Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 379
Filter
Add more filters

Publication year range
1.
J Proteome Res ; 23(10): 4538-4552, 2024 Oct 04.
Article in English | MEDLINE | ID: mdl-39265992

ABSTRACT

Protein S deficiency (PSD) is an autosomal dominant disorder characterized by congenital thrombophilia. Studies on PSD are limited yet, resulting in a lack of clarity about molecular changes during abnormal coagulation. Proteomics and metabolomics analyses were conducted on the plasma of PSD patients based on liquid and gas chromatography-mass spectrometry (LC- and GC-MS). Differential proteins and metabolites of PSD were then filtered by univariate statistical analysis and subjected to network analysis using the ingenuity pathway analysis (IPA) platform. The proteome and metabolome of PSD were obviously disturbed, and the biological pathway of coagulation and complement cascades was the most affected. During PSD, overall levels of anticoagulant protein decreased and negative regulation of thrombin production was reduced, causing the formation of fibrin clots and platelet aggregation. Furthermore, 9 differential proteins correlated significantly with protein S, comprising A2M, AGT, APOE, FGG, GPLD1, IGHV1-69, CFHR5, CPN2, and CA1. The biological networks suggested that the pathways of acute phase response, FXR/RXR activation, serotonin receptor signaling, and p70S6K signaling were associated with PSD, indicating an interaction disorder of inflammatory immune and lipid metabolism. The findings may contribute to knowledge of available functional molecules and biological pathways of familial PSD and help with treatment improvement. Data are available via ProteomeXchange with identifier PXD055111 and MetaboLights with reference number MTBLS2653.


Subject(s)
Metabolome , Protein S Deficiency , Proteome , Humans , Proteome/metabolism , Proteome/analysis , Protein S Deficiency/genetics , Protein S Deficiency/blood , Female , Male , Adult , Protein S/metabolism , Protein S/genetics , Proteomics/methods , Blood Coagulation , Gas Chromatography-Mass Spectrometry , Metabolomics/methods , Chromatography, Liquid
2.
Ann Hematol ; 103(2): 653-662, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38175252

ABSTRACT

We report three heterozygous PROS1 mutations that caused type I protein S deficiency in three unrelated Chinese families. We measured protein S activity and antigen levels for all participants, screened them for mutations in the PROS1 gene. And we employed the calibrated automated thrombin generation (CAT) method to investigate thrombin generation. Numerous bioinformatics tools were utilized to analyze the conservation, pathogenicity of mutation, and spatial structure of the protein S. Phenotyping analysis indicated that all three probands exhibited simultaneous reduced levels of PS:A, TPS:Ag, and FPS:Ag. Genetic testing revealed that proband A harbored a heterozygous c.458_458delA (p.Lys153Serfs*6) mutation in exon 5, proband B carried a heterozygous c.1687C>T (p.Gln563stop) mutation in exon 14, and proband C exhibited a heterozygous c.200A>C (p.Glu67Ala) mutation in exon 2. Bioinformatic analysis predicted that the p.Lys153Serfs*6 frameshift mutation and the p.Gln563stop nonsense mutation in the protein S were classified as "disease-causing." The identification of the novel mutation p.Lys153Serfs*6 in PROS1 enriches the Human Genome Database. Our research suggests that these three mutations (p.Lys153Serfs*6, p.Gln563stop, and p.Glu67Ala) are possibly responsible for the decreased level of protein S in the three families. Furthermore, the evidence also supports the notion that individuals who are asymptomatic but have a family history of PSD can benefit from genetic analysis of the PROS1 gene.


Subject(s)
Blood Proteins , Protein S Deficiency , Humans , Blood Proteins/genetics , Protein S Deficiency/diagnosis , Protein S Deficiency/genetics , Thrombin , Mutation , China , Pedigree , Protein S/genetics
3.
Pediatr Blood Cancer ; 71(3): e30824, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38155150

