Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 378
Filtrar
1.
BMC Neurol ; 24(1): 182, 2024 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-38822265

RESUMEN

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.


Asunto(s)
Trombosis Intracraneal , Trombosis de la Vena , Humanos , Femenino , Embarazo , Estudios Retrospectivos , Adulto , Trombosis Intracraneal/epidemiología , Factores de Riesgo , Trombosis de la Vena/epidemiología , China/epidemiología , Adulto Joven , Complicaciones Hematológicas del Embarazo/epidemiología , Complicaciones Hematológicas del Embarazo/diagnóstico , Deficiencia de Proteína S/epidemiología , Deficiencia de Proteína S/complicaciones , Deficiencia de Proteína S/diagnóstico , Deficiencia de Proteína S/genética
2.
Clin Lab ; 70(3)2024 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-38469768

RESUMEN

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.


Asunto(s)
Deficiencia de Proteína S , Proteína S , Transporte de Proteínas , Trombosis , Humanos , Células HEK293 , Mutación , Proteína C , Deficiencia de Proteína S/genética , Proteína S/genética , Proteína S/metabolismo
3.
J Med Case Rep ; 18(1): 77, 2024 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-38414076

RESUMEN

BACKGROUND: Lower limb deep vein thrombosis (DVT) concurrent with pulmonary embolism (PE) is perilous, particularly in the elderly, exhibiting heterogeneity with thrombophilia mutations. Tailored treatment is essential, yet sudden deaths complicate causative factor elucidation. This report emphasizes genetic testing necessity in PE patients with thrombophilia indicators, facilitating cause identification, personalized treatment guidance, and family education. CASE PRESENTATION: This study details a 75-year-old Chinese woman with DVT and PE, where genetic testing identified thrombophilia, guiding personalized treatment decisions. RESULTS: Upon admission, the patient, after over 10 days of bed rest, presented chest tightness, shortness of breath, and unilateral leg swelling. Diagnostic measures revealed DVT and a substantial PE. Genetic testing identified a PROS1 gene C200A>C mutation, reducing protein S activity. Following 2 weeks of anticoagulation and inferior vena cava filter insertion, the patient, discharged, initiated lifelong anticoagulant therapy. A 1-year follow-up showed no recurrent thrombotic events. Family members carrying the mutation received informed and educational interventions. CONCLUSION: Genetic testing for thrombophilic predisposition post-PE is crucial, elucidating etiology, guiding individualized treatment, and playing a pivotal role in family education.


Asunto(s)
Deficiencia de Proteína S , Embolia Pulmonar , Trombosis , Filtros de Vena Cava , Trombosis de la Vena , Femenino , Humanos , Anciano , Deficiencia de Proteína S/complicaciones , Deficiencia de Proteína S/genética , Embolia Pulmonar/genética , Embolia Pulmonar/complicaciones , Trombosis de la Vena/genética , Trombosis de la Vena/complicaciones , Trombosis/complicaciones , Mutación , Filtros de Vena Cava/efectos adversos
4.
Ann Hematol ; 103(2): 653-662, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38175252

RESUMEN

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.


Asunto(s)
Proteínas Sanguíneas , Deficiencia de Proteína S , Humanos , Proteínas Sanguíneas/genética , Deficiencia de Proteína S/diagnóstico , Deficiencia de Proteína S/genética , Trombina , Mutación , China , Linaje , Proteína S/genética
5.
Pediatr Blood Cancer ; 71(3): e30824, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38155150

RESUMEN

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.


Asunto(s)
Deficiencia de Antitrombina III , Deficiencia de Proteína C , Deficiencia de Proteína S , Trombofilia , Trombosis , Recién Nacido , Femenino , Embarazo , Humanos , Estudios Retrospectivos , Estudios Prospectivos , Japón/epidemiología , Deficiencia de Proteína S/complicaciones , Deficiencia de Proteína S/diagnóstico , Deficiencia de Proteína S/genética , Trombofilia/complicaciones , Trombosis/etiología , Trombosis/genética , Deficiencia de Proteína C/genética , Deficiencia de Proteína C/complicaciones , Proteína C/genética , Anticoagulantes , Antitrombina III , Antitrombinas
6.
Clin Lab ; 69(12)2023 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-38084685

RESUMEN

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.


Asunto(s)
Síndrome Antifosfolípido , Deficiencia de Proteína S , Embolia Pulmonar , Trombofilia , Trombosis , Niño , Humanos , Trombofilia/diagnóstico , Embolia Pulmonar/diagnóstico , Embolia Pulmonar/genética , Anticoagulantes/uso terapéutico , Trombosis/diagnóstico , Trombosis/genética , Síndrome Antifosfolípido/tratamiento farmacológico , Deficiencia de Proteína S/complicaciones , Deficiencia de Proteína S/diagnóstico , Deficiencia de Proteína S/genética
7.
Zhongguo Yi Xue Ke Xue Yuan Xue Bao ; 45(5): 863-866, 2023 Oct.
Artículo en Chino | MEDLINE | ID: mdl-37621108

RESUMEN

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.


Asunto(s)
Deficiencia de Proteína S , Proteína S , Humanos , Femenino , Proteína S/genética , Deficiencia de Proteína S/genética , Linaje , Mutación
8.
Clin Res Hepatol Gastroenterol ; 47(6): 102141, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37207893

RESUMEN

BACKGROUND: Protein S (PS) is a vitamin K-dependent plasma glycoprotein, and the deficiency of PS increases the risk of venous thromboembolism (VTE). PS deficiency has been found in 1.5-7% of selected groups of thrombophilic patients. However, the reported PS deficiency patients with portal vein thrombosis are scarce. CASE REPORT AND RESULTS: Our case described a 60-year-old male patient presented portal vein thrombosis with protein S deficiency. Imaging findings of the patient revealed extensive thrombosis involving the portal vein and superior mesenteric vein. His medical history revealed lower extremity venous thrombosis 10 years ago. The level of PS activity was greatly reduced (14%, reference: 55-130%). Acquired thrombophilia caused by antiphospholipid syndrome, hyperhomocysteinemia, or malignancy were excluded. Whole exome sequencing revealed a heterozygous missense variation c.1574C>T, p.Ala525Val in the PROS1 gene. The in-silico analysis of the variant was performed by SIFT and PolyPhen-2. The results showed that the variant is a pathogenic and likely pathogenic variation respectively (SIFT, -3.404; PolyPhen-2, 0.892), the amino acid substitution A525V is presumed to result in unstable PS protein which is degraded intracellularly. Mutation site of the proband and his family members was validated by Sanger sequencing. CONCLUSION: According to the clinical manifestation, imaging findings, protein S level, and the genetic results, a diagnosis of portal vein thrombosis with PS deficiency was made. To the best of our knowledge, our case is the second reported PS deficiency patient caused by PROS1 c.1574C>T, p.Ala525Val variant in Asia, and the case is also the only reported case with PROS1 c.1574C>T, p.Ala525Val variant presents portal vein thrombosis.


Asunto(s)
Deficiencia de Proteína S , Trombosis , Trombosis de la Vena , Masculino , Humanos , Persona de Mediana Edad , Deficiencia de Proteína S/complicaciones , Deficiencia de Proteína S/genética , Proteínas Sanguíneas/genética , Proteínas Sanguíneas/metabolismo , Vena Porta , Trombosis de la Vena/complicaciones , Trombosis de la Vena/genética , Trombosis/complicaciones , Proteína S/genética
9.
Zhonghua Xue Ye Xue Za Zhi ; 43(1): 48-53, 2022 Jan 14.
Artículo en Chino | MEDLINE | ID: mdl-35231993

RESUMEN

Objective: To analyze the clinical manifestations and molecular pathogenesis of 18 patients with inherited protein S (PS) deficiency. Methods: Eighteen patients with inherited PS deficiency who were admitted to the Institute of Hematology & Blood Diseases Hospital from June 2016 to February 2019 were analyzed: activity of protein C (PC) and antithrombin (AT) , PS activity were measured for phenotype diagnosis; high throughput sequencing (HTS) was used for screening of coagulation disease-related genes; Sanger sequencing was used to confirm candidate variants; Swiss-model was used for three-dimensional structure analysis. Results: The PS:C of 18 patients ranged from 12.5 to 48.2 U/dL. Among them, 16 cases developed deep vein thrombosis, including 2 cases each with mesenteric vein thrombosis and cerebral infarction, and 1 case each with pulmonary embolism and deep vein thrombosis during pregnancy. A total of 16 PROS1 gene mutations were detected, and 5 nonsense mutations (c.134_162del/p.Leu45*, c.847G>T/p.Glu283*, c.995_996delAT/p.Tyr332*, c.1359G> A/p.Trp453*, c.1474C>T/p.Gln492*) , 2 frameshift mutations (c.1460delG/p.Gla487Valfs*9 and c.1747_1750delAATC/p.Asn583Wfs*9) and 1 large fragment deletion (exon9 deletion) were reported for the first time. In addition, the PS:C of the deep vein thrombosis during pregnancy case was 55.2 U/dL carrying PROC gene c.565C>T/p.Arg189Trp mutation. Conclusion: The newly discovered gene mutations enriched the PROS1 gene mutation spectrum which associated with inherited PS deficiency.


