trondheim, Norway
- Department of Public Health and Nursing, HUNT Research Centre, NTNU - Norwegian University of Science and Technology, 7600 Levanger, Norway
| | - Caroline Hayward
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Cristen J Willer
- Department of Public Health and Nursing, K.G. Jebsen Center for Genetic Epidemiology, NTNU - Norwegian University of Science and Technology, 7491 Trondheim, Norway
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Human Genetics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Ben M Brumpton
- Department of Public Health and Nursing, K.G. Jebsen Center for Genetic Epidemiology, NTNU - Norwegian University of Science and Technology, 7491 Trondheim, Norway
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol BS8 2BN, UK
- Clinic of Thoracic and Occupational Medicine, St. Olavs Hospital, Trondheim University Hospital, 7006 Trondheim, Norway
| | - Torbjørn Omland
- Division of Research and Innovation, Akershus University Hospital, 1478 Lørenskog, Norway
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, 0316 Oslo, Norway
- Division of Medicine, Department of Cardiology, Akershus University Hospital, 1478 Lørenskog, Norway
| |
Collapse
|
12
|
Romano AVC, Barnabé A, Gadelha TB, Guerra JCDC, Secolin R, Orsi FLDA, Campanate GDCS, Wolosker N, Annichino-Bizzacchi JM. Gene Variants Associated With Venous Thrombosis: A Replication Study in a Brazilian Multicentre Study. Clin Appl Thromb Hemost 2021; 26:1076029620962225. [PMID: 33119405 PMCID: PMC7607786 DOI: 10.1177/1076029620962225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Single nucleotide polymorphisms (SNP) associated with Venous Thromboembolism (VTE) risk have been identified in European and American populations. Replicate SNPs associated with VTE in a Brazilian multicenter case-control study of the Southeast region. Patients with previous VTE assisted at the Outpatient Clinics of 3 centers of the Southeast Brazilian region were compared to normal controls of the same geographic region. We evaluated 29 SNPs associated with VTE risk in other populations, and 90 SNPs for stratification analysis of the population. Due to high admixture of Brazilian population and lack of previous studies, the calculation of the sample power was performed after genotyping. Sample size, allelic frequency and Hardy-Weinberg equilibrium were estimated. The association and odds ratio analyses were estimated by logistic regression and the results were adjusted for multiple tests using Bonferroni correction. The evaluation of the genetic structure similarity in the cases and controls was performed by AMOVA. 436 cases and 430 controls were included. It was demonstrated that this sample has a statistical power to detect a genetic association of 79.4%. AMOVA showed that the genetic variability between groups was 0.0% and 100% within each group. None of the SNPs showed association with VTE in our population. A Brazilian multicenter case-control study with adequate sample power, high genetic variability though no stratification between groups, showed no replication of SNPs associated with VTE. The high admixture of Brazilian population may be responsible for these results, emphasizing the influence of the population genetic structure in association studies.
Collapse
Affiliation(s)
| | - Aline Barnabé
- Hematology and Hemotherapy Center, 28132University of Campinas (UNICAMP), Campinas, Sao Paulo, Brazil
| | | | | | - Rodrigo Secolin
- Hematology and Hemotherapy Center, Faculty of Medical Sciences, 28132University of Campinas-Unicamp, Campinas, Brazil
| | | | | | - Nelson Wolosker
- Vascular Surgery, Israelite Hospital Albert Einstein, São Paulo, Brazil
| | | |
Collapse
|
13
|
Cis-Segregation of c.1171C>T Stop Codon (p.R391*) in SERPINC1 Gene and c.1691G>A Transition (p.R506Q) in F5 Gene and Selected GWAS Multilocus Approach in Inherited Thrombophilia. Genes (Basel) 2021; 12:genes12060934. [PMID: 34207366 PMCID: PMC8234447 DOI: 10.3390/genes12060934] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 06/12/2021] [Accepted: 06/15/2021] [Indexed: 12/21/2022] Open
Abstract
Inherited thrombophilia (e.g., venous thromboembolism, VTE) is due to rare loss-of-function mutations in anticoagulant factors genes (i.e., SERPINC1, PROC, PROS1), common gain-of-function mutations in procoagulant factors genes (i.e., F5, F2), and acquired risk conditions. Genome Wide Association Studies (GWAS) recently recognized several genes associated with VTE though gene defects may unpredictably remain asymptomatic, so calculating the individual genetic predisposition is a challenging task. We investigated a large family with severe, recurrent, early-onset VTE in which two sisters experienced VTE during pregnancies characterized by a perinatal in-utero thrombosis in the newborn and a life-saving pregnancy-interruption because of massive VTE, respectively. A nonsense mutation (CGA > TGA) generating a premature stop-codon (c.1171C>T; p.R391*) in the exon 6 of SERPINC1 gene (1q25.1) causing Antithrombin (AT) deficiency and the common missense mutation (c.1691G>A; p.R506Q) in the exon 10 of F5 gene (1q24.2) (i.e., FV Leiden; rs6025) were coinherited in all the symptomatic members investigated suspecting a cis-segregation further confirmed by STR-linkage-analyses [i.e., SERPINC1 IVS5 (ATT)5–18, F5 IVS2 (AT)6–33 and F5 IVS11 (GT)12–16] and SERPINC1 intragenic variants (i.e., rs5878 and rs677). A multilocus investigation of blood-coagulation balance genes detected the coexistence of FV Leiden (rs6025) in trans with FV HR2-haplotype (p.H1299R; rs1800595) in the aborted fetus, and F11 rs2289252, F12 rs1801020, F13A1 rs5985, and KNG1 rs710446 in the newborn and other members. Common selected gene variants may strongly synergize with less common mutations tuning potential life-threatening conditions when combined with rare severest mutations. Merging classic and newly GWAS-identified gene markers in at risk families is mandatory for VTE risk estimation in the clinical practice, avoiding partial risk score evaluation in unrecognized at risk patients.
Collapse
|
14
|
Fredenburgh JC, Weitz JI. New anticoagulants: Moving beyond the direct oral anticoagulants. J Thromb Haemost 2021; 19:20-29. [PMID: 33047462 DOI: 10.1111/jth.15126] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 09/21/2020] [Accepted: 09/28/2020] [Indexed: 01/23/2023]
Abstract
Although anticoagulants have been in use for more than 80 years, heparin and vitamin K antagonists were the sole available options until recently. Although these agents revolutionized the prevention and treatment of thrombotic diseases, their use has been hampered by the necessity for coagulation monitoring and by bleeding complications resulting in part from their multiple sites of action. Owing to advances in basic science, animal models, and epidemiology, the arsenal of available anticoagulants has expanded in the past two decades. This evolution has yielded many novel compounds that target single coagulation enzymes. Initially, thrombin and factor Xa were targeted because of their critical roles in coagulation. However, attention has now shifted to compounds that target upstream reactions, particularly those catalyzed by factors XIIa and XIa, which are part of the contact system. This shift is predicated on epidemiological and experimental evidence suggesting that these factors are more important for thrombosis than for hemostasis. With the goal of developing a new class of anticoagulants associated with a lower risk of bleeding than currently available agents, dozens of drugs targeting the contact system are now in development. This article focuses on the rationale, development, and testing of these new agents with a concentration on those that have reached or completed phase 2 evaluation for at least one indication.
Collapse
Affiliation(s)
- James C Fredenburgh
- Thrombosis and Atherosclerosis Research Institute, McMaster University, Hamilton, ON, Canada
- Departments of Medicine, McMaster University, Hamilton, ON, Canada
| | - Jeffrey I Weitz
- Thrombosis and Atherosclerosis Research Institute, McMaster University, Hamilton, ON, Canada
- Departments of Medicine, McMaster University, Hamilton, ON, Canada
- Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
15
|
Hong MG, Dodig-Crnković T, Chen X, Drobin K, Lee W, Wang Y, Edfors F, Kotol D, Thomas CE, Sjöberg R, Odeberg J, Hamsten A, Silveira A, Hall P, Nilsson P, Pawitan Y, Uhlén M, Pedersen NL, Hägg S, Magnusson PK, Schwenk JM. Profiles of histidine-rich glycoprotein associate with age and risk of all-cause mortality. Life Sci Alliance 2020; 3:3/10/e202000817. [PMID: 32737166 PMCID: PMC7409555 DOI: 10.26508/lsa.202000817] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 07/22/2020] [Accepted: 07/22/2020] [Indexed: 12/11/2022] Open
Abstract
Despite recognizing aging as a common risk factor of many human diseases, little is known about its molecular traits. To identify age-associated proteins circulating in human blood, we screened 156 individuals aged 50-92 using exploratory and multiplexed affinity proteomics assays. Profiling eight additional study sets (N = 3,987), performing antibody validation, and conducting a meta-analysis revealed a consistent age association (P = 6.61 × 10-6) for circulating histidine-rich glycoprotein (HRG). Sequence variants of HRG influenced how the protein was recognized in the immunoassays. Indeed, only the HRG profiles affected by rs9898 were associated with age and predicted the risk of mortality (HR = 1.25 per SD; 95% CI = 1.12-1.39; P = 6.45 × 10-5) during a follow-up period of 8.5 yr after blood sampling (IQR = 7.7-9.3 yr). Our affinity proteomics analysis found associations between the particular molecular traits of circulating HRG with age and all-cause mortality. The distinct profiles of this multipurpose protein could serve as an accessible and informative indicator of the physiological processes related to biological aging.
Collapse
Affiliation(s)
- Mun-Gwan Hong
- Department of Protein Science, Science for Life Laboratory, KTH-Royal Institute of Technology, Solna, Sweden
| | - Tea Dodig-Crnković
- Department of Protein Science, Science for Life Laboratory, KTH-Royal Institute of Technology, Solna, Sweden
| | - Xu Chen
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Kimi Drobin
- Department of Protein Science, Science for Life Laboratory, KTH-Royal Institute of Technology, Solna, Sweden
| | - Woojoo Lee
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden.,Department of Public Health Science, Graduate School of Public Health, Seoul National University, Seoul, Korea
| | - Yunzhang Wang
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Fredrik Edfors
- Department of Protein Science, Science for Life Laboratory, KTH-Royal Institute of Technology, Solna, Sweden
| | - David Kotol
- Department of Protein Science, Science for Life Laboratory, KTH-Royal Institute of Technology, Solna, Sweden
| | - Cecilia Engel Thomas
- Department of Protein Science, Science for Life Laboratory, KTH-Royal Institute of Technology, Solna, Sweden
| | - Ronald Sjöberg
- Department of Protein Science, Science for Life Laboratory, KTH-Royal Institute of Technology, Solna, Sweden
| | - Jacob Odeberg
- Department of Protein Science, Science for Life Laboratory, KTH-Royal Institute of Technology, Solna, Sweden.,Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Solna, Sweden
| | - Anders Hamsten
- Department of Medicine Solna, Cardiovascular Medicine Unit, Karolinska Institutet, Solna, Sweden
| | - Angela Silveira
- Department of Medicine Solna, Cardiovascular Medicine Unit, Karolinska Institutet, Solna, Sweden
| | - Per Hall
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden.,Department of Oncology, Södersjukhuset, Stockholm, Sweden
| | - Peter Nilsson
- Department of Protein Science, Science for Life Laboratory, KTH-Royal Institute of Technology, Solna, Sweden
| | - Yudi Pawitan
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Mathias Uhlén
- Department of Protein Science, Science for Life Laboratory, KTH-Royal Institute of Technology, Solna, Sweden
| | - Nancy L Pedersen
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Sara Hägg
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Patrik Ke Magnusson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Jochen M Schwenk
- Department of Protein Science, Science for Life Laboratory, KTH-Royal Institute of Technology, Solna, Sweden
| |
Collapse
|
16
|
Pan Y, Wang L, Xie Y, Tan Y, Chang C, Qiu X, Li X. Characterization of differentially expressed plasma proteins in patients with acute myocardial infarction. J Proteomics 2020; 227:103923. [PMID: 32736138 DOI: 10.1016/j.jprot.2020.103923] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Revised: 07/12/2020] [Accepted: 07/24/2020] [Indexed: 12/11/2022]
Abstract
Acute myocardial infarction (AMI) remains a leading cause of morbidity and mortality worldwide. Novel biomarkers are needed to identify NSTEMI in AMI patients. The study objective was to use proteomics to identify novel plasma biomarkers for STEMI and NSTEMI patients. iTRAQ analysis was performed on pooled samples from 8 healthy controls and 12 STEMI and 12 NSTEMI patients. Bioinformatics analysis identified 95 differentially expressed proteins that were differentially expressed in the plasma of AMI patients and healthy controls; 28 of these proteins were found in STEMI/Con (22 upregulated and 6 downregulated), 48 in NSTEMI/Con (12 upregulated and 36 downregulated), and 44 in NSTEMI/STEMI (11 upregulated and 33 downregulated). Protein network analysis was then performed using STRING software. Functional analysis revealed that the identified plasma proteins were mainly involved with carbon metabolism, toll-like receptor signaling pathway, and hypertrophic cardiomyopathy. Nine of the proteins (SSA1, MDH1, FCN2, GPI, S100A8, LBP, vinculin, VDBP, and RBP4) that changed levels during AMI progression were further validated by ELISA. The constructed plasma proteome could reflect the AMI pathogenesis molecular mechanisms and provide a method for the early identification of NSTEMI in AMI patients. SIGNIFICANCE: The aim of this study was to use proteomics to identify novel predictive plasma biomarkers for patients with acute myocardial infarction (AMI), which would allow for either identification of individuals at risk of an infarction, and early identification of NSTEMI in patients with AMI. Using an approach that combined iTRAQ with LC-MS/MS, we found 95 proteins that showed significant differences in expression levels among the AMI patients and healthy controls. The proteins SSA1, MDH1, FCN2, GPI, S100A8, LBP, vinculin, VDBP, and RBP4 were found to play crucial roles in the pathogenesis of AMI. Using bioinformatics analysis, we found that dysregulation of carbon metabolism, toll-like receptor signaling pathway, and hypertrophic cardiomyopathy may be the major driving forces for cardiac damage during myocardial infarction. However, further investigations are needed to verify the mechanisms involved in the development of AMI especially NSTEMI. Taken together, our findings lay the foundation for understanding the molecular mechanisms underlying the pathogenic processes of AMI, and suggest potential applications for specific biomarkers in early diagnosis and determination of prognosis.
Collapse
Affiliation(s)
- Yilong Pan
- Department of Cardiology, Shengjing Hospital of China Medical University, NO.36 Sanhao Street, Heping District, Shenyang 110004, China
| | - Linlin Wang
- Department of Cardiology, Shengjing Hospital of China Medical University, NO.36 Sanhao Street, Heping District, Shenyang 110004, China
| | - Yaofeng Xie
- Department of Cardiology, Shengjing Hospital of China Medical University, NO.36 Sanhao Street, Heping District, Shenyang 110004, China
| | - Yuan Tan
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Cheng Chang
- Department of Cardiology, Shengjing Hospital of China Medical University, NO.36 Sanhao Street, Heping District, Shenyang 110004, China
| | - Xueshan Qiu
- Department of Pathology, The First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Xiaodong Li
- Department of Cardiology, Shengjing Hospital of China Medical University, NO.36 Sanhao Street, Heping District, Shenyang 110004, China.
| |
Collapse
|
17
|
Dodig-Crnković T, Hong MG, Thomas CE, Häussler RS, Bendes A, Dale M, Edfors F, Forsström B, Magnusson PKE, Schuppe-Koistinen I, Odeberg J, Fagerberg L, Gummesson A, Bergström G, Uhlén M, Schwenk JM. Facets of individual-specific health signatures determined from longitudinal plasma proteome profiling. EBioMedicine 2020; 57:102854. [PMID: 32629387 PMCID: PMC7334812 DOI: 10.1016/j.ebiom.2020.102854] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 06/01/2020] [Accepted: 06/09/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Precision medicine approaches aim to tackle diseases on an individual level through molecular profiling. Despite the growing knowledge about diseases and the reported diversity of molecular phenotypes, the descriptions of human health on an individual level have been far less elaborate. METHODS To provide insights into the longitudinal protein signatures of well-being, we profiled blood plasma collected over one year from 101 clinically healthy individuals using multiplexed antibody assays. After applying an antibody validation scheme, we utilized > 700 protein profiles for in-depth analyses of the individuals' short-term health trajectories. FINDINGS We found signatures of circulating proteomes to be highly individual-specific. Considering technical and longitudinal variability, we observed that 49% of the protein profiles were stable over one year. We also identified eight networks of proteins in which 11-242 proteins covaried over time. For each participant, there were unique protein profiles of which some could be explained by associations to genetic variants. INTERPRETATION This observational and non-interventional study identifyed noticeable diversity among clinically healthy subjects, and facets of individual-specific signatures emerged by monitoring the variability of the circulating proteomes over time. To enable more personal hence precise assessments of health states, longitudinal profiling of circulating proteomes can provide a valuable component for precision medicine approaches. FUNDING This work was supported by the Erling Persson Foundation, the Swedish Heart and Lung Foundation, the Knut and Alice Wallenberg Foundation, Science for Life Laboratory, and the Swedish Research Council.
Collapse
Affiliation(s)
- Tea Dodig-Crnković
- Science for Life Laboratory, Department of Protein Science, KTH-Royal Institute of Technology, Tomtebodavägen 23, Stockholm 171 65, Sweden
| | - Mun-Gwan Hong
- Science for Life Laboratory, Department of Protein Science, KTH-Royal Institute of Technology, Tomtebodavägen 23, Stockholm 171 65, Sweden
| | - Cecilia Engel Thomas
- Science for Life Laboratory, Department of Protein Science, KTH-Royal Institute of Technology, Tomtebodavägen 23, Stockholm 171 65, Sweden
| | - Ragna S Häussler
- Science for Life Laboratory, Department of Protein Science, KTH-Royal Institute of Technology, Tomtebodavägen 23, Stockholm 171 65, Sweden
| | - Annika Bendes
- Science for Life Laboratory, Department of Protein Science, KTH-Royal Institute of Technology, Tomtebodavägen 23, Stockholm 171 65, Sweden
| | - Matilda Dale
- Science for Life Laboratory, Department of Protein Science, KTH-Royal Institute of Technology, Tomtebodavägen 23, Stockholm 171 65, Sweden
| | - Fredrik Edfors
- Science for Life Laboratory, Department of Protein Science, KTH-Royal Institute of Technology, Tomtebodavägen 23, Stockholm 171 65, Sweden; Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Björn Forsström
- Science for Life Laboratory, Department of Protein Science, KTH-Royal Institute of Technology, Tomtebodavägen 23, Stockholm 171 65, Sweden
| | - Patrik K E Magnusson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Nobels väg 12A, Stockholm 171 77, Sweden
| | - Ina Schuppe-Koistinen
- Science for Life Laboratory, Department of Protein Science, KTH-Royal Institute of Technology, Tomtebodavägen 23, Stockholm 171 65, Sweden; Center for Translational Microbiome Research, Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm 171 77, Sweden
| | - Jacob Odeberg
- Science for Life Laboratory, Department of Protein Science, KTH-Royal Institute of Technology, Tomtebodavägen 23, Stockholm 171 65, Sweden; Department of Clinical Medicine, K.G. Jebsen Thrombosis Research and Expertise Center (TREC), UiT the Arctic University of Norway, Tromsø 9010, Norway; Coagulation unit, Department of Hematology, Karolinska University Hospital, Stockholm 171 76, Sweden
| | - Linn Fagerberg
- Science for Life Laboratory, Department of Protein Science, KTH-Royal Institute of Technology, Tomtebodavägen 23, Stockholm 171 65, Sweden
| | - Anders Gummesson
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, Gothenburg University, Gothenburg 413 45, Sweden; Region Västra Götaland, Department of Clinical Genetics and Genomics, Sahlgrenska University Hospital, Gothenburg 413 45, Sweden
| | - Göran Bergström
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, Gothenburg University, Gothenburg 413 45, Sweden; Region Västra Götaland, Department of Clinical Physiology, Sahlgrenska University Hospital, Gothenburg 413 45, Sweden
| | - Mathias Uhlén
- Science for Life Laboratory, Department of Protein Science, KTH-Royal Institute of Technology, Tomtebodavägen 23, Stockholm 171 65, Sweden; Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Lyngby 2800, Denmark
| | - Jochen M Schwenk
- Science for Life Laboratory, Department of Protein Science, KTH-Royal Institute of Technology, Tomtebodavägen 23, Stockholm 171 65, Sweden.
| |
Collapse
|
18
|
Bork K, Wulff K, Rossmann H, Steinmüller‐Magin L, Brænne I, Witzke G, Hardt J. Hereditary angioedema cosegregating with a novel kininogen 1 gene mutation changing the N-terminal cleavage site of bradykinin. Allergy 2019; 74:2479-2481. [PMID: 31087670 DOI: 10.1111/all.13869] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Konrad Bork
- Department of Dermatology, University Medical Center Johannes Gutenberg University Mainz Germany
| | - Karin Wulff
- University Medicine Ernst Moritz Arndt University Greifswald Germany
| | - Heidi Rossmann
- Institute of Clinical Chemistry and Laboratory Medicine University Medical Center, Johannes Gutenberg University Mainz Germany
| | | | - Ingrid Brænne
- Center for Public Health Genomics University of Virginia Charlottesville Virginia
| | - Günther Witzke
- Department of Dermatology, University Medical Center Johannes Gutenberg University Mainz Germany
| | - Jochen Hardt
- Department of Medical Psychology and Medical Sociology Johannes Gutenberg University Mainz Germany
| |
Collapse
|
19
|
Folsom AR, Tang W, Basu S, Misialek JR, Couper D, Heckbert SR, Cushman M. Plasma Concentrations of High Molecular Weight Kininogen and Prekallikrein and Venous Thromboembolism Incidence in the General Population. Thromb Haemost 2019; 119:834-843. [PMID: 30780167 PMCID: PMC6499653 DOI: 10.1055/s-0039-1678737] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The kallikrein/kinin system, an intravascular biochemical pathway that includes several proteins involved in the contact activation system of coagulation, renin-angiotensin activation and inflammation, may or may not play a role in venous thromboembolism (VTE) occurrence. Within a large prospective population-based study in the United States, we conducted a nested case-cohort study to test the hypothesis that higher plasma levels of high molecular weight kininogen (HK) or prekallikrein are associated with greater VTE incidence. We related baseline enzyme-linked immunosorbent assay measures of HK and prekallikrein in 1993 to 1995 to incidence VTE of the lower extremity (n = 612) through 2015 (mean follow-up = 18 years). We found no evidence that plasma HK or prekallikrein was associated positively with incident VTE. HK, in fact, was associated inversely and significantly with VTE in most proportional hazards regression models. For example, the hazard ratio of VTE per standard deviation higher HK concentration was 0.88 (95% confidence interval = 0.81, 0.97), after adjustment for several VTE risk factors. Our findings suggest that plasma levels of these factors do not determine the risk of VTE in the general population.
Collapse
Affiliation(s)
- Aaron R. Folsom
- Division of Epidemiology & Community Health, School of Public Health, University of Minnesota, Minneapolis, Minnesota, United States
| | - Weihong Tang
- Division of Epidemiology & Community Health, School of Public Health, University of Minnesota, Minneapolis, Minnesota, United States
| | - Saonli Basu
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, Minnesota, United States
| | - Jeffrey R. Misialek
- Division of Epidemiology & Community Health, School of Public Health, University of Minnesota, Minneapolis, Minnesota, United States
| | - David Couper
- Department of Biostatistics, University of North Carolina, Chapel Hill, North Carolina, United States
| | - Susan R. Heckbert
- Department of Epidemiology, University of Washington, Seattle, Washington, United States
| | - Mary Cushman
- Department of Medicine, University of Vermont, Burlington, Vermont, United States
- Department of Pathology, University of Vermont, Burlington, Vermont, United States
| |
Collapse
|
20
|
Benincasa G, Costa D, Infante T, Lucchese R, Donatelli F, Napoli C. Interplay between genetics and epigenetics in modulating the risk of venous thromboembolism: A new challenge for personalized therapy. Thromb Res 2019; 177:145-153. [DOI: 10.1016/j.thromres.2019.03.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 02/12/2019] [Accepted: 03/13/2019] [Indexed: 02/09/2023]
|
21
|
Assessing the causal association of glycine with risk of cardio-metabolic diseases. Nat Commun 2019; 10:1060. [PMID: 30837465 PMCID: PMC6400990 DOI: 10.1038/s41467-019-08936-1] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Accepted: 02/11/2019] [Indexed: 02/02/2023] Open
Abstract
Circulating levels of glycine have previously been associated with lower incidence of coronary heart disease (CHD) and type 2 diabetes (T2D) but it remains uncertain if glycine plays an aetiological role. We present a meta-analysis of genome-wide association studies for glycine in 80,003 participants and investigate the causality and potential mechanisms of the association between glycine and cardio-metabolic diseases using genetic approaches. We identify 27 genetic loci, of which 22 have not previously been reported for glycine. We show that glycine is genetically associated with lower CHD risk and find that this may be partly driven by blood pressure. Evidence for a genetic association of glycine with T2D is weaker, but we find a strong inverse genetic effect of hyperinsulinaemia on glycine. Our findings strengthen evidence for a protective effect of glycine on CHD and show that the glycine-T2D association may be driven by a glycine-lowering effect of insulin resistance.
Collapse
|
22
|
Rohmann JL, de Haan HG, Algra A, Vossen CY, Rosendaal FR, Siegerink B. Genetic determinants of activity and antigen levels of contact system factors. J Thromb Haemost 2019; 17:157-168. [PMID: 30288888 DOI: 10.1111/jth.14307] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 09/17/2018] [Indexed: 11/27/2022]
Abstract
Essentials Genetic variation may provide valuable insight into the role of the contact system in thrombosis. Explored associations of genetic variants with activity, antigen, and disease in RATIO study. Two novel loci were identified: KLKB1 rs4253243 for prekallikrein; KNG1 rs5029980 for HMWK levels. Contact system variants and haplotypes were not associated with myocardial infarction or stroke. SUMMARY: Background The complex, interdependent contact activation system has been implicated in thrombotic disease, although few genetic determinants of levels of proteins from this system are known. Objectives Our primary aim was to study the influence of common F11, F12, KLKB1, and KNG1 variants on factor (F) XI activity and FXI, FXII, prekallikrein (PK) and high-molecular-weight kininogen (HMWK) antigen levels, as well as the risk of myocardial infarction and ischemic stroke. Patients/methods We analyzed samples from all 630 healthy participants, 182 ischemic stroke patients and 216 myocardial infarction patients in the RATIO case-control study of women aged < 50 years. Forty-three tagging single nucleotide variants (SNVs) were genotyped to represent common genetic variation in the contact system genes. Antigen and activity levels were measured with sandwich-ELISA-based and one-stage clotting assays. We performed single variant, age-adjusted, linear regression analyses per trait and disease phenotype, assuming additive inheritance and determined conditionally independent associations. Haplotypes based on the lead SNV and all conditionally independent SNVs were tested for association with traits and disease. Results We identified two novel associations of KLKB1 SNV rs4253243 with PK antigen (βconditional = -12.38; 95% CI, -20.07 to -4.69) and KNG1 SNV rs5029980 with HMWK antigen (βconditional = 5.86; 95% CI, 2.40-9.32) and replicated previously reported associations in a single study. Further analyses probed whether the observed associations were indicative of linkage, pleiotropic effects or mediation. No individual SNVs or haplotypes were associated with the disease outcomes. Conclusion This study adds to current knowledge of how genetic variation influences contact system protein levels and clarifies interdependencies.
Collapse
Affiliation(s)
- J L Rohmann
- Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Institute of Public Health, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - H G de Haan
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
| | - A Algra
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
- Department of Neurology and Neurosurgery, Brain Center Rudolph Magnus, University Medical Center Utrecht and Utrecht University, Utrecht, the Netherlands
- Julius Centre for Health Sciences and Primary Care, University Medical Center Utrecht and Utrecht University, Utrecht, the Netherlands
| | - C Y Vossen
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
| | - F R Rosendaal
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
| | - B Siegerink
- Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
23
|
Abstract
DNA methylation plays an important role in the regulation of transcription. Genetic control of DNA methylation is a potential candidate for explaining the many identified SNP associations with disease that are not found in coding regions. We replicated 52,916 cis and 2,025 trans DNA methylation quantitative trait loci (mQTL) using methylation from whole blood measured on Illumina HumanMethylation450 arrays in the Brisbane Systems Genetics Study (n = 614 from 177 families) and the Lothian Birth Cohorts of 1921 and 1936 (combined n = 1366). The trans mQTL SNPs were found to be over-represented in 1 Mbp subtelomeric regions, and on chromosomes 16 and 19. There was a significant increase in trans mQTL DNA methylation sites in upstream and 5′ UTR regions. The genetic heritability of a number of complex traits and diseases was partitioned into components due to mQTL and the remainder of the genome. Significant enrichment was observed for height (p = 2.1 × 10−10), ulcerative colitis (p = 2 × 10−5), Crohn’s disease (p = 6 × 10−8) and coronary artery disease (p = 5.5 × 10−6) when compared to a random sample of SNPs with matched minor allele frequency, although this enrichment is explained by the genomic location of the mQTL SNPs.
Collapse
|
24
|
Correlation of Interleukin-18 and High-Sensitivity C-Reactive Protein with Perioperative Deep Vein Thrombosis in Patients with Ankle Fracture. Ann Vasc Surg 2018; 54:282-289. [PMID: 30103053 DOI: 10.1016/j.avsg.2018.06.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 05/31/2018] [Accepted: 06/10/2018] [Indexed: 11/21/2022]
Abstract
BACKGROUND In the present study, we aimed to inspect the correlation of interleukin-18 (IL-18) and high-sensitivity C-reactive protein (hs-CRP), if any present, with the occurrence of perioperative deep vein thrombosis (DVT) in patients suffering from an ankle fracture (AF). METHODS Sixty-seven AF patients with complicated DVT (DVT group) and 448 AF patients without DVT (non-DVT group) were enrolled in the present study. To begin with, hemorheological indexes were detected. Turbidimetric immunoassay and enzyme-linked immunosorbent assay were used for the determination of the expressions of hs-CRP and IL-18, respectively. Coagulation method was used to detect prothrombin time (PT), thrombin time (TT), fibrinogen, and activated partial thromboplastin time (APTT). Logistic regression analysis was used to analyze the independent risk factors for DVT occurrence in AF patients after operation. RESULTS The results revealed that the DVT group presented an increased expression of hs-CRP and IL-18 1 day before operation and 1, 3, and 7 days after operation in comparison with the non-DVT group. The DVT group also had increased levels of PT, APTT, and prolonged TT and fibrinogen at 3 and 7 days after operation compared to the non-DVT group. After operation, it was observed that the patients in the DVT group had increased plasma viscosity, whole blood reduced viscosity, red blood cell (RBC) aggregation index, RBC deformation index, erythrocyte sedimentation rate, and declined erythrocrit. The independent risk factors for the occurrence of DVT were identified to be high-energy injury, TT, fibrinogen, hs-CRP, and IL-18 in AF patients after operation. CONCLUSIONS Based on the results obtained from the study, it was concluded that AF patients with DVT have higher levels of IL-18 and hs-CRP expressions, which is associated with the occurrence and development of DVT. These results may be particularly useful for diagnosis and treatment on DVT occurrence in AF patients.
Collapse
|
25
|
The Influence of Dyslexia Candidate Genes on Reading Skill in Old Age. Behav Genet 2018; 48:351-360. [PMID: 29959602 PMCID: PMC6097729 DOI: 10.1007/s10519-018-9913-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 06/23/2018] [Indexed: 11/27/2022]
Abstract
A number of candidate genes for reading and language impairment have been replicated, primarily in samples of children with developmental disability or delay, although these genes are also supported in adolescent population samples. The present study used a systematic approach to test 14 of these candidate genes for association with reading assessed in late adulthood (two cohorts with mean ages of 70 and 79 years). Gene-sets (14 candidates, axon-guidance and neuron migration pathways) and individual SNPs within each gene of interest were tested for association using imputed data referenced to the 1000 genomes European panel. Using the results from the genome-wide association (GWA) meta-analysis of the two cohorts (N = 1217), a competitive gene-set analysis showed that the candidate gene-set was associated with the reading index (p = .016) at a family wise error rate corrected significance level. Neither axon guidance nor neuron migration pathways were significant. Whereas individual SNP associations within CYP19A1, DYX1C1, CNTNAP2 and DIP2A genes (p < .05) did not reach corrected significance their allelic effects were in the same direction as past available reports. These results suggest that reading skill in normal adults shares the same genetic substrate as reading in adolescents, and clinically disordered reading, and highlights the utility of adult samples to increase sample sizes in the genetic study of developmental disorders.
Collapse
|
26
|
Allyn-Feuer A, Ade A, Luzum JA, Higgins GA, Athey BD. The pharmacoepigenomics informatics pipeline defines a pathway of novel and known warfarin pharmacogenomics variants. Pharmacogenomics 2018; 19:413-434. [PMID: 29400612 PMCID: PMC6021929 DOI: 10.2217/pgs-2017-0186] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 01/16/2018] [Indexed: 12/21/2022] Open
Abstract
AIM 'Pharmacoepigenomics' methods informed by omics datasets and pre-existing knowledge have yielded discoveries in neuropsychiatric pharmacogenomics. Now we evaluate the generality of these methods by discovering an extended warfarin pharmacogenomics pathway. MATERIALS & METHODS We developed the pharmacoepigenomics informatics pipeline, a scalable multi-omics variant screening pipeline for pharmacogenomics, and conducted an experiment in the genomics of warfarin. RESULTS We discovered known and novel pharmacogenomics variants and genes, both coding and regulatory, for warfarin response, including adverse events. Such genes and variants cluster in a warfarin response pathway consolidating known and novel warfarin response variants and genes. CONCLUSION These results can inform a new warfarin test. The pharmacoepigenomics informatics pipeline may be able to discover new pharmacogenomics markers in other drug-disease systems.
Collapse
Affiliation(s)
- Ari Allyn-Feuer
- Department of Computational Medicine & Bioinformatics, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Alex Ade
- Department of Computational Medicine & Bioinformatics, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Jasmine A Luzum
- Department of Clinical Pharmacy, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
| | - Gerald A Higgins
- Department of Computational Medicine & Bioinformatics, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Brian D Athey
- Department of Computational Medicine & Bioinformatics, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Internal Medicine, University of Michigan Health System, Ann Arbor, MI 48109, USA
- Department of Psychiatry, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Michigan Institute for Data Science, University of Michigan Office of Research, Ann Arbor, MI 48109, USA
| |
Collapse
|
27
|
Salloum-Asfar S, de la Morena-Barrio ME, Esteban J, Miñano A, Aroca C, Vicente V, Roldán V, Corral J. Assessment of two contact activation reagents for the diagnosis of congenital factor XI deficiency. Thromb Res 2018; 163:64-70. [DOI: 10.1016/j.thromres.2017.12.023] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 12/14/2017] [Accepted: 12/29/2017] [Indexed: 10/18/2022]
|
28
|
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: 4.8] [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
|
29
|
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: 9.1] [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
|
30
|
Cao Y, Tian M, Fang Q, Wen Z, Wang W, Ding H, Wang DW. Joint Effects of GWAS SNPs in Coagulation System Confer Risk to Hypertensive Intracerebral Hemorrhage. Neuromolecular Med 2017; 19:395-405. [PMID: 28718048 DOI: 10.1007/s12017-017-8453-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2016] [Accepted: 07/12/2017] [Indexed: 10/19/2022]
Abstract
Recent genome-wide association studies (GWAS) have identified numerous single nucleotide polymorphisms (SNPs) associated with coagulation system, including hemostatic factors and hematological phenotypes. However, few articles described the relationships between these SNPs and the risk of hemorrhagic stroke. The aim of our study was to evaluate the roles of these SNPs as risk factors and survival predictors for hemorrhagic stroke. Thirteen SNPs from GWAS in coagulation system were genotyped in a Chinese Han population including 1000 patients with hemorrhagic stroke (intracerebral hemorrhage, ICH = 743; subarachnoid hemorrhage, SAH = 257) and 1044 population-based controls. The associations between the genetics risk score (GRS) and risk of hemorrhagic stroke as well as post-stroke adverse outcomes were determined. No individual SNP was associated with the risk of hemorrhagic stroke. The GRS was calculated by summing the number of risk alleles of each SNP, and a total of 13 SNPs were included. Meanwhile, the GRS cutoffs values were defined to be close to quartiles or tertiles in control subjects. For quartiles, individuals with GRS about 8-9, 10-11, ≥12 had 1.28 (OR 1.28, 95% CI 0.98-1.68, p = 0.067)-, 1.36 (OR 1.36, 95% CI 1.04-1.79, p = 0.026)-, 1.53 (OR 1.53, 95% CI 1.13-2.07, p = 0.006)-fold increase in ICH risk compared to those with GRS ≤7, respectively; for tertiles, individuals with GRS about GRS 9-10, ≥11 had 0.98 (OR 0.98, 95% CI 0.78-1.23, p = 0.067)- and 1.26 (OR 1.26, 95% CI 1.00-1.59, p = 0.048)-fold increase in ICH risk compared to those with GRS ≤8, respectively. Further stratification analyses indicated that this association was only found in hypertensive ICH subjects. However, no statistical difference was found in the volume of hematoma, activities of daily living scale as well as hospital death in the ICH patients based on GRS values. Joint effects of SNPs associated with low coagulation factor levels might confer risk to ICH patients with hypertension. However, the clinical value on risk stratification and survival prediction was limited.
Collapse
Affiliation(s)
- Yanyan Cao
- Division of Cardiology, Departments of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Min Tian
- Division of Cardiology, Departments of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Qin Fang
- Division of Cardiology, Departments of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Zheng Wen
- Division of Cardiology, Departments of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Wei Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Hu Ding
- Division of Cardiology, Departments of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China. .,Institute of Hypertension, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Ave, Wuhan, 430030, People's Republic of China.
| | - Dao Wen Wang
- Division of Cardiology, Departments of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China. .,Genetic Diagnosis Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China. .,Institute of Hypertension, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefang Ave, Wuhan, 430030, People's Republic of China.
| |
Collapse
|
31
|
Whole-exome sequencing in evaluation of patients with venous thromboembolism. Blood Adv 2017; 1:1224-1237. [PMID: 29296762 DOI: 10.1182/bloodadvances.2017005249] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 05/26/2017] [Indexed: 01/05/2023] Open
Abstract
Genetics play a significant role in venous thromboembolism (VTE), yet current clinical laboratory-based testing identifies a known heritable thrombophilia (factor V Leiden, prothrombin gene mutation G20210A, or a deficiency of protein C, protein S, or antithrombin) in only a minority of VTE patients. We hypothesized that a substantial number of VTE patients could have lesser-known thrombophilia mutations. To test this hypothesis, we performed whole-exome sequencing (WES) in 64 patients with VTE, focusing our analysis on a novel 55-gene extended thrombophilia panel that we compiled. Our extended thrombophilia panel identified a probable disease-causing genetic variant or variant of unknown significance in 39 of 64 study patients (60.9%), compared with 6 of 237 control patients without VTE (2.5%) (P < .0001). Clinical laboratory-based thrombophilia testing identified a heritable thrombophilia in only 14 of 54 study patients (25.9%). The majority of WES variants were either associated with thrombosis based on prior reports in the literature or predicted to affect protein structure based on protein modeling performed as part of this study. Variants were found in major thrombophilia genes, various SERPIN genes, and highly conserved areas of other genes with established or potential roles in coagulation or fibrinolysis. Ten patients (15.6%) had >1 variant. Sanger sequencing performed in family members of 4 study patients with and without VTE showed generally concordant results with thrombotic history. WES and extended thrombophilia testing are promising tools for improving our understanding of VTE pathogenesis and identifying inherited thrombophilias.
Collapse
|
32
|
Martin-Fernandez L, Gavidia-Bovadilla G, Corrales I, Brunel H, Ramírez L, López S, Souto JC, Vidal F, Soria JM. Next generation sequencing to dissect the genetic architecture of KNG1 and F11 loci using factor XI levels as an intermediate phenotype of thrombosis. PLoS One 2017; 12:e0176301. [PMID: 28445521 PMCID: PMC5405990 DOI: 10.1371/journal.pone.0176301] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Accepted: 04/07/2017] [Indexed: 01/25/2023] Open
Abstract
Venous thromboembolism is a complex disease with a high heritability. There are significant associations among Factor XI (FXI) levels and SNPs in the KNG1 and F11 loci. Our aim was to identify the genetic variation of KNG1 and F11 that might account for the variability of FXI levels. The KNG1 and F11 loci were sequenced completely in 110 unrelated individuals from the GAIT-2 (Genetic Analysis of Idiopathic Thrombophilia 2) Project using Next Generation Sequencing on an Illumina MiSeq. The GAIT-2 Project is a study of 935 individuals in 35 extended Spanish families selected through a proband with idiopathic thrombophilia. Among the 110 individuals, a subset of 40 individuals was chosen as a discovery sample for identifying variants. A total of 762 genetic variants were detected. Several significant associations were established among common variants and low-frequency variants sets in KNG1 and F11 with FXI levels using the PLINK and SKAT packages. Among these associations, those of rs710446 and five low-frequency variant sets in KNG1 with FXI level variation were significant after multiple testing correction and permutation. Also, two putative pathogenic mutations related to high and low FXI levels were identified by data filtering and in silico predictions. This study of KNG1 and F11 loci should help to understand the connection between genotypic variation and variation in FXI levels. The functional genetic variants should be useful as markers of thromboembolic risk.
Collapse
Affiliation(s)
- Laura Martin-Fernandez
- Unit of Genomics of Complex Diseases, Biomedical Research Institute Sant Pau (IIB-Sant Pau), Barcelona, Spain
| | - Giovana Gavidia-Bovadilla
- Unit of Genomics of Complex Diseases, Biomedical Research Institute Sant Pau (IIB-Sant Pau), Barcelona, Spain
- Department of ESAII, Center for Biomedical Engineering Research (CREB), Universitat Politècnica de Catalunya, Barcelona, Spain
| | - Irene Corrales
- Congenital Coagulopathies, Blood and Tissue Bank, Barcelona, Spain
- Molecular Diagnosis and Therapy, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona (VHIR-UAB), Barcelona, Spain
| | - Helena Brunel
- Unit of Genomics of Complex Diseases, Biomedical Research Institute Sant Pau (IIB-Sant Pau), Barcelona, Spain
| | - Lorena Ramírez
- Congenital Coagulopathies, Blood and Tissue Bank, Barcelona, Spain
- Molecular Diagnosis and Therapy, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona (VHIR-UAB), Barcelona, Spain
| | - Sonia López
- Unit of Genomics of Complex Diseases, Biomedical Research Institute Sant Pau (IIB-Sant Pau), Barcelona, Spain
| | - Juan Carlos Souto
- Unit of Hemostasis and Thrombosis, Department of Hematology, IIB-Sant Pau, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Francisco Vidal
- Congenital Coagulopathies, Blood and Tissue Bank, Barcelona, Spain
- Molecular Diagnosis and Therapy, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona (VHIR-UAB), Barcelona, Spain
- CIBER de Enfermedades Cardiovasculares, Barcelona, Spain
| | - José Manuel Soria
- Unit of Genomics of Complex Diseases, Biomedical Research Institute Sant Pau (IIB-Sant Pau), Barcelona, Spain
- * E-mail:
| |
Collapse
|
33
|
Brunel H, Massanet R, Martinez-Perez A, Ziyatdinov A, Martin-Fernandez L, Souto JC, Perera A, Soria JM. The Central Role of KNG1 Gene as a Genetic Determinant of Coagulation Pathway-Related Traits: Exploring Metaphenotypes. PLoS One 2016; 11:e0167187. [PMID: 28005926 PMCID: PMC5178993 DOI: 10.1371/journal.pone.0167187] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 11/09/2016] [Indexed: 01/12/2023] Open
Abstract
Traditional genetic studies of single traits may be unable to detect the pleiotropic effects involved in complex diseases. To detect the correlation that exists between several phenotypes involved in the same biological process, we introduce an original methodology to analyze sets of correlated phenotypes involved in the coagulation cascade in genome-wide association studies. The methodology consists of a two-stage process. First, we define new phenotypic meta-variables (linear combinations of the original phenotypes), named metaphenotypes, by applying Independent Component Analysis for the multivariate analysis of correlated phenotypes (i.e. the levels of coagulation pathway–related proteins). The resulting metaphenotypes integrate the information regarding the underlying biological process (i.e. thrombus/clot formation). Secondly, we take advantage of a family based Genome Wide Association Study to identify genetic elements influencing these metaphenotypes and consequently thrombosis risk. Our study utilized data from the GAIT Project (Genetic Analysis of Idiopathic Thrombophilia). We obtained 15 metaphenotypes, which showed significant heritabilities, ranging from 0.2 to 0.7. These results indicate the importance of genetic factors in the variability of these traits. We found 4 metaphenotypes that showed significant associations with SNPs. The most relevant were those mapped in a region near the HRG, FETUB and KNG1 genes. Our results are provocative since they show that the KNG1 locus plays a central role as a genetic determinant of the entire coagulation pathway and thrombus/clot formation. Integrating data from multiple correlated measurements through metaphenotypes is a promising approach to elucidate the hidden genetic mechanisms underlying complex diseases.
Collapse
Affiliation(s)
- Helena Brunel
- Unit of Genomics of Complex Diseases, Sant Pau Institute of Biomedical Research (IIB-Sant Pau), Barcelona, Spain
| | - Raimon Massanet
- B2SLab, Departament d’Enginyeria de Sistemes, Automàtica i Informàtica Industrial, Universitat Politècnica de Catalunya (UPC), Barcelona, Spain
| | - Angel Martinez-Perez
- Unit of Genomics of Complex Diseases, Sant Pau Institute of Biomedical Research (IIB-Sant Pau), Barcelona, Spain
| | - Andrey Ziyatdinov
- Unit of Genomics of Complex Diseases, Sant Pau Institute of Biomedical Research (IIB-Sant Pau), Barcelona, Spain
| | - Laura Martin-Fernandez
- Unit of Genomics of Complex Diseases, Sant Pau Institute of Biomedical Research (IIB-Sant Pau), Barcelona, Spain
| | - Juan Carlos Souto
- Thrombosis and Haemostasis Unit, Sant Pau Institute of Biomedical Research (IIB-Sant Pau), Barcelona, Spain
| | - Alexandre Perera
- Thrombosis and Haemostasis Unit, Sant Pau Institute of Biomedical Research (IIB-Sant Pau), Barcelona, Spain
| | - José Manuel Soria
- Unit of Genomics of Complex Diseases, Sant Pau Institute of Biomedical Research (IIB-Sant Pau), Barcelona, Spain
- * E-mail:
| |
Collapse
|
34
|
Identification of ITGA2B and ITGB3 Single-Nucleotide Polymorphisms and Their Influences on the Platelet Function. BIOMED RESEARCH INTERNATIONAL 2016; 2016:5675084. [PMID: 27965976 PMCID: PMC5124636 DOI: 10.1155/2016/5675084] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 04/18/2016] [Indexed: 12/20/2022]
Abstract
The aim of the study was to investigate ITGA2B and ITGB3 genetic polymorphisms and to evaluate the variability in the platelet function in healthy Chinese subjects. The genetic sequence of the entire coding region of the ITGA2B and ITGB3 genes was investigated. Adenosine diphosphate-induced platelet aggregation, glycoprotein IIb/IIIa content, bleeding time, and coagulation indexes were detected. Thirteen variants in the ITGA2B locus and 29 variants in the ITGB3 locus were identified in the Chinese population. The rs1009312 and rs2015049 were associated with the mean platelet volume. The rs70940817 was significantly correlated with the prothrombin time. The rs70940817 and rs112188890 were related with the activated partial thromboplastin time, and ITGB3 rs4642 was correlated with the thrombin time and fibrinogen. The minor alleles of rs56197296 and rs5919 were associated with decreased ADP-induced platelet aggregation, and rs55827077 was related with decreased GPIIb/IIIa per platelet. The rs1009312, rs2015049, rs3760364, rs567581451, rs7208170, and rs117052258 were related with bleeding time. Further studies are needed to explore the clinical importance of ITGA2B and ITGB3 SNPs in the platelet function.
Collapse
|
35
|
Abstract
PURPOSE OF REVIEW New DNA genotyping and sequencing technologies have facilitated the rapid advancement in our knowledge of human genomic variation and a search for the heritable determinants of complex genetic traits. This review highlights findings from recent genetic studies of complex traits primarily related to venous thromboembolism and provides tools to understand and interpret genome-wide association studies and next-generation sequencing studies. RECENT FINDINGS Genome-wide studies of venous thromboembolic disease and the variation of the protein components of the hemostatic system have been reported. The results of these studies have suggested that variants in a diverse set of known and new genes contribute to the heritability of these traits, but that many of the genetic determinants of these traits still remain undiscovered. SUMMARY Next-generation sequencing studies and functional studies of the gene loci that contribute to hemostatic traits are currently underway. Future studies that explore the role of rare genetic variants, regulatory elements of the genome and gene-gene interactions will be required for a more complete understanding of the genetic control of the hemostatic system and for the application of this knowledge to the care of patients with disorders of thrombosis and hemostasis.
Collapse
Affiliation(s)
- Karl C Desch
- Department of Pediatrics and Communicable Disease, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
36
|
Moren X, Lhomme M, Bulla A, Sanchez JC, Kontush A, James RW. Proteomic and lipidomic analyses of paraoxonase defined high density lipoprotein particles: Association of paraoxonase with the anti-coagulant, protein S. Proteomics Clin Appl 2015; 10:230-8. [PMID: 26358807 DOI: 10.1002/prca.201500062] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Revised: 07/31/2015] [Accepted: 09/07/2015] [Indexed: 11/07/2022]
Abstract
PURPOSE Characterizing high density lipoprotein (HDL) particles and their relevance to HDL function is a major research objective. One aim is to identify functionally distinct particles. To try to limit both functional and compositional heterogeneity the present study focused on paraoxonase-1 (PON1) as a target for isolation of a minor HDL subfraction. EXPERIMENTAL DESIGN Immunoaffinity techniques were applied to isolate PON1-containing HDL (P-HDL) and total HDL (T-HDL), which were subsequently characterized and compared. RESULTS Analyses of the lipidomes showed significant differences between the fractions in the relative concentrations of individual lipid subspecies, notably reduced levels of unsaturated lysophosphatidylcholine (p < 0.05) in P-HDL (reflected in a significantly reduced total lysophosphatidylcholine polyunsaturated fatty acid content, p < 0.004). Significant differences were also observed for the proteomes. P-HDL was highly enriched in the anti-coagulant, vitamin K activated protein S (prot S) (p < 0.0001), and alpha2 macroglobulin (p < 0.01), compared to T-HDL. Conversely, procoagulant proteins kininogen 1 and histidine-rich glycoprotein were largely excluded from P-HDL. Immunoabsorption of PON1 from plasma significantly reduced prot S anti-coagulant activity. CONCLUSIONS AND CLINICAL RELEVANCE The P-HDL lipidome and proteome showed significant differences from T-HDL. Enrichment in anti-coagulation proteins indicates complementary functionalities within P-HDL particles and underlines their anti-atherosclerotic potential.
Collapse
Affiliation(s)
- Xenia Moren
- Clinical Diabetes Unit, Department of Medical Specialities, Medical Faculty, University of Geneva, Geneva, Switzerland
| | - Marie Lhomme
- INSERM, UMR-ICAN, 1166, University of Pierre and Marie Curie - Paris 6, Pitié - Salpétrière University Hospital, Paris, France
| | - Alexandre Bulla
- Department of Genetic and Laboratory Medicine, University Hospital, Geneva, Switzerland
| | - Jean-Charles Sanchez
- Translational Biomarker Group (TBG), Department of Human Protein Sciences, University Medical Centre, University of Geneva, Geneva, Switzerland
| | - Anatol Kontush
- INSERM, UMR-ICAN, 1166, University of Pierre and Marie Curie - Paris 6, Pitié - Salpétrière University Hospital, Paris, France
| | - Richard W James
- Clinical Diabetes Unit, Department of Medical Specialities, Medical Faculty, University of Geneva, Geneva, Switzerland
| |
Collapse
|
37
|
Genome-wide association studies identify genetic loci for low von Willebrand factor levels. Eur J Hum Genet 2015; 24:1035-40. [PMID: 26486471 DOI: 10.1038/ejhg.2015.222] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Revised: 08/23/2015] [Accepted: 09/11/2015] [Indexed: 01/14/2023] Open
Abstract
Low von Willebrand factor (VWF) levels are associated with bleeding symptoms and are a diagnostic criterion for von Willebrand disease, the most common inherited bleeding disorder. To date, it is unclear which genetic loci are associated with reduced VWF levels. Therefore, we conducted a meta-analysis of genome-wide association studies to identify genetic loci associated with low VWF levels. For this meta-analysis, we included 31 149 participants of European ancestry from 11 community-based studies. From all participants, VWF antigen (VWF:Ag) measurements and genome-wide single-nucleotide polymorphism (SNP) scans were available. Each study conducted analyses using logistic regression of SNPs on dichotomized VWF:Ag measures (lowest 5% for blood group O and non-O) with an additive genetic model adjusted for age and sex. An inverse-variance weighted meta-analysis was performed for VWF:Ag levels. A total of 97 SNPs exceeded the genome-wide significance threshold of 5 × 10(-8) and comprised five loci on four different chromosomes: 6q24 (smallest P-value 5.8 × 10(-10)), 9q34 (2.4 × 10(-64)), 12p13 (5.3 × 10(-22)), 12q23 (1.2 × 10(-8)) and 13q13 (2.6 × 10(-8)). All loci were within or close to genes, including STXBP5 (Syntaxin Binding Protein 5) (6q24), STAB5 (stabilin-5) (12q23), ABO (9q34), VWF (12p13) and UFM1 (ubiquitin-fold modifier 1) (13q13). Of these, UFM1 has not been previously associated with VWF:Ag levels. Four genes that were previously associated with VWF levels (VWF, ABO, STXBP5 and STAB2) were also associated with low VWF levels, and, in addition, we identified a new gene, UFM1, that is associated with low VWF levels. These findings point to novel mechanisms for the occurrence of low VWF levels.
Collapse
|
38
|
Olson NC, Butenas S, Lange LA, Lange EM, Cushman M, Jenny NS, Walston J, Souto JC, Soria JM, Chauhan G, Debette S, Longstreth WT, Seshadri S, Reiner AP, Tracy RP. Coagulation factor XII genetic variation, ex vivo thrombin generation, and stroke risk in the elderly: results from the Cardiovascular Health Study. J Thromb Haemost 2015; 13:1867-77. [PMID: 26286125 PMCID: PMC4946166 DOI: 10.1111/jth.13111] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 06/12/2015] [Accepted: 08/02/2015] [Indexed: 12/31/2022]
Abstract
BACKGROUND The relationships of thrombin generation (TG) with cardiovascular disease risk are underevaluated in population-based cohorts. OBJECTIVES To evaluate the relationships of TG influenced by the contact and tissue factor coagulation pathways ex vivo with common single-nucleotide polymorphisms (SNPs) and incident cardiovascular disease and stroke. PATIENTS/METHODS We measured peak TG (pTG) in baseline plasma samples of Cardiovascular Health Study participants (n = 5411), both with and without inhibitory anti-factor XIa antibody (pTG/FXIa(-) ). We evaluated their associations with ~ 50 000 SNPs by using the IBCv2 genotyping array, and with incident cardiovascular disease and stroke events over a median follow-up of 13.2 years. RESULTS The minor allele for an SNP in the FXII gene (F12), rs1801020, was associated with lower pTG in European-Americans (β = - 34.2 ± 3.5 nm; P = 3.3 × 10(-22) ; minor allele frequency [MAF] = 0.23) and African-Americans (β = - 31.1 ± 7.9 nm; P = 9.0 × 10(-5) ; MAF = 0.42). Lower FXIa-independent pTG (pTG/FXIa(-) ) was associated with the F12 rs1801020 minor allele, and higher pTG/FXIa(-) was associated with the ABO SNP rs657152 minor allele (β = 16.3 nm; P = 4.3 × 10(-9) ; MAF = 0.37). The risk factor-adjusted ischemic stroke hazard ratios were 1.09 (95% confidence interval CI 1.01-1.17; P = 0.03) for pTG, 1.06 (95% CI 0.98-1.15; P = 0.17) for pTG/FXIa(-) , and 1.11 (95% CI 1.02-1.21; P = 0.02) for FXIa-dependent pTG (pTG/FXIa(+) ), per one standard deviation increment (n = 834 ischemic strokes). In a multicohort candidate gene analysis, rs1801020 was not associated with incident ischemic stroke (β = - 0.02; standard error = 0.08; P = 0.81). CONCLUSIONS These results support the importance of contact activation pathway-dependent TG as a risk factor for ischemic stroke, and indicate the importance of F12 SNPs for TG ex vivo and in vivo.
Collapse
Affiliation(s)
- N C Olson
- Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, VT, USA
| | - S Butenas
- Department of Biochemistry, University of Vermont College of Medicine, Burlington, VT, USA
| | - L A Lange
- Department of Genetics, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - E M Lange
- Department of Genetics, University of North Carolina School of Medicine, Chapel Hill, NC, USA
- Department of Biostatistics, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - M Cushman
- Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, VT, USA
- Department of Medicine, University of Vermont College of Medicine, Burlington, VT, USA
| | - N S Jenny
- Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, VT, USA
| | - J Walston
- Division of Geriatric Medicine and Gerontology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - J C Souto
- Department of Hematology, Institute of Biomedical Research (IIB-Sant Pau), Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - J M Soria
- Unit of Genomics of Complex Diseases, Institute of Biomedical Research (IIB-Sant Pau), Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - G Chauhan
- INSERM U897, University of Bordeaux, Bordeaux, France
- University of Bordeaux, Bordeaux, France
| | - S Debette
- INSERM U897, University of Bordeaux, Bordeaux, France
- University of Bordeaux, Bordeaux, France
- Bordeaux University Hospital, Bordeaux, France
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
| | - W T Longstreth
- Department of Neurology, University of Washington, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - S Seshadri
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
| | - A P Reiner
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - R P Tracy
- Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, VT, USA
- Department of Biochemistry, University of Vermont College of Medicine, Burlington, VT, USA
| |
Collapse
|
39
|
Lakhal-Chaieb L, Oualkacha K, Richards BJ, Greenwood CM. A rare variant association test in family-based designs and non-normal quantitative traits. Stat Med 2015; 35:905-21. [DOI: 10.1002/sim.6750] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Revised: 09/04/2015] [Accepted: 09/05/2015] [Indexed: 12/13/2022]
Affiliation(s)
- Lajmi Lakhal-Chaieb
- Département de mathématiques et statistique; Université Laval; Québec G1V 0A6 Québec Canada
| | - Karim Oualkacha
- Département de mathématiques; Université de Québec À Montréal; Montreal Québec Canada
| | - Brent J. Richards
- Lady Davis Institute for Medical Research; Jewish General Hospital; Montreal Québec Canada
- Department of Epidemiology, Biostatistics and Occupational Health; McGill University; Montreal Québec Canada
- Department of Twin Research; King's College London; London U.K
| | - Celia M.T. Greenwood
- Lady Davis Institute for Medical Research; Jewish General Hospital; Montreal Québec Canada
- Department of Epidemiology, Biostatistics and Occupational Health; McGill University; Montreal Québec Canada
- Departments of Oncology and Human Genetics; McGill University; Montreal Québec Canada
| |
Collapse
|
40
|
Tang W, Cushman M, Green D, Rich SS, Lange LA, Yang Q, Tracy RP, Tofler GH, Basu S, Wilson JG, Keating BJ, Weng LC, Taylor HA, Jacobs DR, Delaney JA, Palmer CD, Young T, Pankow JS, O'Donnell CJ, Smith NL, Reiner AP, Folsom AR. Gene-centric approach identifies new and known loci for FVIII activity and VWF antigen levels in European Americans and African Americans. Am J Hematol 2015; 90:534-40. [PMID: 25779970 PMCID: PMC4747096 DOI: 10.1002/ajh.24005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Revised: 03/06/2015] [Accepted: 03/10/2015] [Indexed: 01/21/2023]
Abstract
Coagulation factor VIII and von Willebrand factor (VWF) are key proteins in procoagulant activation. Higher FVIII coagulant activity (FVIII :C) and VWF antigen (VWF :Ag) are risk factors for cardiovascular disease and venous thromboembolism. Beyond associations with ABO blood group, genetic determinants of FVIII and VWF are not well understood, especially in non European-American populations. We performed a genetic association study of FVIII :C and VWF:Ag that assessed 50,000 gene-centric single nucleotide polymorphisms (SNPs) in 18,556 European Americans (EAs) and 5,047 African Americans (AAs) from five population-based cohorts. Previously unreported associations for FVIII :C were identified in both AAs and EAs with KNG1 (most significantly associated SNP rs710446, Ile581Thr, Ile581Thr, P = 5.10 × 10(-7) in EAs and P = 3.88 × 10(-3) in AAs) and VWF rs7962217 (Gly2705Arg,P = 6.30 × 10(-9) in EAs and P = 2.98 × 10(-2) in AAs. Significant associations for FVIII :C were also observed with F8/TMLHE region SNP rs12557310 in EAs (P = 8.02 × 10(-10) ), with VWF rs1800380 in AAs (P = 5.62 × 10(-11) ), and with MAT1A rs2236568 in AAs (P51.69 × 10(-6) ). We replicated previously reported associations of FVIII :C and VWF :Ag with the ABO blood group, VWF rs1063856(Thr789Ala), rs216321 (Ala852Gln), and VWF rs2229446 (Arg2185Gln). Findings from this study expand our understanding of genetic influences for FVIII :C and VWF :Ag in both EAs and AAs.
Collapse
Affiliation(s)
- Weihong Tang
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, Minnesota
| | - Mary Cushman
- Department of Medicine, University of Vermont, Burlington, Vermont
- Department of Pathology, University of Vermont, Burlington, Vermont
| | - David Green
- Division of Hematology/Oncology, Department of Medicine, Feinberg School of Medicine of Northwestern University, Chicago, Illinois
| | - Stephen S. Rich
- Department of Public Health Sciences and the Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia
| | - Leslie A. Lange
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Qiong Yang
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts
| | - Russell P. Tracy
- Department of Pathology, University of Vermont College of Medicine, Burlington, Vermont
- Department of Biochemistry, University of Vermont College of Medicine, Burlington, Vermont
| | - Geoffrey H. Tofler
- Department of Cardiology, Royal North Shore Hospital, University of Sydney, Australia
| | - Saonli Basu
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, Minnesota
| | - James G. Wilson
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi
| | - Brendan J. Keating
- Division of Transplantation, Department of Surgery, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Lu-Chen Weng
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, Minnesota
| | - Herman A. Taylor
- Department of Medicine, Jackson Heart Study, University of Mississippi Medical Center, Jackson, Mississippi
| | - David R. Jacobs
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, Minnesota
| | - Joseph A. Delaney
- Department of Epidemiology, University of Washington, Seattle, Washington
| | - Cameron D. Palmer
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Taylor Young
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - James S. Pankow
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, Minnesota
| | - Christopher J. O'Donnell
- Department of Cardiovascular Epidemiology and Human Genomics, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Nicholas L. Smith
- Department of Epidemiology, University of Washington, Seattle, Washington
- Group Health Research Institute, Group Health Cooperative, Seattle, Washington
- Seattle Epidemiologic Research and Information Center, VA Office of Research and Development, Seattle, Washington
| | | | - Aaron R. Folsom
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
41
|
Pathak M, Wilmann P, Awford J, Li C, Hamad BK, Fischer PM, Dreveny I, Dekker LV, Emsley J. Coagulation factor XII protease domain crystal structure. J Thromb Haemost 2015; 13:580-91. [PMID: 25604127 PMCID: PMC4418343 DOI: 10.1111/jth.12849] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 01/12/2015] [Indexed: 12/11/2022]
Abstract
BACKGROUND Coagulation factor XII is a serine protease that is important for kinin generation and blood coagulation, cleaving the substrates plasma kallikrein and FXI. OBJECTIVE To investigate FXII zymogen activation and substrate recognition by determining the crystal structure of the FXII protease domain. METHODS AND RESULTS A series of recombinant FXII protease constructs were characterized by measurement of cleavage of chromogenic peptide and plasma kallikrein protein substrates. This revealed that the FXII protease construct spanning the light chain has unexpectedly weak proteolytic activity compared to β-FXIIa, which has an additional nine amino acid remnant of the heavy chain present. Consistent with these data, the crystal structure of the light chain protease reveals a zymogen conformation for active site residues Gly193 and Ser195, where the oxyanion hole is absent. The Asp194 side chain salt bridge to Arg73 constitutes an atypical conformation of the 70-loop. In one crystal form, the S1 pocket loops are partially flexible, which is typical of a zymogen. In a second crystal form of the deglycosylated light chain, the S1 pocket loops are ordered, and a short α-helix in the 180-loop of the structure results in an enlarged and distorted S1 pocket with a buried conformation of Asp189, which is critical for P1 Arg substrate recognition. The FXII structures define patches of negative charge surrounding the active site cleft that may be critical for interactions with inhibitors and substrates. CONCLUSIONS These data provide the first structural basis for understanding FXII substrate recognition and zymogen activation.
Collapse
Affiliation(s)
- M Pathak
- Centre for Biomolecular Sciences, School of Pharmacy, University of Nottingham, Nottingham, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Lopez LM, Hill WD, Harris SE, Valdes Hernandez M, Munoz Maniega S, Bastin ME, Bailey E, Smith C, McBride M, McClure J, Graham D, Dominiczak A, Yang Q, Fornage M, Ikram MA, Debette S, Launer L, Bis JC, Schmidt R, Seshadri S, Porteous DJ, Starr J, Deary IJ, Wardlaw JM. Genes from a translational analysis support a multifactorial nature of white matter hyperintensities. Stroke 2015; 46:341-7. [PMID: 25586835 PMCID: PMC4306534 DOI: 10.1161/strokeaha.114.007649] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Supplemental Digital Content is available in the text. Background and Purpose— White matter hyperintensities (WMH) of presumed vascular origin increase the risk of stroke and dementia. Despite strong WMH heritability, few gene associations have been identified. Relevant experimental models may be informative. Methods— We tested the associations between genes that were differentially expressed in brains of young spontaneously hypertensive stroke–prone rats and human WMH (using volume and visual score) in 621 subjects from the Lothian Birth Cohort 1936 (LBC1936). We then attempted replication in 9361 subjects from the Cohorts for Heart and Aging Research in Genomic Epidemiology (CHARGE). We also tested the subjects from LBC1936 for previous genome-wide WMH associations found in subjects from CHARGE. Results— Of 126 spontaneously hypertensive stroke–prone rat genes, 10 were nominally associated with WMH volume or score in subjects from LBC1936, of which 5 (AFP, ALB, GNAI1, RBM8a, and MRPL18) were associated with both WMH volume and score (P<0.05); 2 of the 10 (XPNPEP1, P=6.7×10−5; FARP1, P=0.024) plus another spontaneously hypertensive stroke–prone rat gene (USMG5, P=0.00014), on chromosomes 10, 13, and 10 respectively, were associated with WMH in subjects from CHARGE. Gene set enrichment showed significant associations for downregulated spontaneously hypertensive stroke–prone rat genes with WMH in humans. In subjects from LBC1936, we replicated CHARGE’s genome-wide WMH associations on chromosomes 17 (TRIM65 and TRIM47) and, for the first time, 1 (PMF1). Conclusions— Despite not passing multiple testing thresholds individually, these genes collectively are relevant to known WMH associations, proposed WMH mechanisms, or dementia: associations with Alzheimer's disease, late-life depression, ATP production, osmotic regulation, neurodevelopmental abnormalities, and cognitive impairment. If replicated further, they suggest a multifactorial nature for WMH and argue for more consideration of vascular contributions to dementia.
Collapse
Affiliation(s)
- Lorna M Lopez
- From the Centre for Cognitive Ageing and Cognitive Epidemiology (L.M.L., M.V.H., S.M.M., M.E.B., J.S., I.J.D., J.M.W.), Division of Neuroimaging Sciences, Brain Research Imaging Centre, (M.V.H., S.M.M., M.E.B., J.M.W.) and Academic Neuropathology (C.S.), Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom; Centre for Cognitive Ageing and Cognitive Epidemiology, Medical Genetics Section, University of Edinburgh Centre for Genomics and Experimental Medicine and MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom (W.D.H., S.E.H., D.J.P.); Department of Bioengineering, Imperial College London, London, United Kingdom (E.B.); BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (M.M., J.M., D.G., A.D.); Department of Biostatistics, Boston University School of Public Health, MA (Q.Y.); The Framingham Heart Study, Boston, MA (Q.Y., S.S.); The Human Genetics Center and Institute of Molecular Medicine, The University of Texas Health Science Center, Houston (M.F.); Departments of Epidemiology, Radiology and Neurology, Erasmus Medical Center, Rotterdam, The Netherlands (M.A.I.); Netherlands Consortium for Healthy Aging, Leiden, The Netherlands (M.A.I.); 12 INSERM U740 (Paris 7 University) and U708 (Bordeaux University), Bordeaux, France (S.D.); Department of Neurology, Lariboisière Hospital, 7 University, DHU Neurovasc Paris Sorbonne, Paris, France (S.D.); University of Versailles Saint-Quentin-en-Yvelines, Versailles, France (S.D.); Department of Neurology, Boston University School of Medicine, MA (S.D., S.S.); Laboratory of Epidemiology and Population Sciences, Intramural Research Program, National Institute on Aging, National Institutes of Health, Bethesda, MD (L.L.); Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle (J.C.B.); and Clinical Division of Neurogeri
| | - W David Hill
- From the Centre for Cognitive Ageing and Cognitive Epidemiology (L.M.L., M.V.H., S.M.M., M.E.B., J.S., I.J.D., J.M.W.), Division of Neuroimaging Sciences, Brain Research Imaging Centre, (M.V.H., S.M.M., M.E.B., J.M.W.) and Academic Neuropathology (C.S.), Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom; Centre for Cognitive Ageing and Cognitive Epidemiology, Medical Genetics Section, University of Edinburgh Centre for Genomics and Experimental Medicine and MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom (W.D.H., S.E.H., D.J.P.); Department of Bioengineering, Imperial College London, London, United Kingdom (E.B.); BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (M.M., J.M., D.G., A.D.); Department of Biostatistics, Boston University School of Public Health, MA (Q.Y.); The Framingham Heart Study, Boston, MA (Q.Y., S.S.); The Human Genetics Center and Institute of Molecular Medicine, The University of Texas Health Science Center, Houston (M.F.); Departments of Epidemiology, Radiology and Neurology, Erasmus Medical Center, Rotterdam, The Netherlands (M.A.I.); Netherlands Consortium for Healthy Aging, Leiden, The Netherlands (M.A.I.); 12 INSERM U740 (Paris 7 University) and U708 (Bordeaux University), Bordeaux, France (S.D.); Department of Neurology, Lariboisière Hospital, 7 University, DHU Neurovasc Paris Sorbonne, Paris, France (S.D.); University of Versailles Saint-Quentin-en-Yvelines, Versailles, France (S.D.); Department of Neurology, Boston University School of Medicine, MA (S.D., S.S.); Laboratory of Epidemiology and Population Sciences, Intramural Research Program, National Institute on Aging, National Institutes of Health, Bethesda, MD (L.L.); Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle (J.C.B.); and Clinical Division of Neurogeri
| | - Sarah E Harris
- From the Centre for Cognitive Ageing and Cognitive Epidemiology (L.M.L., M.V.H., S.M.M., M.E.B., J.S., I.J.D., J.M.W.), Division of Neuroimaging Sciences, Brain Research Imaging Centre, (M.V.H., S.M.M., M.E.B., J.M.W.) and Academic Neuropathology (C.S.), Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom; Centre for Cognitive Ageing and Cognitive Epidemiology, Medical Genetics Section, University of Edinburgh Centre for Genomics and Experimental Medicine and MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom (W.D.H., S.E.H., D.J.P.); Department of Bioengineering, Imperial College London, London, United Kingdom (E.B.); BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (M.M., J.M., D.G., A.D.); Department of Biostatistics, Boston University School of Public Health, MA (Q.Y.); The Framingham Heart Study, Boston, MA (Q.Y., S.S.); The Human Genetics Center and Institute of Molecular Medicine, The University of Texas Health Science Center, Houston (M.F.); Departments of Epidemiology, Radiology and Neurology, Erasmus Medical Center, Rotterdam, The Netherlands (M.A.I.); Netherlands Consortium for Healthy Aging, Leiden, The Netherlands (M.A.I.); 12 INSERM U740 (Paris 7 University) and U708 (Bordeaux University), Bordeaux, France (S.D.); Department of Neurology, Lariboisière Hospital, 7 University, DHU Neurovasc Paris Sorbonne, Paris, France (S.D.); University of Versailles Saint-Quentin-en-Yvelines, Versailles, France (S.D.); Department of Neurology, Boston University School of Medicine, MA (S.D., S.S.); Laboratory of Epidemiology and Population Sciences, Intramural Research Program, National Institute on Aging, National Institutes of Health, Bethesda, MD (L.L.); Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle (J.C.B.); and Clinical Division of Neurogeri
| | - Maria Valdes Hernandez
- From the Centre for Cognitive Ageing and Cognitive Epidemiology (L.M.L., M.V.H., S.M.M., M.E.B., J.S., I.J.D., J.M.W.), Division of Neuroimaging Sciences, Brain Research Imaging Centre, (M.V.H., S.M.M., M.E.B., J.M.W.) and Academic Neuropathology (C.S.), Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom; Centre for Cognitive Ageing and Cognitive Epidemiology, Medical Genetics Section, University of Edinburgh Centre for Genomics and Experimental Medicine and MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom (W.D.H., S.E.H., D.J.P.); Department of Bioengineering, Imperial College London, London, United Kingdom (E.B.); BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (M.M., J.M., D.G., A.D.); Department of Biostatistics, Boston University School of Public Health, MA (Q.Y.); The Framingham Heart Study, Boston, MA (Q.Y., S.S.); The Human Genetics Center and Institute of Molecular Medicine, The University of Texas Health Science Center, Houston (M.F.); Departments of Epidemiology, Radiology and Neurology, Erasmus Medical Center, Rotterdam, The Netherlands (M.A.I.); Netherlands Consortium for Healthy Aging, Leiden, The Netherlands (M.A.I.); 12 INSERM U740 (Paris 7 University) and U708 (Bordeaux University), Bordeaux, France (S.D.); Department of Neurology, Lariboisière Hospital, 7 University, DHU Neurovasc Paris Sorbonne, Paris, France (S.D.); University of Versailles Saint-Quentin-en-Yvelines, Versailles, France (S.D.); Department of Neurology, Boston University School of Medicine, MA (S.D., S.S.); Laboratory of Epidemiology and Population Sciences, Intramural Research Program, National Institute on Aging, National Institutes of Health, Bethesda, MD (L.L.); Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle (J.C.B.); and Clinical Division of Neurogeri
| | - Susana Munoz Maniega
- From the Centre for Cognitive Ageing and Cognitive Epidemiology (L.M.L., M.V.H., S.M.M., M.E.B., J.S., I.J.D., J.M.W.), Division of Neuroimaging Sciences, Brain Research Imaging Centre, (M.V.H., S.M.M., M.E.B., J.M.W.) and Academic Neuropathology (C.S.), Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom; Centre for Cognitive Ageing and Cognitive Epidemiology, Medical Genetics Section, University of Edinburgh Centre for Genomics and Experimental Medicine and MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom (W.D.H., S.E.H., D.J.P.); Department of Bioengineering, Imperial College London, London, United Kingdom (E.B.); BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (M.M., J.M., D.G., A.D.); Department of Biostatistics, Boston University School of Public Health, MA (Q.Y.); The Framingham Heart Study, Boston, MA (Q.Y., S.S.); The Human Genetics Center and Institute of Molecular Medicine, The University of Texas Health Science Center, Houston (M.F.); Departments of Epidemiology, Radiology and Neurology, Erasmus Medical Center, Rotterdam, The Netherlands (M.A.I.); Netherlands Consortium for Healthy Aging, Leiden, The Netherlands (M.A.I.); 12 INSERM U740 (Paris 7 University) and U708 (Bordeaux University), Bordeaux, France (S.D.); Department of Neurology, Lariboisière Hospital, 7 University, DHU Neurovasc Paris Sorbonne, Paris, France (S.D.); University of Versailles Saint-Quentin-en-Yvelines, Versailles, France (S.D.); Department of Neurology, Boston University School of Medicine, MA (S.D., S.S.); Laboratory of Epidemiology and Population Sciences, Intramural Research Program, National Institute on Aging, National Institutes of Health, Bethesda, MD (L.L.); Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle (J.C.B.); and Clinical Division of Neurogeri
| | - Mark E Bastin
- From the Centre for Cognitive Ageing and Cognitive Epidemiology (L.M.L., M.V.H., S.M.M., M.E.B., J.S., I.J.D., J.M.W.), Division of Neuroimaging Sciences, Brain Research Imaging Centre, (M.V.H., S.M.M., M.E.B., J.M.W.) and Academic Neuropathology (C.S.), Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom; Centre for Cognitive Ageing and Cognitive Epidemiology, Medical Genetics Section, University of Edinburgh Centre for Genomics and Experimental Medicine and MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom (W.D.H., S.E.H., D.J.P.); Department of Bioengineering, Imperial College London, London, United Kingdom (E.B.); BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (M.M., J.M., D.G., A.D.); Department of Biostatistics, Boston University School of Public Health, MA (Q.Y.); The Framingham Heart Study, Boston, MA (Q.Y., S.S.); The Human Genetics Center and Institute of Molecular Medicine, The University of Texas Health Science Center, Houston (M.F.); Departments of Epidemiology, Radiology and Neurology, Erasmus Medical Center, Rotterdam, The Netherlands (M.A.I.); Netherlands Consortium for Healthy Aging, Leiden, The Netherlands (M.A.I.); 12 INSERM U740 (Paris 7 University) and U708 (Bordeaux University), Bordeaux, France (S.D.); Department of Neurology, Lariboisière Hospital, 7 University, DHU Neurovasc Paris Sorbonne, Paris, France (S.D.); University of Versailles Saint-Quentin-en-Yvelines, Versailles, France (S.D.); Department of Neurology, Boston University School of Medicine, MA (S.D., S.S.); Laboratory of Epidemiology and Population Sciences, Intramural Research Program, National Institute on Aging, National Institutes of Health, Bethesda, MD (L.L.); Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle (J.C.B.); and Clinical Division of Neurogeri
| | - Emma Bailey
- From the Centre for Cognitive Ageing and Cognitive Epidemiology (L.M.L., M.V.H., S.M.M., M.E.B., J.S., I.J.D., J.M.W.), Division of Neuroimaging Sciences, Brain Research Imaging Centre, (M.V.H., S.M.M., M.E.B., J.M.W.) and Academic Neuropathology (C.S.), Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom; Centre for Cognitive Ageing and Cognitive Epidemiology, Medical Genetics Section, University of Edinburgh Centre for Genomics and Experimental Medicine and MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom (W.D.H., S.E.H., D.J.P.); Department of Bioengineering, Imperial College London, London, United Kingdom (E.B.); BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (M.M., J.M., D.G., A.D.); Department of Biostatistics, Boston University School of Public Health, MA (Q.Y.); The Framingham Heart Study, Boston, MA (Q.Y., S.S.); The Human Genetics Center and Institute of Molecular Medicine, The University of Texas Health Science Center, Houston (M.F.); Departments of Epidemiology, Radiology and Neurology, Erasmus Medical Center, Rotterdam, The Netherlands (M.A.I.); Netherlands Consortium for Healthy Aging, Leiden, The Netherlands (M.A.I.); 12 INSERM U740 (Paris 7 University) and U708 (Bordeaux University), Bordeaux, France (S.D.); Department of Neurology, Lariboisière Hospital, 7 University, DHU Neurovasc Paris Sorbonne, Paris, France (S.D.); University of Versailles Saint-Quentin-en-Yvelines, Versailles, France (S.D.); Department of Neurology, Boston University School of Medicine, MA (S.D., S.S.); Laboratory of Epidemiology and Population Sciences, Intramural Research Program, National Institute on Aging, National Institutes of Health, Bethesda, MD (L.L.); Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle (J.C.B.); and Clinical Division of Neurogeri
| | - Colin Smith
- From the Centre for Cognitive Ageing and Cognitive Epidemiology (L.M.L., M.V.H., S.M.M., M.E.B., J.S., I.J.D., J.M.W.), Division of Neuroimaging Sciences, Brain Research Imaging Centre, (M.V.H., S.M.M., M.E.B., J.M.W.) and Academic Neuropathology (C.S.), Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom; Centre for Cognitive Ageing and Cognitive Epidemiology, Medical Genetics Section, University of Edinburgh Centre for Genomics and Experimental Medicine and MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom (W.D.H., S.E.H., D.J.P.); Department of Bioengineering, Imperial College London, London, United Kingdom (E.B.); BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (M.M., J.M., D.G., A.D.); Department of Biostatistics, Boston University School of Public Health, MA (Q.Y.); The Framingham Heart Study, Boston, MA (Q.Y., S.S.); The Human Genetics Center and Institute of Molecular Medicine, The University of Texas Health Science Center, Houston (M.F.); Departments of Epidemiology, Radiology and Neurology, Erasmus Medical Center, Rotterdam, The Netherlands (M.A.I.); Netherlands Consortium for Healthy Aging, Leiden, The Netherlands (M.A.I.); 12 INSERM U740 (Paris 7 University) and U708 (Bordeaux University), Bordeaux, France (S.D.); Department of Neurology, Lariboisière Hospital, 7 University, DHU Neurovasc Paris Sorbonne, Paris, France (S.D.); University of Versailles Saint-Quentin-en-Yvelines, Versailles, France (S.D.); Department of Neurology, Boston University School of Medicine, MA (S.D., S.S.); Laboratory of Epidemiology and Population Sciences, Intramural Research Program, National Institute on Aging, National Institutes of Health, Bethesda, MD (L.L.); Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle (J.C.B.); and Clinical Division of Neurogeri
| | - Martin McBride
- From the Centre for Cognitive Ageing and Cognitive Epidemiology (L.M.L., M.V.H., S.M.M., M.E.B., J.S., I.J.D., J.M.W.), Division of Neuroimaging Sciences, Brain Research Imaging Centre, (M.V.H., S.M.M., M.E.B., J.M.W.) and Academic Neuropathology (C.S.), Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom; Centre for Cognitive Ageing and Cognitive Epidemiology, Medical Genetics Section, University of Edinburgh Centre for Genomics and Experimental Medicine and MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom (W.D.H., S.E.H., D.J.P.); Department of Bioengineering, Imperial College London, London, United Kingdom (E.B.); BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (M.M., J.M., D.G., A.D.); Department of Biostatistics, Boston University School of Public Health, MA (Q.Y.); The Framingham Heart Study, Boston, MA (Q.Y., S.S.); The Human Genetics Center and Institute of Molecular Medicine, The University of Texas Health Science Center, Houston (M.F.); Departments of Epidemiology, Radiology and Neurology, Erasmus Medical Center, Rotterdam, The Netherlands (M.A.I.); Netherlands Consortium for Healthy Aging, Leiden, The Netherlands (M.A.I.); 12 INSERM U740 (Paris 7 University) and U708 (Bordeaux University), Bordeaux, France (S.D.); Department of Neurology, Lariboisière Hospital, 7 University, DHU Neurovasc Paris Sorbonne, Paris, France (S.D.); University of Versailles Saint-Quentin-en-Yvelines, Versailles, France (S.D.); Department of Neurology, Boston University School of Medicine, MA (S.D., S.S.); Laboratory of Epidemiology and Population Sciences, Intramural Research Program, National Institute on Aging, National Institutes of Health, Bethesda, MD (L.L.); Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle (J.C.B.); and Clinical Division of Neurogeri
| | - John McClure
- From the Centre for Cognitive Ageing and Cognitive Epidemiology (L.M.L., M.V.H., S.M.M., M.E.B., J.S., I.J.D., J.M.W.), Division of Neuroimaging Sciences, Brain Research Imaging Centre, (M.V.H., S.M.M., M.E.B., J.M.W.) and Academic Neuropathology (C.S.), Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom; Centre for Cognitive Ageing and Cognitive Epidemiology, Medical Genetics Section, University of Edinburgh Centre for Genomics and Experimental Medicine and MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom (W.D.H., S.E.H., D.J.P.); Department of Bioengineering, Imperial College London, London, United Kingdom (E.B.); BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (M.M., J.M., D.G., A.D.); Department of Biostatistics, Boston University School of Public Health, MA (Q.Y.); The Framingham Heart Study, Boston, MA (Q.Y., S.S.); The Human Genetics Center and Institute of Molecular Medicine, The University of Texas Health Science Center, Houston (M.F.); Departments of Epidemiology, Radiology and Neurology, Erasmus Medical Center, Rotterdam, The Netherlands (M.A.I.); Netherlands Consortium for Healthy Aging, Leiden, The Netherlands (M.A.I.); 12 INSERM U740 (Paris 7 University) and U708 (Bordeaux University), Bordeaux, France (S.D.); Department of Neurology, Lariboisière Hospital, 7 University, DHU Neurovasc Paris Sorbonne, Paris, France (S.D.); University of Versailles Saint-Quentin-en-Yvelines, Versailles, France (S.D.); Department of Neurology, Boston University School of Medicine, MA (S.D., S.S.); Laboratory of Epidemiology and Population Sciences, Intramural Research Program, National Institute on Aging, National Institutes of Health, Bethesda, MD (L.L.); Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle (J.C.B.); and Clinical Division of Neurogeri
| | - Delyth Graham
- From the Centre for Cognitive Ageing and Cognitive Epidemiology (L.M.L., M.V.H., S.M.M., M.E.B., J.S., I.J.D., J.M.W.), Division of Neuroimaging Sciences, Brain Research Imaging Centre, (M.V.H., S.M.M., M.E.B., J.M.W.) and Academic Neuropathology (C.S.), Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom; Centre for Cognitive Ageing and Cognitive Epidemiology, Medical Genetics Section, University of Edinburgh Centre for Genomics and Experimental Medicine and MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom (W.D.H., S.E.H., D.J.P.); Department of Bioengineering, Imperial College London, London, United Kingdom (E.B.); BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (M.M., J.M., D.G., A.D.); Department of Biostatistics, Boston University School of Public Health, MA (Q.Y.); The Framingham Heart Study, Boston, MA (Q.Y., S.S.); The Human Genetics Center and Institute of Molecular Medicine, The University of Texas Health Science Center, Houston (M.F.); Departments of Epidemiology, Radiology and Neurology, Erasmus Medical Center, Rotterdam, The Netherlands (M.A.I.); Netherlands Consortium for Healthy Aging, Leiden, The Netherlands (M.A.I.); 12 INSERM U740 (Paris 7 University) and U708 (Bordeaux University), Bordeaux, France (S.D.); Department of Neurology, Lariboisière Hospital, 7 University, DHU Neurovasc Paris Sorbonne, Paris, France (S.D.); University of Versailles Saint-Quentin-en-Yvelines, Versailles, France (S.D.); Department of Neurology, Boston University School of Medicine, MA (S.D., S.S.); Laboratory of Epidemiology and Population Sciences, Intramural Research Program, National Institute on Aging, National Institutes of Health, Bethesda, MD (L.L.); Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle (J.C.B.); and Clinical Division of Neurogeri
| | - Anna Dominiczak
- From the Centre for Cognitive Ageing and Cognitive Epidemiology (L.M.L., M.V.H., S.M.M., M.E.B., J.S., I.J.D., J.M.W.), Division of Neuroimaging Sciences, Brain Research Imaging Centre, (M.V.H., S.M.M., M.E.B., J.M.W.) and Academic Neuropathology (C.S.), Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom; Centre for Cognitive Ageing and Cognitive Epidemiology, Medical Genetics Section, University of Edinburgh Centre for Genomics and Experimental Medicine and MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom (W.D.H., S.E.H., D.J.P.); Department of Bioengineering, Imperial College London, London, United Kingdom (E.B.); BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (M.M., J.M., D.G., A.D.); Department of Biostatistics, Boston University School of Public Health, MA (Q.Y.); The Framingham Heart Study, Boston, MA (Q.Y., S.S.); The Human Genetics Center and Institute of Molecular Medicine, The University of Texas Health Science Center, Houston (M.F.); Departments of Epidemiology, Radiology and Neurology, Erasmus Medical Center, Rotterdam, The Netherlands (M.A.I.); Netherlands Consortium for Healthy Aging, Leiden, The Netherlands (M.A.I.); 12 INSERM U740 (Paris 7 University) and U708 (Bordeaux University), Bordeaux, France (S.D.); Department of Neurology, Lariboisière Hospital, 7 University, DHU Neurovasc Paris Sorbonne, Paris, France (S.D.); University of Versailles Saint-Quentin-en-Yvelines, Versailles, France (S.D.); Department of Neurology, Boston University School of Medicine, MA (S.D., S.S.); Laboratory of Epidemiology and Population Sciences, Intramural Research Program, National Institute on Aging, National Institutes of Health, Bethesda, MD (L.L.); Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle (J.C.B.); and Clinical Division of Neurogeri
| | - Qiong Yang
- From the Centre for Cognitive Ageing and Cognitive Epidemiology (L.M.L., M.V.H., S.M.M., M.E.B., J.S., I.J.D., J.M.W.), Division of Neuroimaging Sciences, Brain Research Imaging Centre, (M.V.H., S.M.M., M.E.B., J.M.W.) and Academic Neuropathology (C.S.), Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom; Centre for Cognitive Ageing and Cognitive Epidemiology, Medical Genetics Section, University of Edinburgh Centre for Genomics and Experimental Medicine and MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom (W.D.H., S.E.H., D.J.P.); Department of Bioengineering, Imperial College London, London, United Kingdom (E.B.); BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (M.M., J.M., D.G., A.D.); Department of Biostatistics, Boston University School of Public Health, MA (Q.Y.); The Framingham Heart Study, Boston, MA (Q.Y., S.S.); The Human Genetics Center and Institute of Molecular Medicine, The University of Texas Health Science Center, Houston (M.F.); Departments of Epidemiology, Radiology and Neurology, Erasmus Medical Center, Rotterdam, The Netherlands (M.A.I.); Netherlands Consortium for Healthy Aging, Leiden, The Netherlands (M.A.I.); 12 INSERM U740 (Paris 7 University) and U708 (Bordeaux University), Bordeaux, France (S.D.); Department of Neurology, Lariboisière Hospital, 7 University, DHU Neurovasc Paris Sorbonne, Paris, France (S.D.); University of Versailles Saint-Quentin-en-Yvelines, Versailles, France (S.D.); Department of Neurology, Boston University School of Medicine, MA (S.D., S.S.); Laboratory of Epidemiology and Population Sciences, Intramural Research Program, National Institute on Aging, National Institutes of Health, Bethesda, MD (L.L.); Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle (J.C.B.); and Clinical Division of Neurogeri
| | - Myriam Fornage
- From the Centre for Cognitive Ageing and Cognitive Epidemiology (L.M.L., M.V.H., S.M.M., M.E.B., J.S., I.J.D., J.M.W.), Division of Neuroimaging Sciences, Brain Research Imaging Centre, (M.V.H., S.M.M., M.E.B., J.M.W.) and Academic Neuropathology (C.S.), Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom; Centre for Cognitive Ageing and Cognitive Epidemiology, Medical Genetics Section, University of Edinburgh Centre for Genomics and Experimental Medicine and MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom (W.D.H., S.E.H., D.J.P.); Department of Bioengineering, Imperial College London, London, United Kingdom (E.B.); BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (M.M., J.M., D.G., A.D.); Department of Biostatistics, Boston University School of Public Health, MA (Q.Y.); The Framingham Heart Study, Boston, MA (Q.Y., S.S.); The Human Genetics Center and Institute of Molecular Medicine, The University of Texas Health Science Center, Houston (M.F.); Departments of Epidemiology, Radiology and Neurology, Erasmus Medical Center, Rotterdam, The Netherlands (M.A.I.); Netherlands Consortium for Healthy Aging, Leiden, The Netherlands (M.A.I.); 12 INSERM U740 (Paris 7 University) and U708 (Bordeaux University), Bordeaux, France (S.D.); Department of Neurology, Lariboisière Hospital, 7 University, DHU Neurovasc Paris Sorbonne, Paris, France (S.D.); University of Versailles Saint-Quentin-en-Yvelines, Versailles, France (S.D.); Department of Neurology, Boston University School of Medicine, MA (S.D., S.S.); Laboratory of Epidemiology and Population Sciences, Intramural Research Program, National Institute on Aging, National Institutes of Health, Bethesda, MD (L.L.); Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle (J.C.B.); and Clinical Division of Neurogeri
| | - M Arfan Ikram
- From the Centre for Cognitive Ageing and Cognitive Epidemiology (L.M.L., M.V.H., S.M.M., M.E.B., J.S., I.J.D., J.M.W.), Division of Neuroimaging Sciences, Brain Research Imaging Centre, (M.V.H., S.M.M., M.E.B., J.M.W.) and Academic Neuropathology (C.S.), Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom; Centre for Cognitive Ageing and Cognitive Epidemiology, Medical Genetics Section, University of Edinburgh Centre for Genomics and Experimental Medicine and MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom (W.D.H., S.E.H., D.J.P.); Department of Bioengineering, Imperial College London, London, United Kingdom (E.B.); BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (M.M., J.M., D.G., A.D.); Department of Biostatistics, Boston University School of Public Health, MA (Q.Y.); The Framingham Heart Study, Boston, MA (Q.Y., S.S.); The Human Genetics Center and Institute of Molecular Medicine, The University of Texas Health Science Center, Houston (M.F.); Departments of Epidemiology, Radiology and Neurology, Erasmus Medical Center, Rotterdam, The Netherlands (M.A.I.); Netherlands Consortium for Healthy Aging, Leiden, The Netherlands (M.A.I.); 12 INSERM U740 (Paris 7 University) and U708 (Bordeaux University), Bordeaux, France (S.D.); Department of Neurology, Lariboisière Hospital, 7 University, DHU Neurovasc Paris Sorbonne, Paris, France (S.D.); University of Versailles Saint-Quentin-en-Yvelines, Versailles, France (S.D.); Department of Neurology, Boston University School of Medicine, MA (S.D., S.S.); Laboratory of Epidemiology and Population Sciences, Intramural Research Program, National Institute on Aging, National Institutes of Health, Bethesda, MD (L.L.); Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle (J.C.B.); and Clinical Division of Neurogeri
| | - Stephanie Debette
- From the Centre for Cognitive Ageing and Cognitive Epidemiology (L.M.L., M.V.H., S.M.M., M.E.B., J.S., I.J.D., J.M.W.), Division of Neuroimaging Sciences, Brain Research Imaging Centre, (M.V.H., S.M.M., M.E.B., J.M.W.) and Academic Neuropathology (C.S.), Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom; Centre for Cognitive Ageing and Cognitive Epidemiology, Medical Genetics Section, University of Edinburgh Centre for Genomics and Experimental Medicine and MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom (W.D.H., S.E.H., D.J.P.); Department of Bioengineering, Imperial College London, London, United Kingdom (E.B.); BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (M.M., J.M., D.G., A.D.); Department of Biostatistics, Boston University School of Public Health, MA (Q.Y.); The Framingham Heart Study, Boston, MA (Q.Y., S.S.); The Human Genetics Center and Institute of Molecular Medicine, The University of Texas Health Science Center, Houston (M.F.); Departments of Epidemiology, Radiology and Neurology, Erasmus Medical Center, Rotterdam, The Netherlands (M.A.I.); Netherlands Consortium for Healthy Aging, Leiden, The Netherlands (M.A.I.); 12 INSERM U740 (Paris 7 University) and U708 (Bordeaux University), Bordeaux, France (S.D.); Department of Neurology, Lariboisière Hospital, 7 University, DHU Neurovasc Paris Sorbonne, Paris, France (S.D.); University of Versailles Saint-Quentin-en-Yvelines, Versailles, France (S.D.); Department of Neurology, Boston University School of Medicine, MA (S.D., S.S.); Laboratory of Epidemiology and Population Sciences, Intramural Research Program, National Institute on Aging, National Institutes of Health, Bethesda, MD (L.L.); Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle (J.C.B.); and Clinical Division of Neurogeri
| | - Lenore Launer
- From the Centre for Cognitive Ageing and Cognitive Epidemiology (L.M.L., M.V.H., S.M.M., M.E.B., J.S., I.J.D., J.M.W.), Division of Neuroimaging Sciences, Brain Research Imaging Centre, (M.V.H., S.M.M., M.E.B., J.M.W.) and Academic Neuropathology (C.S.), Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom; Centre for Cognitive Ageing and Cognitive Epidemiology, Medical Genetics Section, University of Edinburgh Centre for Genomics and Experimental Medicine and MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom (W.D.H., S.E.H., D.J.P.); Department of Bioengineering, Imperial College London, London, United Kingdom (E.B.); BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (M.M., J.M., D.G., A.D.); Department of Biostatistics, Boston University School of Public Health, MA (Q.Y.); The Framingham Heart Study, Boston, MA (Q.Y., S.S.); The Human Genetics Center and Institute of Molecular Medicine, The University of Texas Health Science Center, Houston (M.F.); Departments of Epidemiology, Radiology and Neurology, Erasmus Medical Center, Rotterdam, The Netherlands (M.A.I.); Netherlands Consortium for Healthy Aging, Leiden, The Netherlands (M.A.I.); 12 INSERM U740 (Paris 7 University) and U708 (Bordeaux University), Bordeaux, France (S.D.); Department of Neurology, Lariboisière Hospital, 7 University, DHU Neurovasc Paris Sorbonne, Paris, France (S.D.); University of Versailles Saint-Quentin-en-Yvelines, Versailles, France (S.D.); Department of Neurology, Boston University School of Medicine, MA (S.D., S.S.); Laboratory of Epidemiology and Population Sciences, Intramural Research Program, National Institute on Aging, National Institutes of Health, Bethesda, MD (L.L.); Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle (J.C.B.); and Clinical Division of Neurogeri
| | - Joshua C Bis
- From the Centre for Cognitive Ageing and Cognitive Epidemiology (L.M.L., M.V.H., S.M.M., M.E.B., J.S., I.J.D., J.M.W.), Division of Neuroimaging Sciences, Brain Research Imaging Centre, (M.V.H., S.M.M., M.E.B., J.M.W.) and Academic Neuropathology (C.S.), Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom; Centre for Cognitive Ageing and Cognitive Epidemiology, Medical Genetics Section, University of Edinburgh Centre for Genomics and Experimental Medicine and MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom (W.D.H., S.E.H., D.J.P.); Department of Bioengineering, Imperial College London, London, United Kingdom (E.B.); BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (M.M., J.M., D.G., A.D.); Department of Biostatistics, Boston University School of Public Health, MA (Q.Y.); The Framingham Heart Study, Boston, MA (Q.Y., S.S.); The Human Genetics Center and Institute of Molecular Medicine, The University of Texas Health Science Center, Houston (M.F.); Departments of Epidemiology, Radiology and Neurology, Erasmus Medical Center, Rotterdam, The Netherlands (M.A.I.); Netherlands Consortium for Healthy Aging, Leiden, The Netherlands (M.A.I.); 12 INSERM U740 (Paris 7 University) and U708 (Bordeaux University), Bordeaux, France (S.D.); Department of Neurology, Lariboisière Hospital, 7 University, DHU Neurovasc Paris Sorbonne, Paris, France (S.D.); University of Versailles Saint-Quentin-en-Yvelines, Versailles, France (S.D.); Department of Neurology, Boston University School of Medicine, MA (S.D., S.S.); Laboratory of Epidemiology and Population Sciences, Intramural Research Program, National Institute on Aging, National Institutes of Health, Bethesda, MD (L.L.); Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle (J.C.B.); and Clinical Division of Neurogeri
| | - Reinhold Schmidt
- From the Centre for Cognitive Ageing and Cognitive Epidemiology (L.M.L., M.V.H., S.M.M., M.E.B., J.S., I.J.D., J.M.W.), Division of Neuroimaging Sciences, Brain Research Imaging Centre, (M.V.H., S.M.M., M.E.B., J.M.W.) and Academic Neuropathology (C.S.), Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom; Centre for Cognitive Ageing and Cognitive Epidemiology, Medical Genetics Section, University of Edinburgh Centre for Genomics and Experimental Medicine and MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom (W.D.H., S.E.H., D.J.P.); Department of Bioengineering, Imperial College London, London, United Kingdom (E.B.); BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (M.M., J.M., D.G., A.D.); Department of Biostatistics, Boston University School of Public Health, MA (Q.Y.); The Framingham Heart Study, Boston, MA (Q.Y., S.S.); The Human Genetics Center and Institute of Molecular Medicine, The University of Texas Health Science Center, Houston (M.F.); Departments of Epidemiology, Radiology and Neurology, Erasmus Medical Center, Rotterdam, The Netherlands (M.A.I.); Netherlands Consortium for Healthy Aging, Leiden, The Netherlands (M.A.I.); 12 INSERM U740 (Paris 7 University) and U708 (Bordeaux University), Bordeaux, France (S.D.); Department of Neurology, Lariboisière Hospital, 7 University, DHU Neurovasc Paris Sorbonne, Paris, France (S.D.); University of Versailles Saint-Quentin-en-Yvelines, Versailles, France (S.D.); Department of Neurology, Boston University School of Medicine, MA (S.D., S.S.); Laboratory of Epidemiology and Population Sciences, Intramural Research Program, National Institute on Aging, National Institutes of Health, Bethesda, MD (L.L.); Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle (J.C.B.); and Clinical Division of Neurogeri
| | - Sudha Seshadri
- From the Centre for Cognitive Ageing and Cognitive Epidemiology (L.M.L., M.V.H., S.M.M., M.E.B., J.S., I.J.D., J.M.W.), Division of Neuroimaging Sciences, Brain Research Imaging Centre, (M.V.H., S.M.M., M.E.B., J.M.W.) and Academic Neuropathology (C.S.), Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom; Centre for Cognitive Ageing and Cognitive Epidemiology, Medical Genetics Section, University of Edinburgh Centre for Genomics and Experimental Medicine and MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom (W.D.H., S.E.H., D.J.P.); Department of Bioengineering, Imperial College London, London, United Kingdom (E.B.); BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (M.M., J.M., D.G., A.D.); Department of Biostatistics, Boston University School of Public Health, MA (Q.Y.); The Framingham Heart Study, Boston, MA (Q.Y., S.S.); The Human Genetics Center and Institute of Molecular Medicine, The University of Texas Health Science Center, Houston (M.F.); Departments of Epidemiology, Radiology and Neurology, Erasmus Medical Center, Rotterdam, The Netherlands (M.A.I.); Netherlands Consortium for Healthy Aging, Leiden, The Netherlands (M.A.I.); 12 INSERM U740 (Paris 7 University) and U708 (Bordeaux University), Bordeaux, France (S.D.); Department of Neurology, Lariboisière Hospital, 7 University, DHU Neurovasc Paris Sorbonne, Paris, France (S.D.); University of Versailles Saint-Quentin-en-Yvelines, Versailles, France (S.D.); Department of Neurology, Boston University School of Medicine, MA (S.D., S.S.); Laboratory of Epidemiology and Population Sciences, Intramural Research Program, National Institute on Aging, National Institutes of Health, Bethesda, MD (L.L.); Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle (J.C.B.); and Clinical Division of Neurogeri
| | - David J Porteous
- From the Centre for Cognitive Ageing and Cognitive Epidemiology (L.M.L., M.V.H., S.M.M., M.E.B., J.S., I.J.D., J.M.W.), Division of Neuroimaging Sciences, Brain Research Imaging Centre, (M.V.H., S.M.M., M.E.B., J.M.W.) and Academic Neuropathology (C.S.), Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom; Centre for Cognitive Ageing and Cognitive Epidemiology, Medical Genetics Section, University of Edinburgh Centre for Genomics and Experimental Medicine and MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom (W.D.H., S.E.H., D.J.P.); Department of Bioengineering, Imperial College London, London, United Kingdom (E.B.); BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (M.M., J.M., D.G., A.D.); Department of Biostatistics, Boston University School of Public Health, MA (Q.Y.); The Framingham Heart Study, Boston, MA (Q.Y., S.S.); The Human Genetics Center and Institute of Molecular Medicine, The University of Texas Health Science Center, Houston (M.F.); Departments of Epidemiology, Radiology and Neurology, Erasmus Medical Center, Rotterdam, The Netherlands (M.A.I.); Netherlands Consortium for Healthy Aging, Leiden, The Netherlands (M.A.I.); 12 INSERM U740 (Paris 7 University) and U708 (Bordeaux University), Bordeaux, France (S.D.); Department of Neurology, Lariboisière Hospital, 7 University, DHU Neurovasc Paris Sorbonne, Paris, France (S.D.); University of Versailles Saint-Quentin-en-Yvelines, Versailles, France (S.D.); Department of Neurology, Boston University School of Medicine, MA (S.D., S.S.); Laboratory of Epidemiology and Population Sciences, Intramural Research Program, National Institute on Aging, National Institutes of Health, Bethesda, MD (L.L.); Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle (J.C.B.); and Clinical Division of Neurogeri
| | - John Starr
- From the Centre for Cognitive Ageing and Cognitive Epidemiology (L.M.L., M.V.H., S.M.M., M.E.B., J.S., I.J.D., J.M.W.), Division of Neuroimaging Sciences, Brain Research Imaging Centre, (M.V.H., S.M.M., M.E.B., J.M.W.) and Academic Neuropathology (C.S.), Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom; Centre for Cognitive Ageing and Cognitive Epidemiology, Medical Genetics Section, University of Edinburgh Centre for Genomics and Experimental Medicine and MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom (W.D.H., S.E.H., D.J.P.); Department of Bioengineering, Imperial College London, London, United Kingdom (E.B.); BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (M.M., J.M., D.G., A.D.); Department of Biostatistics, Boston University School of Public Health, MA (Q.Y.); The Framingham Heart Study, Boston, MA (Q.Y., S.S.); The Human Genetics Center and Institute of Molecular Medicine, The University of Texas Health Science Center, Houston (M.F.); Departments of Epidemiology, Radiology and Neurology, Erasmus Medical Center, Rotterdam, The Netherlands (M.A.I.); Netherlands Consortium for Healthy Aging, Leiden, The Netherlands (M.A.I.); 12 INSERM U740 (Paris 7 University) and U708 (Bordeaux University), Bordeaux, France (S.D.); Department of Neurology, Lariboisière Hospital, 7 University, DHU Neurovasc Paris Sorbonne, Paris, France (S.D.); University of Versailles Saint-Quentin-en-Yvelines, Versailles, France (S.D.); Department of Neurology, Boston University School of Medicine, MA (S.D., S.S.); Laboratory of Epidemiology and Population Sciences, Intramural Research Program, National Institute on Aging, National Institutes of Health, Bethesda, MD (L.L.); Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle (J.C.B.); and Clinical Division of Neurogeri
| | - Ian J Deary
- From the Centre for Cognitive Ageing and Cognitive Epidemiology (L.M.L., M.V.H., S.M.M., M.E.B., J.S., I.J.D., J.M.W.), Division of Neuroimaging Sciences, Brain Research Imaging Centre, (M.V.H., S.M.M., M.E.B., J.M.W.) and Academic Neuropathology (C.S.), Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom; Centre for Cognitive Ageing and Cognitive Epidemiology, Medical Genetics Section, University of Edinburgh Centre for Genomics and Experimental Medicine and MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom (W.D.H., S.E.H., D.J.P.); Department of Bioengineering, Imperial College London, London, United Kingdom (E.B.); BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (M.M., J.M., D.G., A.D.); Department of Biostatistics, Boston University School of Public Health, MA (Q.Y.); The Framingham Heart Study, Boston, MA (Q.Y., S.S.); The Human Genetics Center and Institute of Molecular Medicine, The University of Texas Health Science Center, Houston (M.F.); Departments of Epidemiology, Radiology and Neurology, Erasmus Medical Center, Rotterdam, The Netherlands (M.A.I.); Netherlands Consortium for Healthy Aging, Leiden, The Netherlands (M.A.I.); 12 INSERM U740 (Paris 7 University) and U708 (Bordeaux University), Bordeaux, France (S.D.); Department of Neurology, Lariboisière Hospital, 7 University, DHU Neurovasc Paris Sorbonne, Paris, France (S.D.); University of Versailles Saint-Quentin-en-Yvelines, Versailles, France (S.D.); Department of Neurology, Boston University School of Medicine, MA (S.D., S.S.); Laboratory of Epidemiology and Population Sciences, Intramural Research Program, National Institute on Aging, National Institutes of Health, Bethesda, MD (L.L.); Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle (J.C.B.); and Clinical Division of Neurogeri
| | - Joanna M Wardlaw
- From the Centre for Cognitive Ageing and Cognitive Epidemiology (L.M.L., M.V.H., S.M.M., M.E.B., J.S., I.J.D., J.M.W.), Division of Neuroimaging Sciences, Brain Research Imaging Centre, (M.V.H., S.M.M., M.E.B., J.M.W.) and Academic Neuropathology (C.S.), Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom; Centre for Cognitive Ageing and Cognitive Epidemiology, Medical Genetics Section, University of Edinburgh Centre for Genomics and Experimental Medicine and MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom (W.D.H., S.E.H., D.J.P.); Department of Bioengineering, Imperial College London, London, United Kingdom (E.B.); BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (M.M., J.M., D.G., A.D.); Department of Biostatistics, Boston University School of Public Health, MA (Q.Y.); The Framingham Heart Study, Boston, MA (Q.Y., S.S.); The Human Genetics Center and Institute of Molecular Medicine, The University of Texas Health Science Center, Houston (M.F.); Departments of Epidemiology, Radiology and Neurology, Erasmus Medical Center, Rotterdam, The Netherlands (M.A.I.); Netherlands Consortium for Healthy Aging, Leiden, The Netherlands (M.A.I.); 12 INSERM U740 (Paris 7 University) and U708 (Bordeaux University), Bordeaux, France (S.D.); Department of Neurology, Lariboisière Hospital, 7 University, DHU Neurovasc Paris Sorbonne, Paris, France (S.D.); University of Versailles Saint-Quentin-en-Yvelines, Versailles, France (S.D.); Department of Neurology, Boston University School of Medicine, MA (S.D., S.S.); Laboratory of Epidemiology and Population Sciences, Intramural Research Program, National Institute on Aging, National Institutes of Health, Bethesda, MD (L.L.); Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle (J.C.B.); and Clinical Division of Neurogeri
| |
Collapse
|
43
|
Weng LC, Cushman M, Pankow JS, Basu S, Boerwinkle E, Folsom AR, Tang W. A genetic association study of activated partial thromboplastin time in European Americans and African Americans: the ARIC Study. Hum Mol Genet 2014; 24:2401-8. [PMID: 25552651 DOI: 10.1093/hmg/ddu732] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Reduced activated partial thromboplastin time (aPTT) is a risk marker for incident and recurrent venous thromboembolism (VTE). Genetic factors influencing aPTT are not well understood, especially in populations of non-European ancestry. The present study aimed to identify aPTT-related gene variants in both European Americans (EAs) and African Americans (AAs). We conducted a genetic association study for aPTT in 9719 EAs and 2799 AAs from the Atherosclerosis Risk in Communities (ARIC) study. Using the Candidate Gene Association Resource (CARe) consortium candidate gene array, the analyses were based on ∼50 000 SNPs in ∼2000 candidate genes. In EAs, the analyses identified a new independent association for aPTT in F5 (rs2239852, P-value = 1.9 × 10(-8)), which clusters with a coding variant rs6030 (P-value = 7.8 × 10(-7)). The remaining significant signals were located on F5, HRG, KNG1, F11, F12 and ABO and have been previously reported in EA populations. In AAs, significant signals were identified in KNG1, HRG, F12, ABO and VWF, with the leading variants in KNG1, HRG and F12 being the same as in the EAs; the significant variant in VWF (rs2229446, P-value = 1.2 × 10(-6)) was specific to the AA sample (minor allele frequency = 19% in AAs and 0.2% in EAs) and has not been previously reported. This is the first study to report aPTT-related genetic variants in AAs. Our findings in AAs demonstrate transferability of previously reported associations with KNG1, HRG and F12 in EAs. We also identified new associations at F5 in EAs and VWF in AAs that have not been previously reported for aPTT.
Collapse
Affiliation(s)
- Lu-Chen Weng
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN 55454, USA
| | - Mary Cushman
- Departments of Medicine and Pathology, University of Vermont, Burlington, VT 05405, USA
| | - James S Pankow
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN 55454, USA
| | - Saonli Basu
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, MN 55455, USA and
| | - Eric Boerwinkle
- Human Genetics Center and Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Aaron R Folsom
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN 55454, USA
| | - Weihong Tang
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN 55454, USA,
| |
Collapse
|
44
|
Freyburger G, Labrouche S, Hubert C, Bauduer F. Haemostaseome-associated SNPs: has the thrombotic phenotype a greater influence than ethnicity? GMT study from Aquitaine including Basque individuals. Thromb Haemost 2014; 113:66-76. [PMID: 25374097 DOI: 10.1160/th14-02-0189] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 08/15/2014] [Indexed: 11/05/2022]
Abstract
The Genetic Markers for Thrombosis (GMT) study compared the relative influence of ethnicity and thrombotic phenotype regarding the distribution of SNPs implicated in haemostasis pathophysiology ("haemostaseome"). We assessed 384 SNPs in three groups, each of 480 subjects: 1) general population of Aquitaine region (Southwestern France) used as control; 2) patients with venous thromboembolism from the same area; and 3) autochthonous Basques, a genetic isolate, who demonstrate unusual characteristics regarding the coagulation system. This study sought to evaluate i) the value of looking for a large number of genes in order to identify new genetic markers of thrombosis, ii) the value of investigating low risk factors and potential preferential associations, iii) the impact of ethnicity on the characterisation of markers for thrombosis. We did not detect any previously unrecognised SNP significantly associated with thrombosis risk or any preferential associations of low-risk factors in patients with thrombosis. The sum of ϰ² values for our 110 significant SNPs demonstrated a smaller genetic distance between patients and controls (321 cumulated ϰ² value) than between Basques and controls (1,570 cumulated ϰ² value). Hence, our study confirms the genetic particularity of Basques especially regarding a significantly lower expression of the non-O blood group (p< 0.0004). This is mitigated by a higher prevalence of factor II Leiden (p< 0.02) while factor V Leiden prevalence does not differ. Numerous other differences covering a wide range of proteins of the haemostaseome may result in an overall different genetic risk for venous thromboembolism.
Collapse
Affiliation(s)
- Geneviève Freyburger
- Geneviève Freyburger, Laboratory for Hematology, CHU Pellegrin, 33076 Bordeaux cedex, France, Tel.: +335 57 820 206, Fax: +335 56 79 60 20, E-mail:
| | | | | | | |
Collapse
|
45
|
Alfred T, Ben-Shlomo Y, Cooper R, Hardy R, Cooper C, Deary IJ, Elliott J, Gunnell D, Harris SE, Kivimaki M, Kumari M, Martin RM, Power C, Sayer AA, Starr JM, Kuh D, Day INM. Associations between APOE and low-density lipoprotein cholesterol genotypes and cognitive and physical capability: the HALCyon programme. AGE (DORDRECHT, NETHERLANDS) 2014; 36:9673. [PMID: 25073452 PMCID: PMC4150901 DOI: 10.1007/s11357-014-9673-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2013] [Accepted: 06/18/2014] [Indexed: 06/03/2023]
Abstract
The APOE ε2/3/4 genotype has been associated with low-density lipoprotein cholesterol (LDL-C) and Alzheimer disease. However, evidence for associations with measures of cognitive performance in adults without dementia has been mixed, as it is for physical performance. Associations may also be evident in other genotypes implicated in LDL-C levels. As part of the Healthy Ageing across the Life Course (HALCyon) collaborative research programme, genotypic information was obtained for APOE ε2/3/4, rs515135 (APOB), rs2228671 (LDLR) and rs629301 (SORT1) from eight cohorts of adults aged between 44 and 90 + years. We investigated associations with four measures of cognitive (word recall, phonemic fluency, semantic fluency and search speed) and physical capability (grip strength, get up and go/walk speed, timed chair rises and ability to balance) using meta-analyses. Overall, little evidence for associations between any of the genotypes and measures of cognitive capability was observed (e.g. pooled beta for APOE ε4 effect on semantic fluency z score = -0.02; 95 % CI = -0.05 to 0.02; p value = 0.3; n = 18,796). However, there was borderline evidence within studies that negative effects of APOE ε4 on nonverbal ability measures become more apparent with age. Few genotypic associations were observed with physical capability measures. The findings from our large investigation of middle-aged to older adults in the general population suggest that effects of APOE on cognitive capability are at most modest and are domain- and age-specific, while APOE has little influence on physical capability. In addition, other LDL-C-related genotypes have little impact on these traits.
Collapse
Affiliation(s)
- Tamuno Alfred
- School of Social and Community Medicine, University of Bristol, Canynge Hall, 39 Whatley Road, Bristol, BS8 2PS, UK,
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Ritchie SJ, Bates TC, Corley J, McNeill G, Davies G, Liewald DC, Starr JM, Deary IJ. Alcohol consumption and lifetime change in cognitive ability: a gene × environment interaction study. AGE (DORDRECHT, NETHERLANDS) 2014; 36:9638. [PMID: 24652602 PMCID: PMC4082597 DOI: 10.1007/s11357-014-9638-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Accepted: 02/26/2014] [Indexed: 06/03/2023]
Abstract
Studies of the effect of alcohol consumption on cognitive ability are often confounded. One approach to avoid confounding is the Mendelian randomization design. Here, we used such a design to test the hypothesis that a genetic score for alcohol processing capacity moderates the association between alcohol consumption and lifetime change in cognitive ability. Members of the Lothian Birth Cohort 1936 completed the same test of intelligence at age 11 and 70 years. They were assessed for recent alcohol consumption in later life and genotyped for a set of four single-nucleotide polymorphisms in three alcohol dehydrogenase genes. These variants were unrelated to late-life cognition or to socioeconomic status. We found a significant gene × alcohol consumption interaction on lifetime cognitive change (p = 0.007). Individuals with higher genetic ability to process alcohol showed relative improvements in cognitive ability with more consumption, whereas those with low processing capacity showed a negative relationship between cognitive change and alcohol consumption with more consumption. The effect of alcohol consumption on cognitive change may thus depend on genetic differences in the ability to metabolize alcohol.
Collapse
Affiliation(s)
- Stuart J Ritchie
- Department of Psychology, The University of Edinburgh, 7 George Square, Edinburgh, EH8 9JZ, UK,
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Balfoussia E, Skenderi K, Tsironi M, Anagnostopoulos AK, Parthimos N, Vougas K, Papassotiriou I, Tsangaris GT, Chrousos GP. A proteomic study of plasma protein changes under extreme physical stress. J Proteomics 2013; 98:1-14. [PMID: 24345577 DOI: 10.1016/j.jprot.2013.12.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Revised: 12/03/2013] [Accepted: 12/09/2013] [Indexed: 01/18/2023]
Abstract
UNLABELLED The Spartathlon race (brisk walking a distance of 246km in less than 36h) was employed as a model of severe physical stress to investigate proteomic alterations in the plasma of athletes at the start (Athens) and finish (Sparta) of the race, as well as 48h after the race (Post). The athletes' plasma was analyzed by 2D gel electrophoresis (2-DE) and the differentially expressed proteins were identified by matrix-assisted laser desorption ionization-time of flight (MALDI-TOF) mass spectrometry (MS). The ProteoSeek™ Albumin/IgG removal kit and the ProteoMiner™ enrichment kit were utilized to detect medium- and low-abundance proteins, whose expression may be masked due to high-abundance proteins. Our results were confirmed by Western blot and biochemical analyses. Overall fifty-two proteins were differentially expressed between the starting point, the finishing line and two days after the end of the race. Of these, thirty proteins were involved in inflammation, while the rest concerned anti-oxidation, anti-coagulation and iron and vitamin D transport. These results indicate that prolonged physical stress affects circulating stress-related proteins, which might be employed as biomarkers of stress-related diseases. BIOLOGICAL SIGNIFICANCE The current study employed the Spartathlon, as a model of prolonged endurance exercise, to identify and isolate putative biomarkers of inflammation under extreme physical stress conditions. These protein quantitative variations may pave the way to exploration and understanding of stress-related physiological processes, the stress response itself and diseases whose onset appears to be linked to stress.
Collapse
Affiliation(s)
- Edily Balfoussia
- Proteomics Research Unit, Center of Basic Research II, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Katerina Skenderi
- Laboratory of Nutrition and Clinical Dietetics, "Harokopio" University, Athens, Greece
| | - Maria Tsironi
- Department of Internal Medicine, University of Peloponnesus, School of Nursing, Sparta, Greece
| | - Athanasios K Anagnostopoulos
- Proteomics Research Unit, Center of Basic Research II, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Nikolaos Parthimos
- First Department of Pediatrics, University of Athens Medical School, Athens, "Aghia Sophia" Children's Hospital, Athens, Greece
| | - Kostantinos Vougas
- Proteomics Research Unit, Center of Basic Research II, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Ioannis Papassotiriou
- Department of Clinical Biochemistry, "Aghia Sophia" Children's Hospital, Athens, Greece
| | - George Th Tsangaris
- Proteomics Research Unit, Center of Basic Research II, Biomedical Research Foundation of the Academy of Athens, Athens, Greece.
| | - George P Chrousos
- First Department of Pediatrics, University of Athens Medical School, Athens, "Aghia Sophia" Children's Hospital, Athens, Greece
| |
Collapse
|
48
|
Alfred T, Ben-Shlomo Y, Cooper R, Hardy R, Deary IJ, Elliott J, Harris SE, Kivimaki M, Kumari M, Power C, Starr JM, Kuh D, Day INM. Associations between a polymorphism in the pleiotropic GCKR and Age-related phenotypes: the HALCyon programme. PLoS One 2013; 8:e70045. [PMID: 23894584 PMCID: PMC3720952 DOI: 10.1371/journal.pone.0070045] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2012] [Accepted: 06/17/2013] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND The glucokinase regulatory protein encoded by GCKR plays an important role in glucose metabolism and a single nucleotide polymorphism (SNP) rs1260326 (P446L) in the gene has been associated with several age-related biomarkers, including triglycerides, glucose, insulin and apolipoproteins. However, associations between SNPs in the gene and other ageing phenotypes such as cognitive and physical capability have not been reported. METHODS As part of the Healthy Ageing across the Life Course (HALCyon) collaborative research programme, men and women from five UK cohorts aged between 44 and 90+ years were genotyped for rs1260326. Meta-analysis was used to pool within-study genotypic associations between the SNP and several age-related phenotypes, including body mass index (BMI), blood lipid levels, lung function, and cognitive and physical capability. RESULTS We confirm the associations between the minor allele of the SNP and higher triglycerides and lower glucose levels. We also observed a triglyceride-independent association between the minor allele and lower BMI (pooled beta on z-score= -0.04, p-value=0.0001, n=16,251). Furthermore, there was some evidence for gene-environment interactions, including physical activity attenuating the effects on triglycerides. However, no associations were observed with measures of cognitive and physical capability. CONCLUSION Findings from middle-aged to older adults confirm associations between rs1260326 GCKR and triglycerides and glucose, suggest possible gene-environment interactions, but do not provide evidence that its relevance extends to cognitive and physical capability.
Collapse
Affiliation(s)
- Tamuno Alfred
- School of Social and Community Medicine, University of Bristol, Bristol, United Kingdom.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Morange PE, Trégouët DA. Current knowledge on the genetics of incident venous thrombosis. J Thromb Haemost 2013; 11 Suppl 1:111-21. [PMID: 23809115 DOI: 10.1111/jth.12233] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The genetic burden underlying venous thrombosis (VT) is characterized by a sibling relative risk of 2.5 and a strong heritability whose estimates varied from 35% to 60% according to different studies. However, the genetic factors identified so far only explain about 5% of VT heritability and just 16 genes have been robustly associated with the susceptibility to VT, most of them affecting the coagulation cascade. Eight of these have been identified during the last 5 years, thanks to the development of high-throughput micro-array genotyping technologies, which have radically changed the research landscape in human genetics. The present work is aimed at providing a historical review of the known genetic factors contributing to VT risk, as well as discussing future research strategies to follow to disentangle the whole spectrum of genetic variants associated with VT.
Collapse
Affiliation(s)
- P-E Morange
- INSERM, UMR_S1062, Nutrition Obesity and Risk of Thrombosis, Aix-Marseille University, Marseille, France.
| | | |
Collapse
|
50
|
Alfred T, Ben-Shlomo Y, Cooper R, Hardy R, Deary IJ, Elliott J, Harris SE, Hyppönen E, Kivimaki M, Kumari M, Maddock J, Power C, Starr JM, Kuh D, Day IN. Genetic variants influencing biomarkers of nutrition are not associated with cognitive capability in middle-aged and older adults. J Nutr 2013; 143:606-12. [PMID: 23468552 PMCID: PMC3738233 DOI: 10.3945/jn.112.171520] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Several investigations have observed positive associations between good nutritional status, as indicated by micronutrients, and cognitive measures; however, these associations may not be causal. Genetic polymorphisms that affect nutritional biomarkers may be useful for providing evidence for associations between micronutrients and cognitive measures. As part of the Healthy Ageing across the Life Course (HALCyon) program, men and women aged between 44 and 90 y from 6 UK cohorts were genotyped for polymorphisms associated with circulating concentrations of iron [rs4820268 transmembrane protease, serine 6 (TMPRSS6) and rs1800562 hemochromatosis (HFE)], vitamin B-12 [(rs492602 fucosyltransferase 2 (FUT2)], vitamin D ([rs2282679 group-specific component (GC)] and β-carotene ([rs6564851 beta-carotene 15,15'-monooxygenase 1 (BCMO1)]. Meta-analysis was used to pool within-study effects of the associations between these polymorphisms and the following measures of cognitive capability: word recall, phonemic fluency, semantic fluency, and search speed. Among the several statistical tests conducted, we found little evidence for associations. We found the minor allele of rs1800562 was associated with poorer word recall scores [pooled β on Z-score for carriers vs. noncarriers: -0.05 (95% CI: -0.09, -0.004); P = 0.03, n = 14,105] and poorer word recall scores for the vitamin D-raising allele of rs2282679 [pooled β per T allele: -0.03 (95% CI: -0.05, -0.003); P = 0.03, n = 16,527]. However, there was no evidence for other associations. Our findings provide little evidence to support associations between these genotypes and cognitive capability in older adults. Further investigations are required to elucidate whether the previous positive associations from observational studies between circulating measures of these micronutrients and cognitive performance are due to confounding and reverse causality.
Collapse
Affiliation(s)
- Tamuno Alfred
- School of Social and Community Medicine, University of Bristol, Bristol, UK.
| | - Yoav Ben-Shlomo
- School of Social and Community Medicine, University of Bristol, Bristol, UK
| | - Rachel Cooper
- MRC Unit for Lifelong Health and Ageing and Division of Population Health, University College London, London, UK
| | - Rebecca Hardy
- MRC Unit for Lifelong Health and Ageing and Division of Population Health, University College London, London, UK
| | - Ian J. Deary
- Centre for Cognitive Ageing and Cognitive Epidemiology, and,Department of Psychology, University of Edinburgh, Edinburgh, UK
| | - Jane Elliott
- Centre for Longitudinal Studies, Department of Quantitative Social Sciences, Institute of Education, London, UK
| | - Sarah E. Harris
- Centre for Cognitive Ageing and Cognitive Epidemiology, and,Medical Genetics Section, University of Edinburgh, Edinburgh, UK
| | - Elina Hyppönen
- MRC Centre of Epidemiology for Child Health/Centre for Paediatric Epidemiology and Biostatistics, UCL Institute of Child Health, London, UK
| | - Mika Kivimaki
- Department of Epidemiology and Public Health, University College London, London, UK
| | - Meena Kumari
- Department of Epidemiology and Public Health, University College London, London, UK
| | - Jane Maddock
- MRC Centre of Epidemiology for Child Health/Centre for Paediatric Epidemiology and Biostatistics, UCL Institute of Child Health, London, UK
| | - Chris Power
- MRC Centre of Epidemiology for Child Health/Centre for Paediatric Epidemiology and Biostatistics, UCL Institute of Child Health, London, UK
| | - John M. Starr
- Centre for Cognitive Ageing and Cognitive Epidemiology, and,Alzheimer Scotland Dementia Research Centre, University of Edinburgh, Edinburgh, UK; and
| | - Diana Kuh
- MRC Unit for Lifelong Health and Ageing and Division of Population Health, University College London, London, UK
| | - Ian N.M. Day
- School of Social and Community Medicine, University of Bristol, Bristol, UK,MRC Centre for Causal Analyses in Translational Epidemiology, School of Social and Community Medicine, University of Bristol, Bristol, UK
| | | |
Collapse
|