ABSTRACT

OBJECTIVES: To determine the optimal management for early-onset thrombophilia (EOT), the genetic and clinical features of protein C (PC)-, protein S (PS)-, or antithrombin (AT)-deficient patients of ≤20 years of age were studied in Japan. METHODS/RESULTS: Clinical and genetic information of all genetically diagnosed cases was collected through the prospective, retrospective study, and literature review. One-hundred-one patients had PC (n = 55), PS (n = 29), or AT deficiency (n = 18). One overlapping case had PC- and PS-monoallelic variant. Fifty-five PC-deficient patients (54%) had 26 monoallelic or 29 biallelic variant(s), and 29 (29%) PS-deficient patients had 20 monoallelic or nine biallelic variant(s). None of the patients had AT-biallelic variants. The frequent low-risk allele p.K193del (PC-Tottori) was found in five patients with monoallelic (19%) but not 29 with biallelic variant(s). The most common low-risk allele p.K196E (PS-Tokushima) was found in five with monoallelic (25%) and six with biallelic variant(s) (67%). One exceptional de novo PC variant was found in 32 families with EOT. Only five parents had a history of thromboembolism. Thrombosis concurrently developed in three mother-newborn pairs (two PC deficiency and one AT deficiency). The prospective cohort revealed the outcomes of 35 patients: three deaths with PC deficiency and 20 complication-free survivors. Neurological complications were more frequently found in patients with PC-biallelic variants than those with PC-, PS-, or AT-monoallelic variants (73% vs. 24%, p = .019). CONCLUSIONS: We demonstrate the need for elective screening for EOT targeting PC deficiency in Japan. Early prenatal diagnosis of PC deficiency in mother-infant pairs may prevent perinatal thrombosis in them.


Subject(s)
Antithrombin III Deficiency , Protein C Deficiency , Protein S Deficiency , Thrombophilia , Thrombosis , Infant, Newborn , Female , Pregnancy , Humans , Retrospective Studies , Prospective Studies , Japan/epidemiology , Protein S Deficiency/complications , Protein S Deficiency/diagnosis , Protein S Deficiency/genetics , Thrombophilia/complications , Thrombosis/etiology , Thrombosis/genetics , Protein C Deficiency/genetics , Protein C Deficiency/complications , Protein C/genetics , Anticoagulants , Antithrombin III , Antithrombins
4.
BMC Neurol ; 24(1): 182, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38822265

ABSTRACT

OBJECTIVES: To investigate the risk factors and underlying causes of pregnancy-related cerebral venous thrombosis (PCVT). METHODS: A retrospective cohort of 16 patients diagnosed with CVT during pregnancy and postpartum (within six weeks after delivery) in a comprehensive hospital in China between 2009 and 2022 were carefully reviewed, focusing on demographic, clinical, and etiological characteristics, especially underlying causes. We matched 16 PCVT patients with 64 pregnant and puerperal women without PCVT to explore risk factors and clinical susceptibility to PCVT. RESULTS: PCVT occurred commonly during the first trimester (43.75%) and the puerperium (37.5%). The frequency of anemia, thrombocytosis and thrombocytopenia during pregnancy, dehydration, and pre-pregnancy anemia was significantly higher in women with PCVT than in those without PCVT (P < 0.05). Among the 16 patients, five were diagnosed with antiphospholipid syndrome and one was diagnosed with systemic lupus erythematosus. Three patients had distinct protein S deficiency and one had protein C deficiency. Whole Exome Sequencing (WES) was performed for five patients and revealed likely pathogenic mutations associated with CVT, including heterozygous PROC c.1218G > A (p. Met406Ile), heterozygous PROS1 c.301C > T (p. Arg101Cys), composite heterozygous mutation in the F8 gene (c.144-1259C > T; c.6724G > A (p. Val2242Met)) and homozygous MTHFR c.677C > T (p. Ala222Val). CONCLUSIONS: The occurrence of anemia, thrombocytopenia and thrombocytosis during pregnancy, dehydration and pre-pregnancy anemia suggested a greater susceptibility to PCVT. For confirmed PCVT patients, autoimmune diseases, hereditary thrombophilia, and hematological disorders were common causes. Screening for potential etiologies should be paid more attention, as it has implications for treatment and long-term management.


