Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.392
Filter
1.
Arq Neuropsiquiatr ; 82(7): 1, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38977266

ABSTRACT

BACKGROUND: Migraine is associated with several genetic or acquired comorbidities. Studies conducted in recent years emphasize that the frequency of thrombophilia is high in migraine, especially migraine with aura (MA). Similarly, the presence of white matter lesions (WMLs) on brain magnetic resonance imaging (MRI) scans has been associated with migraine for many years. OBJECTIVE: Based on the knowledge that both WMLs and thrombophilia variants are frequently observed in MA, we aimed to investigate whether there is a relationship between genetic thrombophilia and the presence of WMLs in these patients. METHODS: The levels of proteins S and C, antithrombin III activities, activated protein C (APC) resistance, antiphospholipid immunoglobulin G/immunoglobulin M (IgG/IgM) and anticardiolipin IgG/IgM antibodies were investigated in 66 MA patients between the ages of 18 and 49 years who presented no cardiovascular risk factors. The presence of WMLs and the Fazekas grade was determined from the brain magnetic resonance imaging (MRI) scans' T2-weighted and fluid-attenuated inversion recovery (FLAIR) sequence taken from the patients. The rates of WMLs were compared in patients with and without thrombophilia. RESULTS: Thrombophilia was detected in 34.8% of the patients, and 27.3% were determined to have WMLs in brain MRI scans. The WMLs were detected in 23.3% of the patients without thrombophilia, in 34.8% of those with thrombophilia, and in 50% of the subjects with multiple thrombophilia disorders. Among the thrombophilia disorders, only APC resistance was significantly more common in patients with WMLs. CONCLUSION: The results of the present study showed that thrombophilia may be a mechanism that should be investigated in the etiology of increased WMLs in MA.


ANTECEDENTES: La migraña se asocia con una serie de comorbilidades genéticas o adquiridas. Los estudios realizados en los últimos años destacan que la frecuencia de trombofilia es elevada en la migraña, especialmente en la migraña con aura (MA). De manera similar, la presencia de lesiones de la sustancia blanca (LSB) en las imágenes por resonancia magnética (RM) del cerebro se ha asociado con la migraña hace muchos años. OBJETIVO: Con base en la información de que se suelen observar tanto LSB como variantes de la trombofilia en MA, nuestro objetivo fue investigar si existe una relación entre la trombofilia genética y la presencia de LSB en estos pacientes. MéTODOS: Se investigaron los niveles de proteína S y de proteína C, actividades de antitrombina III, resistencia a la proteína C activada (PCA), anticuerpos antifosfolípidos inmunoglobulina G/inmunoglobulina M (IgG/IgM) y anticuerpos anticardiolipina IgG/IgM en 66 pacientes con MA entre 18 y 49 años que no presentaban factores de riesgo cardiovascular. Se determinaron la presencia de LSB y el grado de Fazekas a partir de imágenes por RM del cerebro en la secuencia ponderada en T2 y recuperación de la inversión atenuada de fluido (fluid-attenuated inversion recovery, FLAIR, en inglés) obtenidas de los pacientes. Se compararon las tasas de LSB en pacientes con y sin trombofilia. RESULTADOS: Se detectó trombofilia en el 34,8% de los pacientes y LSB en el 27,3%. Las LSB estuvieron presentes en el 23,3% de los pacientes sin trombofilia, en el 34,8% de los que tenían trombofilia, y en el 50% de los que tenían múltiples trastornos trombofílicos. La resistencia a la PCA fue significativamente más común en aquellos pacientes con LSB. CONCLUSIóN: Los resultados del presente estudio mostraron que la trombofilia puede ser un mecanismo que debe investigarse en la etiología del aumento de LSB en MA.


