Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 214
Filter
1.
Expert Rev Hematol ; 16(12): 1063-1076, 2023.
Article in English | MEDLINE | ID: mdl-38100503

ABSTRACT

INTRODUCTION: Inherited bleeding disorders (IBDs) including hemophilia, von Willebrand disease, platelet disorders, mucocutaneous bleeding disorders and coagulation factor deficiencies are rarely found and under-recognized in low and lower-middle-income countries. Some patients succumbed to serious bleeding without diagnosis and treatment during childhood. AREA COVERED: Diagnosis, management, and prevention should be integrated into the existing health care system. Although some countries have not implemented appropriate health care infrastructure, an initiative plan should be set up by cooperation of experienced experts and health care providers. Identification of patients with IBDs should be started in the antenatal setting to search for females at risk of carrier state. The investigations include bleeding assessment, mixing venous clotting time, coagulogram, coagulation factor assay and mutation detection. Genotypic analysis is helpful for confirming the definite diagnosis, carrier detection as well as prenatal diagnosis for females at risk of bearing an offspring with severe bleeding manifestations. Management involves replacement therapy ranging from blood component to virus-inactivated factor concentrate. Appropriate research is an essential backbone for improving patients' care. EXPERT OPINION: Effective national strategic advocacy to manage patients with IBDs requires intensive collaboration among policy makers, health care providers, patients, and family members.


Subject(s)
Blood Coagulation Disorders, Inherited , Hemophilia A , von Willebrand Diseases , Humans , Female , Pregnancy , Developing Countries , Blood Coagulation Disorders, Inherited/diagnosis , Blood Coagulation Disorders, Inherited/genetics , Blood Coagulation Disorders, Inherited/therapy , Hemophilia A/therapy , Hemorrhage/diagnosis , Hemorrhage/etiology , Hemorrhage/prevention & control , Blood Coagulation Factors
2.
Blood Rev ; 59: 101029, 2023 05.
Article in English | MEDLINE | ID: mdl-36369145

ABSTRACT

Rare bleeding disorders (RBDs), including factor (F) I, FII, FV, FVII, combined FV and FVIII (CF5F8), FXI, FXIII and vitamin-K dependent coagulation factors (VKCF) deficiencies, are a heterogeneous group of hemorrhagic disorder with a variable bleeding tendency. RBDs are due to mutation in underlying coagulation factors genes, except for CF5F8 and VKCF deficiencies. FVII deficiency is the most common RBD with >330 variants in the F7 gene, while only 63 variants have been identified in the F2 gene. Most detected variants in the affected genes are missense (>50% of all RBDs), while large deletions are the rarest, having been reported in FVII, FX, FXI and FXIII deficiencies. Most were located in the catalytic and activated domains of FXI, FX, FXIII and prothrombin deficiencies. Understanding the proper molecular basis of RBDs not only can help achieve a timely and cost-effective diagnosis, but also can help to phenotype properties of the disorders.


Subject(s)
Blood Coagulation Disorders, Inherited , Blood Coagulation Disorders , Coagulation Protein Disorders , Hemorrhagic Disorders , Humans , Blood Coagulation Disorders, Inherited/diagnosis , Blood Coagulation Disorders, Inherited/genetics , Blood Coagulation Disorders, Inherited/therapy , Blood Coagulation Factors/genetics , Hemorrhage/etiology , Hemorrhage/genetics , Vitamin K
3.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 39(6): 646-650, 2022 Jun 10.
Article in Chinese | MEDLINE | ID: mdl-35773773

ABSTRACT

γ-glutamyl carboxylase (GGCX), also known as vitamin K-dependent glutamyl carboxylase, catalyzes the posttranslational modification of specific glutamate residues in vitamin K-dependent proteins (VKDPs), and participates multiple biological functions including blood coagulation, bone metabolism, vascular calcification, and cell proliferation. It has been reported originally that GGCX pathogenic variation causes blood coagulation deficiency, which is called as vitamin K-dependent coagulation factor deficiency 1 (VKCFD1). Recently, it has been found that GGCX gene variation results in multiple clinical phenotypes, including dermatological, ophthalmological, skeletal or cardiac abnormalities. Among them, dermatological phenotype is the most common, which is known as pseudoxanthoma elasticum-like syndrome. This paper has reviewed the GGCX pathogenic variation associated phenotypes, in order to increase the recognition of GGCX-related genetic diseases and to help its diagnosis and treatment.