Subject(s)
Intracranial Thrombosis , Venous Thrombosis , Humans , Female , Pregnancy , Retrospective Studies , Adult , Intracranial Thrombosis/epidemiology , Risk Factors , Venous Thrombosis/epidemiology , China/epidemiology , Young Adult , Pregnancy Complications, Hematologic/epidemiology , Pregnancy Complications, Hematologic/diagnosis , Protein S Deficiency/epidemiology , Protein S Deficiency/complications , Protein S Deficiency/diagnosis , Protein S Deficiency/genetics
5.
Clin Lab ; 70(3)2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38469768

ABSTRACT

BACKGROUND: The recently identified PROS1 mutation Protein S Erlangen c.1904T>C, resulting in amino acid exchange F635S, is associated with severe quantitative protein S (PS) deficiency and clinical thrombosis. It was hypothesized that this deficiency is due to a secretion defect [1]. This report aims to further elucidate the potential secretion defect of PS Erlangen. METHODS: Coding sequences (CDS) of wild type (WT) PROS1 (encoding PS) and mutated PROS1c.1904T>C (encoding PSF635S) were cloned in front of the CDS of green fluorescent protein (GFP), and the respective plasmids were introduced into HEK293T cells. PROS1-GFP and PROS1c.1904T>C-GFP expressing HEK293T cell lines were analyzed by confocal laser scanning microscopy and western blot for cellular proteins and proteins secreted to the growth medium. RESULTS: Western blot analysis revealed a significantly reduced secretion of PSF635S compared to WT PS. This observation was confirmed by the detection of mutant PSF635S-GFP fusion exclusively in the endoplasmic reticulum (ER), while PS-GFP passed through the entire secretory pathway, as indicated by the localization within both the ER and Golgi apparatus. CONCLUSIONS: The Protein S Erlangen mutation results in type I PS deficiency caused by a secretion defect.


Subject(s)
Protein S Deficiency , Protein S , Protein Transport , Thrombosis , Humans , HEK293 Cells , Mutation , Protein C , Protein S Deficiency/genetics , Protein S/genetics , Protein S/metabolism
6.
Clin Lab ; 69(12)2023 Dec 01.
Article in English | MEDLINE | ID: mdl-38084685

ABSTRACT

BACKGROUND: Pulmonary embolism is rare in children, and most of them have high-risk factors, such as antiphospholipid syndrome, intravenous catheterization, fracture bed rest, etc. For children with pulmonary embolism without clear inducement, hereditary thrombophilia should be considered. Genetic protein S deficiency (PSD) is a kind of thrombophilia, which is caused by the mutation of PROS 1 gene, resulting in an increased tendency to thrombosis. METHODS: The diagnosis of the two cases was made after detecting based on Thrombophilia screening and Sanger sequencing in clinical laboratory. RESULTS: Sanger sequencing found that case 2 and case 1 genotypes were the same, case 1 sister and grandfather carried c.200a>c (p.e67a) mutation, and case 1 aunt and grandmother did not carry PROS1 gene mutation. Case 1 received anticoagulation therapy for 3 months, and case 2 also received anticoagulation therapy for 3 months. During the 1 year follow-up, no new thrombotic events and no adverse reactions such as bleeding were observed in both patients. CONCLUSIONS: For children with pulmonary embolism without clear risk factors, PSD should be considered, and protein S activity should be tested before receiving anticoagulant drugs.


Subject(s)
Antiphospholipid Syndrome , Protein S Deficiency , Pulmonary Embolism , Thrombophilia , Thrombosis , Child , Humans , Thrombophilia/diagnosis , Pulmonary Embolism/diagnosis , Pulmonary Embolism/genetics , Anticoagulants/therapeutic use , Thrombosis/diagnosis , Thrombosis/genetics , Antiphospholipid Syndrome/drug therapy , Protein S Deficiency/complications , Protein S Deficiency/diagnosis , Protein S Deficiency/genetics
7.
Zhongguo Yi Xue Ke Xue Yuan Xue Bao ; 45(5): 863-866, 2023 Oct.
Article in Zh | MEDLINE | ID: mdl-37621108

ABSTRACT

Reduced protein S activity is one of the high-risk factors for venous thromboembolism.Hereditary protein S deficiency is an autosomal dominant disorder caused by mutations in the PROS1 gene.We reported a female patient with a mutation of c.292 G>T in exon 3 of the PROS1 gene,which was identified by sequencing.The genealogical analysis revealed that the mutation probably originated from the patient's mother.After searching against the PROS1 gene mutation database and the relevant literature,we confirmed that this mutation was reported for the first time internationally.


