Network


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

Hotspot


Dive into the research topics where Mai Uesugi is active.

Publication


Featured researches published by Mai Uesugi.


Molecular Cancer Therapeutics | 2014

Selective Inhibition of EZH2 by EPZ-6438 Leads to Potent Antitumor Activity in EZH2-Mutant Non-Hodgkin Lymphoma

Sarah K. Knutson; Satoshi Kawano; Yukinori Minoshima; Natalie Warholic; Kuan-Chun Huang; Yonghong Xiao; Tadashi Kadowaki; Mai Uesugi; Galina Kuznetsov; Namita Kumar; Tim J. Wigle; Christine R. Klaus; Christina J. Allain; Alejandra Raimondi; Nigel J. Waters; Jesse J. Smith; Margaret Porter-Scott; Richard Chesworth; Mikel P. Moyer; Robert A. Copeland; Victoria M. Richon; Toshimitsu Uenaka; Roy M. Pollock; Kevin Wayne Kuntz; Akira Yokoi; Heike Keilhack

Mutations within the catalytic domain of the histone methyltransferase EZH2 have been identified in subsets of patients with non-Hodgkin lymphoma (NHL). These genetic alterations are hypothesized to confer an oncogenic dependency on EZH2 enzymatic activity in these cancers. We have previously reported the discovery of EPZ005678 and EPZ-6438, potent and selective S-adenosyl-methionine-competitive small molecule inhibitors of EZH2. Although both compounds are similar with respect to their mechanism of action and selectivity, EPZ-6438 possesses superior potency and drug-like properties, including good oral bioavailability in animals. Here, we characterize the activity of EPZ-6438 in preclinical models of NHL. EPZ-6438 selectively inhibits intracellular lysine 27 of histone H3 (H3K27) methylation in a concentration- and time-dependent manner in both EZH2 wild-type and mutant lymphoma cells. Inhibition of H3K27 trimethylation (H3K27Me3) leads to selective cell killing of human lymphoma cell lines bearing EZH2 catalytic domain point mutations. Treatment of EZH2-mutant NHL xenograft-bearing mice with EPZ-6438 causes dose-dependent tumor growth inhibition, including complete and sustained tumor regressions with correlative diminution of H3K27Me3 levels in tumors and selected normal tissues. Mice dosed orally with EPZ-6438 for 28 days remained tumor free for up to 63 days after stopping compound treatment in two EZH2-mutant xenograft models. These data confirm the dependency of EZH2-mutant NHL on EZH2 activity and portend the utility of EPZ-6438 as a potential treatment for these genetically defined cancers. Mol Cancer Ther; 13(4); 842–54. ©2014 AACR.


Cancer Science | 2014

Eribulin mesylate reduces tumor microenvironment abnormality by vascular remodeling in preclinical human breast cancer models

Yasuhiro Funahashi; Kiyoshi Okamoto; Yusuke Adachi; Taro Semba; Mai Uesugi; Yoichi Ozawa; Osamu Tohyama; Taisuke Uehara; Takayuki Kimura; Hideki Watanabe; Makoto Asano; Satoshi Kawano; Xavier Tizon; Paul J. McCracken; Junji Matsui; Ken Aoshima; Kenichi Nomoto; Yoshiya Oda

Eribulin mesylate is a synthetic macrocyclic ketone analog of the marine sponge natural product halichondrin B and an inhibitor of microtubule dynamics. Some tubulin‐binding drugs are known to have antivascular (antiangiogenesis or vascular‐disrupting) activities that can target abnormal tumor vessels. Using dynamic contrast‐enhanced MRI analyses, here we show that eribulin induces remodeling of tumor vasculature through a novel antivascular activity in MX‐1 and MDA‐MB‐231 human breast cancer xenograft models. Vascular remodeling associated with improved perfusion was shown by Hoechst 33342 staining and by increased microvessel density together with decreased mean vascular areas and fewer branched vessels in tumor tissues, as determined by immunohistochemical staining for endothelial marker CD31. Quantitative RT‐PCR analysis of normal host cells in the stroma of xenograft tumors showed that eribulin altered the expression of mouse (host) genes in angiogenesis signaling pathways controlling endothelial cell–pericyte interactions, and in the epithelial–mesenchymal transition pathway in the context of the tumor microenvironment. Eribulin also decreased hypoxia‐associated protein expression of mouse (host) vascular endothelial growth factor by ELISA and human CA9 by immunohistochemical analysis. Prior treatment with eribulin enhanced the anti‐tumor activity of capecitabine in the MDA‐MB‐231 xenograft model. These findings suggest that eribulin‐induced remodeling of abnormal tumor vasculature leads to a more functional microenvironment that may reduce the aggressiveness of tumors due to elimination of inner tumor hypoxia. Because abnormal tumor microenvironments enhance both drug resistance and metastasis, the apparent ability of eribulin to reverse these aggressive characteristics may contribute to its clinical benefits.


Journal of Pharmacological and Toxicological Methods | 2014

Low-density plating is sufficient to induce cardiac hypertrophy and electrical remodeling in highly purified human iPS cell-derived cardiomyocytes.

Mai Uesugi; Atsuko Ojima; Tomohiko Taniguchi; Norimasa Miyamoto; Kohei Sawada

INTRODUCTION Cardiac hypertrophy is a leading cause of many cardiovascular diseases, including heart failure, but its pathological mechanism is not fully understood. This study used highly purified human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes to produce an in vitro hypertrophy model and characterize its gene expression and electrophysiological properties. METHODS For 7 days we cultured hiPSC-derived cardiomyocytes plated at high (2800-4800 cells/mm(2)) or low (500-1200 cells/mm(2)) cell density and assessed their cell size with confocal and fluorescence microscopy, their electrophysiological and pharmacological responses with multi-electrode array systems, and their gene expression patterns by using DNA microarray technology and quantitative PCR. We used quantitative PCR and Western blotting to compare the expression of potassium-channel genes between the hiPSC-derived cardiomyocytes and human fetal and adult hearts. RESULTS The hiPSC-derived cardiomyocytes showed spontaneous beating and similar pattern of α-actinin molecules regardless of plating density. However, cells plated at low density had the following characteristics compared with those at high density: 1) significant enlargement in size; 2) significant increase or decrease in expression of the cardiac hypertrophy-characteristic genes NPPA, ATP2A2, ANKRD1 and MYL2 in accordance with the progression of hypertrophy; 3) significant reduction in responses to the inhibitors of cardiac slow delayed-rectifier K(+) current (IKs), chromanol 293B and HMR1556, in a cell-density-dependent manner; and 4) significant reduction in the expression of the KCNQ1 and KCNJ2 genes coding the K(+) ion channels conducting each IKs and cardiac inward rectifier outward K(+) current (IK1). DISCUSSION The enlargement, hypertrophy-characteristic and potassium ion channels gene expression of hiPSC-derived cardiomyocytes suggest that low-density plating was sufficient to induce cardiac hypertrophy. This model may be useful in elucidating mechanisms underlying the onset and progress of cardiac hypertrophy, because these cells can be cultured for several weeks.


Journal of Biological Chemistry | 2010

N-type Calcium Channel in the Pathogenesis of Experimental Autoimmune Encephalomyelitis

Naoki Tokuhara; Kana Namiki; Mai Uesugi; Chihiro Miyamoto; Makoto Ohgoh; Katsutoshi Ido; Takashi Yoshinaga; Toshihiko Yamauchi; Junro Kuromitsu; Sadao Kimura; Norimasa Miyamoto; Yoshitoshi Kasuya

One of the family of voltage-gated calcium channels (VGCC), the N-type Ca2+ channel, is located predominantly in neurons and is associated with a variety of neuronal responses, including neurodegeneration. A precise mechanism for how the N-type Ca2+ channel plays a role in neurodegenerative disease, however, is unknown. In this study, we immunized N-type Ca2+ channel α1B-deficient (α1B−/−) mice and their wild type (WT) littermates with myelin oligodendrocyte glycoprotein 35–55 and analyzed the progression of experimental autoimmune encephalomyelitis (EAE). The neurological symptoms of EAE in the α1B−/− mice were less severe than in the WT mice. In conjunction with these results, sections of the spinal cord (SC) from α1B−/− mice revealed a reduction in both leukocytic infiltration and demyelination compared with WT mice. No differences were observed in the delayed-type hypersensitivity response, spleen cell proliferation, or cytokine production from splenocytes between the two genotypes. On the other hand, Western blot array analysis and RT-PCR revealed that a typical increase in the expression of MCP-1 in the SC showed a good correlation with the infiltration of leukocytes into the SC. Likewise, immunohistochemical analysis showed that the predominant source of MCP-1 was activated microglia. The cytokine-induced production of MCP-1 in primary cultured microglia from WT mice was significantly higher than that from α1B−/− mice and was significantly inhibited by a selective N-type Ca2+ channel antagonist, ω-conotoxin GVIA or a withdrawal of extracellular Ca2+. These results suggest that the N-type Ca2+ channel is involved in the pathogenesis of EAE at least in part by regulating MCP-1 production by microglia.


Nature Chemical Biology | 2017

Selective degradation of splicing factor CAPER[alpha] by anticancer sulfonamides

Taisuke Uehara; Yukinori Minoshima; Koji Sagane; Naoko Hata Sugi; Kaoru Ogawa Mitsuhashi; Noboru Yamamoto; Hiroshi Kamiyama; Kentaro Takahashi; Yoshihiko Kotake; Mai Uesugi; Akira Yokoi; Atsushi Inoue; Taku Yoshida; Miyuki Mabuchi; Akito Tanaka; Takashi Owa

Target-protein degradation is an emerging field in drug discovery and development. In particular, the substrate-receptor proteins of the cullin-ubiquitin ligase system play a key role in selective protein degradation, which is an essential component of the anti-myeloma activity of immunomodulatory drugs (IMiDs), such as lenalidomide. Here, we demonstrate that a series of anticancer sulfonamides NSC 719239 (E7820), indisulam, and NSC 339004 (chloroquinoxaline sulfonamide, CQS) induce proteasomal degradation of the U2AF-related splicing factor coactivator of activating protein-1 and estrogen receptors (CAPERα) via CRL4DCAF15 mediated ubiquitination in human cancer cell lines. Both CRISPR-Cas9-based knockout of DCAF15 and a single amino acid substitution of CAPERα conferred resistance against sulfonamide-induced CAPERα degradation and cell-growth inhibition. Thus, these sulfonamides represent selective chemical probes for disrupting CAPERα function and designate DCAFs as promising drug targets for promoting selective protein degradation in cancer therapy.


Journal of Cardiovascular Pharmacology | 2012

Chronic probucol treatment decreases the slow component of the delayed-rectifier potassium current in CHO cells transfected with KCNQ1 and KCNE1: a novel mechanism of QT prolongation.

Tomohiko Taniguchi; Mai Uesugi; Toru Arai; Takashi Yoshinaga; Norimasa Miyamoto; Kohei Sawada

Abstract: Indirect effects of drugs on ion channel expression levels on plasma membrane are focused as the cause of QT prolongation, and we explored the chronic effects of QT-prolonging drugs on the slow component of the delayed-rectifier potassium current (IKs). Chinese Hamster Ovary cells expressing IKs channels were constructed by transfecting KCNQ1/KCNE1 genes, and the IKs values were measured using IonWorks Quattro in the population patch-clamp mode. After 24 hours of treatment with IKs blockers (HMR1556, L-768673, or chromanol 293B) or human Ether-à-go-go related gene channel trafficking inhibitors (amiodarone,17-AAG, brefeldin A, pentamidine, thioridazine, or probucol), brefeldin A, pentamidine, and probucol decreased IKs. Probucol, which is a cholesterol-lowering drug and clinically reported to cause QT prolongation, potently inhibited the IKs in a concentration-dependent manner, with a half maximal inhibitory concentration of 149.1 nM. A reduction in the IKs by 1 &mgr;M of probucol was observed beginning 2 hours after treatment, and the current was reduced by about 80% at 24 hours. The activation and deactivation time constants of residual IKs currents became faster compared with that in the vehicle-treatment group. Acute application of probucol did not directly inhibit IKs channels at concentrations of up to 10 &mgr;M. Western blotting analysis indicated the reduction of multimeric complex of KCNQ1 proteins by probucol treatment but not monomeric form. These results suggest that chronic probucol treatment may contribute to QT prolongation in humans by decreasing the functional IKs channel complexes.


Journal of Receptors and Signal Transduction | 2013

Prediction of relaxin-3-induced downstream pathway resulting in anxiolytic-like behaviors in rats based on a microarray and peptidome analysis

Chihiro Miyamoto Nakazawa; Kohdoh Shikata; Mai Uesugi; Hiroyuki Katayama; Ken Aoshima; Kazuhiro Tahara; Eiki Takahashi; Takayuki Hida; Hisashi Shibata; Hiroo Ogura; Takashi Seiki; Yoshiya Oda; Junro Kuromitsu; Norimasa Miyamoto

Abstract The effect of the intracerebroventricular (i.c.v.) injection of relaxin-3 (RLX3) was evaluated using anxiety-related behavioral tests in rats. RLX3-injected animals showed normal locomotion activity in a habituated environment and declined anxiety cognition in the elevated plus maze test and the shock probe-burying test. The measurement of spontaneous locomotor activity in a novel environment also suggested that RLX3 reduced the stress response. To elucidate the regulatory mechanisms of the downstream signaling pathways underlying RLX3 activity and its relation to anxiolytic and hyperphagic behavior phenotypes, RLX3-i.c.v.-injected rat hypothalamic responses were examined using a microarray analysis. Ingenuity Pathway Analysis software listed the phenotype-relating genes and they showed characteristic expression patterns in the rat hypothalamus. When peptidome data sets for the same listed genes was analyzed using a semi-quantitative approach, the expressions of two neuropeptides were found to have increased. One of these neuropeptides, oxytocin (Oxt), exhibited increased expression in both the microarray and the peptidomic analysis, and a Western blot analysis validated the mass spectrometry results. A cross-omics data analysis is useful for predicting downstream signaling pathways, and the anxiolytic-like behavior of RLX3 may be mediated by an oxytocin signaling pathway in rats. These results suggest that RLX3 acts as an anxiolytic peptide and that the downstream pathways mediated by its receptors may be potential candidates for the treatment of anxieties in the future.


Toxicology and Applied Pharmacology | 2017

Hsp90 inhibitor geldanamycin attenuates the cytotoxicity of sunitinib in cardiomyocytes via inhibition of the autophagy pathway

Takayuki Kimura; Mai Uesugi; Kazuma Takase; Norimasa Miyamoto; Kohei Sawada

&NA; Sunitinib malate (sunitinib) is an orally available, multitargeted tyrosine kinase inhibitor with antitumor and antiangiogenic activities. Although sunitinib is effective for the treatment of patients with gastrointestinal stromal tumor, advanced renal cell carcinoma, or pancreatic neuroendocrine tumor, adverse cardiac events associated with sunitinib administration have been reported. Here, we examined the effect of geldanamycin, an inhibitor of heat shock protein (Hsp) 90, on sunitinib‐induced cytotoxicity in cardiomyocytes. First, we found that treatment with geldanamycin or other Hsp90 inhibitors (tanespimycin, ganetespib, or BIIB021) significantly attenuated sunitinib‐induced cytotoxicity in rat H9c2 cardiomyocytes, suggesting a drug‐class effect of Hsp90 inhibitors. We then examined the mechanisms underlying sunitinib‐induced cytotoxicity and found that sunitinib induced autophagy in H9c2 cells and that pretreatment with geldanamycin inhibited the induction of autophagy by promoting degradation of the autophagy‐related proteins Atg7, Beclin‐1, and ULK1. Pharmacological assessment with autophagy inhibitors confirmed that geldanamycin attenuated the cytotoxicity of sunitinib by interfering with autophagy. In addition, we found that the molecular chaperone Hsp70, which is induced by geldanamycin, was not involved in the attenuation of sunitinib‐induced cytotoxicity. Finally, to provide more clinically relevant data, we confirmed that geldanamycin attenuated sunitinib‐induced cytotoxicity in human induced pluripotent stem cell‐derived cardiomyocytes. Together, these data suggest that geldanamycin attenuates sunitinib‐induced cytotoxicity in cardiomyocytes by inhibiting the autophagy pathway. Thus, the further investigation of combination or sequential treatment with an Hsp90 inhibitor and sunitinib is warranted as a potential strategy of attenuating the cardiotoxicity associated with sunitinib administration in the clinical setting. HighlightsHsp90 inhibitors attenuated sunitinib‐induced cytotoxicity in cardiomyocytes.Sunitinib induced autophagic flux in cardiomyocytes.Geldanamycin degraded autophagy regulator proteins in cardiomyocytes.Inhibition of the autophagy pathway attenuated sunitinib‐induced cardiotoxicity.Hsp70 induction was not involved in attenuating sunitinib‐induced cardiotoxicity.


Cancer Research | 2013

Abstract 1413: Eribulin caused re-modeling of tumor vasculature altering gene expression profiling in angiogenesis and Epithelial Mesenchymal Transition (EMT) signaling pathway of host cells within human breast cancer cell (BCC) xenografts in nude mice.

Junji Matsui; Osamu Toyama; Mitsuhiro Ino; Taro Semba; Mai Uesugi; Hiroki Muto; Judith Oestreicher; Kentaro Takahashi; Kentaro Matsuura; Yoshiaki Sato; Taisuke Uehara; Takayuki Kimura; Hideki Watanabe; Yoichi Ozawa; Makoto Asano; Yusuke Adachi; Ken Aoshima; Yasuhiro Funahashi

Objective: Eribulin mesylate (ERI) is a synthetic macrocyclic ketone analog of the marine sponge natural product halichondrin B and an inhibitor of microtubule dynamics. The objective of this study was to examine the effect of ERI on tumor vasculature with immunohistchemical (IHC) analysis and gene expression profiling (GEP) in normal host cells, such as endothelial cells and vascular mural cells within tumor microenvironments in human BCC xenograft models Methods: Anti-tumor activity of ERI was examined at doses of 1.5 and 3.0 mg/kg, i.v. at day 1, in human BCC MX-1, MDA-MB-231 and MDA-MB-453 sc xenografts in nude mice. For IHC and GEP analysis, tumor tissues were collected at day 4 and day 8. IHC analysis was performed using mouse CD31 antibody to stain endothelial cells. Microvessel density (MVD) and vessel perimeter were determined by using Aperio Image Scope. GEP analysis for mouse host and human tumor cells within tumor tissues was done by using mouse and human TaqMan Low Density Arrays (TLDAs) consisting of a set of 92 genes related to angiogenesis, metastasis/EMT and cell differentiation signal pathways. Results shows % of non-treatment group (NT). Results: ERI showed significant anti-tumor activity against all three human BCC xenografts in a dose dependent manner. IHC analysis showed that ERI altered morphology of tumor vasculature day 8 after treatments and increased number of vessels with small size of perimeter ( 300um) in both MX-1 and MDA-MB-231 xenograft models (p Conclusions: ERI induced re-modeling of tumor vasculature in human BCC xenograft models. GEP related to angiogenesis and EMT/metastasis pathway was significantly affected with ERI treatment in host cells under tumor microenvironments. ERI might cause remodeling of tumor vasculature by regulating GEP in host cells. Further investigation may be warranted to examine if the activity of ERI against host cells in tumor tissues contributed to anti-tumor activity of ERI. Citation Format: Junji Matsui, Osamu Toyama, Mitsuhiro Ino, Taro Semba, Mai Uesugi, Hiroki Muto, Judith L. Oestreicher, Kentaro Takahashi, Kentaro Matsuura, Yoshiaki Sato, Taisuke Uehara, Takayuki Kimura, Hideki Watanabe, Yoichi Ozawa, Makoto Asano, Yusuke Adachi, Ken Aoshima, Yasuhiro Funahashi. Eribulin caused re-modeling of tumor vasculature altering gene expression profiling in angiogenesis and Epithelial Mesenchymal Transition (EMT) signaling pathway of host cells within human breast cancer cell (BCC) xenografts in nude mice. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 1413. doi:10.1158/1538-7445.AM2013-1413


Cancer Research | 2013

Abstract 3830: Eribulin and Paclitaxel differentially affect gene expression profiling of blood vessel cells and in vitro angiogenesis in co-cultures of human endothelial cells with pericytes.

Sergei Agoulnik; Judith Oestreicher; Noel Taylor; Mai Uesugi; Hiroki Muto; Satoshi Kawano; Kentaro Takahashi; Kentaro Matsuura; Ken Aoshima; Junji Matsui; Yasuhiro Funahashi

Proceedings: AACR 104th Annual Meeting 2013; Apr 6-10, 2013; Washington, DC Objectives: Eribulin mesylate (ERI) is a simplified synthetic macrocyclic ketone analog of the marine sponge natural product halichondrin B. We examined effects of ERI and Paclitaxel (PTX) on blood vessel cells by gene expression profiling (GEP) in mono- and co-culture assays of pericytes and endothelial cells. Activity of ERI and PTX against cell growth inhibition and pericyte-driven in vitro angiogenesis was also studied. Methods: We first assessed IC50s of ERI and PTX in 4-day growth inhibition assay in isolated primary human brain vascular pericytes (HBVP) and human umbilical vein endothelial cells (HUVEC). Based on identified IC50s, cells were treated with 10X IC50s of ERI or PTX and GEP was analyzed at 24 h using either Affimetrix Human Genome U133 Plus 2.0 arrays or custom TaqMan Low Density Cards (TLDA) designed with 92 genes related to angiogenesis, metastasis/ Epithelial Mesenchymal Transition (EMT) and cell differentiation signal pathways. Inhibitory activity of drugs on the length of capillary-like networks was analyzed in co-culture of HUVEC with HBVP. Results: ERI and PTX inhibited cell growth of HBVP at IC50s of 1.2 and 3.1nM and HUVEC was more sensitive than HBVP by 1.9 and 3.3 fold in cell growth assay, respectively. In HUVEC, most genes were down-regulated by both ERI and PTX treatments, while in HBVP about equal number of genes were up- or down-regulated with microarray analysis. Interestingly, 63% affected genes in HUVECs overlapped for both treatments. In HBVPs, altered gene signatures were drug-dependent and an overlap was limited by 16%. ERI specifically affected genes in HIF1 and caveolar-mediated signaling pathways while PTX regulated genes in HER2, PI3K/AKT and HGF signaling pathways among others. We confirmed obtained altered GEP using TLDA and identified 42 significant genes differentially regulated by ERI and PTX in HBVP. To examine effects on pericyte-driven in vitro angiogenesis, we compared length of capillary-like networks in co-culture of HUVEC with HBVP. ERI disrupted capillary-like networks starting at about 2 nM, while PTX showed limited inhibitory activity by less than 50% even at 100-1000 nM for 4 days treatments. In co-culture assay, TLDA data showed decreased expression levels of angiogenesis-related genes DLL4 (14% compare to control), PDGFRB (70%) and metastasis/EMT-related genes ZEB1 (53%), TGFB3 (54 %), VIM (62 %) after treatment with 10X IC50s of ERI (p<0.05). Conclusions: Endothelial cells and pericytes responded differently to ERI and PTX treatments and effects of ERI on GEP of pericytes were distinct from PTX. ERI inhibited pericyte-driven in vitro angiogenesis at sub nM using co-culture assay of HUVEC with HBVP. Further analysis of the role of ERI on GEP of pericytes and pericyte-driven angiogenesis in anti-tumor activity will be warranted. Citation Format: Sergei I. Agoulnik, Judith L. Oestreicher, Noel H. Taylor, Mai Uesugi, Hiroki Muto, Satoshi Kawano, Kentaro Takahashi, Kentaro Matsuura, Ken Aoshima, Junji Matsui, Yasuhiro Funahashi. Eribulin and Paclitaxel differentially affect gene expression profiling of blood vessel cells and in vitro angiogenesis in co-cultures of human endothelial cells with pericytes. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 3830. doi:10.1158/1538-7445.AM2013-3830

Collaboration


Dive into the Mai Uesugi's collaboration.

Researchain Logo
Decentralizing Knowledge