Subject(s)
Magnetic Resonance Imaging , Migraine with Aura , Thrombophilia , White Matter , Humans , Adult , Female , Male , Thrombophilia/blood , Middle Aged , Migraine with Aura/diagnostic imaging , Migraine with Aura/blood , Young Adult , White Matter/diagnostic imaging , White Matter/pathology , Adolescent , Antithrombin III/analysis , Protein S/analysis , Risk Factors , Antibodies, Anticardiolipin/blood , Protein C/analysis , Immunoglobulin G/blood , Antibodies, Antiphospholipid/blood
2.
Sci Rep ; 14(1): 13855, 2024 06 15.
Article in English | MEDLINE | ID: mdl-38879576

ABSTRACT

Data on the pathophysiological mechanisms of hemostatic alterations in the thrombotic events that occur during Ramadan intermittent fasting (RIF), particularly in the natural coagulation inhibitors, are very limited. Thus, our objective was to evaluate the effect of RIF on the natural anticoagulants level, antithrombin, protein C, and total and free protein S (PS) in healthy participants. Participants were divided into two groups. Group I consisted of 29 healthy fasting participants whose blood samples were taken after 20 days of fasting. Group II included 40 healthy non-fasting participants whose blood samples were taken 2-4 weeks before the month of Ramadan. Coagulation screening tests including prothrombin time (PT), activated partial thromboplastin time (APTT) and plasma fibrinogen level, natural anticoagulants; antithrombin, protein C, free and total PS and C4 binding protein (C4BP) levels were evaluated in the two groups. High levels of total and free PS without change in antithrombin, protein C, and C4BP levels were noted in the fasting group as compared with non-fasting ones (p < 0.05). PT and APTT showed no difference between the two groups. However, the fibrinogen level was higher in the fasting group. In conclusion, RIF was found to be associated with improved anticoagulant activity in healthy participants, which may provide temporal physiological protection against the development of thrombosis in healthy fasting people.


Subject(s)
Anticoagulants , Blood Coagulation , Fasting , Islam , Humans , Fasting/blood , Male , Adult , Female , Case-Control Studies , Blood Coagulation/drug effects , Anticoagulants/administration & dosage , Protein C/metabolism , Protein S/metabolism , Protein S/analysis , Blood Coagulation Tests , Healthy Volunteers , Fibrinogen/metabolism , Middle Aged , Young Adult , Prothrombin Time , Antithrombins , Partial Thromboplastin Time , Intermittent Fasting
3.
Int J Mol Sci ; 25(12)2024 Jun 16.
Article in English | MEDLINE | ID: mdl-38928335

ABSTRACT

Among the myriad of existing tyrosine kinase receptors, the TAM family-abbreviated from Tyro3, Axl, and Mer tyrosine kinase (MerTK)-has been extensively studied with an outstanding contribution from the team of Prof. Greg Lemke. MerTK activity is implicated in a wide variety of functions involving the elimination of apoptotic cells and has recently been linked to cancers, auto-immune diseases, and atherosclerosis/stroke. In the retina, MerTK is required for the circadian phagocytosis of oxidized photoreceptor outer segments by the retinal-pigment epithelial cells, a function crucial for the long-term maintenance of vision. We previously showed that MerTK ligands carry the opposite role in vitro, with Gas6 inhibiting the internalization of photoreceptor outer segments while Protein S acts conversely. Using site-directed mutagenesis and ligand-stimulated phagocytosis assays on transfected cells, we presently demonstrate, for the first time, that Gas6 and Protein S recognize different amino acids on MerTK Ig-like domains. In addition, MerTK's function in retinal-pigment epithelial cells is rhythmic and might thus rely on the respective stoichiometry of both ligands at different times of the day. Accordingly, we show that ligand bioavailability varies during the circadian cycle using RT-qPCR and immunoblots on retinal and retinal-pigment epithelial samples from control and beta5 integrin knockout mice where retinal phagocytosis is arrhythmic. Taken together, our results suggest that Gas6 and Protein S might both contribute to refine the acute regulation of MerTK in time for the daily phagocytic peak.


Subject(s)
Intercellular Signaling Peptides and Proteins , Phagocytosis , Protein S , c-Mer Tyrosine Kinase , Animals , Mice , c-Mer Tyrosine Kinase/metabolism , c-Mer Tyrosine Kinase/genetics , Circadian Rhythm/physiology , Intercellular Signaling Peptides and Proteins/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Ligands , Protein S/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Retina/metabolism , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/cytology
4.
J Thromb Thrombolysis ; 57(6): 1018-1030, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38722521