Subject(s)
Blood Coagulation Disorders, Inherited , Carbon-Carbon Ligases , Blood Coagulation Disorders, Inherited/diagnosis , Blood Coagulation Disorders, Inherited/enzymology , Blood Coagulation Disorders, Inherited/genetics , Blood Coagulation Disorders, Inherited/metabolism , Carbon-Carbon Ligases/genetics , Carbon-Carbon Ligases/metabolism , Carboxy-Lyases/genetics , Carboxy-Lyases/metabolism , Humans , Phenotype , Vitamin K/metabolism , Vitamin K 1
4.
J Thromb Haemost ; 20(7): 1568-1575, 2022 07.
Article in English | MEDLINE | ID: mdl-35621921

ABSTRACT

Individuals with inherited bleeding disorders (IBDs) have higher bleeding risk during pregnancy, childbirth, and the postpartum period. Clinical management requires recognition of the IBD as high risk for postpartum hemorrhage and a personalized multidisciplinary approach that includes the patient in decision making. When the fetus is known or at risk to inherit a bleeding disorder, fetal and neonatal bleeding risk also need to be considered. In pregnant IBD patients, it is common for providers to need to make decisions in the absence of high level of certainty evidence. We here present the case of a pregnant von Willebrand disease patient that reached multiple decision points where there is currently clinical ambiguity due to a lack of high level of certainty evidence. For each stage of her care, from diagnosis to the postpartum period, we discuss current literature and describe our approach. This is followed by a brief overview of considerations in other IBDs and pregnancy.


Subject(s)
Blood Coagulation Disorders, Inherited , Hemorrhagic Disorders , Inflammatory Bowel Diseases , Postpartum Hemorrhage , von Willebrand Diseases , Blood Coagulation Disorders, Inherited/complications , Blood Coagulation Disorders, Inherited/diagnosis , Blood Coagulation Disorders, Inherited/genetics , Female , Humans , Infant, Newborn , Postpartum Hemorrhage/diagnosis , Postpartum Hemorrhage/etiology , Postpartum Hemorrhage/therapy , Pregnancy , von Willebrand Diseases/complications , von Willebrand Diseases/diagnosis , von Willebrand Diseases/therapy
5.
Int J Mol Sci ; 23(2)2022 Jan 12.
Article in English | MEDLINE | ID: mdl-35054981

ABSTRACT

Vitamin K dependent coagulation factor deficiency type 1 (VKCFD1) is a rare hereditary bleeding disorder caused by mutations in γ-Glutamyl carboxylase (GGCX) gene. The GGCX enzyme catalyzes the γ-carboxylation of 15 different vitamin K dependent (VKD) proteins, which have function in blood coagulation, calcification, and cell signaling. Therefore, in addition to bleedings, some VKCFD1 patients develop diverse non-hemorrhagic phenotypes such as skin hyper-laxity, skeletal dysmorphologies, and/or cardiac defects. Recent studies showed that GGCX mutations differentially effect γ-carboxylation of VKD proteins, where clotting factors are sufficiently γ-carboxylated, but not certain non-hemostatic VKD proteins. This could be one reason for the development of diverse phenotypes. The major manifestation of non-hemorrhagic phenotypes in VKCFD1 patients are mineralization defects. Therefore, the mechanism of regulation of calcification by specific VKD proteins as matrix Gla protein (MGP) and Gla-rich protein (GRP) in physiological and pathological conditions is of high interest. This will also help to understand the patho-mechanism of VKCFD1 phenotypes and to deduce new treatment strategies. In the present review article, we have summarized the recent findings on the function of GRP and MGP and how these proteins influence the development of non-hemorrhagic phenotypes in VKCFD1 patients.


Subject(s)
Blood Coagulation Disorders, Inherited/diagnosis , Blood Coagulation Disorders, Inherited/etiology , Calcium-Binding Proteins/metabolism , Extracellular Matrix Proteins/metabolism , Mutation , Phenotype , Alleles , Animals , Biomarkers , Blood Coagulation , Blood Coagulation Disorders, Inherited/genetics , Blood Coagulation Disorders, Inherited/metabolism , Calcification, Physiologic/genetics , Carrier Proteins/metabolism , Disease Models, Animal , Genetic Association Studies , Genetic Predisposition to Disease , Genotype , Humans , Protein Binding , Matrix Gla Protein
6.
Platelets ; 33(2): 320-323, 2022 Feb 17.
Article in English | MEDLINE | ID: mdl-33616470

ABSTRACT

Familial Platelet Disorder with associated Myeloid Malignancy (FPDMM) is a rare inherited disorder confirmed with the presence of a pathogenic germline RUNX1 variant and is thought to be heavily underdiagnosed. RUNX1 has also been found to be mutated in up to 10% of adult AML cases and other cell malignancies. We performed targeted next-generation sequencing and subsequent MLPA analysis in a kindred with multiple affected individuals with low platelet counts and a bleeding history. We detected a novel heterozygous exon 3-7 large deletion in the RUNX1 gene in all affected family members which is predicted to remove all of the Runt-homology DNA-binding domain and a portion of the Activation domain. Our results show that the combination of targeted NGS and MLPA analysis is an effective way to detect copy number variants (CNVs) which would be missed by conventional sequencing methods. This precise diagnosis offers the possibility of accurate counseling and clinical management in such patients who could go onto develop other cell malignancies.


Subject(s)
Blood Coagulation Disorders, Inherited/genetics , Blood Platelet Disorders/genetics , Core Binding Factor Alpha 2 Subunit/genetics , Exons/genetics , High-Throughput Nucleotide Sequencing/methods , Leukemia, Myeloid, Acute/genetics , Adult , Aged , Genetic Predisposition to Disease , Humans , Male , Young Adult
7.
Hum Mutat ; 43(1): 42-55, 2022 01.
Article in English | MEDLINE | ID: mdl-34816548

ABSTRACT

γ-Glutamyl carboxylase (GGCX) catalyzes the γ-carboxylation of 15 different vitamin K dependent (VKD) proteins. Pathogenic variants in GGCX cause a rare hereditary bleeding disorder called Vitamin K dependent coagulation factor deficiency type 1 (VKCFD1). In addition to bleedings, some VKCFD1 patients develop skin laxity and skeletal dysmorphologies. However, the pathophysiological mechanisms underlying these non-hemorrhagic phenotypes remain elusive. Therefore, we have analyzed 20 pathogenic GGCX variants on their ability to γ-carboxylate six non-hemostatic VKD proteins in an in vitro assay, where GGCX variants were expressed in GGCX-/- cells and levels of γ-carboxylated co-expressed VKD proteins were detected by a functional ELISA. We observed that GGCX variants causing markedly reduced γ-carboxylation of Gla rich protein (GRP) in vitro were reported in patients with skin laxity. Reduced levels of γ-carboxylated Matrix gla protein (MGP) are not exclusive for causing skeletal dysmorphologies in VKCFD1 patients. In silico docking of vitamin K hydroquinone on a GGCX model revealed a binding site, which was validated by in vitro assays. GGCX variants affecting this site result in disability to γ-carboxylate VKD proteins and hence are involved in the most severe phenotypes. This genotype-phenotype analysis will help to understand the development of non-hemorrhagic phenotypes and hence improve treatment in VKCFD1 patients.


Subject(s)
Blood Coagulation Disorders, Inherited , Carbon-Carbon Ligases , Blood Coagulation Disorders, Inherited/genetics , Carbon-Carbon Ligases/chemistry , Carbon-Carbon Ligases/genetics , Carbon-Carbon Ligases/metabolism , Carboxy-Lyases , Humans , Mutation
8.
Genes (Basel) ; 12(11)2021 11 18.
Article in English | MEDLINE | ID: mdl-34828413

ABSTRACT

Inherited bleeding disorders (IBDs) are the most frequent congenital diseases in the Colombian population; three of them are hemophilia A (HA), hemophilia B (HB), and von Willebrand Disease (VWD). Currently, diagnosis relies on multiple clinical laboratory assays to assign a phenotype. Due to the lack of accessibility to these tests, patients can receive an incomplete diagnosis. In these cases, genetic studies reinforce the clinical diagnosis. The present study characterized the molecular genetic basis of 11 HA, three HB, and five VWD patients by sequencing the F8, F9, or the VWF gene. Twelve variations were found in HA patients, four in HB patients, and 19 in WVD patients. From these variations a total of 25 novel variations were found. Disease-causing variations were used as positive controls for validation of the high-resolution melting (HRM) variant-scanning technique. This approach is a low-cost genetic diagnostic method proposed to be incorporated in developing countries. For the data analysis, we developed an accessible open-source code in Python that improves HRM data analysis with better sensitivity of 95% and without bias when using different HRM equipment and software. Analysis of amplicons with a length greater than 300 bp can be performed by implementing an analysis by denaturation domains.


Subject(s)
Blood Coagulation Disorders, Inherited/diagnosis , Computational Biology/methods , Factor IX/genetics , Genetic Testing/methods , Hemophilia A/genetics , von Willebrand Factor/genetics , Blood Coagulation Disorders, Inherited/genetics , Colombia , Computational Biology/economics , Computational Biology/standards , Costs and Cost Analysis , Factor IX/chemistry , Genetic Testing/economics , Genetic Testing/standards , Hemophilia A/diagnosis , Humans , Protein Domains , Sensitivity and Specificity , von Willebrand Factor/chemistry
10.
Indian J Pathol Microbiol ; 64(4): 741-745, 2021.
Article in English | MEDLINE | ID: mdl-34673595

ABSTRACT

OBJECTIVES: The spectrum of thrombophilia in women with recurrent pregnancy loss (RPL) is different in Indian ethnicity as reported by few studies. We aimed to study the prevalence of thrombophilia in RPL patients referred to hematology department of a tertiary centre. MATERIAL AND METHODS: This is an observational study of 112 RPL patients with no apparent cause after extensive workup for non-hematological causes. The investigations performed were routine coagulogram, APLA workup, plasma homocysteine, MTHFRC677T polymorphisms, Protein C, free Protein S, Anti-thrombin III levels, test for Activated Protein C resistance (APC-R) ,Factor V Leiden and Prothrombin gene G20210A mutation. RESULTS: Of 112 patients, at least one thrombophilia was identified in 70.5% and combined thrombophilia in 12.5% patients. Hyperhomocysteinemia (30.4%) and APLA (25.9%) were the commonest thrombophilia whereas anticoagulant defects were seen in 12.5% of the population. Protein C deficiency (5.35%) was the commonest anticoagulant defect followed by APCR (3.6%). Mutational analysis revealed MTHFRC677T polymorphism in 20.5% whereas Factor V Leiden heterozygous in 1.8% patients. None of the patients had homozygous Factor V Leiden or Prothrombin gene G20210A mutation. Hyperhomocysteinemia, MTHFRC677T and Protein C deficiency were more associated with early pregnancy losses whereas Protein S deficiency, Factor V Leiden and APLA caused both early and late losses. Patients with greater number of losses were positive for homozygous MTHFRC677T, factor V Leiden and APLA. CONCLUSION: The approach to investigating Indian women with RPL should be based on the prevalence of thrombophilia which is unique to Indian ethnicity.


Subject(s)
Abortion, Habitual/genetics , Asian People/genetics , Blood Coagulation Disorders, Inherited/genetics , Ethnicity/genetics , Thrombophilia/diagnosis , Thrombophilia/epidemiology , Thrombophilia/genetics , Adolescent , Adult , Asian People/statistics & numerical data , Case-Control Studies , Ethnicity/statistics & numerical data , Female , Genetic Predisposition to Disease , Humans , India/epidemiology , Pregnancy , Prothrombin , Thrombophilia/physiopathology , Young Adult
11.
Int J Mol Sci ; 22(18)2021 Sep 08.
Article in English | MEDLINE | ID: mdl-34575869

ABSTRACT

Factor V is an essential clotting factor that plays a key role in the blood coagulation cascade on account of its procoagulant and anticoagulant activity. Eighty percent of circulating factor V is produced in the liver and the remaining 20% originates in the α-granules of platelets. In humans, the factor V gene is about 80 kb in size; it is located on chromosome 1q24.2, and its cDNA is 6914 bp in length. Furthermore, nearly 190 mutations have been reported in the gene. Factor V deficiency is an autosomal recessive coagulation disorder associated with mutations in the factor V gene. This hereditary coagulation disorder is clinically characterized by a heterogeneous spectrum of hemorrhagic manifestations ranging from mucosal or soft-tissue bleeds to potentially fatal hemorrhages. Current treatment of this condition consists in the administration of fresh frozen plasma and platelet concentrates. This article describes the cases of two patients with severe factor V deficiency, and of their parents. A high level of mutational heterogeneity of factor V gene was identified, nonsense mutations, frameshift mutations, missense changes, synonymous sequence variants and intronic changes. These findings prompted the identification of a new mutation in the human factor V gene, designated as Jaén-1, which is capable of altering the procoagulant function of factor V. In addition, an update is provided on the prospects for the treatment of factor V deficiency on the basis of yet-to-be-developed recombinant products or advanced gene and cell therapies that could potentially correct this hereditary disorder.


Subject(s)
DNA Mutational Analysis , Factor V Deficiency/genetics , Factor V Deficiency/therapy , Factor V/genetics , Adolescent , Blood Coagulation , Blood Coagulation Disorders, Inherited/genetics , Blood Coagulation Tests , Blood Platelets/metabolism , Child, Preschool , Codon, Nonsense , DNA, Complementary/metabolism , Family Health , Female , Frameshift Mutation , Humans , Male , Pakistan , Recombinant Proteins/chemistry , Sequence Analysis, DNA , Spain
12.
Angiology ; 72(9): 861-866, 2021 10.
Article in English | MEDLINE | ID: mdl-33783233

ABSTRACT

Autologous cell therapy (ACT) is a new treatment for patients with no-option critical limb ischemia (NO-CLI). We evaluated the factors involved in the nonresponse to ACT in patients with CLI and diabetic foot. Diabetic patients (n = 72) with NO-CLI treated using ACT in our foot clinic over a period of 8 years were divided into responders (n = 57) and nonresponders (n = 15). Nonresponder was defined as an insufficient increase in transcutaneous oxygen pressure by <5 mm Hg, 3 months after ACT. Patient demographics, diabetes duration and treatment, and comorbidities as well as a cellular response to ACT, limb-related factors, and the presence of inherited thrombotic disorders were compared between the 2 groups. The main independent predictors for an impaired response to ACT were heterozygote Leiden mutation (OR 10.5; 95% CI, 1.72-4) and homozygote methylenetetrahydrofolate reductase (MTHFR 677) mutation (OR 3.36; 95% CI, 1.0-14.3) in stepwise logistic regression. Univariate analysis showed that lower mean protein C levels (P = .041) were present in nonresponders compared with responders. In conclusion, the significant predictors of an impaired response to ACT in diabetic patients with NO-CLI were inherited thrombotic disorders.


Subject(s)
Blood Coagulation Disorders, Inherited/complications , Cell Transplantation , Diabetic Foot/surgery , Ischemia/surgery , Activated Protein C Resistance/complications , Activated Protein C Resistance/genetics , Aged , Blood Coagulation Disorders, Inherited/diagnosis , Blood Coagulation Disorders, Inherited/genetics , Cell Transplantation/adverse effects , Critical Illness , Diabetic Foot/complications , Diabetic Foot/diagnosis , Factor V/genetics , Female , Heterozygote , Homozygote , Humans , Ischemia/complications , Ischemia/diagnosis , Male , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Middle Aged , Mutation , Risk Assessment , Risk Factors , Transplantation, Autologous , Treatment Failure
13.
J Hum Genet ; 66(8): 753-759, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33564083

ABSTRACT

The arachidonic acid (AA) cascade plays a significant role in platelet aggregation. AA released from membrane phospholipids is metabolized by cyclooxygenase (COX) pathway to thromboxane A2 (TXA2) or by 12S-lipoxygenase (ALOX12) to 12-hydroperoxyeicosatetraenoic acid (12-HPETE). In contrast to a well-known role of the COX pathway in platelet aggregation, the role of ALOX12 is not well understood. Platelets of ALOX12-deficient mice exhibit increased sensitivity for ADP-induced aggregation. However, recent evidence strongly suggests a significant role of ALOX12 in platelet aggregation and calcium signaling. 12-HPETE potentiates thrombin- and thromboxane-induced platelet aggregation, and calcium signaling. Inhibition experiments of ALOX12 demonstrated decreased platelet aggregation and calcium signaling in stimulated platelets. We studied a family with a dominantly inherited bleeding diathesis using next-generation sequencing analysis. Platelet aggregation studies revealed that the proband's platelets had defective aggregation responses to ADP, TXA2 mimetic U46619, collagen, and AA, normal affinity of TXA2 receptor for U46619, and normal induction of GTPase activity upon stimulation with U46619. However, the production of inositol 1,4,5-triphosphate (IP3) was only increased up to 30% of the control upon U46619 stimulation, suggesting a defect in phospholipase C-ß2 (PLCB2) activation downstream from TXA2 receptors. Affected family members had no mutation of PLCB2, but had a heterozygous c.1946A > G (p.Tyr649Cys) mutation of ALOX12. ALOX12 activity in platelets from the affected members was decreased to 25-35% of the control. Our data strongly suggested that a heterozygous c.1946A > G ALOX12 mutation was a disease-causing mutation; however, further experiments are required to confirm the pathogenesis of ALOX12 mutation in platelet aggregation.


Subject(s)
Arachidonate 12-Lipoxygenase/genetics , Blood Coagulation Disorders, Inherited/genetics , Blood Platelets/physiology , Genetic Predisposition to Disease , Hemorrhage/genetics , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/metabolism , Arachidonic Acid/metabolism , Calcium/metabolism , Disease Susceptibility , GTP Phosphohydrolases/metabolism , Hemorrhage/metabolism , High-Throughput Nucleotide Sequencing , Humans , Inositol 1,4,5-Trisphosphate/metabolism , Mutation , Pedigree , Phospholipase C beta/metabolism , Platelet Aggregation , Prostaglandin-Endoperoxide Synthases/metabolism , Signal Transduction , Thromboxane A2/metabolism
14.
Blood Coagul Fibrinolysis ; 31(1S): S6-S8, 2020 Dec 01.
Article in English | MEDLINE | ID: mdl-33351494

ABSTRACT

Hemophilia and other hereditary coagulopathies tend to be associated with a huge negative impact both for individuals who suffer the disease and for their families. In this respect, hemophilia carriers feel the need to make reproductive decisions which will inevitably affect their children, their families and from themselves. Genetic and reproductive counseling is of the essence to alleviate these women's distress. Prenatal diagnosis and preimplantation genetic diagnosis (PGD) allow couples at high-risk of transmitting genetic diseases like hemophilia and other hereditary coagulopathies to prevent the birth of children with the disease. The main difference between prenatal diagnosis and PGD is related to the time at which diagnosis is made. Prenatal diagnosis is done when the woman is pregnant, and both the performance of the technique and its result can affect the course of pregnancy. PGD is a diagnostic procedure in which embryos created in vitro are analyzed for genetic defects before being transferred to the uterus. Performance of both prenatal diagnosis and PGD is subject to a few prerequisites: the establishment of an exact clinical diagnosis, an understanding of the parental genetic alterations that are responsible for the disease and technical feasibility of genetic diagnosis. These couples should be provided with complete, up-to-date and easy-to-understand information.


Subject(s)
Blood Coagulation Disorders, Inherited/genetics , Fertilization in Vitro , Genetic Testing , Prenatal Diagnosis , Female , Genetic Counseling/methods , Genetic Testing/methods , Hemophilia A/genetics , Humans , Male , Preimplantation Diagnosis/methods , Prenatal Diagnosis/methods
16.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 37(8): 811-814, 2020 Aug 10.
Article in Chinese | MEDLINE | ID: mdl-32761584

ABSTRACT

OBJECTIVE: To develop a cell-based system for the diagnosis of vitamin K-dependent coagulation factor deficiency 1 (VKCFD1). METHODS: In HEK293 cells stably expressing the reporter gene FIX-Gla-PC, the gamma-glutamyl carboxylase (GGCX) gene was knocked out by using CRISPR/Cas9 technology. Enzyme-linked immunosorbent assay (ELISA), DNA sequencing and Western blotting were used to identify the GGCX gene knockout cells. A quickchange point variant method was used to construct the GGCX variant. ELISA was used to assess the influence of GGCX variant on the activity of reporter gene. RESULTS: Two monoclonal cell lines with no reporter activity by ELISA was identified. Edition and knockout of the GGCX gene was confirmed by DNA sequencing and Western blotting. The activity of the reporter gene was recovered by transfection of the wild-type GGCX gene. Thereby two monoclonal cells with GGCX knockout were obtained. By comparing the wild-type and pathogenic GGCX variants, the reporter activity was decreased in the pathogenic variants significantly. CONCLUSION: A cell-based system for the detection of GGCX activity was successfully developed, which can be used for the diagnosis of VKCFD1 caused by GGCX variants.


Subject(s)
Blood Coagulation Disorders, Inherited/genetics , Carbon-Carbon Ligases/genetics , Vitamin K 1 , Base Sequence , Blood Coagulation Disorders, Inherited/diagnosis , CRISPR-Cas Systems , Gene Knockout Techniques , HEK293 Cells , Humans
17.
J Clin Invest ; 130(10): 5302-5312, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32663190

ABSTRACT

Tissue factor (TF) is the primary initiator of blood coagulation in vivo and the only blood coagulation factor for which a human genetic defect has not been described. As there are no routine clinical assays that capture the contribution of endogenous TF to coagulation initiation, the extent to which reduced TF activity contributes to unexplained bleeding is unknown. Using whole genome sequencing, we identified a heterozygous frameshift variant (p.Ser117HisfsTer10) in F3, the gene encoding TF, causing premature termination of TF (TFshort) in a woman with unexplained bleeding. Routine hematological laboratory evaluation of the proposita was normal. CRISPR-edited human induced pluripotent stem cells recapitulating the variant were differentiated into vascular smooth muscle and endothelial cells that demonstrated haploinsufficiency of TF. The variant F3 transcript is eliminated by nonsense-mediated decay. Neither overexpression nor addition of exogenous recombinant TFshort inhibited factor Xa or thrombin generation, excluding a dominant-negative mechanism. F3+/- mice provide an animal model of TF haploinsufficiency and exhibited prolonged bleeding times, impaired thrombus formation, and reduced survival following major injury. Heterozygous TF deficiency is present in at least 1 in 25,000 individuals and could limit coagulation initiation in undiagnosed individuals with abnormal bleeding but a normal routine laboratory evaluation.


Subject(s)
Blood Coagulation Disorders, Inherited/blood , Blood Coagulation Disorders, Inherited/genetics , Frameshift Mutation , Thromboplastin/deficiency , Thromboplastin/genetics , Animals , Base Sequence , Codon, Nonsense , Disease Models, Animal , Female , Gene Editing , Haploinsufficiency , Heterozygote , Humans , Induced Pluripotent Stem Cells , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Peptide Chain Termination, Translational , Phenotype
19.
Cytometry B Clin Cytom ; 98(6): 464-475, 2020 11.
Article in English | MEDLINE | ID: mdl-32516490

ABSTRACT

Inherited platelet function disorders are rare hemorrhagic diseases. The gold standard for their exploration is optical aggregometry; however, investigations by flow cytometry (FCM) are being increasingly used. In this review, the physiology of platelets is first recalled, setting the stage for the compartments of platelets that can be apprehended by specific and appropriate labeling. As this requires some pre-analytical precautions and specific analytical settings, a second part focuses on these characteristic aspects, based on literature and on the authors' experience in the field, for qualitative or quantitative explorations. Membrane labeling with antibodies to CD42a or CD41, respectively, useful to assess the genetic-related defects of Glanzmann thrombocytopenia and Bernard Soulier syndrome are then described. Platelet degranulation disorders are detailed in the next section, as they can be explored, upon platelet activation, by measuring the expression of surface P-Selectin (CD62P) or CD63. Mepacrin uptake and release after activation is another test allowing to explore the function of dense granules. Finally, the flip-flop anomaly related to Scott syndrome is depicted. Tables summarizing possible FCM assays, and characteristic histograms are provided as reference for flow laboratories interested in developing platelet exploration.


Subject(s)
Blood Coagulation Disorders, Inherited/blood , Blood Platelet Disorders/blood , Flow Cytometry , Immunophenotyping , Blood Coagulation Disorders, Inherited/drug therapy , Blood Coagulation Disorders, Inherited/genetics , Blood Coagulation Disorders, Inherited/immunology , Blood Platelet Disorders/drug therapy , Blood Platelet Disorders/immunology , Blood Platelet Disorders/pathology , Blood Platelets/drug effects , Blood Platelets/immunology , Blood Platelets/pathology , Humans , Platelet Activation/genetics , Platelet Activation/immunology , Quinacrine/therapeutic use
20.
J Thromb Haemost ; 18(8): 1839-1845, 2020 08.
Article in English | MEDLINE | ID: mdl-32521110

ABSTRACT

Inherited bleeding and platelet disorders (BPD) are highly heterogeneous and their diagnosis involves a combination of clinical investigations, laboratory tests, and genetic screening. This review will outline some of the challenges that geneticists and experts in clinical hemostasis face when implementing high-throughput sequencing (HTS) for patient care. We will provide an overview of the strengths and limitations of the different HTS techniques that can be used to diagnose BPD. An HTS test is cost-efficient and expected to increase the diagnostic rate with a possibility to detect unexpected diagnoses and decrease the turnaround time to diagnose patients. On the other hand, technical shortcomings, variant interpretation difficulties, and ethical issues related to HTS for BPD will also be documented. Delivering a genetic diagnosis to patients is highly desirable to improve clinical management and allow family counseling, but making incorrect assumptions about variants and providing insufficient information to patients before initiating the test could be harmful. Data-sharing and improved HTS guidelines are essential to limit these major drawbacks of HTS.


Subject(s)
Blood Coagulation Disorders, Inherited , Blood Platelet Disorders , Blood Coagulation Disorders, Inherited/diagnosis , Blood Coagulation Disorders, Inherited/genetics , Blood Platelet Disorders/diagnosis , Blood Platelet Disorders/genetics , Blood Platelets , Genetic Testing , High-Throughput Nucleotide Sequencing , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...