Asunto(s)
Deficiencia de Proteína S , Antitrombina III/genética , Femenino , Pruebas Genéticas , Humanos , Mutación , Embarazo , Proteína C/genética , Proteína S/genética , Deficiencia de Proteína S/diagnóstico , Deficiencia de Proteína S/genética
10.
Zhonghua Er Ke Za Zhi ; 60(2): 134-138, 2022 Feb 02.
Artículo en Chino | MEDLINE | ID: mdl-35090231

RESUMEN

Objective: To explore the clinical characteristics and genotype of PROS1 gene related hereditary protein S deficiency (PSD) with the onset of pulmonary embolism in children. Methods: A family with pulmonary embolism was diagnosed as hereditary PSD in the Department of Pediatrics of Peking University First Hospital in November 2020, and the clinical data, including clinical manifestations, laboratory tests, imaging and genetic results, were collected for a retrospective research. The family members were also screened for protein S activity and PROS1 gene mutations. A literature search with "PROS1" "protein S deficiency" "homozygous" and "complex heterozygous" as key words was conducted at PubMed, China National Knowledge Infrastructure, and Wanfang Data Knowledge Service Platform (up to October 2021). Case reports of patients with PROS1 gene homozygous or complex heterozygous variants and related clinical features, protein S activity, and genotype were reviewed and analyzed. Results: The proband, a 14-year-old girl, was admitted to the hospital for a 9-day history of coughing and a 4-day history of chest pain in November 2020. After admission, laboratory tests showed that D-dimer was 8.38 mg/L (reference:<0.24 mg/L). An urgent CT pulmonary angiography confirmed bilateral pulmonary embolism and right lower pulmonary infarction, while an ultrasonography showed deep vein thrombosis in her left leg. Further examination revealed that protein S activity was less than 10%. The proband's second sister, a 12-year-old girl, was admitted to the hospital in December 2020. Her protein S activity was 8% and an ultrasonography showed deep vein thrombosis in her right leg. The protein S activity of the proband's father and mother were 36% and 26%, respectively. Trio-whole-exome sequencing detected compound heterozygous PROS1 gene variants (c.-168C>T and c.200A>C (p.E67A)) for the proband and her second sister, that were inherited from her father and mother, respectively. The proband's third sister's protein S activity was 28%; she and the proband's grandfather both carried c.200A>C (p.E67A) variants. The proband and her younger sister were treated with rivaroxaban and responded well during the 3-month follow-up. A total of 1 Chinese report in literature and 18 English literature were retrieved and 14 patients with protein S deficiency caused by homozygous or complex heterozygous variants of PROS1 gene were enrolled, including 8 male and 6 female patients. The ages ranged from 4 days to 35 years. Three patients experienced fulminant purpura or severe intracranial hemorrhage in early neonatal-period, while the remaining 11 patients developed venous thromboembolism in adolescence. Protein S activity was examined in 11 patients, and all showed less than 10% of activity. Missense variants was the most common type of gene variants. Conclusions: For children with pulmonary embolism, if there are no clear risk factors for thrombosis, hereditary protein S deficiency should be considered, and protein S activity should be examined before oral anticoagulant drugs. If protein S activity is less than 10%, protein S deficiency caused by homozygous or complex heterozygous variants should be considered.


Asunto(s)
Deficiencia de Proteína S , Embolia Pulmonar , Adolescente , Niño , Preescolar , Femenino , Humanos , Recién Nacido , Masculino , Linaje , Proteína S/genética , Deficiencia de Proteína S/genética , Embolia Pulmonar/diagnóstico , Embolia Pulmonar/genética , Estudios Retrospectivos
11.
Blood Coagul Fibrinolysis ; 33(4): 224-227, 2022 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-34939974

RESUMEN

The members of a Caucasian family were genetically analyzed on suspicion of hereditary protein S deficiency. A novel mutation, c.1904T>C, associated with severe quantitative protein S deficiency was found. The novel PROS1 mutation was identified by sequencing of the PROS1 gene coding sequence. The identified c.1904T>C point mutation results in p.Phe635Ser amino acid exchange, which is located in the Laminin G-like 2 domain of protein S. Computational analysis indicates that this amino acid exchange affects the correct folding of the protein S antigen. Furthermore, this mutation is located in a region of the Laminin G-like 2 domain where changes in the amino acid sequence often result in decreased secretion. We postulate that the novel p.Phe635Ser mutation might lead to an incorrect folding, and thus, to a strongly impaired secretion of this protein S variant. We named this novel variant protein.


Asunto(s)
Deficiencia de Proteína S , Aminoácidos/genética , Humanos , Laminina/genética , Mutación , Linaje , Proteína S/genética , Deficiencia de Proteína S/genética
13.
J Pediatr Hematol Oncol ; 44(2): e442-e443, 2022 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-34486549

RESUMEN

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.


Asunto(s)
Deficiencia de Proteína S , Tromboembolia Venosa , Trombosis de la Vena , Adolescente , Niño , Femenino , Humanos , Proteína S/genética , Proteína S/metabolismo , Deficiencia de Proteína S/complicaciones , Deficiencia de Proteína S/genética , Trombofilia , Tromboembolia Venosa/tratamiento farmacológico , Tromboembolia Venosa/genética , Trombosis de la Vena/tratamiento farmacológico , Trombosis de la Vena/genética
14.
BMJ Case Rep ; 14(10)2021 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-34667051

RESUMEN

Inherited thrombophilic disorders are well-established predisposing factors for venous thromboembolism, but their role in arterial ischaemic stroke is uncertain. The exact mechanism of arterial thrombosis in thrombophilias remains elusive. Herein, we report a case of a 30-year-old woman who was admitted to our facility with sudden-onset right-sided ptosis and ophthalmoplegia. Detailed clinical features, neuroimaging and laboratory evaluation clinched the diagnosis of ischaemic stroke in midbrain due to microvascular obstruction associated with isolated protein S deficiency. She was treated with oral anticoagulant (warfarin) and physiotherapy; without any improvement of her symptoms at 2 months of follow-up. A high index of clinical suspicion is needed in any case of young ischaemic stroke in absence of common cardiac and vascular risk factors, to recognise the presence of inherited thrombophilia.


Asunto(s)
Isquemia Encefálica , Deficiencia de Proteína S , Accidente Cerebrovascular , Trombofilia , Adulto , Femenino , Humanos , Infarto , Mesencéfalo , Deficiencia de Proteína S/complicaciones , Deficiencia de Proteína S/genética , Factores de Riesgo , Accidente Cerebrovascular/diagnóstico , Accidente Cerebrovascular/etiología , Trombofilia/complicaciones , Trombofilia/genética
16.
Zhonghua Jie He He Hu Xi Za Zhi ; 44(4): 360-364, 2021 Apr 12.
Artículo en Chino | MEDLINE | ID: mdl-33832024

RESUMEN

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.


Asunto(s)
Deficiencia de Proteína S , Consanguinidad , Exones , Familia , Humanos , Masculino , Mutación , Linaje , Deficiencia de Proteína S/genética
17.
Eur J Clin Invest ; 51(5): e13546, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33738814
18.
Intern Emerg Med ; 16(4): 997-1003, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33619677

RESUMEN

BACKGROUND: Whether the carriership of inherited antithrombin (AT), protein C (PC), and protein S (PS) deficiency increases the risk of arterial thromboembolic events (ATE) is controversial. This information has the potential to inform the management of family members of probands with inherited deficiency of natural anticoagulants. PATIENTS/METHODS: We conducted a large prospective family cohort study in 640 subjects (of whom 341 carriers and 299 non-carriers) belonging to 86 families with inherited deficiency of AT, PC, or PS. RESULTS: A total of 4240 and 3810 patient-years were available for carriers and non-carriers, respectively. Risk factors for atherosclerosis were similarly distributed in the two groups. Of the 26 ATE that were recorded, 19 occurred in carriers (5.6%), as compared to 7 in non-carriers (2.3%) [p = 0.07]. After adjusting for confounders, the hazard ratio (HR) for ATE was 4.9 (95% CI 1.5-16.3) in carriers as compared to non-carriers. CONCLUSIONS: Among family members of probands with an inherited deficiency of natural anticoagulants, carriers exhibit a risk of ATE that is almost five times higher than in non-carriers.


Asunto(s)
Deficiencia de Antitrombina III/genética , Deficiencia de Proteína C/genética , Deficiencia de Proteína S/genética , Trombosis/genética , Adulto , Femenino , Predisposición Genética a la Enfermedad , Humanos , Italia/epidemiología , Masculino , Linaje , Estudios Prospectivos , Factores de Riesgo , Trombosis/epidemiología
19.
Eur Rev Med Pharmacol Sci ; 25(1): 353-361, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33506924

RESUMEN

OBJECTIVE: To explore the clinical and prognostic features of CVT caused by PROS1 gene mutations and to provide clinical experience for new oral anticoagulants, such as rivaroxaban, in the treatment of CVT with a high risk of thrombosis. PATIENTS AND METHODS: The CVT patient's clinical symptoms were described, and the brain imaging and blood coagulation tests were performed to confirm the diagnosis of CVT. The patient's family members were recruited to receive blood coagulation tests and ultrasonic examination of lower limb vessels. Genetic analysis on the pedigree was carried out to identify the responsible gene for PS deficiency. We followed-up with this patient for 24 months to evaluate the clinical outcomes, laboratory results and imaging performances of CVT. RESULTS: The patient presented with typical CVT symptoms, including headache and epilepsy. Brain CT showed hemorrhage in the bilateral frontal lobe and left occipital lobe, while MRV demonstrated that thrombus had occurred. It was reviewed that the patient and his mother had a history of bilateral leg deep vein thrombosis. Gene tests revealed that the patient and two family members carried a heterozygous mutation of PROS1 (c.751_752delAT, p.M251Vfs*17). During 24 months of follow-up study, the patient was treated with rivaroxaban continuously and recovered well, supported by an mRS score that remained below 2. Blood coagulation tests were within normal limits, and MRV revealed partial recanalization of the cerebral venous sinus. CONCLUSIONS: The frame shift mutation in the PROS1 gene (c.751_752delAT) may greatly affect the function of protein S and lead to a severe phenotype of CVT. Rivaroxaban showed a satisfying therapeutic effect in this CVT patient with hereditary thrombophilia.


Asunto(s)
Anticoagulantes/farmacología , Deficiencia de Proteína S/tratamiento farmacológico , Proteína S/genética , Rivaroxabán/farmacología , Trombofilia/tratamiento farmacológico , Trombosis de la Vena/tratamiento farmacológico , Administración Oral , Adulto , Anticoagulantes/administración & dosificación , Coagulación Sanguínea/efectos de los fármacos , Estudios de Seguimiento , Humanos , Masculino , Mutación , Linaje , Proteína S/metabolismo , Deficiencia de Proteína S/genética , Deficiencia de Proteína S/metabolismo , Rivaroxabán/administración & dosificación , Trombofilia/genética , Trombofilia/metabolismo , Trombosis de la Vena/genética , Trombosis de la Vena/metabolismo
20.
J Thromb Haemost ; 19(1): 68-74, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33405382

RESUMEN

Hereditary deficiencies of protein S (PS) increase the risk of venous thrombosis; however, assessing the plasma levels of PS can be difficult because of its complex physiological interactions in plasma, sample-related preanalytical variables, and numerous acquired disease processes. Reliable laboratory assays are essential for accurate evaluation of PS when diagnosing a congenital deficiency based on the plasma phenotype alone. This report presents the current evidence-based recommendations for clinical PS assays as well as when to test for PS abnormalities.


Asunto(s)
Deficiencia de Proteína S , Trombosis de la Vena , Coagulación Sanguínea , Técnicas de Laboratorio Clínico , Comunicación , Humanos , Proteína S , Deficiencia de Proteína S/diagnóstico , Deficiencia de Proteína S/genética , Trombosis de la Vena/diagnóstico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...