ABSTRACT

COVID-19 has been associated with alterations in coagulation. Recent reports have shown that protein C and S activities are altered in COVID-19. This may affect the complications and outcome of the disease. However, their exact role in COVID-19 remains uncertain. The aim of the current study was therefore to analyze all papers in the literature on protein C and S activities in COVID-19. We searched three medical electronic databases. Of the 2442 papers, 28 studies were selected for the present meta-analysis. For the meta-analysis, means ± standard deviations with 95% confidence intervals (CI) for protein C and S activities were extracted. Pooled p values were calculated using STATA software. Protein C and S activities were significantly lower in COVID-19 patients than in healthy controls (pooled p values: 0.04 and 0.02, respectively). Similarly, protein C activities were considerably lower in nonsurviving patients (pooled p value = 0.00). There was no association between proteins C or S and thrombosis risk or ICU admission in COVID-19 patients (p value > 0.05). COVID-19 patients may exhibit lower activities of the C and S proteins, which might affect disease outcome; however, additional attention should be given when considering therapeutic strategies for these patients.


Subject(s)
COVID-19 , Protein C , Protein S , COVID-19/blood , Humans , Protein C/metabolism , Protein S/metabolism , Protein S/analysis , SARS-CoV-2 , Thrombosis/blood , Thrombosis/etiology , Blood Coagulation
6.
Aging (Albany NY) ; 16(8): 6883-6897, 2024 04 10.
Article in English | MEDLINE | ID: mdl-38613800

ABSTRACT

BACKGROUND: Diabetic foot ulcers (DFUs) pose a serious long-term threat because of elevated mortality and disability risks. Research on its biomarkers is still, however, very limited. In this paper, we have effectively identified biomarkers linked with macrophage excretion in diabetic foot ulcers through the application of bioinformatics and machine learning methodologies. These findings were subsequently validated using external datasets and animal experiments. Such discoveries are anticipated to offer novel insights and approaches for the early diagnosis and treatment of DFU. METHODS: In this work, we used the Gene Expression Omnibus (GEO) database's datasets GSE68183 and GSE80178 as the training dataset to build a gene model using machine learning methods. After that, we used the training and validation sets to validate the model (GSE134431). On the model genes, we performed enrichment analysis using both gene set variant analysis (GSVA) and gene set enrichment analysis (GSEA). Additionally, the model genes were subjected to immunological association and immune function analyses. RESULTS: In this study, PROS1 was identified as a potential key target associated with macrophage efflux in DFU by machine learning and bioinformatics approaches. Subsequently, the key biomarker status of PROS1 in DFU was also confirmed by external datasets. In addition, PROS1 also plays a key role in macrophage exudation in DFU. This gene may be associated with macrophage M1, CD4 memory T cells, naïve B cells, and macrophage M2, and affects IL-17, Rap1, hedgehog, and JAK-STAT signaling pathways. CONCLUSIONS: PROS1 was identified and validated as a biomarker for DFU. This finding has the potential to provide a target for macrophage clearance of DFU.


Subject(s)
Diabetic Foot , Machine Learning , Macrophages , Protein S , Humans , Biomarkers/metabolism , Computational Biology , Diabetic Foot/genetics , Diabetic Foot/metabolism , Efferocytosis , Macrophages/metabolism , Phagocytosis/genetics , Protein S/genetics
7.
Int J Mol Sci ; 25(8)2024 Apr 17.
Article in English | MEDLINE | ID: mdl-38673989

ABSTRACT

Mertk, a type I receptor tyrosine kinase and member of the TAM family of receptors, has important functions in promoting efferocytosis and resolving inflammation under physiological conditions. In recent years, Mertk has also been linked to pathophysiological roles in cancer, whereby, in several cancer types, including solid cancers and leukemia/lymphomas. Mertk contributes to oncogenic features of proliferation and cell survival as an oncogenic tyrosine kinase. In addition, Mertk expressed on macrophages, including tumor-associated macrophages, promotes immune evasion in cancer and is suggested to act akin to a myeloid checkpoint inhibitor that skews macrophages towards inhibitory phenotypes that suppress host T-cell anti-tumor immunity. In the present study, to better understand the post-translational regulation mechanisms controlling Mertk expression in monocytes/macrophages, we used a PMA-differentiated THP-1 cell model to interrogate the regulation of Mertk expression and developed a novel Mertk reporter cell line to study the intracellular trafficking of Mertk. We show that PMA treatment potently up-regulates Mertk as well as components of the ectodomain proteolytic processing platform ADAM17, whereas PMA differentially regulates the canonical Mertk ligands Gas6 and Pros1 (Gas6 is down-regulated and Pros1 is up-regulated). Under non-stimulated homeostatic conditions, Mertk in PMA-differentiated THP1 cells shows active constitutive proteolytic cleavage by the sequential activities of ADAM17 and the Presenilin/γ-secretase complex, indicating that Mertk is cleaved homeostatically by the combined sequential action of ADAM17 and γ-secretase, after which the cleaved intracellular fragment of Mertk is degraded in a proteasome-dependent mechanism. Using chimeric Flag-Mertk-EGFP-Myc reporter receptors, we confirm that inhibitors of γ-secretase and MG132, which inhibits the 26S proteasome, stabilize the intracellular fragment of Mertk without evidence of nuclear translocation. Finally, the treatment of cells with active γ-carboxylated Gas6, but not inactive Warfarin-treated non-γ-carboxylated Gas6, regulates a distinct proteolytic itinerary-involved receptor clearance and lysosomal proteolysis. Together, these results indicate that pleotropic and complex proteolytic activities regulate Mertk ectodomain cleavage as a homeostatic negative regulatory event to safeguard against the overactivation of Mertk.


Subject(s)
ADAM17 Protein , Amyloid Precursor Protein Secretases , Proteolysis , c-Mer Tyrosine Kinase , Humans , c-Mer Tyrosine Kinase/metabolism , c-Mer Tyrosine Kinase/genetics , ADAM17 Protein/metabolism , ADAM17 Protein/genetics , Amyloid Precursor Protein Secretases/metabolism , Amyloid Precursor Protein Secretases/genetics , Intercellular Signaling Peptides and Proteins/metabolism , THP-1 Cells , Macrophages/metabolism , Protein S/metabolism , Monocytes/metabolism , Tetradecanoylphorbol Acetate/pharmacology
8.
Clin Immunol ; 263: 110202, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38575045

ABSTRACT

Celiac disease (CD) is an immune-driven disease characterized by tissue damage in the small intestine of genetically-susceptible individuals. We evaluated here a crucial immune regulatory pathway involving TYRO3, AXL, and MERTK (TAM) receptors and their ligands PROS1 and GAS6 in duodenal biopsies of controls and CD patients. We found increased GAS6 expression associated with downregulation of PROS1 and variable TAM receptors levels in duodenum tissue of CD patients. Interestingly, CD3+ lymphocytes, CD68+, CD11c+ myeloid and epithelial cells, showed differential expressions of TAM components comparing CD vs controls. Principal component analysis revealed a clear segregation of two groups of CD patients based on TAM components and IFN signaling. In vitro validation demonstrated that monocytes, T lymphocytes and epithelial cells upregulated TAM components in response to IFN stimulation. Our findings highlight a dysregulated TAM axis in CD related to IFN signaling and contribute to a deeper understanding of the pathophysiology of CD.


Subject(s)
Axl Receptor Tyrosine Kinase , Celiac Disease , Duodenum , Intercellular Signaling Peptides and Proteins , Intestinal Mucosa , Protein S , Receptor Protein-Tyrosine Kinases , c-Mer Tyrosine Kinase , Humans , Celiac Disease/immunology , Celiac Disease/metabolism , Celiac Disease/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/immunology , Male , Intestinal Mucosa/metabolism , Intestinal Mucosa/immunology , Female , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Adult , Duodenum/metabolism , Duodenum/immunology , Duodenum/pathology , c-Mer Tyrosine Kinase/genetics , c-Mer Tyrosine Kinase/metabolism , Protein S/metabolism , Protein S/genetics , Middle Aged , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/genetics , Young Adult , Signal Transduction , Adolescent , Interferons/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
9.
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
10.
Transfus Apher Sci ; 63(3): 103918, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38555232

ABSTRACT

INTRODUCTION: Therapeutic plasma exchange (TPE), with solvent/detergent (S/D)-treated plasma as replacement fluid, is an extracorporeal blood purification technique with major impact on both coagulation and lipids. Our previous in vitro study showed that S/D-plasma enhances thrombin generation by lowering intact protein S (PS) levels. AIMS: To evaluate the impact of altered lipid balance on coagulation phenotype during heparin-anticoagulated TPE with S/D-plasma, and to investigate whether the lowered intact PS levels with concomitant procoagulant phenotype, are recapitulated in vivo. METHODS: Coagulation biomarkers, thrombin generation with Calibrated Automated Thrombogram (CAT), and lipid levels were measured before and after the consecutive 1st, 3rd and 5th episodes of TPE performed to six patients with Guillain-Barré syndrome or myasthenia gravis. The effects of in vitro dilution of S/D-plasma on thrombin generation were explored with CAT to mimic TPE. RESULTS: Patients did not have coagulation disorders, except elevated FVIII. Intact PS, lipoproteins, especially LDL, Apolipoprotein CIII (ApoC3) and ApoB/ApoA1 ratio declined (p < 0.05). In contrast, VLDL and triglyceride levels stayed intact. CAT lag time shortened (p < 0.05). In vitro dilution of S/D plasma with co-transfused Ringer's lactate and 4% albumin partially reduced its procoagulant phenotype in CAT, which is mainly seen as peak thrombin, and modestly shortened lag time. CONCLUSIONS: After the five settings of TPE using S/D-plasma in vivo, which associated with heparinization and reduced coagulation factor activities, our observations of declining natural anticoagulant intact PS and apolipoproteins refer to rebalance of the hemostatic and lipid profiles.


Subject(s)
Apolipoproteins , Plasma Exchange , Protein S , Thrombin , Humans , Plasma Exchange/methods , Male , Thrombin/metabolism , Apolipoproteins/blood , Female , Middle Aged , Protein S/metabolism , Adult , Aged
11.
Endocrine ; 85(2): 558-565, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38483687

ABSTRACT

Proteins C and S are vitamin K-dependent anticoagulative factors that also exert a significant influence on bone quality. Clinical studies have linked the deficiency of proteins C and S to lower bone mineral density and the onset of femoral head osteonecrosis in children. Rare foundational studies analyzing this topic have demonstrated that activated protein C, upon binding to the endothelial protein C receptor expressed on the surface of osteoblasts, promotes osteoblast proliferation. It is also established that proteins C and S play crucial roles in proper collagen synthesis and in maintaining the number of osteoclasts and blood vessels. However, the association between protein C and/or S deficiency and the gradual onset of osteoporosis remains largely uninvestigated. Calculations based on data from peer-reviewed journals suggest that approximately one in every 10 individuals may develop osteoporosis due to congenital protein C or S deficiency. Moreover, when secondary causes of protein C and S deficiency are also considered, the proportion likely further increases. In this paper, we discuss the pathophysiological background of the potential relationship between protein C and S deficiency and the genesis of osteoporosis.


Subject(s)
Osteoporosis , Protein C Deficiency , Protein S Deficiency , Humans , Osteoporosis/etiology , Protein C Deficiency/complications , Protein S Deficiency/complications , Protein C/metabolism , Bone Density , Animals , Protein S/metabolism
12.
Int J Hematol ; 119(2): 196-204, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38228939

ABSTRACT

The protein C (PC) pathway involves physiological anticoagulant factors (PC, protein S [PS], and factor V) and performs major anticoagulant functions in adults. Variations in overall PC pathway function due to dynamic changes in PC and PS in early childhood are poorly understood. We aimed to evaluate the contributions of PC pathway function during early childhood by measuring changes in plasma thrombin generation (TG) after administration of the PC activator protac. We evaluated correlations between anticoagulant factors and percentage of protac-induced coagulation inhibition (PiCi%). Before protac addition, TG in newborns (n = 35), infants (n = 42), young children (n = 35), and adults (n = 20) were 525 ± 74, 720 ± 96, 785 ± 53, and 802 ± 64 mOD/min, and PiCi% were 42.1 ± 9.9, 69.8 ± 11.0, 82.9 ± 4.4, and 86.9 ± 3.4%, respectively. The distribution of PiCi% on the two axes of TG (with or without protac) changed continuously with age and differed from that of warfarin-treated plasma and adult PC- or PS-deficient plasma. PiCi% increased dynamically during infancy and correlated with PS levels in newborns and PC levels in young children. Addition of PC or fresh frozen plasma equivalent to approximately 25% PC to PC-deficient plasma improved PiCi%. This automatic measurement requires only a small sample volume and is useful for analysis of developmental hemostasis.


Subject(s)
Protein C , Proteolysis Targeting Chimera , Adult , Child , Child, Preschool , Humans , Infant, Newborn , Anticoagulants/pharmacology , Antithrombins/pharmacology , Blood Coagulation , Protein C/analysis , Protein C/metabolism , Protein C/pharmacology , Protein S/metabolism , Thrombin/metabolism , Infant
13.
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
14.
Vox Sang ; 119(3): 193-202, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38018260

ABSTRACT

BACKGROUND AND OBJECTIVES: Deficiencies of protein C (PC) or protein S (PS) are rare diseases, characterized by mutations in the PC or PS genes, which encode plasma serine proteases with anti-coagulant activity. Severe PC or PS deficiencies manifest in early life as neonatal purpura fulminans, a life-threatening heamorrhagic condition requiring immediate treatment. First-line treatment involves replacement therapy, followed by maintenance with anti-coagulants. Replacement therapy with specific protein concentrates is currently only limited to PC, and therefore, a PC + PS concentrate represents a useful addition to therapeutic options, particularly for severe PS deficiency. Further, the production of a PC + PS concentrate from unused plasma fractionation intermediates would impact favourably on manufacturing costs, and consequently therapy prices for patients and health systems. MATERIALS AND METHODS: Several chromatographic runs were performed on the same unused plasma fractionation intermediates using different supports to obtain a PC/PS concentrate. The best chromatographic mediums were chosen, in terms of specific activity and recovery. A full process of purification including virus inactivation/removal and lyophilization steps was set up. RESULTS: The final freeze-dried product had a mean PC concentration of 47.75 IU/mL with 11% of PS, and a mean specific activity of 202.5 IU/mg protein, corresponding to over 12,000-fold purification from plasma. CONCLUSION: The development of a novel concentrated PC/PS mixture obtained from a waste fraction of other commercial products could be used for its potential therapeutic role in the management of neonatal purpura fulminans pathology.


Subject(s)
Protein C Deficiency , Purpura Fulminans , Infant, Newborn , Humans , Purpura Fulminans/drug therapy , Purpura Fulminans/genetics , Protein C Deficiency/drug therapy , Protein C/analysis , Protein C/therapeutic use , Protein S , Plasma/chemistry
15.
Cell Signal ; 112: 110918, 2023 12.
Article in English | MEDLINE | ID: mdl-37827342

ABSTRACT

BACKGROUND: PROS1 is an encoding gene that can generate protein S. This protein is a glycoprotein found in plasma that conducts physiological functions with vitamin K. However, the impact of its expression remains absent in the progression and prognosis of breast cancer (BC). METHODS: In this study, we comprehensively explored the expression of PROS1 in BC and its relationship with BC patient survival, prognosis, and other clinicopathological features. We investigated how PROS1 influenced the malignant biological behavior of BC cells. A series of enrichment analyses were conducted, and the immune landscape was explored in BC affected by PROS1. We also determined correlations between PROS1 and common drug sensitivities used for BC treatments. RESULTS: PROS1 had low expression in BC, which tended to result in poor survival of BC patients. Overexpressed PROS1 inhibited the migration and invasion of BC cells as well as the epithelial-mesenchymal transition process by downregulating SNAIL. Functional enrichment analyses revealed that PROS1 was more active in extracellular matrix (ECM) organization and structural constituent, ECM-receptor interaction, and other pathways with its related genes. PROS1 was also found to affect immune activity, including various immune cells infiltrating BC. BC patients with high PROS1 expression tended to have lower IC50 values of three common medications and obtained better efficacy. CONCLUSIONS: PROS1 can become a promising prognostic factor and a possible therapeutic target in BC patients and suppress BC cell metastatic potential. In addition, PROS1 is a crucial factor in immune infiltration in BC.


Subject(s)
Breast Neoplasms , Humans , Female , Prognosis , Breast Neoplasms/genetics , Epithelial-Mesenchymal Transition/genetics , Computational Biology , Biomarkers , Protein S
16.
Thromb Res ; 230: 84-93, 2023 10.
Article in English | MEDLINE | ID: mdl-37660436

ABSTRACT

INTRODUCTION: Thrombin, the enzyme which converts fibrinogen into a fibrin clot, is produced by the prothrombinase complex, composed of factor Xa (FXa) and factor Va (FVa). Down-regulation of this process is critical, as excess thrombin can lead to life-threatening thrombotic events. FXa and FVa are inhibited by the anticoagulants tissue factor pathway inhibitor alpha (TFPIα) and activated protein C (APC), respectively, and their common cofactor protein S (PS). However, prothrombinase is resistant to either of these inhibitory systems in isolation. MATERIALS AND METHODS: We hypothesized that these anticoagulants function best together, and tested this hypothesis using purified proteins and plasma-based systems. RESULTS: In plasma, TFPIα had greater anticoagulant activity in the presence of APC and PS, maximum PS activity required both TFPIα and APC, and antibodies against TFPI and APC had an additive procoagulant effect, which was mimicked by an antibody against PS alone. In purified protein systems, TFPIα dose-dependently inhibited thrombin activation by prothrombinase, but only in the presence of APC, and this activity was enhanced by PS. Conversely, FXa protected FVa from cleavage by APC, even in the presence of PS, and TFPIα reversed this protection. However, prothrombinase assembled on platelets was still protected from inhibition, even in the presence of TFPIα, APC, and PS. CONCLUSIONS: We propose a model of prothrombinase inhibition through combined targeting of both FXa and FVa, and that this mechanism enables down-regulation of thrombin activation outside of a platelet clot. Platelets protect prothrombinase from inhibition, however, supporting a procoagulant environment within the clot.


Subject(s)
Protein C , Protein S , Thrombin , Humans , Anticoagulants , Blood Coagulation , Factor V/metabolism , Factor Va/metabolism , Factor Xa/metabolism , Protein C/metabolism , Protein S/metabolism , Thrombin/metabolism , Thromboplastin/metabolism
17.
Zhongguo Yi Xue Ke Xue Yuan Xue Bao ; 45(5): 863-866, 2023 Oct.
Article in Chinese | 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
18.
Medicine (Baltimore) ; 102(28): e34338, 2023 Jul 14.
Article in English | MEDLINE | ID: mdl-37443483

ABSTRACT

The objective of this retrospective cohort study was to investigate the correlation between serum 25-OH-VitD levels, coagulation function, immune factors, and the risk of spontaneous abortion in the first trimester of pregnancy. Additionally, independent risk factors for spontaneous abortion in the first trimester of pregnancy were identified. A total of 412 pregnant women who attended Hubei Maternal and Child Health Care Hospital between October 2021 and February 2022 were included in the study. Of these, 221 met the eligibility criteria and were categorized into the early spontaneous abortion case group (n = 107) or the normal pregnancy control group (n = 114). The serum levels of 25-OH-VitD, CD3 + CD19- T lymphocytes, CD3-CD19 + B lymphocytes, NK (Natural Killer) cells, TNF-α (tumor necrosis factor-α) and coagulation factors (D-dimer, Protein C, Protein S) in both these groups were measured during early pregnancy (within 12 weeks) and evaluated using logistic regression analysis. Compared to the control group, body mass index, Protein S, CD19 + CD3-B lymphocytes, and 25-OH-VitD were significantly lower in the spontaneous abortion group during early pregnancy (P = .001; P = .004; P = .009; P = .001), blood glucose (fasting) and TNF-α significantly increased (P = .001; P = .046). Logistic regression analysis of potential mixed factors showed that fasting blood glucose and TNF-α were significantly different from the control group and were positively correlated (P = .001, P = .038). Fasting blood glucose 0R value is 2.264, 95% confidence interval is 0.043~0.25, TNF-α 0R value is 0.126, 95% confidence interval is 0.800~0.972. CD19 + CD3-B cells and 25-OH-VitD were correlated with spontaneous abortion (P = .005; P = .001), respectively 0R value and 95% confidence interval being -0.007 (1.002~1.012), -0.179 (1.139~1.256). Risk factors for spontaneous abortion in the first trimester (<12 weeks) of pregnancy include fasting glucose tolerance, decreased CD19 + CD3-energy B lymphocytes and 25-OH-VitD, and abnormal increase of TNF-α. Therefore, it is recommended that women with fertility needs be examined as early as possible to avoid adverse outcomes.


Subject(s)
Abortion, Spontaneous , Child , Pregnancy , Female , Humans , Abortion, Spontaneous/epidemiology , Pregnant Women , Protein S , Retrospective Studies , Blood Glucose , Tumor Necrosis Factor-alpha , Vitamin D , B-Lymphocytes , Vitamins
19.
J Agric Food Chem ; 71(29): 11150-11157, 2023 Jul 26.
Article in English | MEDLINE | ID: mdl-37435821

ABSTRACT

This study aimed to investigate the effect of pre-slaughter transport stress on the protein S-nitrosylation levels of pork during 0, 3, and 6 days of aging. Pigs (N = 16) were randomly selected and divided into two treatments: 3 h transport (transport stress, TS) and 3 h transport followed by 3 h resting (control, CON). Results showed that the TS group at 0 and 3 days presented a higher nitric oxide synthase (NOS) activity and neuronal NOS (nNOS) expression than the CON group (P < 0.05). Additionally, nNOS was not only abundantly localized in the membrane but also observed in small amounts in the cytoplasm. The immunoblot of overall S-nitrosylated protein showed that protein S-nitrosylation levels in the TS group were greater than in the CON group during postmortem aging (P < 0.05). This work can deliver novel insights into the mechanism of meat quality changes in response to pre-slaughter stress.


Subject(s)
Pork Meat , Red Meat , Animals , Swine , Protein S , Proteins , Nitric Oxide/metabolism
20.
Int J Biol Macromol ; 250: 126027, 2023 Oct 01.
Article in English | MEDLINE | ID: mdl-37506796

ABSTRACT

BACKGROUND: Class 2 uveal melanomas are associated with the inactivation of the BRCA1 ((breast cancer type 1 susceptibility protein)-associated protein 1 (BAP1)) gene. Inactivation of BAP1 promotes the upregulation of vitamin K-dependent protein S (PROS1), which interacts with the tyrosine-protein kinase Mer (MERTK) receptor on M2 macrophages to induce an immunosuppressive environment. METHODS: We simulated the interaction of PROS1 with MERTK with ColabFold. We evaluated PROS1 and MERTK for the presence of intrinsically disordered protein regions (IDPRs) and disorder-to-order (DOT) regions to understand their protein-protein interaction (PPI). We first evaluated the structure of each protein with AlphaFold. We then analyzed specific sequence-based features of the PROS1 and MERTK with a suite of bioinformatics tools. RESULTS: With high-resolution, moderate confidence, we successfully modeled the interaction between PROS1 and MERTK (predicted local distance difference test score (pDLLT) = 70.68). Our structural analysis qualitatively demonstrated IDPRs (i.e., spaghetti-like entities) in PROS1 and MERK. PROS1 was 23.37 % disordered, and MERTK was 23.09 % disordered, classifying them as moderately disordered and flexible proteins. PROS1 was significantly enriched in cysteine, the most order-promoting residue (p-value <0.05). Our IUPred analysis demonstrated that there are two disorder-to-order transition (DOT) regions in PROS1. MERTK was significantly enriched in proline, the most disorder-promoting residue (p-value <0.05), but did not contain DOT regions. Our STRING analysis demonstrated that the PPI network between PROS1 and MERTK is more complex than their assumed one-to-one binding (p-value <2.0 × 10-6). CONCLUSION: Our findings present a novel prediction for the interaction between PROS1 and MERTK. Our findings show that PROS1 and MERTK contain elements of intrinsic disorder. PROS1 has two DOT regions that are attractive immunotherapy targets. We recommend that IDPRs and DOT regions found in PROS1 and MERTK should be considered when developing immunotherapies targeting this PPI.


Subject(s)
Melanoma , Uveal Neoplasms , Humans , c-Mer Tyrosine Kinase/genetics , c-Mer Tyrosine Kinase/metabolism , Melanoma/genetics , Uveal Neoplasms/genetics , Carrier Proteins/metabolism , Protein S/genetics , Protein S/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL