Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.381
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
J Proteome Res ; 23(10): 4538-4552, 2024 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-39265992

RESUMO

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.


Assuntos
Metaboloma , Deficiência de Proteína S , Proteoma , Humanos , Proteoma/metabolismo , Proteoma/análise , Deficiência de Proteína S/genética , Deficiência de Proteína S/sangue , Feminino , Masculino , Adulto , Proteína S/metabolismo , Proteína S/genética , Proteômica/métodos , Coagulação Sanguínea , Cromatografia Gasosa-Espectrometria de Massas , Metabolômica/métodos , Cromatografia Líquida
2.
Ann Hematol ; 103(2): 653-662, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38175252

RESUMO

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.


Assuntos
Proteínas Sanguíneas , Deficiência de Proteína S , Humanos , Proteínas Sanguíneas/genética , Deficiência de Proteína S/diagnóstico , Deficiência de Proteína S/genética , Trombina , Mutação , China , Linhagem , Proteína S/genética
3.
Pediatr Blood Cancer ; 71(3): e30824, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38155150

RESUMO

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.


Assuntos
Deficiência de Antitrombina III , Deficiência de Proteína C , Deficiência de Proteína S , Trombofilia , Trombose , Recém-Nascido , Feminino , Gravidez , Humanos , Estudos Retrospectivos , Estudos Prospectivos , Japão/epidemiologia , Deficiência de Proteína S/complicações , Deficiência de Proteína S/diagnóstico , Deficiência de Proteína S/genética , Trombofilia/complicações , Trombose/etiologia , Trombose/genética , Deficiência de Proteína C/genética , Deficiência de Proteína C/complicações , Proteína C/genética , Anticoagulantes , Antitrombina III , Antitrombinas
4.
BMC Neurol ; 24(1): 182, 2024 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-38822265

RESUMO

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.


Assuntos
Trombose Intracraniana , Trombose Venosa , Humanos , Feminino , Gravidez , Estudos Retrospectivos , Adulto , Trombose Intracraniana/epidemiologia , Fatores de Risco , Trombose Venosa/epidemiologia , China/epidemiologia , Adulto Jovem , Complicações Hematológicas na Gravidez/epidemiologia , Complicações Hematológicas na Gravidez/diagnóstico , Deficiência de Proteína S/epidemiologia , Deficiência de Proteína S/complicações , Deficiência de Proteína S/diagnóstico , Deficiência de Proteína S/genética
5.
J Thromb Thrombolysis ; 57(4): 710-720, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38491267

RESUMO

While direct oral anticoagulants (DOACs) are frequently used to treat venous thromboembolism (VTE), the outcomes of patients with inherited thrombophilia (IT) receiving DOACs for VTE remain understudied. We used data from the international RIETE registry to compare the rates of VTE recurrences, major bleeding, and mortality during anticoagulant treatment in VTE patients with and without IT, grouped by the use of DOACs or standard anticoagulant therapy. Among 103,818 enrolled patients, 21,089 (20.3%) were tested for IT, of whom 8422 (39.9%) tested positive: Protein C deficiency 294, Protein S deficiency 726, Antithrombin deficiency 240, Factor V Leiden 2248, Prothrombin gene mutation 1434, combined IT 3480. Overall, 14,189 RIETE patients (6.2% with IT) received DOACs, and 89,629 standard anticoagulation (8.4% with IT), mostly with heparins followed by vitamin K antagonists. Proportions of patients receiving DOACs did not differ between IT-positive and IT-negative patients. Rates of VTE recurrence on anticoagulant treatment were highest in patients with AT deficiency (P < 0.01). Rates of on-treatment major bleeding and all-cause mortality were lowest among patients with Factor V Leiden (FVL) or PT G20210A mutations, compared with patients who tested negative. Patients with IT who received DOACs had lower rates of major bleeding than those receiving standard anticoagulation. Excluding FVL and Protein S deficiency, patients with IT had lower rates of VTE recurrence with DOACs than with standard anticoagulation. DOACs are equally safe and effective in VTE patients with IT, with lower bleeding rates than those on standard anticoagulation.


Assuntos
Deficiência de Proteína S , Trombofilia , Tromboembolia Venosa , Humanos , Tromboembolia Venosa/tratamento farmacológico , Anticoagulantes/uso terapêutico , Trombofilia/genética , Hemorragia/induzido quimicamente , Sistema de Registros , Administração Oral
6.
Clin Lab ; 70(3)2024 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-38469768

RESUMO

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.


Assuntos
Deficiência de Proteína S , Proteína S , Transporte Proteico , Trombose , Humanos , Células HEK293 , Mutação , Proteína C , Deficiência de Proteína S/genética , Proteína S/genética , Proteína S/metabolismo
7.
Clin Lab ; 69(12)2023 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-38084685

RESUMO

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.


Assuntos
Síndrome Antifosfolipídica , Deficiência de Proteína S , Embolia Pulmonar , Trombofilia , Trombose , Criança , Humanos , Trombofilia/diagnóstico , Embolia Pulmonar/diagnóstico , Embolia Pulmonar/genética , Anticoagulantes/uso terapêutico , Trombose/diagnóstico , Trombose/genética , Síndrome Antifosfolipídica/tratamento farmacológico , Deficiência de Proteína S/complicações , Deficiência de Proteína S/diagnóstico , Deficiência de Proteína S/genética
8.
J Stroke Cerebrovasc Dis ; 32(7): 107151, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37116445

RESUMO

Protein-losing gastroenteropathies are characterized by an excessive loss of serum proteins into the gastrointestinal tract, resulting in hypoalbuminemia. Some rare cases are complicated with ischemic stroke. We report a 24-year-old woman who developed acute dysarthria and right hemiplegia 4 months after delivering her first baby by cesarean section. Diffusion-weighted magnetic resonance imaging showed a high-intensity signal in the left anterior cerebral artery territory and middle cerebral artery territory. She had marked hypoalbuminemia and decreased protein S activity. We identified protein-losing gastroenteropathy as the cause of the hypoalbuminemia, and she had a missense mutation of the PROS 1 gene, which was associated with decreased protein S activity. We speculated that the development of protein-losing gastroenteropathy accelerated the decline in protein S activity and caused cerebral infarction.


Assuntos
Hipoalbuminemia , AVC Isquêmico , Deficiência de Proteína S , Acidente Vascular Cerebral , Humanos , Gravidez , Feminino , Adulto Jovem , Adulto , AVC Isquêmico/complicações , Hipoalbuminemia/complicações , Hipoalbuminemia/diagnóstico , Deficiência de Proteína S/complicações , Deficiência de Proteína S/diagnóstico , Cesárea/efeitos adversos , Proteína S , Acidente Vascular Cerebral/complicações , Acidente Vascular Cerebral/diagnóstico por imagem
9.
Medicina (Kaunas) ; 59(5)2023 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-37241140

RESUMO

Introduction: Chronic thromboembolic pulmonary hypertension (CTEPH) is a phenotype of pulmonary hypertension due to chronic and multiple organized thrombus. The therapeutic strategy for patients with CTEPH and comorbid protein S deficiency remains unknown due to its rarity. Case: We encountered a 49-year-old male patient with CTEPH and concomitant mild protein S deficiency (type III). We could successfully perform balloon pulmonary angioplasty without any major complications, including thromboembolism and bleeding, followed by standard-dose oral anticoagulation therapy instead of warfarin. Conclusion: A currently established standard therapeutic strategy for CTEPH, including pulmonary angioplasty, may be safe and effective even in patients with concomitant inherent coagulation abnormalities.


Assuntos
Hipertensão Pulmonar , Deficiência de Proteína S , Embolia Pulmonar , Masculino , Humanos , Hipertensão Pulmonar/complicações , Hipertensão Pulmonar/tratamento farmacológico , Artéria Pulmonar , Embolia Pulmonar/complicações , Embolia Pulmonar/tratamento farmacológico , Deficiência de Proteína S/complicações , Angioplastia/efeitos adversos , Anticoagulantes/uso terapêutico , Doença Crônica
10.
Zhongguo Yi Xue Ke Xue Yuan Xue Bao ; 45(5): 863-866, 2023 Oct.
Artigo em Zh | MEDLINE | ID: mdl-37621108

RESUMO

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.


Assuntos
Deficiência de Proteína S , Proteína S , Humanos , Feminino , Proteína S/genética , Deficiência de Proteína S/genética , Linhagem , Mutação
11.
BMC Neurol ; 22(1): 164, 2022 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-35501720

RESUMO

OBJECTIVE: To describe the characteristics of patients with cerebral venous sinus thrombosis (CVST) and dural arteriovenous fistula (AVF) associated with protein S (PS) deficiency. METHODS: We conducted a search of medical records in Hainan General Hospital from January 2000 to December 2020 for coexistence of CVST and dural AVF associated with PS deficiency and searched PubMed、Embase and Chinese biomedical databases (CBM) for all literature describing CVST and dural AVF with PS. We analyzed clinical characteristics, location, sequence of CVST and dural AVF, level of PS, therapeutic methods and prognosis. RESULTS: We presented 1 patient in our hospital's database combined CVST and dural AVF associated with PS, plus 5 cases reported in literature. The most common symptoms were headache, generalized seizure, disturbance of consciousness. The most frequent location of CVST was at internal cerebral vein, while transverse sinus, sigmoid sinus, parietal region in dural AVF. Two patients developed dural AVF several months or years after CVST. Clinical characteristics and level of PS were summarized. CONCLUSION: These findings alert physicians to consider PS deficiency in patients who suffer from CVST, especially those combined with dural AVF.


Assuntos
Malformações Vasculares do Sistema Nervoso Central , Deficiência de Proteína S , Trombose dos Seios Intracranianos , Malformações Vasculares do Sistema Nervoso Central/complicações , Cavidades Cranianas/diagnóstico por imagem , Cefaleia/complicações , Humanos , Deficiência de Proteína S/complicações , Trombose dos Seios Intracranianos/complicações
12.
J Pediatr Hematol Oncol ; 44(2): e442-e443, 2022 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-34486549

RESUMO

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.


Assuntos
Deficiência de Proteína S , Tromboembolia Venosa , Trombose Venosa , Adolescente , Criança , Feminino , Humanos , Proteína S/genética , Proteína S/metabolismo , Deficiência de Proteína S/complicações , Deficiência de Proteína S/genética , Trombofilia , Tromboembolia Venosa/tratamento farmacológico , Tromboembolia Venosa/genética , Trombose Venosa/tratamento farmacológico , Trombose Venosa/genética
13.
J Pediatr Hematol Oncol ; 44(8): 479-481, 2022 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-35700409

RESUMO

Idiopathic purpura fulminans (PF) is rare but has been reported in pediatric patients, commonly following infections. We present a case of a 5-year-old boy, heterozygous for factor V Leiden, with no history of recent infections, who presented with PF secondary to acquired protein S deficiency. Despite initial supportive treatment, the patient required surgical fasciotomy and extensive skin grafts. The protein S level normalized 4 months following the presentation. In this context, an autoimmune component with transient anti-protein S antibodies was believed to be involved. This case report highlights the course of idiopathic PF due to noninfectious acquired protein S deficiency.


Assuntos
Vasculite por IgA , Deficiência de Proteína S , Púrpura Fulminante , Trombofilia , Masculino , Humanos , Criança , Pré-Escolar , Púrpura Fulminante/complicações , Vasculite por IgA/complicações , Deficiência de Proteína S/complicações , Trombofilia/complicações
14.
Transfus Apher Sci ; 61(3): 103345, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-34963615

RESUMO

BACKGROUND: Protein S deficiency is associated with increased risk of venous thromboembolism, complicating the perioperative management of such patients. We present a patient with sickle cell disease (Hb SC genotype) and inherited protein S deficiency who underwent a living-donor renal transplant. To minimize thrombotic risk and sickle cell complications, both plasma exchange and red blood cell (RBC) exchange transfusion were performed pre-operatively. METHODS AND MATERIALS: Plasma exchange was utilized to increase protein S levels and to reduce the risk of post-operative venous thromboembolism, including allograft thrombosis, while RBC exchange was performed to reduce the risk of acute post-operative sickle cell disease complications. RESULTS: With the use of combined pre-operative plasma exchange and RBC exchange transfusion, this patient with protein S deficiency and Hb SC underwent a successful renal transplant without acute sickle cell complications or thrombotic complications. CONCLUSIONS: This case demonstrates the potential use of pre-operative plasma exchange in patients with protein S deficiency undergoing high thrombotic risk procedures.


Assuntos
Anemia Falciforme , Doença da Hemoglobina SC , Transplante de Rim , Deficiência de Proteína S , Tromboembolia Venosa , Anemia Falciforme/terapia , Transfusão de Eritrócitos/métodos , Eritrócitos , Humanos , Troca Plasmática , Complicações Pós-Operatórias
15.
J Thromb Thrombolysis ; 54(1): 156-161, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-35032256

RESUMO

Protein S deficiency is associated with an increased risk of thromboembolism, which may be caused by hereditary deficiency and several physiological and pathologic conditions, such as pregnancy, contraceptive use, liver diseases, inflammatory disease, and certain viruses infections. However, monoclonal immunoglobulin-mediated Protein S deficiency is rarely reported. Here we described a 49-year-old woman with a history of recurrent painful swelling in both lower extremities due to venous thrombosis for 7 years, accompanied by recurrent gross hematuria and multiple painful necrotic purpuras for 5 years, who was then diagnosed with acquired Protein S deficiency induced by the monoclonal immunoglobulin. Then she was successfully treated with rituximab combined with anticoagulation therapy. This case highlights the rare manifestations of Protein S deficiency and the influence of the monoclonal immunoglobulin produced by monoclonal B lymphocytes and monoclonal plasma cells on the activity of Protein S, which can be treated effectively with rituximab combined with anticoagulation therapy.


Assuntos
Deficiência de Proteína S , Púrpura , Anticoagulantes/uso terapêutico , Feminino , Hematúria , Humanos , Pessoa de Meia-Idade , Necrose/complicações , Gravidez , Deficiência de Proteína S/complicações , Púrpura/complicações , Rituximab/uso terapêutico
16.
Clin Lab ; 68(8)2022 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-35975485

RESUMO

BACKGROUND: Protein S is a central regulator of coagulation as it critically participates in down-regulation of both extrinsic and intrinsic pathways of the coagulation cascade. In this review, we aim to provide an update on protein S and its anticoagulant functions as a central hemostatic regulator. METHODS: Electronic databases including, Google, Google Scholar, PMC, PubMed, Science Direct, and Scopus were rigorously searched using the terms protein S, hemostasis, natural anticoagulants, regulators of coagulation, and coagulation inhibitors for the completion of this descriptive review. RESULTS: Literature review shows that protein S is a potent cofactor for activated protein C (APC) in the regulation of the intrinsic pathway and a cofactor for tissue factor pathway inhibitor (TFPI) in the regulation of the extrinsic pathway. The strong association between protein S deficiency either hereditary or acquired and increased risk for venous thrombosis indicates the important and central role of protein S in controlling the initiation and propagation phase of coagulation cascade and that protein S is an important determinant for optimal activity of both APC and TFPI in coagulation regulation. CONCLUSIONS: Available evidence suggests that the role of protein S in the down-regulation of blood coagulation is mainly mediated through its high affinity binding to negatively charged phospholipid surfaces. This high affinity binding to negatively charged phospholipids helps bring the anticoagulant proteins to the membranes, resulting in efficient and targeted regulation of coagulation. In the shade of current COVID-19 pandemic, protein S deficiency has been found to be a leading cause of thrombotic complications associated with COVID-19.


Assuntos
Coagulação Sanguínea , Deficiência de Proteína S , Proteína S , Anticoagulantes/farmacologia , COVID-19 , Humanos , Proteína S/fisiologia
17.
J Obstet Gynaecol ; 42(6): 2069-2074, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-35048758

RESUMO

The aim of this study was to analyse the risk factors of pregnancy loss of patients with recurrent spontaneous abortion (RSA) and develop a scoring system to predict RSA. Clinical data of 242 cases, with RSA who were treated at Fujian Provincial Maternity and Children's Hospital, were selected. The factors of pregnancy loss for RSA patients were evaluated by univariate and multivariate analyses. There were 242 RSA patients, of whom 34 (14.0%) developed pregnancy loss. A multivariate analysis showed the following adverse risk factors for RSA: antinuclear antibody spectrum, protein s deficiency and antiphospholipid antibodies. The pregnancy loss rates of antinuclear antibody spectrum group, protein S deficiency group and antiphospholipid antibodies group were 25.0%, 22.5% and 19.4%, respectively. Each of these factors contributed 1 point to the risk score. The pregnancy loss rates were 6.3%, 24.6%, 50% for the low-, intermediate- and high-risk categories, respectively (p < .001). The area under the receiver operating characteristic curve for the score of RSA was .733. Our findings suggest that this validated and simple scoring system could accurately predict the risk of pregnancy loss of RSA patients. The score might be helpful in the selection of risk-adapted interventions to decrease the incidence. Impact StatementWhat is already known on this subject? The live birth rate increases to 80%-90% after anticoagulant and/or immunosuppressive treatment in patients with RSA. However, there is still a high rate of re-abortion even after active treatment.What do the results of this study add? Antinuclear antibody spectrum, protein s deficiency and antiphospholipid antibodies were independent risk factors for pregnancy loss. A novel predictive model based on these factors was then established and validated.What are the implications of these findings for clinical practice and/or further research? The newly developed score might be helpful in the selection of risk-adapted interventions to decrease the incidence. For patients in the intermediate-risk and high-risk groups, we should conduct more targeted studies and formulate corresponding therapies to improve the success rate of treatment.