Subject(s)
Protein S Deficiency , Protein S , Humans , Female , Protein S/genetics , Protein S Deficiency/genetics , Pedigree , Mutation
8.
J Pediatr Hematol Oncol ; 44(2): e442-e443, 2022 Mar 01.
Article in English | MEDLINE | ID: mdl-34486549

ABSTRACT

Hereditary protein S (PS) deficiency is a rare autosomal dominant disorder with increased risk of venous thromboembolism. The PS Heerlen polymorphism at codon 501 of the PROS1 gene is considered a variant of uncertain significance. It has since been shown that PS Heerlen has a reduced half-life, resulting in reduced levels of free PS. We report a case of an adolescent female with May Thurner syndrome and heterozygous PS Heerlen mutation resulting in a mild PS deficiency and venous thromboembolism. With this nonmodifiable risk factor, the patient received prolonged anticoagulation with strong consideration for lifelong prophylaxis.


Subject(s)
Protein S Deficiency , Venous Thromboembolism , Venous Thrombosis , Adolescent , Child , Female , Humans , Protein S/genetics , Protein S/metabolism , Protein S Deficiency/complications , Protein S Deficiency/genetics , Thrombophilia , Venous Thromboembolism/drug therapy , Venous Thromboembolism/genetics , Venous Thrombosis/drug therapy , Venous Thrombosis/genetics
9.
Acta Haematol ; 144(2): 222-226, 2021.
Article in English | MEDLINE | ID: mdl-32653888

ABSTRACT

Protein S (PS) is an important anticoagulant. Its main function is to act as a non-enzymatical cofactor of activated protein C. PS deficiency is defined as low plasma levels of PS and/or loss of function associated with variable risk of venous thromboembolism (VTE). We report 2 novel variants in the PS gene (PROS1) which are associated with PS deficiency and severe thrombophilic diathesis in 2 patients. Patient 1 suffered from 3 VTE events, including a spontaneous VTE at the age of 19. Patient 2 suffered from 2 provoked VTE events. In both patients decreased plasma levels of PS antigen as well as decreased PS activity were found. Gene sequencing results showed a heterozygous deletion of 8 base pairs (c.938_945delTAAAATTT, p.Leu313Serfs13*) in exon 9 of the PROS1 gene in patient 1 and a missense variant (c.1613C>T, p.Ser538Phe) in patient 2. Due to the clinically proven history of recurrent VTE events in both patients, genetic testing of first-degree relatives is discussed.


Subject(s)
Protein S Deficiency/diagnosis , Protein S/genetics , Venous Thromboembolism/diagnosis , Anticoagulants/therapeutic use , Exons , Factor V/genetics , Female , Gene Deletion , Heterozygote , Homozygote , Humans , Middle Aged , Mutation, Missense , Polymorphism, Single Nucleotide , Protein S Deficiency/complications , Protein S Deficiency/genetics , Venous Thromboembolism/drug therapy , Venous Thromboembolism/etiology
10.
Zhonghua Jie He He Hu Xi Za Zhi ; 44(4): 360-364, 2021 Apr 12.
Article in Zh | MEDLINE | ID: mdl-33832024

ABSTRACT

Objective: To observe the clinical feature of familiar hereditary protein S deficiency, and to explore the related gene mutation. Methods: The blood samples were obtained from the proband and the family memebers(3 generations,6 persons). PROS1 gene of the proband and the family members was analyzed. The 15 exons and flanking sequence of PROS1 gene were analyzed by PCR and DNA sequencing. Results: Five out of 6 family members were diagnosed as having hereditary protein S deficiency. The proband suffered from pulmonary embolism. The others had no obvious thrombotic event. The gene sequencing revealed that the proband carried a c.-168C>T homozygous variant in the promoter of exon 1. His parents, brother and son all carried c.-168C>T heterozygosis variant at the same position. The gene of his wife was a wild type. Conclusion: A gene variant (c.-168C>T) of PROS1 was discovered in this Chinese family. Gene variant of PROS1 may result in protein S deficiency. Patients with protein S deficiency may suffer from vein thrombosis and(or) pulmonary embolism.


Subject(s)
Protein S Deficiency , Consanguinity , Exons , Family , Humans , Male , Mutation , Pedigree , Protein S Deficiency/genetics
11.
Clin Sci (Lond) ; 134(10): 1181-1190, 2020 05 29.
Article in English | MEDLINE | ID: mdl-32426810

ABSTRACT

Autosomal dominant inherited Protein S deficiency (PSD) (MIM 612336) is a rare disorder caused by rare mutations, mainly located in the coding sequence of the structural PROS1 gene, and associated with an increased risk of venous thromboembolism. To identify the molecular defect underlying PSD observed in an extended French pedigree with seven PSD affected members in whom no candidate deleterious PROS1 mutation was detected by Sanger sequencing of PROS1 exons and their flanking intronic regions or via an multiplex ligation-dependent probe amplification (MLPA) approach, a whole genome sequencing strategy was adopted. This led to the identification of a never reported C to T substitution at c.-39 from the natural ATG codon of the PROS1 gene that completely segregates with PSD in the whole family. This substitution ACG→ATG creates a new start codon upstream of the main ATG. We experimentally demonstrated in HeLa cells that the variant generates a novel overlapping upstream open reading frame (uORF) and inhibits the translation of the wild-type PS. This work describes the first example of 5'UTR PROS1 mutation causing PSD through the creation of an uORF, a mutation that is not predicted to be deleterious by standard annotation softwares, and emphasizes the need for better exploration of such type of non-coding variations in clinical genomics.


Subject(s)
5' Untranslated Regions/genetics , Codon, Initiator/genetics , Mutation/genetics , Protein Biosynthesis , Protein S Deficiency/genetics , Protein S/genetics , Base Sequence , Female , HeLa Cells , Humans , Male , Pedigree , Young Adult
12.
J Clin Lab Anal ; 34(4): e23111, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31743498

ABSTRACT

BACKGROUND: Thrombophilia is becoming a more frequently reported disorder these years. Hereditary protein S deficiency is one of the anticoagulant deficiencies that eventually results in thrombophilia. CASE PRESENTATION: A 24-year-old male patient was suffering from unexplained thrombosis for the second time with a family history of deep venous thrombosis. Screening tests for anticoagulant proteins found the activity of protein S markedly lowered (5.0%). The patient was discharged after anticoagulation treatment. Four years later, the review still showed the activity of protein S in his plasma decreased (16.0%). Molecular genetic analysis revealed him homozygous for a missense mutation, c.664G>A, in the exon7 of PROS1. The mutation discovered here is the first mutation affecting the codon 222 of PROS1. This mutation results in the replacement of the glycine at the codon 222 of protein S with arginine, leading to a reduction of protein S function. CONCLUSIONS: The finding of this mutation may help with the understanding of the mechanism of protein S deficiency, especially in the Chinese population.


Subject(s)
Mutation, Missense , Protein S Deficiency/genetics , Protein S/genetics , Pulmonary Embolism/genetics , Amino Acid Substitution , Female , Heterozygote , Humans , Male , Pedigree , Protein S Deficiency/complications , Pulmonary Embolism/blood , Pulmonary Embolism/complications , Young Adult
13.
Rinsho Ketsueki ; 60(3): 171-183, 2019.
Article in Japanese | MEDLINE | ID: mdl-31068512

ABSTRACT

Protein S (PS) gene (PROS1) is found on chromosome 3 (3q11.1). To date, the reported detection rate of causative gene mutations in patients suspected of PS deficiency is only approximately 50%. To improve the detection rate of causative mutations, an exhaustive analysis of PROS1 was attempted using the next-generation sequencing method (NGS) to analyze the entire nucleotide sequence of PROS1 without analyzing those affected by pseudogenes. A total of 10 different mutations (three males and six females (52.9%) out of 17 patients (3 males and 14 females) with clinical PS deficiency were identified in this study. Remarkable improvements in the detection rate of causative mutations could not be obtained even with NGS analysis. These results suggested that the rate of diagnosis did not improve even after performing an exhaustive genetic analysis in patients clinically diagnosed with low PS antigen level and/or low PS activity. Although no reports were found on the gender gap in the rate of gene diagnosis for PS deficiency, the fluctuation of estrogen levels especially in women might cause a lower rate of diagnosis.


Subject(s)
Blood Proteins/genetics , High-Throughput Nucleotide Sequencing , Mutation , Protein S Deficiency/genetics , DNA Mutational Analysis , Female , Humans , Male , Protein S
14.
Eur Arch Otorhinolaryngol ; 275(8): 1971-1977, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29948264

ABSTRACT

PURPOSE: To describe the prevalent clinical, laboratory, and radiological features of otogenic lateral sinus thrombosis (OLST) in children; to identify clinical predictors of outcome; to propose a management algorithm derived from experience. METHODS: A retrospective review was conducted of the clinical records of patients with OLST, treated in a single tertiary care referral center for pediatric disease from 2006 to 2017. The inclusion criteria were pediatric age (0-16 years) and OLST diagnosis confirmed by a pre- and post-contrast CT or venography-MRI scan. Primary outcome measures were early (1-2 months) and late (6 months) sinus recanalization assessed by means of neuroimaging. RESULTS: Twenty-five patients (8 females and 17 males; mean age = 6 ± 3 years) were included. A genetic abnormality associated with thrombophilia was found in 24 (96%) patients. At diagnosis, anticoagulant treatment with low-molecular-weight heparin (LMWH) was started in all subjects, while surgical treatment (mastoidectomy and tympanostomy tube insertion) was performed in 16/25 (64%) patients. Follow-up neuroimaging showed lateral sinus recanalization in 12/25 (48%) patients after 1-2 months and in 17/25 (68%) after 6 months. At multivariate logistic regression analysis, no significant predictors of the early and late neuroradiological outcome were found. CONCLUSIONS: All children with OLST should be screened for thrombophilia to decide on treatment duration and to assess the need for future antithrombotic prophylaxis. Immediately after diagnosis, anticoagulant treatment with LMWH should be started according to the international guidelines. Instead, our experience suggests that surgical treatment should not be indicated in all patients, but decided on a case-to-case basis.


Subject(s)
Decision Support Techniques , Lateral Sinus Thrombosis/diagnosis , Lateral Sinus Thrombosis/therapy , Adolescent , Anticoagulants , Child , Child, Preschool , Consciousness Disorders/etiology , Cranial Nerve Diseases/etiology , Factor V/genetics , Female , Headache/etiology , Heparin, Low-Molecular-Weight , Humans , Infant , Male , Mastoidectomy , Mastoiditis/complications , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Middle Ear Ventilation , Mutation , Otitis Media/complications , Protein S Deficiency/genetics , Retrospective Studies , Thrombophilia/diagnosis , Thrombophilia/genetics
16.
Pediatr Blood Cancer ; 64(5)2017 05.
Article in English | MEDLINE | ID: mdl-27748013

ABSTRACT

The prevalence of protein S (PS) deficiency in Asian patients with venous thromboembolism is around 8-30%, higher than that in Caucasian populations. The present study reports the genotypes (including one novel mutation) and phenotypes of children with PS deficiency at a tertiary care institute. A total of six patients were included, three with arterial ischemic stroke, two with cerebral venous sinus thrombosis, and one with deep vein thrombosis. PS mutations were identified in four patients: p.R355C, p.G336D, p.E67A, and p.N188KfsX9. p.N188KfsX9 is a novel mutation with less than 20% PS activity noted in heterozygotes.


Subject(s)
Protein S Deficiency/epidemiology , Protein S Deficiency/genetics , Sinus Thrombosis, Intracranial/genetics , Stroke/genetics , Thromboembolism/epidemiology , Thromboembolism/genetics , Venous Thrombosis/genetics , Adolescent , Child , Child, Preschool , Female , Genotype , Humans , Male , Phenotype , Polymerase Chain Reaction , Protein S Deficiency/pathology , Retrospective Studies , Thailand/epidemiology
17.
Adv Exp Med Biol ; 906: 253-272, 2017.
Article in English | MEDLINE | ID: mdl-27638626

ABSTRACT

Genetic risk factors predispose to thrombophilia and play the most important etiopathogenic role in venous thromboembolism (VTE) in people younger than 50 years old. At least one inherited risk factor could be found in about half of the cases with a first episode of idiopathic VTE.Roughly, genetic risk factors are classified into two main categories: loss of function mutations (such as deficiencies of antithrombin, protein C, protein S) and gain of function mutations, (such as prothrombin mutation G20210A, factor V Leiden). A revolutionary contribution to the genetic background of VTE was brought by the achievements of the genome-wide association studies which analyze the association of a huge number of polymorphisms in large sample size.Hereditary thrombophilia testing should be done only in selected cases. The detection of hereditary thrombophilia has impact on the management of the anticoagulation in children with purpura fulminans, pregnant women at risk of VTE and may be useful in the assessment of the risk for recurrent thrombosis in patients presenting an episode of VTE at a young age (<40 years) and in cases with positive family history regarding thrombosis.


Subject(s)
Antithrombin III Deficiency/genetics , Protein C Deficiency/genetics , Protein S Deficiency/genetics , Thrombophilia/genetics , Venous Thromboembolism/genetics , Antithrombin III Deficiency/complications , Antithrombin III Deficiency/diagnosis , Antithrombin III Deficiency/epidemiology , Europe/epidemiology , Factor V/genetics , Gene Expression , Genetic Predisposition to Disease , Humans , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Mutation , Prevalence , Protein C Deficiency/complications , Protein C Deficiency/diagnosis , Protein C Deficiency/epidemiology , Protein S Deficiency/complications , Protein S Deficiency/diagnosis , Protein S Deficiency/epidemiology , Prothrombin/genetics , Risk Factors , Thrombophilia/complications , Thrombophilia/diagnosis , Thrombophilia/epidemiology , Venous Thromboembolism/diagnosis , Venous Thromboembolism/epidemiology , Venous Thromboembolism/etiology
18.
Vasa ; 46(3): 227-230, 2017 May.
Article in English | MEDLINE | ID: mdl-28102772
19.
Eur J Clin Invest ; 51(5): e13546, 2021 May.
Article in English | MEDLINE | ID: mdl-33738814
20.
Pediatr Res ; 79(1-1): 81-6, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26372516

ABSTRACT

BACKGROUND: The early diagnosis of inherited thrombophilia in children is challenging because of the rarity and hemostatic maturation. METHODS: We explored protein C (PC), protein S (PS), and antithrombin (AT) deficiencies in 306 thromboembolic patients aged ≤20 y using the screening of plasma activity and genetic analysis. RESULTS: Reduced activities were determined in 122 patients (40%). Low PC patients were most frequently found in the lowest age group (0-2 y, 45%), while low PS or low AT patients were found in the highest age group (16-20 y; PS: 30% and AT: 20%). Genetic study was completed in 62 patients having no other causes of thromboembolism. Mutations were determined in 18 patients (8 PC, 8 PS, and 2 AT genes). Six of eight patients with PC gene mutation were found in age 0-2 y (75%), while six of eight patients with PS gene mutation were in 7-20 y. Two AT gene-mutated patients were older than 4 y. Four PC-deficient and two PS-deficient patients carried compound heterozygous mutations. All but one PC gene-mutated patient suffered from intracranial thromboembolism, while PS/AT gene-mutated patients mostly developed extracranial venous thromboembolism. CONCLUSION: Stroke in low PC infants and deep vein thrombosis in low PS/AT school age children could be targeted for genetic screening of pediatric thrombophilias.


Subject(s)
Activated Protein C Resistance/epidemiology , Antithrombin III Deficiency/epidemiology , Protein C Deficiency/epidemiology , Protein S Deficiency/epidemiology , Thromboembolism/etiology , Thrombophilia/genetics , Activated Protein C Resistance/blood , Activated Protein C Resistance/diagnosis , Activated Protein C Resistance/genetics , Adolescent , Age of Onset , Antithrombin III/analysis , Antithrombin III/genetics , Antithrombin III Deficiency/blood , Antithrombin III Deficiency/diagnosis , Antithrombin III Deficiency/genetics , Cerebrovascular Disorders/epidemiology , Cerebrovascular Disorders/etiology , Child , Child, Preschool , DNA Mutational Analysis , Factor V/genetics , Female , Genotype , Humans , Infant , Japan/epidemiology , Male , Promoter Regions, Genetic/genetics , Protein C/analysis , Protein C/genetics , Protein C Deficiency/blood , Protein C Deficiency/diagnosis , Protein C Deficiency/genetics , Protein S/analysis , Protein S/genetics , Protein S Deficiency/blood , Protein S Deficiency/diagnosis , Protein S Deficiency/genetics , Prothrombin/genetics , Thromboembolism/epidemiology , Thrombophilia/blood , Thrombophilia/diagnosis , Thrombophilia/epidemiology
SELECTION OF CITATIONS
SEARCH DETAIL