1
|
Galyfos G, Chamzin A, Charalampopoulos G, Liasis N, Sigala F, Filis K. Direct oral anticoagulants for deep vein thrombosis among patients with hereditary thrombophilia-A cohort study. Phlebology 2023; 38:599-604. [PMID: 37647589 DOI: 10.1177/02683555231199004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
OBJECTIVES to evaluate direct oral anticoagulants (DOACs) in patients with hereditary thrombophilia and deep venous thrombosis (DVT). METHODS This is a retrospective observational study. RESULTS In total, 45 patients were treated between 01/2012 and 12/2022 (mean follow-up: 1.5 +/- 0.3 years). The most frequent thrombophilias were heterozygous V Leiden (20%), heterozygous MTHFR C677T (37.8%), heterozygous MTHFR A1298C (24.4%), and hyperhomocysteinemia (26.7%). The patients received rivaroxaban (n = 19), apixaban (n = 15), and dabigatran (n = 11). Three cases presented symptoms' recurrence without evidence of thrombosis' recurrence (two under rivaroxaban and one under apixaban; p > .05). These patients improved under parenteral anticoagulation and were further treated with dabigatran. No other event or major bleeding occurred during the follow-up. The presence of more than two factors was associated with acute recurrence of symptoms (OR = 25.9; 95% CI [1.454-461.262]; p = .026). CONCLUSIONS DOACs seem to be safe and efficient for patients with hereditary thrombophilia and DVT. The presence of more than two thrombophilia factors is associated with a higher risk for symptom recurrence. Although statistically non-significant, symptoms' recurrence was also observed more frequently among patients under anti-Xa inhibitors than antithrombin inhibitors. This should be verified in larger comparative studies.
Collapse
Affiliation(s)
- George Galyfos
- First Department of Propedeutic Surgery, Vascular Surgery Unit, National and Kapodistrian University of Athens, Hippocration Hospital, Athens, Greece
| | - Alexandros Chamzin
- First Department of Propedeutic Surgery, Vascular Surgery Unit, National and Kapodistrian University of Athens, Hippocration Hospital, Athens, Greece
| | - Georgios Charalampopoulos
- First Department of Propedeutic Surgery, Vascular Surgery Unit, National and Kapodistrian University of Athens, Hippocration Hospital, Athens, Greece
| | - Nikolaos Liasis
- First Department of Propedeutic Surgery, Vascular Surgery Unit, National and Kapodistrian University of Athens, Hippocration Hospital, Athens, Greece
| | - Frangiska Sigala
- First Department of Propedeutic Surgery, Vascular Surgery Unit, National and Kapodistrian University of Athens, Hippocration Hospital, Athens, Greece
| | - Konstantinos Filis
- First Department of Propedeutic Surgery, Vascular Surgery Unit, National and Kapodistrian University of Athens, Hippocration Hospital, Athens, Greece
| |
Collapse
|
2
|
Tang LV, Tao Y, Feng Y, Ma J, Lin W, Zhang Y, Zhang Y, Wu T, Cai Y, Lu H, Wei J, Corral J, Hu Y. Gene editing of human iPSCs rescues thrombophilia in hereditary antithrombin deficiency in mice. Sci Transl Med 2022; 14:eabq3202. [PMID: 36449603 DOI: 10.1126/scitranslmed.abq3202] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Hereditary antithrombin deficiency is caused by SERPINC1 gene mutations and predisposes to recurrent venous thromboembolism that can be life-threatening. Therefore, lifelong anticoagulation is required, which has side effects and may not be effective. In this study, peripheral blood mononuclear cells from a patient with severe antithrombin deficiency were reprogrammed into induced pluripotent stem cells (iPSCs). The mutation was corrected using CRISPR-Cas9 and Cre/LoxP genome editing. iPSCs were differentiated into hepatocytes, which were injected into the spleen of antithrombin knockout mice to restore the activity of antithrombin and reduce the thrombophilic state. Human iPSC-differentiated hepatocytes colonized mice and secreted antithrombin stably, normalizing antithrombin in plasma (activity: from 46.8 ± 5.7% to 88.6 ± 7.6%, P < 0.0001; antigen: from 146.9 ± 19.5 nanograms per milliliter to 390.7 ± 16.1 nanograms per milliliter, P < 0.0001). In venous thrombosis model, the rate of thrombosis in mice treated with edited hepatocytes, parental hepatocytes, and wild-type mice were 60, 90, and 70%, respectively. The thrombus weight was much lighter in mice treated with edited hepatocytes compared with parental hepatocytes (7.25 ± 2.00 milligrams versus 15.32 ± 2.87 milligrams, P = 0.0025) and showed no notable difference compared with that in wild-type mice (10.41 ± 2.91 milligrams). The activity and concentration of antithrombin remained high for 3 weeks after injection. The liver and kidney function markers showed no obvious abnormality during the observation period. This study provides a proof of principle for correction of mutations in patient-derived iPSCs and potential therapeutic applications for hereditary thrombophilia.
Collapse
Affiliation(s)
- Liang V Tang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yanyi Tao
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yuanzheng Feng
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jiewen Ma
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Wenyi Lin
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yuyang Zhang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yi Zhang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Tingting Wu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yaohua Cai
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Hui Lu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jun Wei
- iRegene Therapeutics Co. Ltd., Wuhan 430070, PR China
| | - Javier Corral
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia, IMIB-Arrixaca, CIBERER, Ronda de Garay S/N, 30003 Murcia, Spain
| | - Yu Hu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
3
|
Bonnesen K, Schmidt M, Horváth-Puhó E, Sørensen HT. The Interaction Effect between Comorbidity Burden and Venous Thromboembolism on Mortality: A Nationwide Cohort Study. Thromb Haemost 2022; 122:578-589. [PMID: 34116582 DOI: 10.1055/a-1527-6215] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
BACKGROUND Comorbidity influences venous thromboembolism (VTE) mortality, but it is unknown whether this is due to comorbidity alone or whether biological interaction exists. OBJECTIVES We examined whether comorbidity and VTE interact to increase VTE mortality beyond their individual effects. METHODS This nationwide population-based cohort study included all VTE patients ≥18 years during 2000 to 2016, and an age-, sex-, and comorbidity-matched comparison cohort of individuals without VTE. We computed age-standardized mortality rates and examined interaction on the additive scale using interaction contrasts (difference in rate differences). RESULTS After 30-day follow-up, the mortality rate per 1,000 person-years among individuals with no comorbidity was 419 (95% confidence interval [CI]: 391-447) in the VTE and 16 (95% CI: 13-18) in the comparison cohort (rate difference: 403). The corresponding mortality rate increased to 591 (95% CI: 539-643) in the VTE cohort and 38 (95% CI: 33-44) in the comparison cohort among individuals with low comorbidity (rate difference: 553). The interaction contrast (150) showed that 25% (150/591) of mortality was explained by the interaction in individuals with low comorbidity. This percentage increased to 56% for moderate and 63% for severe comorbidity. Interaction effects were largest within 30-day follow-up, for provoked VTE, in young individuals, and in individuals noncompliant to anticoagulant therapy. Dose-response patterns for interaction effects were also observed after 31-365-day and >1-5-year follow-up (p < 0.0001). Interaction effects varied between individual comorbidities. CONCLUSION Biological interaction between comorbidity and VTE explained a substantial proportion of VTE mortality. The interaction effect increased with comorbidity burden.
Collapse
Affiliation(s)
- Kasper Bonnesen
- Department of Clinical Epidemiology, Aarhus University Hospital, Aarhus, Denmark
| | - Morten Schmidt
- Department of Clinical Epidemiology, Aarhus University Hospital, Aarhus, Denmark.,Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
| | | | - Henrik T Sørensen
- Department of Clinical Epidemiology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
4
|
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
|
5
|
Samelson-Jones BJ, Acord MR, Raffini L. Comment on: Bivalirudin during thrombolysis with catheter-directed tPA in a heparin-refractory patient: A case report. Pediatr Blood Cancer 2020; 67:e28518. [PMID: 32618428 DOI: 10.1002/pbc.28518] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 06/03/2020] [Indexed: 11/11/2022]
Affiliation(s)
- Benjamin J Samelson-Jones
- Department of Pediatrics, Division of Hematology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania.,Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Michael R Acord
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania.,Department of Radiology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Leslie Raffini
- Department of Pediatrics, Division of Hematology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania.,Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
6
|
Kjaergaard AD, Larsen OH, Hvas AM, Nissen PH. SERPINC1 variants causing hereditary antithrombin deficiency in a Danish population. Thromb Res 2019; 175:68-75. [PMID: 30721820 DOI: 10.1016/j.thromres.2019.01.022] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 01/08/2019] [Accepted: 01/30/2019] [Indexed: 01/05/2023]
Abstract
INTRODUCTION Antithrombin deficiency is associated with increased risk of venous thromboembolism (VTE). We aimed to identify variants causing antithrombin deficiency in a Danish population. MATERIALS AND METHODS We performed Sanger sequencing and, in relevant cases, multiplex ligation-dependent probe amplification analyses, in 46 individuals (23 index cases) with and 9 relatives without antithrombin deficiency. Furthermore, in order to explore whether a combination of antithrombin type II heparin binding site (HBS) deficiency and factor V Leiden single nucleotide variant (SNV) conferred a higher risk of VTE than either risk factor alone, we performed genotyping for factor V Leiden in most of the carriers of type II HBS deficiency (n = 25). RESULTS We detected causal variants in all 46 carriers: three large and two small deletions, all causing type I antithrombin deficiency, and seven SNVs: one causing type I, one causing type II reactive site (RS), four causing type II HBS and one causing pleiotropic effect (PE) type II antithrombin deficiency. None of the relatives without antithrombin deficiency had the family variant. All detected SNVs have been reported previously. Majority (n = 27) of carriers had type II HBS deficiency, most often caused by the p.(Pro73Leu) SNV (n = 19). Heterozygosity for factor V Leiden was observed in three (3/25 = 12%) carriers of type II HBS deficiency. Only four (4/25 = 16%) carriers of type II HBS antithrombin deficiency experienced VTE, and two of these were heterozygous for factor V Leiden. CONCLUSIONS In a systematic search to identify variants causing hereditary antithrombin deficiency in a Danish population, we achieved a variant detection rate of 100%.
Collapse
Affiliation(s)
- Alisa D Kjaergaard
- Department of Clinical Epidemiology, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark.
| | - Ole Halfdan Larsen
- Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark; Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark.
| | - Anne-Mette Hvas
- Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark.
| | - Peter H Nissen
- Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark.
| |
Collapse
|
7
|
Kim J, Kraft P, Hagan KA, Harrington LB, Lindstroem S, Kabrhel C. Interaction of a genetic risk score with physical activity, physical inactivity, and body mass index in relation to venous thromboembolism risk. Genet Epidemiol 2018. [PMID: 29520861 DOI: 10.1002/gepi.22118] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Venous thromboembolism (VTE) is highly heritable. Physical activity, physical inactivity and body mass index (BMI) are also risk factors, but evidence of interaction between genetic and environmental risk factors is limited. METHODS Data on 2,134 VTE cases and 3,890 matched controls were obtained from the Nurses' Health Study (NHS), Nurses' Health Study II (NHS II), and Health Professionals Follow-up Study (HPFS). We calculated a weighted genetic risk score (wGRS) using 16 single nucleotide polymorphisms associated with VTE risk in published genome-wide association studies (GWAS). Data on three risk factors, physical activity (metabolic equivalent [MET] hours per week), physical inactivity (sitting hours per week) and BMI, were obtained from biennial questionnaires. VTE cases were incident since cohort inception; controls were matched to cases on age, cohort, and genotype array. Using conditional logistic regression, we assessed joint effects and interaction effects on both additive and multiplicative scales. We also ran models using continuous wGRS stratified by risk-factor categories. RESULTS We observed a supra-additive interaction between wGRS and BMI. Having both high wGRS and high BMI was associated with a 3.4-fold greater risk of VTE (relative excess risk due to interaction = 0.69, p = 0.046). However, we did not find evidence for a multiplicative interaction with BMI. No interactions were observed for physical activity or inactivity. CONCLUSION We found a synergetic effect between a genetic risk score and high BMI on the risk of VTE. Intervention efforts lowering BMI to decrease VTE risk may have particularly large beneficial effects among individuals with high genetic risk.
Collapse
Affiliation(s)
- Jihye Kim
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Peter Kraft
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA.,Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Kaitlin A Hagan
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Laura B Harrington
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Sara Lindstroem
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Christopher Kabrhel
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.,Department of Emergency Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
8
|
Reitsma PH. Genetics in thrombophilia. Hamostaseologie 2017; 35:47-51. [DOI: 10.5482/hamo-14-11-0062] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 11/14/2014] [Indexed: 01/05/2023] Open
Abstract
SummaryVenous thromboembolism (VTE) poses a worldwide health burden affecting millions of people each year. The annual incidence of symptomatic VTE, the collective term used here for deep venous thrombosis, pulmonary embolism or both, is 2–3 per thousand inhabitants. The one-year mortality is 20% after a first VTE. Of the surviving patients 15–25% will experience a recurrent episode of VTE in the three years after the first event. Primary and secondary prevention is key to reducing death and disability from VTE. How to make use of our current knowledge of inherited risk of VTE for primary and secondary disease prevention is not straightforward. This despite the fact that in the past two or three decades we have made major strides in enlarging our understanding of inherited VTE risk, and that new inherited risk factors continue to be identified.For primary prevention of VTE genetic testing is not likely to play a role in the future. Genetic variations also determine recurrence risk, albeit that the effect sizes for individual genetic variations are invariably lower than those for first VTE events. Multilocus genetic risk scores improve risk classification, and it is now possible to stratify patients who have had a first venous thrombosis, into subgroups with a high and low risk of recurrence. Whether this approach can be used to tailor intensity and duration of treatment remains to be established.
Collapse
|
9
|
Westrick RJ, Tomberg K, Siebert AE, Zhu G, Winn ME, Dobies SL, Manning SL, Brake MA, Cleuren AC, Hobbs LM, Mishack LM, Johnston AJ, Kotnik E, Siemieniak DR, Xu J, Li JZ, Saunders TL, Ginsburg D. Sensitized mutagenesis screen in Factor V Leiden mice identifies thrombosis suppressor loci. Proc Natl Acad Sci U S A 2017; 114:9659-9664. [PMID: 28827327 PMCID: PMC5594664 DOI: 10.1073/pnas.1705762114] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Factor V Leiden (F5L ) is a common genetic risk factor for venous thromboembolism in humans. We conducted a sensitized N-ethyl-N-nitrosourea (ENU) mutagenesis screen for dominant thrombosuppressor genes based on perinatal lethal thrombosis in mice homozygous for F5L (F5L/L ) and haploinsufficient for tissue factor pathway inhibitor (Tfpi+/- ). F8 deficiency enhanced the survival of F5L/LTfpi+/- mice, demonstrating that F5L/LTfpi+/- lethality is genetically suppressible. ENU-mutagenized F5L/L males and F5L/+Tfpi+/- females were crossed to generate 6,729 progeny, with 98 F5L/LTfpi+/- offspring surviving until weaning. Sixteen lines, referred to as "modifier of Factor 5 Leiden (MF5L1-16)," exhibited transmission of a putative thrombosuppressor to subsequent generations. Linkage analysis in MF5L6 identified a chromosome 3 locus containing the tissue factor gene (F3). Although no ENU-induced F3 mutation was identified, haploinsufficiency for F3 (F3+/- ) suppressed F5L/LTfpi+/- lethality. Whole-exome sequencing in MF5L12 identified an Actr2 gene point mutation (p.R258G) as the sole candidate. Inheritance of this variant is associated with suppression of F5L/LTfpi+/- lethality (P = 1.7 × 10-6), suggesting that Actr2p.R258G is thrombosuppressive. CRISPR/Cas9 experiments to generate an independent Actr2 knockin/knockout demonstrated that Actr2 haploinsufficiency is lethal, supporting a hypomorphic or gain-of-function mechanism of action for Actr2p.R258G Our findings identify F8 and the Tfpi/F3 axis as key regulators in determining thrombosis balance in the setting of F5L and also suggest a role for Actr2 in this process.
Collapse
Affiliation(s)
- Randal J Westrick
- Department of Biological Sciences, Oakland University, Rochester, MI 48309;
- Center for Data Science and Big Data Analysis, Oakland University, Rochester, MI 48309
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109
| | - Kärt Tomberg
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109
- Department of Human Genetics, University of Michigan, Ann Arbor, MI 48109
| | - Amy E Siebert
- Department of Biological Sciences, Oakland University, Rochester, MI 48309
| | - Guojing Zhu
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109
| | - Mary E Winn
- Bioinformatics and Biostatistics Core, Van Andel Research Institute, Grand Rapids, MI 49503
| | - Sarah L Dobies
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109
| | - Sara L Manning
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109
| | - Marisa A Brake
- Department of Biological Sciences, Oakland University, Rochester, MI 48309
| | - Audrey C Cleuren
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109
| | - Linzi M Hobbs
- Department of Biological Sciences, Oakland University, Rochester, MI 48309
| | - Lena M Mishack
- Department of Biological Sciences, Oakland University, Rochester, MI 48309
| | | | - Emilee Kotnik
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109
| | - David R Siemieniak
- Howard Hughes Medical Institute, University of Michigan, Ann Arbor, MI 48109
| | - Jishu Xu
- Department of Human Genetics, University of Michigan, Ann Arbor, MI 48109
| | - Jun Z Li
- Department of Human Genetics, University of Michigan, Ann Arbor, MI 48109
| | - Thomas L Saunders
- Transgenic Animal Model Core, University of Michigan, Ann Arbor, MI 48109
| | - David Ginsburg
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109;
- Department of Human Genetics, University of Michigan, Ann Arbor, MI 48109
- Howard Hughes Medical Institute, University of Michigan, Ann Arbor, MI 48109
- Department of Internal Medicine, Ann Arbor, MI 48109
- Department of Pediatrics, University of Michigan, Ann Arbor, MI 48109
| |
Collapse
|
10
|
Causative genetic mutations for antithrombin deficiency and their clinical background among Japanese patients. Int J Hematol 2016; 105:287-294. [PMID: 27858332 DOI: 10.1007/s12185-016-2142-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 11/07/2016] [Accepted: 11/07/2016] [Indexed: 10/20/2022]
Abstract
We summarize causative genetic mutations for antithrombin (AT) deficiency and their clinical background in Japanese patients. A total of 19 mutations, including seven novel mutations, were identified. We also summarize clinical symptoms of thrombosis, age at onset, family history, and contributing factors for thrombosis, and review the use of prophylactic anticoagulation in pregnant women with heterozygous type II heparin binding site defects (HBS) AT deficiency. The prevalence of thrombosis in probands with type I AT deficiency (88%) was double that observed in those with type II AT deficiency (50%). The prevalence of thrombotic episodes among family members was also higher for type I AT deficiency subjects (82%) than for those with type II AT deficiency (0%). The most common contributing factor for thrombosis among women with type I AT deficiency was pregnancy. Forty-five percent of women with type I AT deficiency developed thrombotic events before the 20th week of gestation. In contrast, women with type II (HBS) AT deficiency appear to be at a lower risk of thrombosis during pregnancy. In conclusion, thrombotic risk varies among different subtypes. Risk assessments based on genetic/clinical backgrounds may contribute to appropriate diagnosis, treatment, and prophylaxis for patients with AT deficiency.
Collapse
|
11
|
Abstract
The hypercoagulable states consist of a group of prothrombotic clinical disorders associated with an increased risk for thromboembolic events. The abnormalities lead to inappropriate thrombus formation. After a review of the coagulation process, inherited disorders (including antithrombin-III deficiency, protein CS system deficiencies, disorders of plasmin generation, dysfibrinogenemias, and homocysteinuria) and acquired disorders (including responses to surgery, cancer, drugs, and the antiphospholipid syndrome) are described. Screening and management methods are discussed. Copyright © 1997 by W. B. Saunders Company.
Collapse
Affiliation(s)
- Peter Robbins
- Department of Anaesthesia, University College London and Royal Free School of Anaesthesia, London, UK
| | - Mark Forrest
- Department of Anaesthesia, Charing Cross Hospital and Hammersmith School of Anaesthesia, London, UK
| | | |
Collapse
|
12
|
Moore GW, de Jager N, Cutler JA. Development of a novel, rapid assay for detection of heparin-binding defect antithrombin deficiencies: the heparin-antithrombin binding (HAB) ratio. Thromb Res 2015; 135:161-6. [DOI: 10.1016/j.thromres.2014.10.023] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Revised: 09/25/2014] [Accepted: 10/27/2014] [Indexed: 10/24/2022]
|
13
|
The early-onset preeclampsia is associated with MTHFR and FVL polymorphisms. Arch Gynecol Obstet 2014; 291:1303-12. [DOI: 10.1007/s00404-014-3561-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Accepted: 11/27/2014] [Indexed: 10/24/2022]
|
14
|
Abstract
PURPOSE OF REVIEW This article provides an overview of the evaluation and management of ischemic stroke in young adults, with an emphasis on cervical artery dissection, patent foramen ovale, and hypercoagulable states. RECENT FINDINGS The incidence of ischemic stroke in young patients is increasing, although the reasons remain unclear. Patients with ischemic stroke at a young age are more likely to die at an early age than their peers. Well-established vascular risk factors are prevalent in young stroke patients. Recent studies have informed the treatment of dissection and patent foramen ovale among others. The utility of testing for hypercoagulable states in ischemic stroke is unclear. SUMMARY Ischemic stroke in young adults is a major public health problem. A wide range of etiologies of ischemic stroke is found in this age group. A careful history, thorough examination, and methodical workup are essential. Specific management is predicated on identification of the underlying etiology.
Collapse
|
15
|
Influence of proband’s characteristics on the risk for venous thromboembolism in relatives with factor V Leiden or prothrombin G20210A polymorphisms. Blood 2013; 122:2555-61. [DOI: 10.1182/blood-2013-05-503649] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Key Points
The risk for venous thromboembolism in relatives with factor V Leiden or prothrombin G20210A differs according to the probands’ selection. Relatives belonging to families with heterozygous probands with venous thromboembolism have the highest risk for venous thromboembolism.
Collapse
|
16
|
Cooper DN, Krawczak M, Polychronakos C, Tyler-Smith C, Kehrer-Sawatzki H. Where genotype is not predictive of phenotype: towards an understanding of the molecular basis of reduced penetrance in human inherited disease. Hum Genet 2013; 132:1077-130. [PMID: 23820649 PMCID: PMC3778950 DOI: 10.1007/s00439-013-1331-2] [Citation(s) in RCA: 423] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Accepted: 06/15/2013] [Indexed: 02/06/2023]
Abstract
Some individuals with a particular disease-causing mutation or genotype fail to express most if not all features of the disease in question, a phenomenon that is known as 'reduced (or incomplete) penetrance'. Reduced penetrance is not uncommon; indeed, there are many known examples of 'disease-causing mutations' that fail to cause disease in at least a proportion of the individuals who carry them. Reduced penetrance may therefore explain not only why genetic diseases are occasionally transmitted through unaffected parents, but also why healthy individuals can harbour quite large numbers of potentially disadvantageous variants in their genomes without suffering any obvious ill effects. Reduced penetrance can be a function of the specific mutation(s) involved or of allele dosage. It may also result from differential allelic expression, copy number variation or the modulating influence of additional genetic variants in cis or in trans. The penetrance of some pathogenic genotypes is known to be age- and/or sex-dependent. Variable penetrance may also reflect the action of unlinked modifier genes, epigenetic changes or environmental factors. At least in some cases, complete penetrance appears to require the presence of one or more genetic variants at other loci. In this review, we summarize the evidence for reduced penetrance being a widespread phenomenon in human genetics and explore some of the molecular mechanisms that may help to explain this enigmatic characteristic of human inherited disease.
Collapse
Affiliation(s)
- David N. Cooper
- Institute of Medical Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff, CF14 4XN UK
| | - Michael Krawczak
- Institute of Medical Informatics and Statistics, Christian-Albrechts University, 24105 Kiel, Germany
| | | | - Chris Tyler-Smith
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, CB10 1SA UK
| | | |
Collapse
|
17
|
Great discrepancy in antithrombin activity measured using five commercially available functional assays. Thromb Res 2013; 132:132-7. [DOI: 10.1016/j.thromres.2013.05.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Revised: 04/26/2013] [Accepted: 05/10/2013] [Indexed: 11/20/2022]
|
18
|
Águila S, Martínez-Martínez I, Collado M, Llamas P, Antón AI, Martínez-Redondo C, Padilla J, Miñano A, de la Morena-Barrio ME, Garcia-Avello Á, Vicente V, Corral J. Compound heterozygosity involving Antithrombin Cambridge II (p.Ala416Ser) in antithrombin deficiency. Thromb Haemost 2013; 109:556-8. [PMID: 23329010 DOI: 10.1160/th12-09-0707] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2012] [Accepted: 11/27/2012] [Indexed: 11/05/2022]
|
19
|
Miller VM, Petterson TM, Jeavons EN, Lnu AS, Rider DN, Heit JA, Cunningham JM, Huggins GS, Hodis HN, Budoff MJ, Santoro N, Hopkins PN, Lobo RA, Manson JE, Naftolin F, Taylor HS, Harman SM, de Andrade M. Genetic polymorphisms associated with carotid artery intima-media thickness and coronary artery calcification in women of the Kronos Early Estrogen Prevention Study. Physiol Genomics 2013; 45:79-88. [PMID: 23188791 PMCID: PMC3546410 DOI: 10.1152/physiolgenomics.00114.2012] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2012] [Accepted: 11/25/2012] [Indexed: 12/15/2022] Open
Abstract
Menopausal hormone treatment (MHT) may limit progression of cardiovascular disease (CVD) but poses a thrombosis risk. To test targeted candidate gene variation for association with subclinical CVD defined by carotid artery intima-media thickness (CIMT) and coronary artery calcification (CAC), 610 women participating in the Kronos Early Estrogen Prevention Study (KEEPS), a clinical trial of MHT to prevent progression of CVD, were genotyped for 13,229 single nucleotide polymorphisms (SNPs) within 764 genes from anticoagulant, procoagulant, fibrinolytic, or innate immunity pathways. According to linear regression, proportion of European ancestry correlated negatively, but age at enrollment and pulse pressure correlated positively with CIMT. Adjusting for these variables, two SNPs, one on chromosome 2 for MAP4K4 gene (rs2236935, β = 0.037, P value = 2.36 × 10(-06)) and one on chromosome 5 for IL5 gene (rs739318, β = 0.051, P value = 5.02 × 10(-05)), associated positively with CIMT; two SNPs on chromosome 17 for CCL5 (rs4796119, β = -0.043, P value = 3.59 × 10(-05); rs2291299, β = -0.032, P value = 5.59 × 10(-05)) correlated negatively with CIMT; only rs2236935 remained significant after correcting for multiple testing. Using logistic regression, when we adjusted for waist circumference, two SNPs (rs11465886, IRAK2, chromosome 3, OR = 3.91, P value = 1.10 × 10(-04); and rs17751769, SERPINA1, chromosome 14, OR = 1.96, P value = 2.42 × 10(-04)) associated positively with a CAC score of >0 Agatston unit; one SNP (rs630014, ABO, OR = 0.51, P value = 2.51 × 10(-04)) associated negatively; none remained significant after correcting for multiple testing. Whether these SNPs associate with CIMT and CAC in women randomized to MHT remains to be determined.
Collapse
|
20
|
Bucciarelli P, Passamonti SM, Biguzzi E, Gianniello F, Franchi F, Mannucci PM, Martinelli I. Low borderline plasma levels of antithrombin, protein C and protein S are risk factors for venous thromboembolism. J Thromb Haemost 2012; 10:1783-91. [PMID: 22812604 DOI: 10.1111/j.1538-7836.2012.04858.x] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Inherited deficiencies of antithrombin (AT), protein C (PC) and protein S (PS) are risk factors for venous thromboembolism (VTE). They are usually defined by laboratory cut-offs (in our setting 81, 70 and 63 IU dL(-1), respectively), which give only a rough idea of the VTE risk associated with plasma levels of these proteins. OBJECTIVES We investigated whether the risk of VTE associated with the plasma deficiencies of AT, PC or PS has a dose-response effect, and whether low borderline levels of these proteins are associated with an increased risk of VTE, both in the whole study population and separately in carriers of either factor V Leiden or G20210A prothrombin gene mutation. PATIENTS/METHODS A case-control study of 1401 patients with a first objectively-documented VTE and 1847 healthy controls has been carried out. RESULTS A dose-response effect on the VTE risk was observed for all the three anticoagulant proteins. Compared with individuals with AT, PC or PS levels > 100 IU/dL, the adjusted odds ratio (95% CI) of VTE was 2.00 (1.44-2.78) for AT levels between 76 and 85 IUdL(-1) , 2.21 (1.54-3.18) and 1.84 (1.31-2.59) for PC and PS levels between 61 and 75 IUdL(-1) . The risk of unprovoked VTE in factor V Leiden or prothrombin G20210A carriers appears 2 to 3-fold increased when levels of AT or PS are low borderline. CONCLUSIONS Low borderline plasma levels of AT, PC and PS are associated with a 2-fold increased risk of VTE and should be considered in the assessment of the individual VTE risk.
Collapse
Affiliation(s)
- P Bucciarelli
- A. Bianchi Bonomi Hemophilia and Thrombosis Center, Department of Medicine and Medical Specialties, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico and University of Milan, Milan.
| | | | | | | | | | | | | |
Collapse
|
21
|
Korteweg FJ, Folkeringa N, Brouwer JLP, Erwich JJHM, Holm JP, van der Meer J, Veeger NJGM. Fetal loss in women with hereditary thrombophilic defects and concomitance of other thrombophilic defects: a retrospective family study. BJOG 2012; 119:422-30. [DOI: 10.1111/j.1471-0528.2011.03264.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
22
|
|
23
|
Chaireti R, Jennersjö C, Lindahl TL. Thrombin generation and D-dimer concentrations in a patient cohort investigated for venous thromboembolism. Relations to venous thrombosis, factor V Leiden and prothrombin G20210A. The LIST study. Thromb Res 2009; 124:178-84. [DOI: 10.1016/j.thromres.2008.12.033] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2008] [Revised: 10/22/2008] [Accepted: 12/09/2008] [Indexed: 11/24/2022]
|
24
|
Middeldorp S, van Hylckama Vlieg A. Does thrombophilia testing help in the clinical management of patients? Br J Haematol 2008; 143:321-35. [PMID: 18710381 DOI: 10.1111/j.1365-2141.2008.07339.x] [Citation(s) in RCA: 99] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Saskia Middeldorp
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands.
| | | |
Collapse
|
25
|
Abstract
Inherited thrombophilia can be defined as a genetically determined predisposition to the development of thromboembolic complications. Since the discovery of activated protein C resistance in 1993, several additional disorders have been described and, at present, it is possible to identify an inherited predisposition in about 60 to 70% of patients with such complications. These inherited prothrombotic risk factors include qualitative or quantitative defects of coagulation factor inhibitors, increased levels or function of coagulation factors, defects of the fibrinolytic system, altered platelet function, and hyperhomocysteinemia. In this review, the main inherited prothrombotic risk factors are analyzed from epidemiological, laboratory, clinical, and therapeutic points of view. Finally, we discuss the synergism between genetic and acquired prothrombotic risk factors in particular conditions such as childhood and pregnancy.
Collapse
Affiliation(s)
- Massimo Franchini
- Servizio di Immunoematologia e Trasfusione, Azienda Ospedaliera di Verona, Verona, Italy.
| | | | | | | | | |
Collapse
|
26
|
Abstract
Thromboembolism occurs more frequently in newborns than in older infants or children. The developing hemostasis system of neonates has decreased concentrations of procoagulant proteins and the naturally occurring anticoagulants and hemostatic control proteins. Overall, neonatal hemostasis provides protection from excessive bleeding at the expense of an increased risk for thromboembolism. Intensive medical care for premature and ill infants often requires central vascular assess, and the most frequent risk factor for thromboembolism is the presence of an indwelling vascular catheter. Management of venous thromboembolism in the newborn period varies depending on the location and extent of the thrombus as well as the risk for acute embolic complications and later vascular compromise. Therapeutic decisions are guided by practitioners' past experience, published case reports and case series, several large registries, and extrapolation from results of clinical trials in adults with thromboembolic disease. Valuable consensus guidelines have been compiled by the AACP Conference on Antithrombotic and Thrombolytic Therapy. Heparin, either unfractionated or a low molecular weight preparation, is the most commonly utilized anticoagulant to treat thromboembolism in newborn infants. Thrombolytic therapy may be considered if the thrombus is life or limb threatening and there is no hemorrhagic contraindication. Multicenter, prospective, controlled clinical trials in this important patient population are needed to provide evidence-based data to better inform optimal management.
Collapse
Affiliation(s)
- Diana S Beardsley
- Department of Pediatrics, Yale Women and Children's Center for Blood Disorders, Yale University School of Medicine, New Haven, CT 06520-3209, USA.
| |
Collapse
|
27
|
Abstract
The discovery of common genetic polymorphisms that predispose to venous thrombosis has led to the widespread availability of molecular testing for genetic thrombophilia traits. In terms of consent, genetic tests differ significantly from other types of laboratory test. We demonstrate the need for genetic counseling before and after genetic thrombophilia testing, but emphasize that such counseling need not be delivered by a specialist. We describe the potential advantages, limitations, and disadvantages of genetic testing for the common thrombophilic mutations that should be borne in mind when explaining testing to symptomatic individuals and asymptomatic relatives. In the vast majority of cases, genetic testing for thrombophilia is of limited value to the symptomatic patient, and provides minimal benefit over and above the family history when it comes to counseling at-risk family members.
Collapse
Affiliation(s)
- Paul Lochhead
- North of Scotland Regional Genetics Service, Clinical Genetics Centre, Aberdeen, UK
| | | |
Collapse
|
28
|
|
29
|
Miller VM, Jayachandran M, Heit JA, Owen WG. Estrogen therapy and thrombotic risk. Pharmacol Ther 2006; 111:792-807. [PMID: 16473411 DOI: 10.1016/j.pharmthera.2006.01.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2005] [Accepted: 01/04/2006] [Indexed: 11/25/2022]
Abstract
Post-menopausal hormone therapy increases the risk for venous thrombosis, and possibly myocardial infarction (MI) and ischemic stroke. However, most women using hormone therapy do not suffer thrombosis, and to date our ability to identify women at risk is limited. Thrombosis, arterial or venous, has 2 requisites: a vascular anomaly and a response of the hemostasis system to the anomaly. Consequently, experimental approaches to understand the pathophysiology of thrombosis require definition of vascular anatomy and function as well as characteristics of the blood within the context of genetic background, lifestyle choices and environmental exposures, which influence gene expression. Defining interactions among factors that affect individual propensity to thrombosis will allow physicians to better identify at-risk individuals, for example a woman contemplating estrogen therapy for symptoms of menopause, and prevent adverse thrombotic events.
Collapse
Affiliation(s)
- Virginia M Miller
- Department of Surgery, Mayo Clinic College of Medicine, Rochester, MN 55905, United States.
| | | | | | | |
Collapse
|
30
|
Tripodi A. Issues concerning the laboratory investigation of inherited thrombophilia. ACTA ACUST UNITED AC 2006; 9:181-6. [PMID: 16392896 DOI: 10.1007/bf03260089] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Inherited thrombophilia, defined as an increased familial tendency to develop thrombosis, may be due to congenital deficiencies or abnormalities of antithrombin, protein C or protein S; to the presence of a point mutation in the factor V gene (G1691A, factor V Leiden) leading to a poor anticoagulant response to activated protein C; or to the presence of a mutation in the prothrombin gene (G20210A) leading to increased plasma levels of prothrombin. The laboratory investigation of inherited thrombophilia should be limited to patients with a history of venous thromboembolism and, if positive, to their family members even though they are still asymptomatic. There is no indication for indiscriminate screening of the general population or screening of asymptomatic women before prescribing oral contraceptives. Testing should be based on the phenotype for antithrombin, protein C and protein S; on the phenotype and genotype (factor V Leiden mutation) for activated protein C resistance; and on the genotype (G20210A mutation) for hyperprothrombinemia. Phenotypic testing should be performed no sooner than three months after acute thrombotic events and at least 2 weeks after discontinuation of oral anticoagulant treatment.
Collapse
Affiliation(s)
- Armando Tripodi
- Angelo Bianchi Bonomi Hemophilia and Thrombosis Center, Department of Internal Medicine and Dermatology, University and IRCCS Maggiore Hospital, Milan, Italy.
| |
Collapse
|
31
|
Page C, Rubin LE, Gusberg RJ, Dardik A. Arterial thrombosis associated with heterozygous factor V Leiden disorder, hyperhomocysteinemia, and peripheral arterial disease: Importance of synergistic factors. J Vasc Surg 2005; 42:1014-8. [PMID: 16275464 DOI: 10.1016/j.jvs.2005.06.019] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2005] [Accepted: 06/22/2005] [Indexed: 11/29/2022]
Abstract
A 47-year-old man with heterozygous factor V Leiden disorder and intermittent hyperhomocysteinemia developed spontaneous acute popliteal artery thrombosis. Homocysteine levels were above normal limits at presentation. Intra-arterial thrombolysis was used successfully to treat the acute thrombosis; long-term treatment included anticoagulation, folic acid, and risk factor modification. Although factor V Leiden is strongly associated with deep venous thrombosis, additional cofactors such as hyperhomocysteinemia may predispose to an increased risk of acute arterial thrombosis in areas of pre-existing peripheral arterial disease.
Collapse
Affiliation(s)
- Cameron Page
- Department of Surgery, VA Connecticut Healthcare System, Yale University School of Medicine, New Haven, CT 06519, USA
| | | | | | | |
Collapse
|
32
|
Tormene D, Fortuna S, Tognin G, Gavasso S, Pagnan A, Prandoni P, Simioni P. The incidence of venous thromboembolism in carriers of antithrombin, protein C or protein S deficiency associated with the HR2 haplotype of factor V: a family cohort study. J Thromb Haemost 2005; 3:1414-20. [PMID: 15978097 DOI: 10.1111/j.1538-7836.2005.01308.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
In order to assess whether the HR2 haplotype of the factor V gene (HR2) increases the risk of venous thromboembolism (VTE) in carriers of antithrombin (AT), protein C (PC) or S (PS) defects, we performed this determination in 336 subjects, who were family members of 66 symptomatic patients with clotting inhibitors defects. We first assessed the presence of previous VTE, and then followed prospectively subjects without prior VTE. VTE episodes had occurred in 26 individuals: 18 in 139 carriers of clotting inhibitors defects alone (annual incidence, 0.55%), four in 33 carriers of clotting inhibitors defects combined with HR2 (0.52%) and four in 151 non-carriers (0.1%), resulting in a relative risk (RR) for VTE of 4.9 (95% CI: 1.7-14.4) and 4.62 (95% CI: 1.2-18.4), respectively. After an overall follow-up of 2557 patient-years, VTE episodes developed in 12 subjects: nine in 121 carriers of clotting inhibitors defects alone (annual incidence, 0.92%), three in 29 carriers of clotting inhibitors defects combined with HR2 (1.0%) and none in 147 non-carriers. In family members of patients with AT, PC or PS defects the coinheritance of HR2 haplotype does not seem to increase the thromboembolic risk.
Collapse
Affiliation(s)
- D Tormene
- Department of Medical and Surgical Sciences, 2nd Chair of Internal Medicine, University of Padua Medical School, Padua, Italy
| | | | | | | | | | | | | |
Collapse
|
33
|
O'Donnell J, Riddell A, Owens D, Handa A, Pasi J, Hamilton G, Perry DJ. Role of the Thrombelastograph as an adjunctive test in thrombophilia screening. Blood Coagul Fibrinolysis 2005; 15:207-11. [PMID: 15060415 DOI: 10.1097/00001721-200404000-00002] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The Thrombelastograph (TEG) provides a global assessment of haemostatic function. We have investigated the potential role of TEG in thrombophilia testing by studying 87 consecutive patients (all with personal or family history of thrombosis). For each patient, routine thrombophilia screening tests and TEG were performed in parallel. In keeping with previous reports, at least one thrombophilic trait was identified in 34% of the patients. Among those patients with previous thrombosis, the yield from screening was higher (54% with prothrombotic trait). Overall, a hypercoaguable TEG was demonstrated in 45% of patients. However, TEG and standard thrombophilia testing did not necessarily identify the same subset of individuals. Of 39 patients with hypercoaguable TEG, only 17 had a demonstrable thrombophilic trait. Conversely, of 30 patients with proven thrombophilia, 17 also had a hypercoaguable TEG. We conclude that TEG cannot be used as a sole initial screening test in patients referred for thrombophilia testing, as it fails to identify 43% of underlying thrombophilic traits. However, TEG may be a useful adjunctive test, particularly in patients in whom the regular thrombophilia screen proves unremarkable. Prospective studies are warranted to determine whether a hypercoaguable TEG can be used to predict a subset of patients at increased risk for recurrent thrombotic events.
Collapse
Affiliation(s)
- James O'Donnell
- Department of Haematology, Imperial College School of Medicine, Hammersmith Hospital, London, UK.
| | | | | | | | | | | | | |
Collapse
|
34
|
Abstract
Abstract
Over the last decade we have witnessed an avalanche of newly identified risk factors for venous thrombosis. This has advanced our knowledge of its etiology, because more determinants have been described and because the underlying concepts have received a new and broader understanding.
Venous thrombosis is a common multicausal disease occurring as the result of interacting genetic, environmental and behavioral risk factors. Some of these have been known since medieval times, such as the increased risk of thrombosis during immobilization in pregnancy and after childbirth (although retained milk of the breast-feeding mother was seen as the primary cause for the latter). Pregnancy and puerperium still cause thrombosis, as do exogenous hormones in oral contraceptives and hormonal replacement therapy. Furthermore, the immobilization in the puerperium of the old days translates directly to situations of immobilization in current times, such as prolonged travel in airplanes or excessive electronic gaming.
While pedigrees with abundant thrombosis were observed in the early 1900s, the first cause of heritable thrombophilia (antithrombin deficiency) was discovered in 1965, with the subsequent identification of deficiencies of protein C and protein S in the early 1980s. These were uncommon and strong risk factors, whereas the more recently discovered genetic variants are common and weak, and cause disease only in the presence of other factors.
Collapse
|
35
|
Rosendaal FR. Venous thrombosis: the role of genes, environment, and behavior. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2005:1-12. [PMID: 16304352 DOI: 10.1182/asheducation-2005.1.1] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Over the last decade we have witnessed an avalanche of newly identified risk factors for venous thrombosis. This has advanced our knowledge of its etiology, because more determinants have been described and because the underlying concepts have received a new and broader understanding. Venous thrombosis is a common multicausal disease occurring as the result of interacting genetic, environmental and behavioral risk factors. Some of these have been known since medieval times, such as the increased risk of thrombosis during immobilization in pregnancy and after childbirth (although retained milk of the breast-feeding mother was seen as the primary cause for the latter). Pregnancy and puerperium still cause thrombosis, as do exogenous hormones in oral contraceptives and hormonal replacement therapy. Furthermore, the immobilization in the puerperium of the old days translates directly to situations of immobilization in current times, such as prolonged travel in airplanes or excessive electronic gaming. While pedigrees with abundant thrombosis were observed in the early 1900s, the first cause of heritable thrombophilia (antithrombin deficiency) was discovered in 1965, with the subsequent identification of deficiencies of protein C and protein S in the early 1980s. These were uncommon and strong risk factors, whereas the more recently discovered genetic variants are common and weak, and cause disease only in the presence of other factors.
Collapse
Affiliation(s)
- Frits R Rosendaal
- Leiden Univ. Med.Center, PO Box 9600, C9-P, 2300 RC Leiden, Netherlands.
| |
Collapse
|
36
|
Gonthier A, Bogousslavsky J. Infarctus cérébraux artériels d’origine hématologique : expérience lausannoise et revue de la littérature. Rev Neurol (Paris) 2004; 160:1029-39. [PMID: 15602345 DOI: 10.1016/s0035-3787(04)71140-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
INTRODUCTION Hematological diseases are seldom found as the etiology of ischemic strokes, but are frequently investigated by expensive laboratory tests after a first cerebral vascular event. METHODS In the Lausanne Stroke Registry, we retrospectively reviewed the cases of patients hospitalized between 1979 and 2001 for a first ischemic arterial stroke which was attributed to a hematological etiology. Of 4697 patients, 22 (0.47 per cent) had a stroke due to one of the following hematological pathology: polycythemia vera (4), secondary polycythemia (4), essential thrombocytemia (2), secondary thrombocytosis (4), multiple myeloma (1), CIVD (1), protein S deficiency (1), antiphospholipid antibody syndrome (4), moderate homocysteinemia (1). A literature review was undertaken for each hemopathy. CONCLUSION In light of the results of these data, we concluded that a complete blood count provides sufficient hematological screening for the majority of patients hospitalized for an arterial stroke. The antiphospholipid antibody syndrome is a rare cause of cerebral infarction, which needs to be investigated in young patients, in cases of multiple or recurring stroke or in the presence of a typical history. Inherited thrombophilias are not a significant risk factor for arterial cerebral infarction and their investigation is only warranted for a sub-group of young patients with a cryptogenic stroke, in which group the prevalence is slightly increased. Moderate homocysteinemia must be considered as a cerebrovascular risk factor of minor importance, but potentially treatable by a substitution of vitamin B12, B6 and folates. The efficacy of this substitution in the prevention of cardiovascular events needs yet to be demonstrated.
Collapse
Affiliation(s)
- A Gonthier
- Service de Neurologie, CHUV, Lausanne, Suisse.
| | | |
Collapse
|
37
|
Sartori MT, Danesin C, Saggiorato G, Tormene D, Simioni P, Spiezia L, Patrassi GM, Girolami A. The PAI-1 gene 4G/5G polymorphism and deep vein thrombosis in patients with inherited thrombophilia. Clin Appl Thromb Hemost 2004; 9:299-307. [PMID: 14653439 DOI: 10.1177/107602960300900405] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Genetic and acquired factors may influence phenotypic expression of inherited thrombophilia. Hypofibrinolysis due to excess PAI-1 can be found in patients with deep vein thrombosis (DVT) and 4G/5G polymorphism of the PAI-1 gene may modulate the inhibitor's synthesis. In 149 patients with inherited thrombophilia, the possible thrombotic contribution of both 4G/5G polymorphism and PAI-1 plasma levels was evaluated. Sixty-seven patients with idiopathic DVT and 98 normal subjects were also studied. By comparison with controls, a significantly higher prevalence of 4G/4G genotype was seen in idiopathic DVT and in thrombophilia patients, although in this latter group the difference only remained significant in cases symptomatic for thrombosis (p = 0.01). The 4G/4G genotype was associated with a greater risk of thrombosis both in symptomatic thrombophilia patients (OR 2.85, 95% CI 1.26-6.46) and in idiopathic DVT patients (OR 3.1, 95% CI 1.26-7.59). The greater frequency of 4G allele in symptomatic thrombophilia patients with respect to controls was statistically significant (p = 0.04). Compared to healthy subjects, PAI-1:Ag levels were higher in symptomatic thrombophilia patients and related to the 4G/5G polymorphism, with significantly higher values in the 4G/4G carriers. In conclusion, PAI-1 4G/5G polymorphism may influence PAI-1 expression and thrombotic risk in patients with inherited thrombophilia.
Collapse
Affiliation(s)
- Maria Teresa Sartori
- Department of Medical and Surgical Sciences, University of Padua Medical School, Padua, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Vossen CY, Conard J, Fontcuberta J, Makris M, Van Der Meer FJM, Pabinger I, Palareti G, Preston FE, Scharrer I, Souto JC, Svensson P, Walker ID, Rosendaal FR. Familial thrombophilia and lifetime risk of venous thrombosis. J Thromb Haemost 2004; 2:1526-32. [PMID: 15333025 DOI: 10.1111/j.1538-7836.2004.00852.x] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND We started a large multicenter prospective follow-up study to provide reliable risk estimates of venous thrombosis in families with various thrombophilic defects. OBJECTIVES This paper describes data collected at study entry on venous events experienced before study inclusion, i.e. the baseline data. PATIENTS/METHODS All individuals (probands, relatives) registered in nine European thrombosis centers with the factor (F)V Leiden mutation, a deficiency of antithrombin, protein C or protein S, or a combination of these defects, were enrolled between March 1994 and September 1997. As control individuals, partners, friends or acquaintances of the thrombophilic participants were included. Incidence and relative risk of objectively confirmed venous thrombotic events (VTEs) prior to entry were calculated for the relatives with thrombophilia and the controls. RESULTS Of the 846 relatives with thrombophilia (excluding probands), 139 (16%) had experienced a VTE with an incidence of 4.4 per 1000 person years. Of the controls, 15 of the 1212 (1%) controls had experienced a VTE with an incidence of 0.3 per 1000 person years. The risk of venous thrombosis associated with familial thrombophilia was 15.7 (95% CI 9.2-26.8) and remained similar after adjustment for regional and sex-effects (16.4; 95% CI 9.6-28.0). The highest incidence per 1000 person years was found in relatives with combined defects (8.4; 95% CI 5.6-12.2), and the lowest incidence was found in those with the FV Leiden mutation (1.5; 95% CI 0.8-2.6). CONCLUSIONS Considerable differences in the lifetime risk of VTE were observed among individuals with different thrombophilia defects.
Collapse
Affiliation(s)
- C Y Vossen
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Abstract
This article about antithrombotic therapy in children is part of the 7th American College of Chest Physicians Conference on Antithrombotic and Thrombolytic Therapy: Evidence-Based Guidelines. Grade 1 recommendations are strong and indicate that the benefits do, or do not, outweigh the risks, burden, and costs. Grade 2 suggests that individual patients' values may lead to different choices (for a full understanding of the grading see Guyatt et al, CHEST 2004; 126:179S-187S). Among the key recommendations in this article are the following. In neonates with venous thromboembolism (VTE), we suggest treatment with either unfractionated heparin or low-molecular-weight heparin (LMWH), or radiographic monitoring and anticoagulation therapy if extension occurs (Grade 2C). We suggest that clinicians not use thrombolytic therapy for treating VTE in neonates, unless there is major vessel occlusion that is causing the critical compromise of organs or limbs (Grade 2C). For children (ie, > 2 months of age) with an initial VTE, we recommend treatment with i.v. heparin or LMWH (Grade 1C+). We suggest continuing anticoagulant therapy for idiopathic thromboembolic events (TEs) for at least 6 months using vitamin K antagonists (target international normalized ratio [INR], 2.5; INR range, 2.0 to 3.0) or alternatively LMWH (Grade 2C). We suggest that clinicians not use thrombolytic therapy routinely for VTE in children (Grade 2C). For neonates and children requiring cardiac catheterization (CC) via an artery, we recommend i.v. heparin prophylaxis (Grade 1A). We suggest the use of heparin doses of 100 to 150 U/kg as a bolus and that further doses may be required in prolonged procedures (both Grade 2 B). For prophylaxis for CC, we recommend against aspirin therapy (Grade 1B). For neonates and children with peripheral arterial catheters in situ, we recommend the administration of low-dose heparin through a catheter, preferably by continuous infusion to prolong the catheter patency (Grade 1A). For children with a peripheral arterial catheter-related TE, we suggest the immediate removal of the catheter (Grade 2C). For prevention of aortic thrombosis secondary to the use of umbilical artery catheters in neonates, we suggest low-dose heparin infusion (1 to 5 U/h) (Grade 2A). In children with Kawasaki disease, we recommend therapy with aspirin in high doses initially (80 to 100 mg/kg/d during the acute phase, for up to 14 days) and then in lower doses (3 to 5 mg/kg/d for > or = 7 weeks) [Grade 1C+], as well as therapy with i.v. gammaglobulin within 10 days of the onset of symptoms (Grade 1A).
Collapse
Affiliation(s)
- Paul Monagle
- Division of Laboratory Services, Royal Children's Hospital, Department of Paediatrics, University of Melbourne, Flemington Rd, Parkville, Melbourne, VIC, Australia 3052.
| | | | | | | | | |
Collapse
|
40
|
David D, Ribeiro S, Ferrão L, Gago T, Crespo F. Molecular basis of inherited antithrombin deficiency in Portuguese families: identification of genetic alterations and screening for additional thrombotic risk factors. Am J Hematol 2004; 76:163-71. [PMID: 15164384 DOI: 10.1002/ajh.20067] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Antithrombin (AT), the most important coagulation serine proteases inhibitor, plays an important role in maintaining the hemostatic balance. Inherited AT deficiency, mainly characterized by predisposition to recurrent venous thromboembolism, is transmitted in an autosomal dominant manner. In this study, we analyzed the underlying genetic alterations in 12 unrelated Portuguese thrombophilic families with AT deficiency. At the same time, the modulating effect of the FV Leiden mutation, PT 20210A, PAI-1 4G, and MTHFR 677T allelic variants, on the thrombotic risk of AT deficient patients was also evaluated. Three novel frameshift alterations, a 4-bp deletion in exon 4 and two 1-bp insertions in exon 6, were identified in six unrelated type I AT deficient families. A novel missense mutation in exon 3a, which changes the highly conserved F147 residue, and a novel splice site mutation in the invariant acceptor AG dinucleotide of intron 2 were also identified in unrelated type I AT deficient families. In addition to these, two previously reported missense mutations changing the AT reactive site bond (R393-S394) and leading to type II-RS deficiency, and a previously reported cryptic splice site mutation (IVS4-14G-->A), were also identified. In these families, increased thrombotic risk associated with co-inheritance of the FV Leiden mutation and of the PAI-1 4G variant was also observed. In conclusion, we present the first data regarding the underlying genetic alterations in Portuguese thrombophilic families with AT deficiency, and confirm that the FV Leiden mutation and probably the PAI-1 4G variant represent additional thrombotic risk factors in these families.
Collapse
Affiliation(s)
- Dezsö David
- Centro de Genética Humana, Instituto Nacional de Saúde Dr. Ricardo Jorge, Lisboa, Portugal.
| | | | | | | | | |
Collapse
|
41
|
Deitcher SR, Gomes MPV. Hypercoagulable state testing and malignancy screening following venous thromboembolic events. Vasc Med 2003; 8:33-46. [PMID: 12866610 DOI: 10.1191/1358863x03vm461ra] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Mounting interest in hypercoagulability, increased availability of hypercoagulable state test 'panels' and enhanced ability to identify abnormalities in tested patients have prompted widespread testing. Testing for acquired and inherited hypercoagulable states uncovers an abnormality in over 50% of patients presenting with an initial venous thromboembolic event (VTE) but may have minimal actual impact on management in most of these patients. Such laboratory screening should be reserved for patients in whom the results of individual tests will significantly impact the choice of anticoagulant agent, intensity of anticoagulant therapy, therapeutic monitoring, family screening, family planning, prognosis determination, and most of all duration of therapy. Testing 'just to know' is neither cost-effective nor clinically appropriate. The most important testing in patients following acute VTE may be age- and gender-specific cancer screening. Cancer screening following VTE seems most prudent in older individuals and in those with idiopathic VTE and no laboratory evidence for an inherited hypercoagulable state. Cancer screening should focus on identification of treatable cancers and those where diagnosis in an early stage favorably impacts patient survival. Extensive searches for occult malignancy employing whole-body computed tomography and serum tumor markers may identify more cancers but without affecting patient outcome. We advocate that physicians should focus their attention more on VTE prophylaxis and proper treatment and less on costly and, at times, invasive testing of questionable value.
Collapse
Affiliation(s)
- Steven R Deitcher
- Department of Hematology and Medical Oncology, The Cleveland Clinic Foundation, Cleveland, OH 44195, USA.
| | | |
Collapse
|
42
|
Abstract
The propensity for both arterial and venous thrombotic disorders involves a genetic predetermination that operates In concert with environmental factors or triggers. Appropriate clinical assessment and therapeutic recommendations for patients with thrombosis requires a thorough knowledge of genetic variables that influence this propensity. This review focuses on the pathophysiology, natural history, and molecular biology of defined thrombophilic risk factors relevant to the care of patients with thrombotic disorders.
Collapse
Affiliation(s)
- Robert D. McBane
- Division of Cardiovascular Medicine, Section of Hematology Research, Mayo Clinic and Foundation for Education and Research, Rochester, Minnesota
| |
Collapse
|
43
|
Samama MM, Rached RA, Horellou MH, Aquilanti S, Mathieux VG, Plu-Bureau G, Elalamy I, Conard J. Pregnancy-associated venous thromboembolism (VTE) in combined heterozygous factor V Leiden (FVL) and prothrombin (FII) 20210 A mutation and in heterozygous FII single gene mutation alone. Br J Haematol 2003; 123:327-34. [PMID: 14531916 DOI: 10.1046/j.1365-2141.2003.04582.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The risk of venous thromboembolism (VTE) in the absence of prophylaxis was evaluated in a retrospective study of 47 women (84 pregnancies) with combined thrombophilia [heterozygous factor V Leiden (FVL) plus prothrombin (FII) 20210A mutation (group I)] and in 82 women (193 pregnancies) with the FII alone (group II). VTE was more frequent in group I than in group II [17.8% versus 6.2%, P = 0.003, relative risk (RR) 2.9, 95% confidence interval (CI) 1.4-5.9], ante partum (7.1% and 2.1%) and post partum (11.5% and 4.2%). The risk was higher in index cases than in family members (RR 2.5, 95% CI 1.2-5.2 and RR 2.1, 95% CI 0.2-22.3 respectively) Even women who had no history of VTE before pregnancy had an increased risk (RR 2.2, 95% CI 1.0-4.8). Our results suggest that, during ante partum, prophylaxis is indicated in women with combined thrombophilia and with a VTE before pregnancy. In those without VTE before pregnancy, prophylaxis might be decided for each individual case, taking into consideration all risk factors. In women with the FII mutation alone, the low risk may not justify prophylaxis in the absence of previous VTE. In post partum, prophylaxis is indicated in all cases.
Collapse
|
44
|
Affiliation(s)
- Elizabeth G Nabel
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Md 20892, USA.
| |
Collapse
|
45
|
Abstract
It is now possible to identify acquired and hereditary risk factors in a substantial percentage of patients presenting with a venous thrombotic event. Discovery of the factor V Leiden and prothrombin G20210A mutations has greatly increased the percentage of patients in whom venous thrombosis can be attributed to hereditary thrombophilia. There is, however, considerable uncertainty as to how this information should be used in patient management. Although prolonged anticoagulation at an international normalized ratio of 2-3 is highly effective in preventing thrombotic recurrences, this benefit is partially offset by major bleeding which occurs at an average rate of 2%-3% per year. A decision as to the overall benefit of extended anticoagulation in the individual patient requires assessment of the risk of recurrence in the absence of treatment vs. the bleeding risk associated with prolonged anticoagulation. Low-intensity warfarin therapy or novel anticoagulants such as oral direct thrombin inhibitors may prove effective strategies for preventing recurrent venous thromboembolism in patients with thrombophilia.
Collapse
Affiliation(s)
- K A Bauer
- VA Boston Healthcare System, Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
46
|
Hayden PJ, O'Brien M, Collins B, O'Farrelly C, Murphy K. Inherited thrombotic tendency in a pedigree with antithrombin La Rochelle (N405K) and factor V Leiden. CLINICAL AND LABORATORY HAEMATOLOGY 2003; 25:191-3. [PMID: 12755798 DOI: 10.1046/j.1365-2257.2003.00514.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
An Irish pedigree with a strong thrombotic tendency, due to the novel association of antithrombin La Rochelle (N405K) and factor V Leiden is described. Thromboses occurred at a median age of 16 years in four family members, carrying both mutant alleles. Molecular diagnosis was facilitated by the creation of an additional restriction enzyme site (Mnl1) in exon 6 of the antithrombin gene. Both the genes for antithrombin and factor V are located at chromosome 1q23. The molecular data and phenotypic behaviour in this family are strongly suggestive of allelic co-segregation of the mutant alleles.
Collapse
Affiliation(s)
- P J Hayden
- Department of Haematology, St Vincent's University Hospital, Dublin, Ireland.
| | | | | | | | | |
Collapse
|
47
|
Abstract
Within the past decade, the identification of two mutations that are relatively prevalent among the white population (the factor V Leiden and prothrombin G20210A gene mutations) has paved the way for a number of large cohort studies that have greatly advanced our understanding of the pathogenesis of venous thromboembolism (VTE). VTE is clearly a multigenic disorder, with well-characterized examples of gene-gene and gene-environment interactions underlying its pathogenesis. Increasing numbers of patients are being referred for testing, and many more diagnoses of inherited thrombophilia are being made. The purpose of this article is to discuss the practical applications of both diagnostic testing and genetic counseling for the major inherited thrombophilias: inherited resistance to activated protein C/factor V Leiden, prothrombin G20210A mutation, protein C deficiency, protein S deficiency, and antithrombin deficiency. A description of each entity is included along with a discussion of the indications for testing, selection of the most appropriate screening test, and proper interpretation of test results. Informed consent for testing, screening of asymptomatic individuals in special circumstances (such as during pregnancy or before initiation of estrogen therapy), screening of family members, and posttest education are also addressed. This article emphasizes that these polymorphisms should be regarded as risk factors for thrombosis whose clinical expression generally depends on the coexistence of additional thrombophilic mutations or environmental conditions that provoke the development of VTE.
Collapse
Affiliation(s)
- Laura M Reich
- Vascular Medicine Program, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | | | | |
Collapse
|
48
|
Endler G, Mannhalter C. Polymorphisms in coagulation factor genes and their impact on arterial and venous thrombosis. Clin Chim Acta 2003; 330:31-55. [PMID: 12636925 DOI: 10.1016/s0009-8981(03)00022-6] [Citation(s) in RCA: 77] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Arterial and venous thromboses, with their clinical manifestations such as stroke, myocardial infarction (MI), or pulmonary embolism, are the major causes of death in developed countries. Several studies in twins and siblings have shown that genetic factors contribute significantly to the development of these diseases. Since the advent of molecular genetics in medicine, it has been a focus of interest to elucidate the role of mutations in various candidate genes and their impact on hemostatic disorders such as arterial and venous thromboses. In this article, we review the current knowledge of the contribution of polymorphisms in coagulation factors to the development of thrombotic diseases. We show that in arterial thrombosis, results are controversial. Only for factor XIII 34Leu a protective effect on the development of myocardial infarction has been demonstrated in several studies. No other single polymorphism in a coagulation factor could be confirmed as a relevant risk factor, although there is evidence for a role of factor V Arg506Gln, factor VII Arg353Gln, and vWF Thr789Ala polymorphisms in patient subgroups. Further studies will be necessary to confirm the value of testing for genetic polymorphisms in arterial thrombosis. A large body of data is available on the role of factor V Arg506Gln and the prothrombin G20210A mutation in venous thrombosis. Some papers already recommend diagnosis and treatment strategies. We will discuss these recent publications on venous thrombosis in our review.
Collapse
Affiliation(s)
- Georg Endler
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Molecular Biology Division, University Vienna Medical School, Austria
| | | |
Collapse
|
49
|
Abstract
Thrombophilia is the predisposition to venous thromboembolism and is caused by inherited and acquired factors, alone or in combination. With the discovery of APC resistance and the prothrombin gene mutation, more than half of all patients with clinical characteristics of thrombophilia are now diagnosed with an inherited disorder. The hypercoagulable work-up of patients with venous thromboembolism is important, because the causes can influence the duration and management of anticoagulation therapy, as well as affect other decisions regarding life and health issues.
Collapse
Affiliation(s)
- Stephanie L Perry
- Division of Medical Oncology, Division of Hematology, Duke University Medical Center, Trent Drive, Durham, NC 27710, USA
| | | |
Collapse
|
50
|
Chen TY, Su WC, Tsao CJ. Incidence of thrombophilia detected in southern Taiwanese patients with venous thrombosis. Ann Hematol 2003; 82:114-117. [PMID: 12601491 DOI: 10.1007/s00277-002-0603-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2002] [Accepted: 12/08/2002] [Indexed: 11/26/2022]
Abstract
In order to analyze the incidence of thrombophilia in southern Taiwan, we studied the prevalence of antithrombin (AT), protein C (PC), and protein S (PS) deficiencies, the prevalence of factor V Leiden mutation, and the presence of acquired lupus anticoagulant (LA) and anticardiolipin antibody (ACA) in 56 patients < or =65 years old with deep venous thrombosis (DVT). Of 56 patients, 30 were male, 26 female, and the mean age of the patients was 43 years (18-65 years). None had factor V mutation or activated PC resistance; 21 patients (37.5%) showed abnormal results: 4 (7.1%) had AT deficiencies, 6 (10.7%) PC deficiencies, 6 (10.7%) PS deficiencies, 2 (3.6%) a combined PC and PS deficiency, and 3 (5.4%) LA and ACA. Only PC and PS deficiencies were significantly associated with increased risk for the development of thrombosis with an odds ratio of 4.2 (95% confidence interval: 1.2-15.0, P=0.018) and 8.1 (95% confidence interval: 1.6-40.6, P=0.003), respectively. We concluded that the prevalence of heritable thrombophilia (34.0%) in Taiwan is higher than that in Western countries, but that it is lower than previously reported in Hong Kong and Taiwan. We attribute this to selection bias.
Collapse
Affiliation(s)
- T-Y Chen
- Division of Hematology/Oncology, Department of Internal Medicine, National Cheng-Kung University Hospital, 138 Sheng-Li Road, 704, Tainan, Taiwan.
| | - W-C Su
- Division of Hematology/Oncology, Department of Internal Medicine, National Cheng-Kung University Hospital, 138 Sheng-Li Road, 704, Tainan, Taiwan
| | - C-J Tsao
- Division of Hematology/Oncology, Department of Internal Medicine, National Cheng-Kung University Hospital, 138 Sheng-Li Road, 704, Tainan, Taiwan
| |
Collapse
|