Assuntos
Aborto Habitual , Aborto Espontâneo , Deficiência de Proteína S , Aborto Habitual/epidemiologia , Aborto Habitual/etiologia , Aborto Espontâneo/epidemiologia , Aborto Espontâneo/etiologia , Anticorpos Antinucleares/uso terapêutico , Anticorpos Antifosfolipídeos/uso terapêutico , Anticoagulantes/uso terapêutico , Criança , Feminino , Humanos , Gravidez , Deficiência de Proteína S/complicações , Fatores de Risco
18.
Acta Haematol ; 144(2): 222-226, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-32653888

RESUMO

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.


Assuntos
Deficiência de Proteína S/diagnóstico , Proteína S/genética , Tromboembolia Venosa/diagnóstico , Anticoagulantes/uso terapêutico , Éxons , Fator V/genética , Feminino , Deleção de Genes , Heterozigoto , Homozigoto , Humanos , Pessoa de Meia-Idade , Mutação de Sentido Incorreto , Polimorfismo de Nucleotídeo Único , Deficiência de Proteína S/complicações , Deficiência de Proteína S/genética , Tromboembolia Venosa/tratamento farmacológico , Tromboembolia Venosa/etiologia
19.
J Obstet Gynaecol Res ; 47(9): 3008-3033, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34169611

RESUMO

Hereditary thrombophilia is a condition in which individuals are susceptible to the formation of thrombi due to a hereditary deficiency in anticoagulant factors, antithrombin (AT), protein C (PC), or protein S (PS). Many Japanese thrombophilia patients have PS deficiency, especially PS p.K196E (also called as PS Tokushima), which is exclusive to the Japanese population, and thrombosis sometimes occurs during pregnancy. At present, no management guidelines for pregnancy and delivery in thrombophilia patients have been developed. The Study Group for Hereditary Thrombophilia, one of the research groups of blood coagulation abnormalities in the Research Program on Rare and Intractable Diseases supported with the Research Grants of the Ministry of Health, Labour and Welfare Science, has therefore developed this clinical guidance to provide healthcare workers with necessary information on safe pregnancy, parturition and neonatal management, adopting a format of responses to seven clinical questions (CQ). At the end of each answer, the corresponding Recommendation Level (A, B, C) is indicated.


Assuntos
Deficiência de Proteína C , Deficiência de Proteína S , Trombofilia , Trombose , Feminino , Humanos , Recém-Nascido , Período Periparto , Gravidez , Trombofilia/complicações , Trombofilia/genética , Trombofilia/terapia
20.
Rev Gastroenterol Peru ; 41(1): 48-51, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34347772

RESUMO

Portal vein thrombosis (PVT) is a rare condition in the general population that develops serious complications if left untreated for long time. We present a case of a 29-year-old woman who developed PVT due to protein S deficiency versus neonatal funiculitis. Over time, the patient developed upper gastrointestinal bleeding due to esophageal varices and hypersplenism with splenic sequestration that caused minor bleeding episodes. Laparoscopic splenectomy and proximal splenorenal shunt with distal pancreatectomy due to aneurysmal dilatations of the splenic artery were successfully performed to avoid mayor progression of portal hypertension. Patient was discharged with indefinite anticoagulation and after surgery platelets raised up to 200x103/mm3. Laparoscopic splenectomy and proximal splenorenal shunt for portal hypertension due to portal vein thrombosis is an adequate surgery procedure which should be applied in these medical cases.


Assuntos
Varizes Esofágicas e Gástricas , Hipertensão Portal , Laparoscopia , Deficiência de Proteína S , Derivação Esplenorrenal Cirúrgica , Trombose Venosa , Adulto , Feminino , Hemorragia Gastrointestinal , Humanos , Hipertensão Portal/complicações , Recém-Nascido , Veia Porta/cirurgia , Esplenectomia , Trombose Venosa/complicações , Trombose Venosa/cirurgia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA