Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Akiko Sekiya is active.

Publication


Featured researches published by Akiko Sekiya.


Thrombosis Research | 2012

Carbon monoxide (CO)-releasing molecule-derived CO regulates tissue factor and plasminogen activator inhibitor type 1 in human endothelial cells

Keiko Maruyama; Eriko Morishita; Takeo Yuno; Akiko Sekiya; Hidesaku Asakura; Shigeki Ohtake; Akihiro Yachie

INTRODUCTION Heme oxygenase-1 (HO-1) is the rate limiting enzyme that catalyzes the conversion of heme into biliverdin, free iron, and carbon monoxide (CO). The first human case of HO-1 deficiency showed abnormalities in blood coagulation and the fibrinolytic system. Thus, HO-1 or HO-1 products, such as CO, might regulate coagulation and the fibrinolytic system. This study examined whether tricarbonyldichlororuthenium (II) dimer (CORM-2), which liberates CO, modulates the expression of tissue factor (TF) and plasminogen activator inhibitor type 1 (PAI-1) in human umbilical vein endothelial cells (HUVECs), and TF expression in peripheral blood mononuclear cells (PBMCs). Additionally, we examined the mechanism by which CO exerts its effects. MATERIALS AND METHODS HUVECs were pretreated with 50 μM CORM-2 for 3 hours, and stimulated with tumor necrosis factor-α (TNF-α, 10 ng/ml) for an additional 0-5 hours. PBMCs were pretreated with 50-100 μM CORM-2 for 1 hour followed by stimulating with lipopolysaccharid (LPS, 10 ng/ml) for additional 0-9 hours. The mRNA and protein levels were determined by RT-PCR and western blotting, respectively. RESULTS Pretreatment with CORM-2 significantly inhibited TNF-α-induced TF and PAI-1 up-regulation in HUVECs, and LPS-induced TF expression in PBMCs. CORM-2 inhibited TNF-α-induced activation of p38 MAPK, ERK1/2, JNK, and NF-κB signaling pathways in HUVECs. CONCLUSIONS CORM-2 suppresses TNF-α-induced TF and PAI-1 up-regulation, and MAPKs and NF-κB signaling pathways activation by TNF-α in HUVECs. CORM-2 suppresses LPS-induced TF up-regulation in PBMCs. Therefore, we envision that the antithrombotic activity of CORM-2 might be used as a pharmaceutical agent for the treatment of various inflammatory conditions.


PLOS ONE | 2015

ELISA-Based Detection System for Protein S K196E Mutation, a Genetic Risk Factor for Venous Thromboembolism

Keiko Maruyama; Masashi Akiyama; Koichi Kokame; Akiko Sekiya; Eriko Morishita; Toshiyuki Miyata

Protein S (PS) acts as a cofactor for activated protein C in the plasma anticoagulant system. PS Lys196-to-Glu (K196E) mutation is a genetic risk factor for venous thromboembolism in Japanese individuals. Because of the substantial overlap in PS anticoagulant activity between KK (wild-type) and KE (heterozygous) genotypes, it is difficult to identify PS K196E carriers by measuring PS activity. Here, we generated monoclonal antibodies specific to the PS K196E mutant and developed a simple and reliable method for the identification of PS K196E carriers. We immunized mice with a keyhole limpet hemocyanin-conjugated synthetic peptide with Glu196. The hybridoma cells were screened for the binding ability of the produced antibodies to recombinant mutant EGF-like domains of PS (Ile117–Glu283). We obtained three hybridoma cell lines producing PS K196E mutation-specific antibodies. We established a sandwich enzyme-linked immunosorbent assay (ELISA) system in which the PS K196E mutation-specific monoclonal antibody was used as a detection antibody. We measured human plasma samples by using this system and successfully discriminated 11 individuals with the KE genotype from 122 individuals with the KK genotype. The ELISA system using the PS K196E mutation-specific antibody is a useful tool for the rapid identification of PS K196E carriers, who are at a higher risk for venous thromboembolism.


Thrombosis Research | 2013

Antithrombin deficiency in three Japanese families: one novel and two reported point mutations in the antithrombin gene.

Keiko Maruyama; Eriko Morishita; Megumi Karato; Tadaaki Kadono; Akiko Sekiya; Yukie Goto; Tomomi Sato; Haruka Nomoto; Wataru Omi; Sachie Tsuzura; Hidenori Imai; Hidesaku Asakura; Shigeki Ohtake; Shinji Nakao

INTRODUCTION Inherited antithrombin (AT) deficiency is associated with a predisposition to familial venous thromboembolic disease. We analyzed the AT gene in three unrelated patients with an AT deficiency who developed thrombosis. MATERIALS AND METHODS We analyzed the SERPINC1 gene in three patients. Additionally, we expressed the three mutants in the COS-1 cells and compared their secretion rates and levels of AT activity with those of the wild-type (WT). RESULTS We identified three distinct heterozygous mutations of c.2534C>T: p.56Arginine → Cysteine (R56C), c.13398C>A: p.459Alanine → Aspartic acid (A459D) and c.2703C>G: p.112 Proline → Arginine (P112R). In the in vitro expression experiments, the AT antigen levels in the conditioned media (CM) of the R56C mutant were nearly equal to those of WT. In contrast, the AT antigen levels in the CM of the A459D and P112R mutants were significantly decreased. The AT activity of R56C was decreased in association with a shorter incubation time in a FXa inhibition assay and a thrombin inhibition-based activity test. However, the AT activity of R56C was comparable to that of WT when the incubation time was increased. CONCLUSIONS We concluded that the R56C mutant is responsible for type II HBS deficiency. We considered that the A459D and P112R mutants can be classified as belonging to the type I AT deficiency.


International Journal of Hematology | 2011

Two case reports of inherited antithrombin deficiency: a novel frameshift mutation and a large deletion including all seven exons detected using two methods.

Akiko Sekiya; Eriko Morishita; Megumi Karato; Keiko Maruyama; Itsumi Shimogawara; Mika Omote; Yoshiyuki Wakugawa; Moeko Shinohara; Tomoe Hayashi; Yasuko Kadohira; Hidesaku Asakura; Shinji Nakao; Shigeki Ohtake

An inherited antithrombin deficiency is an autosomal dominant thrombotic disorder. We identified two pedigrees of inherited type I antithrombin deficiency and two responsible mutations in each. A novel 21–22delAA appeared to have caused a frameshift with a premature termination at amino acid +63 in one patient and a large deletion including all seven exons was identified by multiplex ligation-dependent probe amplification in the other. Some asymptomatic relatives of the second patient had the same mutation. The present findings support the value of using more than one method of gene analysis and of studying the families of probands with inherited thrombotic disorders.


Thrombosis Research | 2015

Late onset thrombosis in two Japanese patients with compound heterozygote protein S deficiency

Fumina Taniguchi; Eriko Morishita; Akiko Sekiya; Daisuke Yamaguchi; Haruka Nomoto; Erina Kobayashi; Mao Takata; Ikuko Kosugi; Nobuyasu Takeuchi; Hidesaku Asakura; Shigeki Ohtake

Protein S (PS; gene symbol PROS1; MIM #176880) is a vitamin Kdependent plasma glycoprotein that acts as a nonenzymic cofactor for activated protein C (APC) in the degradation of activated coagulation factors V and VIII [1]. PS is a single-chain protein of 676 amino acids that consists of a γ-carboxyglutamic acid (Gla) domain, a thrombinsensitive region (TSR), four epidermal growth factor (EGF)-like domains and a carboxy-terminal region homologous to sex hormone binding globulins (SHBG-like domain). Hereditary PS deficiency is an autosomal dominant disorder associated with venous thromboembolism (VTE) [2]. The prevalence of PS deficiency is 0.03-0.13% in the general Caucasian population [3], whereas it is estimated to be 1.12% in the Japanese population [4]. In particular, the Lys196Glu classified qualitative PS deficiency has been reported to be a polymorphism in the Japanese population, known as PS Tokushima [5]. A homozygous or a compound heterozygote mutation for PROS1 gene has been reported in several families, but it is rare [6]. Generally, patients with a homozygote or compound heterozygote mutation for PROS1 developed life-threatening thrombosis, for example, neonatal purpura fulminans. Two patients with late-onset thrombosis who had compound heterozygote PS deficiency of non PS Tokushima type, which is reported to have a high frequency in Japanese, are reported. Proband 1 was a 14 year-old Japanese girl who suffered from deep vein thrombosis (DVT) from the right calf to the right common iliac vein and pulmonary embolism (PE) involving bilateral lungs (Fig. 1). She had been thrombosis-free for 14 years. Although the trigger of her thrombosis was unclear, dehydration due to exercise might cause development of thrombosis because she belonged to an ice hockey team. Plasma levels of antithrombin activity, protein C activity, PS activity (Staclot Protein S; Clot, Roche, Basel, Switzerland), total PS antigen (Asserachrom Total Protein S;Roche) and free PS antigen (STA(R)Liatest(R)Free Protein S; Roche) were 128%, 78%, b10%, 30%, and 8%, respectively. Tests were donewhile the patientwas onwarfarin therapy. Concentration of antiphospholipid antibodies (including anticardiolipin and anti-beta-2-glycoprotein I antibodies) were below the respective cut off values, and screening tests for lupus anticoagulant (LA) were negative. Her parents and her two brothers showed low PS activities (Fig. 2A). However, her family historywas negative for thrombotic episodes, but her mother had three miscarriages and her younger brother has epilepsy. Proband 2 was an 18-year-old Japanese man who suffered from cerebral venous sinus thrombosis. The trigger of thrombosis was unclear. Plasma levels of antithrombin activity, protein C activity, PS activity, total PS antigen and free antigen were 115%, 110%, b10%, 47% and 9% respectively. Concentration of antiphospholipid antibodies (including anti-cardiolipin and anti-beta-2-glycoprotein I antibodies)


Clinical and Applied Thrombosis-Hemostasis | 2017

Thrombosis Prediction Based on Reference Ranges of Coagulation-Related Markers in Different Stages of Pregnancy

Akiko Sekiya; Tomoe Hayashi; Yasuko Kadohira; Masami Shibayama; Tomohide Tsuda; Xiuri Jin; Haruka Nomoto; Hidesaku Asakura; Takashi Wada; Shigeki Ohtake; Eriko Morishita

Introduction: Careful monitoring of the hypercoagulable state is required during pregnancy. However, coagulation and fibrinolysis markers are not fully utilized because there are no reference values reflective of coagulation and fibrinolysis dynamics during pregnancy, which differ from the nonpregnant state. Methods: Changes in antithrombin (AT), fibrinogen (Fbg), prothrombin fragment 1+2 (F1+2), thrombin–antithrombin complex (TAT), soluble fibrin (SF), D-dimer (DD), and protein S (PS) were investigated in healthy pregnant women, and reference ranges in the early, mid, late, and end stages of pregnancy were established. Results: The AT was essentially constant throughout pregnancy. The Fbg, F1+2, TAT, and DD increased significantly as pregnancy progressed. In contrast, SF did not show a significant increase throughout the entire pregnancy period. Total PS antigen and total PS activity showed a corresponding decrease from early gestation. When test data in 3 cases in which deep vein thrombosis or intrauterine fetal death occurred during pregnancy were compared to the established reference ranges, all of the cases had multiple markers with values that exceeded the reference ranges. Conclusion: Establishing reference ranges for each week could potentially make it possible to evaluate abnormalities of the coagulation and fibrinolysis systems during pregnancy. Of note, SF might be a useful marker that reflects thrombus formation during pregnancy. Larger-scale studies will be required to establish reference ranges for every gestational week.


International Journal of Hematology | 2017

Causative genetic mutations for antithrombin deficiency and their clinical background among Japanese patients

Akiko Sekiya; Fumina Taniguchi; Daisuke Yamaguchi; Sayaka Kamijima; Shonosuke Kaneko; Shiori Katsu; Miho Hanamura; Mao Takata; Haruka Nakano; Hidesaku Asakura; Shigeki Ohtake; Eriko Morishita

We summarize causative genetic mutations for antithrombin (AT) deficiency and their clinical background in Japanese patients. A total of 19 mutations, including seven novel mutations, were identified. We also summarize clinical symptoms of thrombosis, age at onset, family history, and contributing factors for thrombosis, and review the use of prophylactic anticoagulation in pregnant women with heterozygous type II heparin binding site defects (HBS) AT deficiency. The prevalence of thrombosis in probands with type I AT deficiency (88%) was double that observed in those with type II AT deficiency (50%). The prevalence of thrombotic episodes among family members was also higher for type I AT deficiency subjects (82%) than for those with type II AT deficiency (0%). The most common contributing factor for thrombosis among women with type I AT deficiency was pregnancy. Forty-five percent of women with type I AT deficiency developed thrombotic events before the 20th week of gestation. In contrast, women with type II (HBS) AT deficiency appear to be at a lower risk of thrombosis during pregnancy. In conclusion, thrombotic risk varies among different subtypes. Risk assessments based on genetic/clinical backgrounds may contribute to appropriate diagnosis, treatment, and prophylaxis for patients with AT deficiency.


Journal of Atherosclerosis and Thrombosis | 2015

Fluvastatin Upregulates the Expression of Tissue Factor Pathway Inhibitor in Human Umbilical Vein Endothelial Cells.

Akiko Sekiya; Eriko Morishita; Keiko Maruyama; Hiroki Torishima; Shigeki Ohtake

AIM 3-Hydroxy-3-methylglutaryl coenzyme A reductase inhibitors (statins) are cholesterol-lowering drugs with a variety of pleiotropic effects including antithrombotic properties. Tissue factor pathway inhibitor (TFPI), which is produced predominantly in endothelial cells and platelets, inhibits the initiating phase of clot formation. We investigated the effect of fluvastatin on TFPI expression in cultured endothelial cells. METHODS Human umbilical vein endothelial cells (HUVECs) were treated with fluvastatin (0-10μM). The expression of TFPI mRNA and antigen were detected by RT-PCR and western blotting, respectively. The effects of mevalonate intermediates, small GTP-binding inhibitors, and signal transduction inhibitors were also evaluated to identify which pathway was involved. A luciferase reporter assay was performed to evaluate the effect of fluvastatin on TFPI transcription. The stability of TFPI mRNA was estimated by quantitating its levels after actinomycin D treatment. RESULTS Fluvastatin increased TFPI mRNA expression and antigen in HUVECs. Fluvastatin-induced TFPI expression was reversed by co-treatment with mevalonate or geranylgeranylpyrophosphate (GGPP). NSC23766 and Y-27632 had no effect on TFPI expression. SB203580, GF109203, and LY294002 reduced fluvastatin-induced TFPI upregulation. Moreover, fluvastatin did not significantly affect TFPI promoter activity. TFPI mRNA degradation in the presence of actinomycin D was delayed by fluvastatin treatment. CONCLUSIONS Fluvastatin increases endothelial TFPI expression through inhibition of mevalonate-, GGPP-, and Cdc42-dependent signaling pathways, and activation of the p38 MAPK, PI3K, and PKC pathways. This study revealed unknown mechanisms of the anticoagulant effect of statins and gave a new insight to its therapeutic potential for the prevention of thrombotic diseases.


International Journal of Hematology | 2018

Gene analysis of inherited antithrombin deficiency and functional analysis of abnormal antithrombin protein (N87D)

Sayaka Kamijima; Akiko Sekiya; Mao Takata; Haruka Nakano; Morika Murakami; Tomonori Nakazato; Hidesaku Asakura; Eriko Morishita

Inherited antithrombin (AT) deficiency is one of the most clinically significant forms of congenital thrombophilia and follows an autosomal dominant mode of inheritance. We analyzed SERPINC1 in a patient who developed deep-vein thrombosis and low AT activity during pregnancy, and identified a novel missense mutation c.259A>G (p.Asn87Asp; N87D). Surprisingly, analysis of the parents’ DNA showed that they did not possess this mutant, and thus, it may have been due to a de novo mutation. We also expressed this mutant AT protein in COS-1 cells and compared its intracellular localization and intracellular and extracellular antigen levels with that of wild-type AT. The expression experiment did not reveal a significant difference in the antigen levels of the mutant and wild-type AT in the cell lysate, but the mutant AT antigen level was markedly lower than that of its wild-type counterpart in the COS-1 cell supernatant. Immunofluorescence did not indicate any difference between the mutant and wild-type AT in terms of cytoplasmic localization of fluorescence signals. Our findings suggest that the patient’s AT deficiency may have been caused by impaired extracellular secretion of mutant AT protein p.Asn87Asp.


Haemophilia | 2018

Congenital coagulation factor X deficiency: Genetic analysis of five patients and functional characterization of mutant factor X proteins

Satomi Nagaya; Masashi Akiyama; Morika Murakami; Akiko Sekiya; Hidesaku Asakura; Eriko Morishita

Congenital factor X (FX) deficiency is a rare bleeding disorder that is inherited as an autosomal recessive trait. In this study, a genetic analysis of the FX gene was performed in five families with this disorder. Four heterozygous mutations [p.Gly154Arg, p.Val236Met, p.Gly263Val and p.Arg387Cys] and one pair of compound heterozygous FX gene mutations consisting of p.Gly406Ser and p.Val424Phe were identified. Mutant FX proteins containing the identified amino acid substitutions were also expressed in cultured cells. These proteins were analysed by enzyme‐linked immunosorbent assay and pulse‐chase experiments. The results demonstrated normal intracellular synthesis and extracellular secretion of mutant FX proteins carrying the p.Val236Met, p.Arg387Cys and p.Gly406Ser amino acid substitutions. However, the results also showed that the p.Gly154Arg, p.Gly263Val and p.Val424Phe proteins were secreted less efficiently than the wild‐type protein, although they were synthesized normally in the cell. Collectively, these observations suggest that the amino acid substitutions p.Gly154Arg, p.Gly263Val and p.Val424Phe induce protein misfolding, leading to the intracellular degradation of many FX proteins containing any of these mutations, and ultimately to the development of FX deficiency. On the other hand, for the p.Val236Met, p.Arg387Cys and p.Gly406Ser mutant proteins, we hypothesize that secreted FX proteins have impaired coagulant activities due to functional defects caused by these amino acid substitutions.

Collaboration


Dive into the Akiko Sekiya's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mao Takata

Kanazawa Medical University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge