10 research outputs found

    キイロショウジョウバエ由来のチオレドキシン還元酵素のC未端テトラペプチド配列は、ヒト肺由来のチオレドキシン還元酵素では酸化還元活性を示さない

    Get PDF
    The isozymes of mammalian thioredoxin reductase (TrxR) contain the penultimate selenocysteineresidue (SeCys) in the redox-active C-terminal tetrapeptide, -Gly-Cys-SeCys-Gly (end). Amutant form of the mammalian enzyme TrxR-X498C in which SeCys is replaced with Cys showsa dramatically decreased catalytic activity, suggesting that SeCys residue plays an integral role inthe catalysis. In contrast, TrxR of the fruit fly, Drosophila melanogaster, has no selenium in the corresponding C-terminal redox sequence, which instead of SeCys has flanking serine residues in the terminal sequence, -Ser-Cys-Cys-Ser (end). Because the catalytic activity of Dm-TrxR is comparable to that of the mammalian selenoenzyme, we introduced the serine residues at the corresponding positions of the recombinant TrxR-X498C and mimicked the redox center of the fruit fly TrxR. However, the catalysis remained as low as the Cys mutant of the selenoenzyme, suggesting that the additional structural features are still required for the tetrapeptide to function as a redox center. MOPAC calculation suggested that the complete motif might involve the hexapeptide sequence, which includes a proline residue, -Pro-X-Ser-Cys-Cys-Ser (end). The proline-containing motif is conserved among other insect TrxRs such as those of honeybee and fruit fly.ほ乳類チオレドキシン還元酵素はC末端配列-Gly-Cys-SeCys-Gly(end)の後ろから2番目にセレノシステイン(SeCys)残基を持つ.SeCys をシステインに変換すると酵素の活性は大きく低下するので,SeCys 残基が触媒活性に必須であることが分かる.これに対してキイロショウジョウバエのチオレドキシン還元酵素(Dm-TrxR)のC末端配列にはセレンが含まれず,システイン残基の対が2つのセリンに挟まれた配列-Ser-Cys-Cys-Ser (end)を持つ.それでも Dm-TrxR はほ乳類のセレン含有酵素と同程度の触媒能を示す.われわれはヒト肺チオレドキシン還元酵素に Dm-TrxR のC末端テトラペプチド配列を導入してその効果を調べた.しかし,酵素活性はまったく上昇せず,Dm-TrxR のC末端のテトラペプチド配列-Ser-Cys-Cys-Ser だけでは Cys 残基のチオール基を活性化する効果はなかった.そこで,分子軌道計算 MOPAC を用いて酸化還元機能を担うためのC末端配列モチーフを探索した.その結果,テトラペプチドにさらに2つ先のプロリンまでを含めた Pro-X-Ser-Cys-Cys-Ser(end)により初めて酸化還元モチーフとして機能する可能性が示唆された.Pro を含むこの配列モチーフはミツバチや蚊などほかの昆虫の TrxR でも保存されてい

    Increased predominance of the matured ventricular subtype in embryonic stem cell-derived cardiomyocytes in vivo

    Get PDF
    Accumulating evidence suggests that human pluripotent stem cell-derived cardiomyocytes can affect “heart regeneration”, replacing injured cardiac scar tissue with concomitant electrical integration. However, electrically coupled graft cardiomyocytes were found to innately induce transient post-transplant ventricular tachycardia in recent large animal model transplantation studies. We hypothesised that these phenomena were derived from alterations in the grafted cardiomyocyte characteristics. In vitro experiments showed that human embryonic stem cell-derived cardiomyocytes (hESC-CMs) contain nodal-like cardiomyocytes that spontaneously contract faster than working-type cardiomyocytes. When transplanted into athymic rat hearts, proliferative capacity was lower for nodal-like than working-type cardiomyocytes with grafted cardiomyocytes eventually comprising only relatively matured ventricular cardiomyocytes. RNA-sequencing of engrafted hESC-CMs confirmed the increased expression of matured ventricular cardiomyocyte-related genes, and simultaneous decreased expression of nodal cardiomyocyte-related genes. Temporal engraftment of electrical excitable nodal-like cardiomyocytes may thus explain the transient incidence of post-transplant ventricular tachycardia, although further large animal model studies will be required to control post-transplant arrhythmia

    Metronomic S-1 Chemotherapy and Vandetanib: An Efficacious and Nontoxic Treatment for Hepatocellular Carcinoma

    Get PDF
    Background: Metronomic chemotherapy involves frequent, regular administration of cytotoxic drugs at nontoxic doses, usually without prolonged breaks. We investigated the therapeutic efficacies of metronomic S-1, an oral 5-fluorouracil prodrug, and vandetanib, an epidermal growth factor receptor and vascular endothelial growth factor (VEGF) receptor tyrosine kinase inhibitor, in models of hepatocellular carcinoma (HCC). Methods: We compared anti-HCC effects and toxicity in the six treatment groups: control (untreated), maximum tolerated dose (MTD) S-1, metronomic S-1, vandetanib, MTD S-1 with vandetanib, and metronomic S-1 with vandetanib. Tumor microvessel density (MVD) and tumor apoptosis were evaluated by immunohistochemistry. The expression of VEGF and thrombospondin-1, an endogenous inhibitor of angiogenesis, was analyzed by Western blot. Results: Metronomic S-1 significantly inhibited tumor growth, which was enhanced by combination with vandetanib. With respect to toxicities, MTD S-1 caused severe body weight loss and myelosuppression, whereas metronomic S-1 did not cause any overt toxicities. Moreover, metronomic S-1 or metronomic S-1 with vandetanib prolonged survival, the latter treatment providing the greatest benefit. Metronomic S-1 and metronomic S-1 with vandetanib decreased MVDs and increased apoptosis in tumor tissues. The expression of VEGF in tumor tissues was upregulated by vandetanib and metronomic S-1 with vandetanib, whereas the expression of thrombospondin-1 was upregulated by metronomic S-1 and metronomic S-1 with vandetanib. Conclusion: Metronomic S-1 with an antiangiogenic agent seems to be an effective and safe therapeutic strategy for HCC

    Mature human induced pluripotent stem cell-derived cardiomyocytes promote angiogenesis through alpha-B crystallin

    No full text
    Abstract Background Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) can be used to treat heart diseases; however, the optimal maturity of hiPSC-CMs for effective regenerative medicine remains unclear. We aimed to investigate the benefits of long-term cultured mature hiPSC-CMs in injured rat hearts. Methods Cardiomyocytes were differentiated from hiPSCs via monolayer culturing, and the cells were harvested on day 28 or 56 (D28-CMs or D56-CMs, respectively) after differentiation. We transplanted D28-CMs or D56-CMs into the hearts of rat myocardial infarction models and examined cell retention and engraftment via in vivo bioluminescence imaging and histological analysis. We performed transcriptomic sequencing analysis to elucidate the genetic profiles before and after hiPSC-CM transplantation. Results Upregulated expression of mature sarcomere genes in vitro was observed in D56-CMs compared with D28-CMs. In vivo bioluminescence imaging studies revealed increased bioluminescence intensity of D56-CMs at 8 and 12 weeks post-transplantation. Histological and immunohistochemical analyses showed that D56-CMs promoted engraftment and maturation in the graft area at 12 weeks post-transplantation. Notably, D56-CMs consistently promoted microvessel formation in the graft area from 1 to 12 weeks post-transplantation. Transcriptomic sequencing analysis revealed that compared with the engrafted D28-CMs, the engrafted D56-CMs enriched genes related to blood vessel regulation at 12 weeks post-transplantation. As shown by transcriptomic and western blot analyses, the expression of a small heat shock protein, alpha-B crystallin (CRYAB), was significantly upregulated in D56-CMs compared with D28-CMs. Endothelial cell migration was inhibited by small interfering RNA-mediated knockdown of CRYAB when co-cultured with D56-CMs in vitro. Furthermore, CRYAB overexpression enhanced angiogenesis in the D28-CM grafts at 4 weeks post-transplantation. Conclusions Long-term cultured mature hiPSC-CMs promoted engraftment, maturation and angiogenesis post-transplantation in infarcted rat hearts. CRYAB, which was highly expressed in D56-CMs, was identified as an angiogenic factor from mature hiPSC-CMs. This study revealed the benefits of long-term culture, which may enhance the therapeutic potential of hiPSC-CMs

    Additional file 1 of Mature human induced pluripotent stem cell-derived cardiomyocytes promote angiogenesis through alpha-B crystallin

    No full text
    Additional file1. Fig S1. Generation of the Akaluc-expressing hiPSC line. Fig S2. Flow cytometric analysis. Fig S3. qRT-PCR analysis. Fig S4. Immunocytochemistry. Fig S5. Comparison of Luc2- and Akaluc-expressing hiPSCs. Fig S6. Echocardiography. Fig S7. BLI. Fig S8. Assessment of the graft size and composition of hiPSC-CMs at 1 and 12 weeks post-transplantation. Fig S9. Cell proliferation in grafts at 4 and 8 weeks post-transplantation. Fig S10. Apoptosis in grafts at 4 and 8 weeks post-transplantation. Fig S11. Microvessels in grafts and in vitro assays for assessing angiogenesis by hiPSC-CMs. Fig S12. Angiogenesis profiler array and qRT-PCR. Fig S13. Expression of CRYAB in hiPSC-CMs. Fig S14. AAV-mediated CRYAB overexpression. Table S1. Primer sequences for genomic PCR and qRT-PCR in this study. Table S2. Antibodies used for immunohistochemistry. Table S3. Top 10 gene ontology (GO) terms enriched in D56-CM compared with D28-CM at 12 weeks after transplantation. Table S4. Top 20 genes with TPM-normalized counts upregulated by more than 4-fold in D56-CMs compared with D28-CMs before transplantation
    corecore