Network


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

Hotspot


Dive into the research topics where Sachie Hirai is active.

Publication


Featured researches published by Sachie Hirai.


Molecular Therapy | 2003

Adenoviral delivered angiopoietin-1 reduces the infarction and attenuates the progression of cardiac dysfunction in the rat model of acute myocardial infarction

Kazuhiro Takahashi; Yoshinori Ito; Masayuki Morikawa; Masayoshi Kobune; Jianhua Huang; Masaru Tsukamoto; Katsunori Sasaki; Kiminori Nakamura; Hironari Dehari; Katsuya Ikeda; Hiroaki Uchida; Sachie Hirai; Tomio Abe; Hirofumi Hamada

In acute myocardial infarction (AMI), prognosis and mortality rate are closely related to the infarct size and the progression of postinfarction cardiac failure. Angiogenic gene therapy has presented a new approach for the treatment of AMI. Angiopoietin-1 (Ang1) is a critical angiogenic factor for vascular maturation and enhances vascular endothelial growth factor (VEGF)-induced angiogenesis in a complementary manner. We hypothesized that gene therapy using Ang1 for AMI might promote angiogenesis cooperatively with intrinsic VEGF, since high concentrations of circulating VEGF have been reported in AMI. To evaluate our hypothesis, we employed a rat AMI model and adenoviral Ang1 (HGMW-approved gene symbol ANGPT1) gene transfer to the heart. A significant increase in capillary density and reduction in infarct sizes were noted in the infarcted hearts with adenoviral Ang1 gene treatment compared with control infarcted hearts treated with saline or adenoviral vector containing the beta-galactosidase gene. Furthermore, the Ang1 group showed significantly higher cardiac performance in echocardiography (55.0% of ejection fraction, P < 0.05 vs control) than the saline or adenoviral controls (36.0 or 40.5%, respectively) 4 weeks after myocardial infarction. The adenoviral delivery of Ang1 during the acute phase of myocardial infarction would be feasible to attenuate the progression of cardiac dysfunction in the rat model.


Circulation | 2005

Bcl-xL Gene Transfer Inhibits Bax Translocation and Prolongs Cardiac Cold Preservation Time in Rats

Jianhua Huang; Kiminori Nakamura; Yoshinori Ito; Takeshi Uzuka; Masayuki Morikawa; Sachie Hirai; Kei Tomihara; Toshihiro Tanaka; Yukari Masuta; Keiji Ishii; Kazunori Kato; Hirofumi Hamada

Background—Apoptosis is an important cause of early graft loss after heart transplantation. Bcl-xL was reported to protect the heart against normothermic ischemia and reperfusion injury. In this study, we determined whether overexpression of Bcl-xL could inhibit tissue injury resulting from prolonged cold preservation followed by warm reperfusion of heart transplants. Methods and Results—Lewis rat hearts were transduced with an adenovirus vector harboring Bcl-xL cDNA (AxCAhBclxL) 4 days before collection of tissue. After preservation in University of Wisconsin solution at 4°C for 24 hours, the heart was either perfused with a Langendorff device ex vivo or used for heterotopic heart transplantation in vivo. Bcl-xL gene transfer significantly reduced the infarct size (23.0±2.6% versus 47.7±7.0% in saline control and 48.6±6.1% in vector control, P<0.01) after 2-hour reperfusion at 37°C with the Langendorff device and significantly decreased creatine kinase release (0.82±0.27 IU, versus 1.57±0.33 and 1.50±0.37 IU in saline and vector controls, respectively; P<0.05). In heart transplantation, overexpresson of Bcl-xL inhibited Bax translocation from the cytosol to the mitochondria, resulting in decreased cytochrome c release from the mitochondria; it also significantly decreased cardiac cell apoptosis and improved graft survival rate after long cold preservation, followed by warm reperfusion. Conclusions—Bcl-xL gene transfer inhibited the translocation of Bax and prolonged the cold preservation time of cardiac transplants. This may be a potential therapeutic method in clinical practice.


Clinical Cancer Research | 2006

Carcinoembryonic Antigen–Targeted Selective Gene Therapy for Gastric Cancer through FZ33 Fiber-Modified Adenovirus Vectors

Toshihiro Tanaka; Jianhua Huang; Sachie Hirai; Motomu Kuroki; Masahide Kuroki; Naoki Watanabe; Kei Tomihara; Kazunori Kato; Hirofumi Hamada

Purpose: A major problem when using the adenoviral vectors for gene therapy applications is thought to be related to low transduction efficiency in cancer cells or to side effects in normal cells. There is an urgent requirement to improve the specificity of gene delivery in the context of cancer gene therapy. Experimental Design: We constructed a genetically modified adenovirus incorporating an IgG Fc-binding motif from the Staphylococcus protein A, Z33, within the HI loop (Adv-FZ33). A remarkable degree of targeted gene delivery to gastric cancer cells was obtained with Adv-FZ33 with the fully human anti–carcinoembryonic antigen (CEA) monoclonal antibody, C2-45. Results:In vitro LacZ or EGFP gene expression after Adv-FZ33 infection via C2-45 was 20 times higher than control monoclonal antibody in MKN-45 at 1,000 viral particles/cell. We generated Ax3CAUP-FZ33 (UP-FZ33), which is an Adv-FZ33 derivative vector expressing a therapeutic gene (i.e., Escherichia coli uracil phosphoribosyltransferase), which converts 5-fluorouracil (5-FU) directly to 5-fluoro-UMP. UP-FZ33 with C2-45 enhanced the cytotoxicity of 5-FU by 10.5-fold in terms of IC50 against MKN-45 compared with control IgG4. In a nude mouse peritoneal dissemination model, tumor growth in mice treated with UP-FZ33/C2-45/5-FU was significantly suppressed, and tumor volumes were less than one-fourth of those of the control IgG4 group (P < 0.05). The median survival time of the UP-FZ33/C2-45/5-FU group was significantly longer than those treated with PBS or 5-FU only (P < 0.01). Conclusions: These data suggest that CEA-targeted FZ33 mutant adenovirus-mediated gene delivery offers a strong and selective therapeutic modality against CEA-producing cancers.


Clinical Cancer Research | 2011

Epigenetic Modulation Enhances the Therapeutic Effect of Anti–IL-13Rα2 Antibody in Human Mesothelioma Xenografts

Makoto Takenouchi; Sachie Hirai; Naoya Sakuragi; Hideo Yagita; Hirofumi Hamada; Kazunori Kato

Purpose: The interleukin-13 receptor α2 (IL-13Rα2) is expressed by a variety of human malignant cells. Here, we have examined the constitutive surface expression and the epigenetic regulation of IL-13Rα2 by human mesothelioma. We have also investigated the therapeutic effect of the DNA methylation inhibitor 5-aza-2′-deoxycytidine (5-aza-dC) and anti–IL-13Rα2 monoclonal antibody on mesothelioma xenografts. Experimental Design: Cell surface expression of IL-13Rα2 by various lung carcinomas was analyzed using flow cytometry. Therapeutic effects of anti–IL-13Rα2 and 5-aza-dC were investigated using antibody-dependent cellular cytotoxicity and proliferation assays and by monitoring the survival of mesothelioma-bearing mice. Results: We found that human malignant mesotheliomas expressed surface IL-13Rα2 on their surface and that it was upregulated by treatment with 5-aza-dC. This augmented expression of IL-13Rα2 resulted in growth inhibition of the mesothelioma cells when cocultured with anti-IL-13Rα2 and effector cells, such as splenocytes and peritoneal exudate cells. The growth inhibition of mesothelioma cells was mediated by IFN-γ that was only detected in the supernatant when effector cells were exposed to 5-aza-dC–treated tumors in the presence of anti–IL-13Rα2. Compared with the control or either regimen alone, in vivo administration of anti-IL-13Rα2 in combination with 5-aza-dC significantly prolonged the survival of mice with mesothelioma xenografts. Conclusions: These observations indicate a promising role for IL-13Rα2 as a target for antibody treatment in malignant mesothelioma, and, in combination with epigenetic regulation by a DNA methylation inhibitor, suggest the potential for a novel strategy to enhance therapeutic potency. Clin Cancer Res; 17(9); 2819–29. ©2011 AACR.


Laboratory Investigation | 2017

Fibroblastic foci, covered with alveolar epithelia exhibiting epithelial–mesenchymal transition, destroy alveolar septa by disrupting blood flow in idiopathic pulmonary fibrosis

Miki Yamaguchi; Sachie Hirai; Yusuke Tanaka; Toshiyuki Sumi; Masahiro Miyajima; Taijiro Mishina; Gen Yamada; Mitsuo Otsuka; Tadashi Hasegawa; Takashi Kojima; Toshiro Niki; Atsushi Watanabe; Hiroki Takahashi; Yuji Sakuma

Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive interstitial lung disease of unknown cause. IPF has a distinct histopathological pattern of usual interstitial pneumonia in which fibroblastic foci (FF) represent the leading edge of fibrotic destruction of the lung. Currently there are three major hypotheses for how FF are generated: (1) from resident fibroblasts, (2) from bone marrow-derived progenitors of fibroblasts, and (3) from alveolar epithelial cells that have undergone epithelial–mesenchymal transition (EMT). We found that FF dissociated capillary vessels from the alveolar epithelia, the basement membranes of which are fused in normal physiological conditions, and pushed the capillaries and elastic fibers down ~100 μm below the alveolar epithelia. Furthermore, the alveolar epithelial cells covering the FF exhibited a partial EMT phenotype. In addition, normal human alveolar epithelial cells in vitro underwent dynamic EMT in response to transforming growth factor-β signaling within 72 h. Because it seems that resident fibroblasts or bone marrow-derived cells cannot easily infiltrate and form FF between the alveolar epithelia and capillaries in tight contact with each other, FF are more likely to be derived from the epithelial-to-mesenchymal transitioned alveolar epithelia located over them. Moreover, histology and immunohistochemistry suggested that the FF formed in the lung parenchyma disrupt blood flow to the alveolar septa, thus destroying them. Consequently, collapse of the alveolar septa is likely to be the first step toward honeycombing in the lung during late stage IPF. On the basis of these findings, inhibition of transforming growth factor-β signaling, which can suppress EMT of the alveolar epithelial cells in vitro, is a potential strategy for treating IPF.


Biochemical and Biophysical Research Communications | 2014

Development of a sensitive screening method for selecting monoclonal antibodies to be internalized by cells.

Miki Yamaguchi; Yukari Nishii; Kiminori Nakamura; Haruka Aoki; Sachie Hirai; Hiroaki Uchida; Yuji Sakuma; Hirofumi Hamada

Antibody-drug conjugates (ADCs), drugs developed by conjugation of an anticancer agent to a monoclonal antibody (mAb), have lately attracted attention in cancer therapy because ADCs can directly bind cancer cells and kill them. Although mAbs for ADCs must be internalized by the target cells, few methods are available for screening mAbs for their ability to be internalized by cells. We have developed a recombinant protein, termed DT3C, which consists of diphtheria toxin (DT) lacking the receptor-binding domain but containing the C1, C2, and C3 domains of Streptococcus protein G (3C). When a mAb-DT3C conjugate, which functions in vitro like an ADC, reduces the viability of cancer cells, the mAb being tested must have been internalized by the target cells. DT3C can thus be a tool to identify efficiently and easily mAbs that can be internalized by cells, thereby enhancing the development of promising ADCs.


Laboratory Investigation | 2016

Prolyl isomerase Pin1 promotes survival in EGFR -mutant lung adenocarcinoma cells with an epithelial–mesenchymal transition phenotype

Yuji Sakuma; Hirotaka Nishikiori; Sachie Hirai; Miki Yamaguchi; Gen Yamada; Atsushi Watanabe; Tadashi Hasegawa; Takashi Kojima; Toshiro Niki; Hiroki Takahashi

The secondary epidermal growth factor receptor (EGFR) T790M mutation is the most prominent mechanism that confers resistance to first- or second-generation EGFR tyrosine kinase inhibitors (TKIs) in lung cancer treatment. Although third-generation EGFR TKIs can suppress the kinase activity of T790M-positive EGFR, they still cannot eradicate EGFR-mutated cancer cells. We previously reported that a subpopulation of EGFR-mutant lung adenocarcinomas depends on enhanced autophagy, instead of EGFR, for survival, and in this study we explore another mechanism that contributes to TKI resistance. We demonstrate here that an EGFR-mutant lung adenocarcinoma cell line, H1975 (L858R+T790M), has a subset of cells that exhibits an epithelial–mesenchymal transition (EMT) phenotype and can thrive in the presence of third-generation EGFR TKIs. These cells depend on not only autophagy but also on the isomerase Pin1 for survival in vitro, unlike their parental cells. The Pin1 protein was expressed in an EGFR-mutant lung cancer tissue that has undergone partial EMT and acquired resistance to EGFR TKIs, but not its primary tumor. These findings suggest that inhibition of Pin1 activity can be a novel strategy in lung cancer treatment.


Journal of Gene Medicine | 2008

Selective gene transfer into neurons via Na,K-ATPase β1. Targeting gene transfer with monoclonal antibody and adenovirus vector

Keiji Ishii; Kiminori Nakamura; Satoshi Kawaguchi; Rong Li; Sachie Hirai; Naoya Sakuragi; Takuro Wada; Kazunori Kato; Toshihiko Yamashita; Hirofumi Hamada

Neuron‐selective gene transfer is an attractive therapeutic strategy for neurological disorders. However, optimal targets and gene delivery systems remain to be determined.


International Journal of Oncology | 2018

Survivin knockdown induces senescence in TTF‑1-expressing, KRAS-mutant lung adenocarcinomas

Toshiyuki Sumi; Sachie Hirai; Miki Yamaguchi; Yusuke Tanaka; Makoto Tada; Gen Yamada; Tadashi Hasegawa; Yohei Miyagi; Toshiro Niki; Atsushi Watanabe; Hiroki Takahashi; Yuji Sakuma

Survivin plays a key role in regulating the cell cycle and apoptosis, and is highly expressed in the majority of malignant tumors. However, little is known about the roles of survivin in KRAS-mutant lung adenocarcinomas. In the present study, we examined 28 KRAS-mutant lung adenocarcinoma tissues and two KRAS-mutant lung adenocarcinoma cell lines, H358 and H441, in order to elucidate the potential of survivin as a therapeutic target. We found that 19 (68%) of the 28 KRAS-mutant lung adenocarcinomas were differentiated tumors expressing thyroid transcription factor-1 (TTF-1) and E-cadherin. Patients with tumors immunohistochemically positive for survivin (n=18) had poorer outcomes than those with survivin-negative tumors (n=10). In the H358 and H441 cells, which expressed TTF-1 and E-cadherin, survivin knockdown alone induced senescence, not apoptosis. However, in monolayer culture, the H358 cells and H441 cells in which survivin was silenced, underwent significant apoptosis following combined treatment with ABT-263, a Bcl-2 inhibitor, and trametinib, a MEK inhibitor. Importantly, the triple combination of survivin knockdown with ABT-263 and trametinib treatment, clearly induced cell death in a three-dimensional cell culture model and in an in vivo tumor xenograft model. We also observed that the growth of the H358 and H441 cells was slightly, yet significantly suppressed in vitro when TTF-1 was silenced. These findings collectively suggest that the triple combination of survivin knockdown with ABT-263 and trametinib treatment, may be a potential strategy for the treatment of KRAS-mutant lung adenocarcinoma. Furthermore, our findings indicate that the well-differentiated type of KRAS-mutant lung tumors depends, at least in part, on TTF-1 for growth.


Experimental Cell Research | 2018

Characterization of distal airway stem-like cells expressing N-terminally truncated p63 and thyroid transcription factor-1 in the human lung

Yusuke Tanaka; Miki Yamaguchi; Sachie Hirai; Toshiyuki Sumi; Makoto Tada; Atsushi Saito; Hirofumi Chiba; Takashi Kojima; Atsushi Watanabe; Hiroki Takahashi; Yuji Sakuma

Abstract Distal airway stem cells (DASCs) in the mouse lung can differentiate into bronchioles and alveoli. However, it remains unclear whether the same stem cells exist in the human lung. Here, we found that human lung epithelial (HuL) cells, derived from normal, peripheral lung tissue, in monolayer, mostly express both the N‐terminally truncated isoform of p63 (&Dgr;Np63), a marker for airway basal cells, and thyroid transcription factor‐1 (TTF‐1), a marker for alveolar epithelial cells, even though these two molecules are usually expressed in a mutually exclusive way. Three‐dimensionally cultured HuL cells differentiated to form bronchiole‐like and alveolus‐like organoids. We also uncovered a few bronchiolar epithelial cells expressing both &Dgr;Np63 and TTF‐1 in the human lung, suggesting that these cells are the cells of origin for HuL cells. Taken together, &Dgr;Np63+ TTF‐1+ peripheral airway epithelial cells are possibly the human counterpart of mouse DASCs and may offer potential for future regenerative medicine. Graphical abstract Figure. No Caption available. HighlightsSome bronchiolar epithelial basal cells express both &Dgr;Np63 and TTF‐1 in the lung.These specific cells can form bronchiole‐like or alveolus‐like organoids.The dual‐positive cells can survive and differentiate in the mouse lung.They are a distinct type of peripheral lung epithelial stem cell in the human lung.

Collaboration


Dive into the Sachie Hirai's collaboration.

Top Co-Authors

Avatar

Hirofumi Hamada

Sapporo Medical University

View shared research outputs
Top Co-Authors

Avatar

Kazunori Kato

Sapporo Medical University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Miki Yamaguchi

Sapporo Medical University

View shared research outputs
Top Co-Authors

Avatar

Yuji Sakuma

Sapporo Medical University

View shared research outputs
Top Co-Authors

Avatar

Hiroki Takahashi

Sapporo Medical University

View shared research outputs
Top Co-Authors

Avatar

Yoshinori Ito

Sapporo Medical University

View shared research outputs
Top Co-Authors

Avatar

Atsushi Watanabe

Sapporo Medical University

View shared research outputs
Top Co-Authors

Avatar

Jianhua Huang

Sapporo Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge