Updated on 2025/05/21

写真a

 
Shouichi Higashi
 
Organization
Graduate School of Nanobioscience Department of Life and Environmental System Science Professor
School of Science Department of Science
Title
Professor
Profile
大学院時代に血液凝固反応の開始機構の研究を行って以来、一貫してタンパク質の構造-機能相関解析をベースとした疾患関連プロテアーゼの活性発現・調節機構の研究を進めている。これまでに組織因子による血液凝固第VIIa因子の触媒活性増強機構の解明、アミロイド前駆体タンパク質(APP)分子内のMMP-2選択的インヒビター部位の同定とその選択性発現機構の解明、MMP-7と細胞表層コレステロール硫酸との相互作用を介したがん転移促進機構解明等の成果を得た。これら成果を発展させ、がんや血栓症をはじめとした様々な疾患に対する創薬研究に繋げたいと考えている。
External link

Degree

  • Science ( Kyushu University )

Research Interests

  • factor VII

  • マトリックスメタロプロテアーゼ

  • MMP

  • アミロイド前駆体タンパク質

  • 細胞表層プロテオリシス

  • HAI-1

  • マトリプターゼ

  • MT1-MMP

  • コレステロール硫酸

  • MMP-7

  • MMP-9

  • amyloid precursor protein

  • tissue factor

  • cholesterol sulfate

  • selective MMP inhibitor

  • APP

  • APP-IP

  • 高特異性インヒビター

  • 細胞凝集

  • プロセッシング

  • TIMP

  • 分子認識及び相互作用

  • がんの浸潤・転移

  • 酵素活性阻害機構

  • tumor invasion and metastasis

  • MMPインヒビター

Research Areas

  • Life Science / Biomedical engineering

  • Nanotechnology/Materials / Chemistry and chemical methodology of biomolecules

  • Life Science / Tumor biology

  • Life Science / Biomaterials

  • Life Science / Functional biochemistry

  • Life Science / Structural biochemistry

▼display all

Education

  • Kyushu University

    - 1994

      More details

    Country: Japan

    researchmap

  • Kyushu University   Graduate School, Division of Medical Sciences   Molecular Biology

    - 1994

      More details

Research History

  • Yokohama City University International College of Arts and Sciences Life and Environmental Science Graduate School of Nanobioscience Department of Life and Environmental System Science   Professor

      More details

Professional Memberships

Committee Memberships

  • 日本病態プロテアーゼ学会   評議員  

    2013.8   

      More details

    Committee type:Academic society

    researchmap

Papers

  • Expression and Purification of Active Monomeric MMP7. International journal

    Kazuhiro Yamamoto, Moe Isohata, Shouichi Higashi

    Methods in molecular biology (Clifton, N.J.)   2747   67 - 73   2024

     More details

    Language:English   Publishing type:Research paper (scientific journal)  

    MMP7 is the smallest member of the MMP family and plays multiple physiological and pathological roles through interaction with a variety of molecules. Purified MMP7 would be beneficial for studying its function and for the development of inhibitors, which could be potential therapeutics. Due to low levels of endogenously produced MMP7, its recombinant expression and purification using E. coli have been established. Here, we describe an effective method to express and purify an active form of MMP7. Our recent discovery is that adding high concentration of CaCl2 during refolding process prevents nonspecific binding of MMP7 to plastic and its aggregation, significantly improving the yield of active monomeric forms of MMP7.

    DOI: 10.1007/978-1-0716-3589-6_6

    PubMed

    researchmap

  • Matrix metalloproteinase-7 induces homotypic tumor cell aggregation via proteolytic cleavage of the membrane-bound Kunitz-type inhibitor HAI-1 Reviewed

    Tomohiro Ishikawa, Yayoi Kimura, Hisashi Hirano, Shouichi Higashi

    JOURNAL OF BIOLOGICAL CHEMISTRY   292 ( 50 )   20769 - 20784   2017.12

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:AMER SOC BIOCHEMISTRY MOLECULAR BIOLOGY INC  

    Matrix metalloproteinase-7 (MMP-7) plays important roles in tumor progression and metastasis. Our previous studies have demonstrated that MMP-7 binds to colon cancer cells via cell surface-bound cholesterol sulfate and induces significant cell aggregation by cleaving cell-surface protein(s). These aggregated cells exhibit a dramatically enhanced metastatic potential. However, the molecular mechanism inducing this cell-cell adhesion through the proteolytic action of MMP-7 remained to be clarified. Here, we explored MMP-7 substrates on the cell surface; the proteins on the cell surface were first biotinylated, and a labeled protein fragment specifically released from the cells after MMP-7 treatment was analyzed using LC-MS/MS. We found that hepatocyte growth factor activator inhibitor type 1 (HAI-1), a membrane-bound Kunitz-type serine protease inhibitor, is an MMP-7 substrate. We also found that the cell-bound MMP-7 cleaves HAI-1 mainly between Gly(451) and Leu(452) and thereby releases the extracellular region as soluble HAI-1 (sHAI-1). We further demonstrated that this sHAI-1 can induce cancer cell aggregation and determined that the HAI-1 region corresponding to amino acids 141-249, which does not include the serine protease inhibitor domain, has the cell aggregation-inducing activity. Interestingly, a cell-surface cholesterol sulfate-independent proteolytic action of MMP-7 is critical for the sHAI-1-mediated induction of cell aggregation, whereas cholesterol sulfate is needed for the MMP-7-catalyzed generation of sHAI-1. Considering that MMP-7-induced cancer cell aggregation is an important mechanism in cancer metastasis, we propose that sHAI-1 is an essential component of MMP-7-induced stimulation of cancer metastasis and may therefore represent a suitable target for antimetastatic therapeutic strategies.

    DOI: 10.1074/jbc.M117.796789

    Web of Science

    researchmap

  • A novel bacterial transport mechanism of Acinetobacter baumannii via activated human neutrophils through interleukin-8 Reviewed

    Go Kamoshida, Shigeru Tansho-Nagakawa, Takane Kikuchi-Ueda, Ryuichi Nakano, Kenji Hikosaka, Satoshi Nishida, Tsuneyuki Ubagai, Shouichi Higashi, Yasuo Ono

    JOURNAL OF LEUKOCYTE BIOLOGY   100 ( 6 )   1405 - 1412   2016.12

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:FEDERATION AMER SOC EXP BIOL  

    Hospital-acquired infections as a result of Acinetobacter baumannii have become problematic because of high rates of drug resistance. Although neutrophils play a critical role in early protection against bacterial infection, their interactions with A. baumannii remain largely unknown. To elucidate the interactions between A. baumannii and human neutrophils, we cocultured these cells and analyzed them by microscopy and flow cytometry. We found that A. baumannii adhered to neutrophils. We next examined neutrophil and A. baumannii infiltration into Matrigel basement membranes by an in vitro transmigration assay. Neutrophilswere activated by A. baumannii, and invasion was enhanced. More interestingly, A. baumannii was transported together by infiltrating neutrophils. Furthermore, we observed by live cell imaging that A. baumannii and neutrophils moved together. In addition, A. baumannii-activated neutrophils showed increased IL-8 production. The transport of A. baumannii was suppressed by inhibiting neutrophil infiltration by blocking the effect of IL-8. A. baumannii appears to use neutrophils for transport by activating these cells via IL-8. In this study, we revealed a novel bacterial transport mechanism that A. baumannii exploits human neutrophils by adhering to and inducing IL-8 release for bacterial portage. This mechanism might be a new treatment target.

    DOI: 10.1189/jlb.4AB0116-023RR

    Web of Science

    PubMed

    researchmap

  • Small-molecule auxin inhibitors that target YUCCA are powerful tools for studying auxin function Reviewed

    Yusuke Kakei, Chiaki Yamazaki, Masashi Suzuki, Ayako Nakamura, Akiko Sato, Yosuke Ishida, Rie Kikuchi, Shouichi Higashi, Yumiko Kokudo, Takahiro Ishii, Kazuo Soeno, Yukihisa Shimada

    PLANT JOURNAL   84 ( 4 )   827 - 837   2015.11

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:WILEY  

    Auxin is essential for plant growth and development, this makes it difficult to study the biological function of auxin using auxin-deficient mutants. Chemical genetics have the potential to overcome this difficulty by temporally reducing the auxin function using inhibitors. Recently, the indole-3-pyruvate (IPyA) pathway was suggested to be a major biosynthesis pathway in Arabidopsis thaliana L. for indole-3-acetic acid (IAA), the most common member of the auxin family. In this pathway, YUCCA, a flavin-containing monooxygenase (YUC), catalyzes the last step of conversion from IPyA to IAA. In this study, we screened effective inhibitors, 4-biphenylboronic acid (BBo) and 4-phenoxyphenylboronic acid (PPBo), which target YUC. These compounds inhibited the activity of recombinant YUC in vitro, reduced endogenous IAA content, and inhibited primary root elongation and lateral root formation in wild-type Arabidopsis seedlings. Co-treatment with IAA reduced the inhibitory effects. Kinetic studies of BBo and PPBo showed that they are competitive inhibitors of the substrate IPyA. Inhibition constants (Ki) of BBo and PPBo were 67 and 56 nM, respectively. In addition, PPBo did not interfere with the auxin response of auxin-marker genes when it was co-treated with IAA, suggesting that PPBo is not an inhibitor of auxin sensing or signaling. We propose that these compounds are a class of auxin biosynthesis inhibitors that target YUC. These small molecules are powerful tools for the chemical genetic analysis of auxin function.

    DOI: 10.1111/tpj.13032

    Web of Science

    PubMed

    researchmap

  • Amino-terminal fragments of laminin gamma 2 chain stimulate migration of metastatic breast cancer cells by interacting with CD44 Reviewed

    Hiroki Sato, Shouichi Higashi, Kaoru Miyazaki

    CLINICAL & EXPERIMENTAL METASTASIS   32 ( 5 )   405 - 415   2015.6

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:SPRINGER  

    Laminin gamma 2 (Lm gamma 2) chain, a subunit of the basement membrane protein laminin-332, is regarded as a typical cancer invasion marker. The overexpression of Lm gamma 2 chain by invasive cancer cells correlates with poor prognosis of cancer patients, and its forced expression in human cancer cells promotes their invasive growth in a nude mouse model. However, its actual roles in cancer progression, as well as the mechanism of its proinvasive effect, remain unclear. CD44 is known to be an important cancer stem cell marker and support cancer progression and stem cell functions. Here we demonstrate that amino-terminal fragments of Lm gamma 2 interact with CD44 on the membrane of breast cancer cells. Lm gamma 2 highly bound to the metastatic cell line MDA-MB-231 but poorly to the benign cell line MCF-7. The membrane receptor for Lm gamma 2 on MDA-MB-231 cells was identified to be the standard form of CD44 (CD44s) by co-immunoprecipitation, affinity chromatography and direct protein interaction assay. Lm gamma 2 interacted with CD44s through EGF-like repeat 2/3 in the Lm gamma 2 amino-terminus. Amino-terminal fragments of Lm gamma 2 induced the phosphorylation of CD44 cytoplasmic domain and stimulated migration of the cancer cells in a CD44-dependent manner. This migration was blocked by inhibitors of TGF-beta receptor I (TGF-beta RI) kinase. These results suggest that two important tumor markers, Lm gamma 2 and CD44, cooperate for cancer progression and possibly for cancer stem cell functions. TGF-beta RI may be involved in the Lm gamma 2/CD44 interaction.

    DOI: 10.1007/s10585-015-9705-6

    Web of Science

    PubMed

    researchmap

  • Inhibition of transforming growth factor-β signaling potentiates tumor cell invasion into collagen matrix induced by fibroblast-derived hepatocyte growth factor. Reviewed International journal

    Jun Oyanagi, Nako Kojima, Haruki Sato, Shouichi Higashi, Keiji Kikuchi, Katsuya Sakai, Kunio Matsumoto, Kaoru Miyazaki

    Experimental cell research   326 ( 2 )   267 - 79   2014.8

     More details

    Language:English   Publishing type:Research paper (scientific journal)  

    Interaction between tumor cells and stromal fibroblasts plays essential roles in tumor progression. However, its detailed molecular mechanism remains unclear. To understand the mechanism, we investigated molecules mediating this interaction using the three-dimensional (3D) co-culture system of Panc-1 pancreatic carcinoma cells with normal fibroblasts. When the two kinds of cells were placed on the top of collagen gel, the tumor cells scattered into the fibroblast layer, apparently undergoing epithelial-mesenchymal transition. When fibroblasts were placed within collagen gel, Panc-1 cells actively invaded into the collagen gel, extending a microtubule-based long protrusion. Although transforming growth factor-β (TGF-β) and hepatocyte growth factor (HGF) individually stimulated the tumor cell invasion into collagen gel without fibroblasts, TGF-β signaling inhibitors (SB431542 and LY2157299) significantly enhanced the Panc-1 cell invasion in the 3D co-culture with fibroblasts. Experiments with HGF/Met signaling inhibitors or with the fibroblast conditioned medium revealed that HGF was a major invasion-promoting factor secreted from fibroblasts and SB431542 increased the HGF secretion by blocking the HGF-suppressing activity of cancer cell-derived TGF-β. These results indicate that HGF and TGF-β are critical regulators for both tumor-stroma interaction and tumor invasion. The results also suggest that TGF-β signaling inhibitors may promote tumor progression under some pathological conditions.

    DOI: 10.1016/j.yexcr.2014.04.009

    PubMed

    researchmap

  • Pericellular proteolysis by matrix metalloproteinase-7 is differentially modulated by cholesterol sulfate, sulfatide, and cardiolipin Reviewed

    Kazuhiro Yamamoto, Kaoru Miyazaki, Shouichi Higashi

    FEBS JOURNAL   281 ( 15 )   3346 - 3356   2014.8

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:WILEY-BLACKWELL  

    Matrix metalloproteinase (MMP)-7 binds to cell surface cholesterol sulfate (CS) and acts as a membrane-associated protease. We have previously found that CS modulates the substrate preference of MMP-7, thereby regulating its pericellular proteolytic action. MMP-7 potentially associates with the cell surface via sulfatide (SM4) and cardiolipin (CL) when they are overexpressed on the cell surface. Here, we investigated the molecular interaction between these acidic lipids and MMP-7 or its substrates, and their effects on the activity of MMP-7. Studies using MMP-7 variants with low CS-binding ability suggested that these lipids interact with a similar site on MMP-7. The hydroxamate-based MMP inhibitor TAPI-1 markedly reduced the affinity of MMP-7 for CS and CL, whereas that for SM4 was not affected by TAPI-1. These three acidic lipids also had different effects on the hydrolytic activity of MMP-7 towards a small peptide substrate: SM4, CL and CS reduced the activity to 80%, 92%, and 20%, respectively. Nevertheless, SM4 and CS similarly accelerated the MMP-7-catalyzed degradation of fibronectin and laminin-332, whereas CL did not. The increased proteolysis of substrate was observed only when both substrate and enzyme had affinity for the lipid, suggesting that the lipids probably bring the reactants into closer proximity. Furthermore, MMP-7 bound to cell surface SM4 or CS cleaved specific cell surface proteins and released similar fragments, whereas the cleavage was not stimulated by cell surface CL-bound MMP-7. This study provides a novel mechanism by which acidic lipids differentially regulate pericellular proteolysis by MMP-7 through allosteric alteration of the substrate-binding site and their inherent affinities for MMP-7 substrates.

    DOI: 10.1111/febs.12865

    Web of Science

    PubMed

    researchmap

  • Angiomodulin, a marker of cancer vasculature, is upregulated by vascular endothelial growth factor and increases vascular permeability as a ligand of integrin alpha v beta 3 Reviewed

    Eriko Komiya, Hiroki Sato, Naoko Watanabe, Marii Ise, Shouichi Higashi, Yohei Miyagi, Kaoru Miyazaki

    CANCER MEDICINE   3 ( 3 )   537 - 549   2014.6

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:WILEY-BLACKWELL  

    Angiomodulin (AGM) is a member of insulin-like growth factor binding protein (IGFBP) superfamily and often called IGFBP-rP1 or IGFBP-7. AGM was originally identified as a tumor-derived cell adhesion factor, which was highly accumulated in blood vessels of human cancer tissues. AGM is also overexpressed in cancer-associated fibroblasts (CAFs) and activates fibroblasts. However, some studies have shown tumor-suppressing activity of AGM. To understand the roles of AGM in cancer progression, we here investigated the expression of AGM in benign and invasive breast cancers and its functions in cancer vasculature. Immunohistochemical analysis showed that AGM was highly expressed in cancer vasculature even in ductal carcinoma in situ (DCIS) as compared to normal vasculature, while its expression in CAFs was more prominent in invasive carcinomas than DCIS. In vitro analyses showed that AGM was strongly induced by vascular endothelial cell growth factor (VEGF) in vascular endothelial cells. Although AGM stimulated neither the growth nor migration of endothelial cells, it supported efficient adhesion of endothelial cells. Integrin alpha v beta 3 was identified as a novel major receptor for AGM in vascular endothelial cells. AGM retracted endothelial cells by inducing actin stress fibers and loosened their VE-cadherin-mediated intercellular junction. Consequently, AGM increased vascular permeability both in vitro and in vivo. Furthermore, AGM and integrin alpha v beta 3 were highly expressed and colocalized in cancer vasculature. These results suggest that AGM cooperates with VEGF to induce the aberrant functions of cancer vasculature as a ligand of integrin alpha v beta 3.

    DOI: 10.1002/cam4.216

    Web of Science

    PubMed

    researchmap

  • Modulation of matrix metalloproteinase-9 secretion from tumor-associated macrophage-like cells by proteolytically processed laminin-332 (laminin-5) Reviewed

    Go Kamoshida, Takashi Ogawa, Jun Oyanagi, Hiroki Sato, Eriko Komiya, Shouichi Higashi, Kaoru Miyazaki, Tsutomu Tsuji

    CLINICAL & EXPERIMENTAL METASTASIS   31 ( 3 )   285 - 291   2014.3

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:SPRINGER  

    Macrophages infiltrating tumor tissues (tumor-associated macrophages, TAM) affect the malignant behaviors of tumor cells. We previously reported that monocytes were differentiated into TAM-like cells secreting matrix metalloproteinase (MMP)-9 by co-culture with tumor cells, and that cell adhesion to extracellular matrix (ECM) proteins played a critical role in the differentiation. In this study, we found that the monocyte differentiation was promoted by laminin-332 (laminin-5), a major epithelial ECM component. We also demonstrated that the proteolytic processing of the gamma 2 chain of laminin-332 was essential for its activity but that the N-terminal short arm of the gamma 2 chain inhibited MMP-9 secretion. These results indicate that the activity of laminin-332 for monocyte differentiation is dynamically regulated by the proteolytic processing of the gamma 2 chain.

    DOI: 10.1007/s10585-013-9627-0

    Web of Science

    PubMed

    researchmap

  • Amino-terminal fragments of laminin gamma 2 chain retract vascular endothelial cells and increase vascular permeability Reviewed

    Hiroki Sato, Jun Oyanagi, Eriko Komiya, Takashi Ogawa, Shouichi Higashi, Kaoru Miyazaki

    CANCER SCIENCE   105 ( 2 )   168 - 175   2014.2

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:WILEY-BLACKWELL  

    Laminin 2 (Lm2) chain, a subunit of laminin-332, is a typical molecular marker of invading cancer cells, and its expression correlates with poor prognosis of cancer patients. It was previously found that forced expression of Lm2 in cancer cells promotes their invasive growth in nude mice. However, the mechanism of the tumor-promoting activity of Lm2 remains unknown. Here we investigated the interaction between Lm2 and vascular endothelial cells. When treated with an N-terminal proteolytic fragment of 2 (2pf), HUVECs became markedly retracted or shrunken. The overexpression of Lm2 or treatment with 2pf stimulated T-24 bladder carcinoma cells to invade into the HUVEC monolayer and enhanced their transendothelial migration in vitro. Moreover, 2pf increased endothelial permeability in vitro and in vivo. As the possible mechanisms, 2pf activated ERK and p38 MAPK but inactivated Akt in HUVECs. Such effects of 2pf led to prominent actin stress fiber formation in HUVECs, which was blocked by a ROCK inhibitor. In addition, 2pf induced delocalization of VE-cadherin and -catenin from the intercellular junction. As possible receptors, 2pf interacted with heparan sulfate proteoglycans on the surface of HUVECs. Moreover, we localized the active site of 2pf to the N-terminal epidermal growth factor-like repeat. These data suggest that the interaction between 2pf and heparan sulfate proteoglycans induces cytoskeletal changes of endothelial cells, leading to the loss of endothelial barrier function and the enhanced transendothelial migration of cancer cells. These activities of Lm2 seem to support the aberrant growth of cancer cells.

    DOI: 10.1111/cas.12323

    Web of Science

    researchmap

  • Amino-terminal fragments of laminin γ2 chain retract vascular endothelial cells and increase vascular permeability Reviewed

    Hiroki Sato, Jun Oyanagi, Eriko Komiya, Takashi Ogawa, Shouichi Higashi, Kaoru Miyazaki

    Cancer Science   105 ( 2 )   168 - 175   2014.2

     More details

    Language:English   Publishing type:Research paper (scientific journal)  

    Laminin γ2 (Lmγ2) chain, a subunit of laminin-332, is a typical molecular marker of invading cancer cells, and its expression correlates with poor prognosis of cancer patients. It was previously found that forced expression of Lmγ2 in cancer cells promotes their invasive growth in nude mice. However, the mechanism of the tumor-promoting activity of Lmγ2 remains unknown. Here we investigated the interaction between Lmγ2 and vascular endothelial cells. When treated with an N-terminal proteolytic fragment of γ2 (γ2pf), HUVECs became markedly retracted or shrunken. The overexpression of Lmγ2 or treatment with γ2pf stimulated T-24 bladder carcinoma cells to invade into the HUVEC monolayer and enhanced their transendothelial migration in vitro. Moreover, γ2pf increased endothelial permeability in vitro and in vivo. As the possible mechanisms, γ2pf activated ERK and p38 MAPK but inactivated Akt in HUVECs. Such effects of γ2pf led to prominent actin stress fiber formation in HUVECs, which was blocked by a ROCK inhibitor. In addition, γ2pf induced delocalization of VE-cadherin and β-catenin from the intercellular junction. As possible receptors, γ2pf interacted with heparan sulfate proteoglycans on the surface of HUVECs. Moreover, we localized the active site of γ2pf to the N-terminal epidermal growth factor-like repeat. These data suggest that the interaction between γ2pf and heparan sulfate proteoglycans induces cytoskeletal changes of endothelial cells, leading to the loss of endothelial barrier function and the enhanced transendothelial migration of cancer cells. These activities of Lmγ2 seem to support the aberrant growth of cancer cells. © 2013 The Authors.

    DOI: 10.1111/cas.12323

    Scopus

    PubMed

    researchmap

  • Molecular Design of a Highly Selective and Strong Protein Inhibitor against Matrix Metalloproteinase-2 (MMP-2) Reviewed

    Shouichi Higashi, Tomokazu Hirose, Tomoka Takeuchi, Kaoru Miyazaki

    JOURNAL OF BIOLOGICAL CHEMISTRY   288 ( 13 )   9066 - 9076   2013.3

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:AMER SOC BIOCHEMISTRY MOLECULAR BIOLOGY INC  

    Synthetic inhibitors of matrix metalloproteinases (MMPs), designed previously, as well as tissue inhibitors of metalloproteinases (TIMPs) lack enzyme selectivity, which has been a major obstacle for developing inhibitors into safe and effective MMP-targeted drugs. Here we designed a fusion protein named APP-IP-TIMP-2, in which the ten amino acid residue sequence of APP-derived MMP-2 selective inhibitory peptide (APP-IP) is added to the N terminus of TIMP-2. The APP-IP and TIMP-2 regions of the fusion protein are designed to interact with the active site and the hemopexin-like domain of MMP-2, respectively. The reactive site of the TIMP-2 region, which has broad specificity against MMPs, is blocked by the APP-IP adduct. The recombinant APP-IP-TIMP-2 showed strong inhibitory activity toward MMP-2 (K-i(app) = 0.68 pM), whereas its inhibitory activity toward MMP-1, MMP-3, MMP-7, MMP-8, MMP-9, or MT1-MMP was six orders of magnitude or more weaker (IC50 > 1 mu M). The fusion protein inhibited the activation of pro-MMP-2 in the concanavalin A-stimulated HT1080 cells, degradation of type IV collagen by the cells, and the migration of stimulated cells. Compared with the decapeptide APP-IP (t(1/2) = 30 min), APP-IP-TIMP-2 (t(1/2) >> 96 h) showed a much longer half-life in cultured tumor cells. Therefore, the fusion protein may be a useful tool to evaluate contributions of proteolytic activity of MMP-2 in various pathophysiological processes. It may also be developed as an effective anti-tumor drug with restricted side effects.

    DOI: 10.1074/jbc.M112.441758

    Web of Science

    PubMed

    researchmap

  • Angiomodulin (AGM/IGFBP-rP1) is overexpressed in tumor vasculature and regulates adhesion of vascular endothelial cells via integrin alpha v beta 3: Possible roles in tumor angiogenesis Reviewed

    Miyazaki Kaoru, Komiya Eriko, Sato Hiroki, Miyagi Yohei, Higashi Shouichi

    CANCER RESEARCH   73   2013.2

  • Development of a highly selective inhibitor against matrix metalloproteinase-2 (MMP-2)

    63 ( 1 )   51 - 61   2013

  • Epithelial-Mesenchymal Transition Stimulates Human Cancer Cells to Extend Microtubule-based Invasive Protrusions and Suppresses Cell Growth in Collagen Gel Reviewed

    Jun Oyanagi, Takashi Ogawa, Hiroki Sato, Shouichi Higashi, Kaoru Miyazaki

    PLOS ONE   7 ( 12 )   e53209   2012.12

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:PUBLIC LIBRARY SCIENCE  

    Epithelial-mesenchymal transition (EMT) is a crucial event in tumor invasion and metastasis. However, most of past EMT studies have been conducted in the conventional two-dimensional (2D) monolayer culture. Therefore, it remains unclear what invasive phenotypes are acquired by EMT-induced cancer cells. To address this point, we attempted to characterize EMT cells in more physiological, three-dimensional (3D) collagen gel culture. EMT was induced by treating three human carcinoma cell lines (A549, Panc-1 and MKN-1) with TGF-beta. The TGF-beta treatment stimulated these cells to overexpress the invasion markers laminin gamma 2 and MT1-MMP in 2D culture, in addition to the induction of well-known morphological change and EMT marker expression. EMT induction enhanced cell motility and adhesiveness to fibronectin and collagen in 2D culture. Although EMT cells showed comparable cell growth to control cells in 2D culture, their growth rates were extremely suppressed in soft agar and collagen gel cultures. Most characteristically, EMT-induced cancer cells commonly and markedly extended invasive protrusions in collagen gel. These protrusions were mainly supported by microtubules rather than actin cytoskeleton. Snail-introduced, stable EMT cells showed similar protrusions in 3D conditions without TGF-beta. Moreover, these protrusions were suppressed by colchicine or inhibitors of heat shock protein 90 (HSP-90) and protein phosphatase 2A. However, MMP inhibitors did not suppress the protrusion formation. These data suggest that EMT enhances tumor cell infiltration into interstitial stroma by extending microtubule-based protrusions and suppressing cell growth. The elevated cell adhesion to fibronectin and collagen and high cell motility also seem important for the tumor invasion.

    DOI: 10.1371/journal.pone.0053209

    Web of Science

    PubMed

    researchmap

  • Elevated expression of angiomodulin (AGM/IGFBP-rP1) in tumor stroma and its roles in fibroblast activation Reviewed

    Eriko Komiya, Momoko Furuya, Naoko Watanabe, Yohei Miyagi, Shouichi Higashi, Kaoru Miyazaki

    CANCER SCIENCE   103 ( 4 )   691 - 699   2012.4

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:WILEY-BLACKWELL  

    Angiomodulin (AGM/IGFBP-rP1), a glycoprotein of about 30 kDa, is overexpressed in tumor vasculature as well as some human cancer cell lines, but it has been suggested to be a tumor suppressor. To elucidate roles of angiomodulin (AGM) in tumor progression, we here examined distribution of AGM in three types of human cancer tissues by immunohistochemistry. The results showed that AGM was overexpressed in the stroma as well as the vasculature surrounding tumor cells in the human cancer tissues. AGM and a-smooth muscle actin (a-SMA) as an activated fibroblast marker were often colocalized in cancer-associated fibroblasts (CAFs). In vitro analysis indicated that transforming growth factor (TGF)-beta might be an important inducer of AGM in normal human fibroblasts. AGM strongly stimulated the expression of fibronectin and weakly that of alpha-SMA in normal fibroblasts. AGM significantly stimulated the proliferation and migration of fibroblasts. The AGM-induced expression of fibronectin and alpha-SMA was blocked by a TGF-beta signal inhibitor but neither the stimulation of cell growth nor migration. These results imply that AGM activates normal fibroblasts by TGF-beta-dependent and independent mechanisms. These findings also suggest that AGM and TGF-beta cooperatively or complementarily contribute to the stromal activation and connective tissue formation in human cancer tissues, contributing to tumor progression. (Cancer Sci 2012; 103: 691699)

    DOI: 10.1111/j.1349-7006.2012.02203.x

    Web of Science

    PubMed

    researchmap

  • Structural Basis for Matrix Metalloproteinase-2 (MMP-2)-selective Inhibitory Action of beta-Amyloid Precursor Protein-derived Inhibitor Reviewed

    Hiroshi Hashimoto, Tomoka Takeuchi, Kyoko Komatsu, Kaoru Miyazaki, Mamoru Sato, Shouichi Higashi

    JOURNAL OF BIOLOGICAL CHEMISTRY   286 ( 38 )   33236 - 33243   2011.9

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:AMER SOC BIOCHEMISTRY MOLECULAR BIOLOGY INC  

    Unlike other synthetic or physiological inhibitors for matrix metalloproteinases (MMPs), the beta-amyloid precursor protein-derived inhibitory peptide (APP-IP) having an ISYGNDALMP sequence has a high selectivity toward MMP-2. Our previous study identified amino acid residues of MMP-2 essential for its selective inhibition by APP-IP and demonstrated that the N to C direction of the decapeptide inhibitor relative to the substrate-binding cleft of MMP-2 is opposite that of substrate. However, detailed interactions between the two molecules remained to be clarified. Here, we determined the crystal structure of the catalytic domain of MMP-2 in complex with APP-IP. We found that APP-IP in the complex is indeed embedded into the substrate-binding cleft of the catalytic domain in the N to C direction opposite that of substrate. With the crystal structure, it was first clarified that the aromatic side chain of Tyr(3) of the inhibitor is accommodated into the S1' pocket of the protease, and the carboxylate group of Asp(6) of APP-IP coordinates bidentately to the catalytic zinc of the enzyme. The Ala(7) to Pro(10) and Tyr(3) to Ile(1) strands of the inhibitor extend into the nonprime and the prime sides of the cleft, respectively. Therefore, the decapeptide inhibitor has long range contact with the substrate-binding cleft of the protease. This mode of interaction is probably essential for the high MMP-2 selectivity of the inhibitor because MMPs share a common architecture in the vicinity of the catalytic center, but whole structures of their substrate-binding clefts have sufficient variety for the inhibitor to distinguish MMP-2 from other MMPs.

    DOI: 10.1074/jbc.M111.264176

    Web of Science

    PubMed

    researchmap

  • Downregulation of a newly identified laminin, laminin-3B11, in vascular basement membranes of invasive human breast cancers Reviewed

    Taizo Mori, Yoshinobu Kariya, Eriko Komiya, Shouichi Higashi, Yohei Miyagi, Kiyotoshi Sekiguchi, Kaoru Miyazaki

    CANCER SCIENCE   102 ( 5 )   1095 - 1100   2011.5

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:WILEY-BLACKWELL  

    Laminins present in the basement membranes (BM) of blood vessels are involved in angiogenesis and other vascular functions that are critical for tumor growth and metastasis. Two major vascular laminins, the alpha 4 (laminin-411/421) and alpha 5 (laminin-511/521) types, have been well characterized. We recently found a third type of vascular laminin, laminin-3B11, consisting of the alpha 3B, beta 1 and gamma 1 chains, and revealed its biological activity. Laminin-3B11 potently stimulates vascular endothelial cells to extend lamellipodial protrusions. To understand the roles of laminin-3B11 in blood vessel functions and tumor growth, we examined localization of the laminin alpha 3B chain in normal mammary glands and breast cancers, in comparison with the alpha 4 and alpha 5 laminins. In the immunohistochemical analysis, the alpha 3B laminin was co-localized with the alpha 4 and alpha 5 laminins in the BM of venules and capillaries of normal breast tissues, but alpha 3B was scarcely detected in vessels near invasive breast carcinoma cells. In contrast, the alpha 4 laminin was overexpressed in capillaries of invasive carcinomas, where a large number of macrophages were found. The alpha 5 laminin appeared to be weakly downregulated in cancer tissues, especially in capillary vessels. Furthermore, our in vitro analysis indicated that TNF-alpha significantly suppressed the laminin alpha 3B expression in vascular endothelial cells, while it, as well as IL-1 beta and TGF-alpha, upregulated the alpha 4 expression. These results suggest that Lm3B11/3B21 may be required for normal mature vessels and interfere with tumor angiogenesis. (Cancer Sci 2011; 102: 1095-1100).

    DOI: 10.1111/j.1349-7006.2011.01892.x

    Web of Science

    PubMed

    researchmap

  • Laminin-3B11, a Novel Vascular-type Laminin Capable of Inducing Prominent Lamellipodial Protrusions in Microvascular Endothelial Cells Reviewed

    Taizo Mori, Kota Ono, Yoshinobu Kariya, Takashi Ogawa, Shouichi Higashi, Kaoru Miyazaki

    JOURNAL OF BIOLOGICAL CHEMISTRY   285 ( 45 )   35068 - 35078   2010.11

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:AMER SOC BIOCHEMISTRY MOLECULAR BIOLOGY INC  

    The basement membrane (BM) proteins laminins, which consist of alpha, beta, and gamma chains, support tissue structures and cellular functions. To date only alpha 4 and alpha 5 types of laminins have been identified in the BMs of blood vessels. Our recent study suggested the presence of novel alpha 3B-containing laminins in vascular BMs. Here we identified and characterized the third member of vascular laminins, laminin-3B11 (Lm3B11). RT-PCR analysis showed that microvascular endothelial (MVE) cells and umbilical vein endothelial cells expressed the messages for the alpha 3B, beta 1, beta 2, and gamma 1 chains. In the culture of MVE cells, alpha 3B was associated with beta 1 and gamma 1, producing Lm3B11. Recombinant Lm3B11 was overexpressed by introducing the cDNAs of the three chains into HEK-293 cells and purified to homogeneity. Purified Lm3B11 exhibited relatively weak cell adhesion activity through both alpha 3 beta 1 and alpha 6 beta 1 integrins. Most characteristically, Lm3B11 strongly stimulated MVE cells to extend many lamellipodial protrusions. This pseudopodial branching was blocked by an inhibitor for Src or phosphatidylinositol 3-kinase. Consistently, Lm3B11 stimulated the phosphorylation of Src and Akt more strongly than other laminins, suggesting that the integrin-derived signaling is mediated by these factors. The unique activity of Lm3B11 appears to be favorable to the branching of capillaries and venules.

    DOI: 10.1074/jbc.M110.146126

    Web of Science

    PubMed

    researchmap

  • Cholesterol Sulfate Alters Substrate Preference of Matrix Metalloproteinase-7 and Promotes Degradations of Pericellular Laminin-332 and Fibronectin Reviewed

    Kazuhiro Yamamoto, Kaoru Miyazaki, Shouichi Higashi

    JOURNAL OF BIOLOGICAL CHEMISTRY   285 ( 37 )   28862 - 28873   2010.9

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:AMER SOC BIOCHEMISTRY MOLECULAR BIOLOGY INC  

    Localization of secreted matrix metalloproteinases (MMPs) on the cell surface is required not only for processing of cell surface proteins, but also for controlled degradation of the extracellular matrix (ECM). Our previous study demonstrated that binding of MMP-7 (matrilysin) to cell surface cholesterol sulfate (CS) is essential for the cell membrane-associated proteolytic action of this MMP. In this study, we investigated the role of CS in the MMP-7-catalyzed degradation of protein components of ECM. We found that the degradation of laminin-332 (laminin-5) catalyzed by MMP-7 was accelerated dramatically in the presence of CS, whereas the sulfated lipid inhibited the degradation of casein catalyzed by the protease. The MMP-7 catalyzed degradation of fibronectin was partially inhibited in the presence of low concentrations of CS, whereas it was accelerated significantly at high concentrations of the lipid. Therefore, it is likely that CS alters the substrate preference of MMP-7. We also found that the proteins of which MMP-7-catalyzed degradation were accelerated by CS also had affinities for CS, suggesting that CS facilitates the proteolyses by cross-linking MMP-7 to its substrates. Moreover, MMP-7 tethered to cancer cell surface via CS degraded fibronectin and laminin-332 coated on a culture plate. The degradations of the adhesive proteins led to significant detachment of the cells from the plate. Taken together, our findings provide a novel mechanism in which cell surface CS promotes the proteolytic activities of MMP-7 toward selective substrates in the pericellular ECM, thereby contributing to cancer cell migration and metastasis.

    DOI: 10.1074/jbc.M110.136994

    Web of Science

    PubMed

    researchmap

  • Matrilysin (MMP-7) Cleaves C-Type Lectin Domain Family 3 Member A (CLEC3A) on Tumor Cell Surface and Modulates its Cell Adhesion Activity Reviewed

    Jun Tsunezumi, Shouichi Higashi, Kaoru Miyazaki

    JOURNAL OF CELLULAR BIOCHEMISTRY   106 ( 4 )   693 - 702   2009.3

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:WILEY-LISS  

    Matrilysin (MMP-7) plays important roles in tumor progression. Previous studies have suggested that MMP-7 binds to tumor cell surface and promotes their metastatic potential. In this study, we identified C-type lectin domain family 3 member A (CLEC3A) as a membrane-bound substrate of MMP-7. Although this protein is known to be expressed specifically in cartilage, its message was found in normal breast and breast cancer tissues as well as breast and colon cancer cell lines. Because few studies have been done on CLEC3A, we overexpressed its recombinant protein in human cancer cells. CLEC3A was found in the cell membrane, extracellular matrix (ECM), and culture medium of the CLEC3A-expressing cells. CLEC3A has a basic sequence in the NH(2)-terminal domain and showed a strong heparin-binding activity. MMP-7 cleaved the 20-kDa CLEC3A protein, dividing it to a 15-kDa COOH-terminal fragment and an NH(2)-terminal fragment with the basic sequence. The 15-kDa fragment no longer had heparin-binding activity. Treatment of file CLEC3A-expressing cells with MMP-7 released the 15-kDa CLEC3A into file culture supernatant, Furthermore, the 20-kDa CLEC3A promoted cell adhesion to laminin-332 and fibronectin substrates, but this activity was abrogated by the cleavage by MMP-7. These results suggest that CLEC3A binds to heparan sulfate proteoglycans oil cell surface, leading to the enhancement of cell adhesion to integrin ligands on ECM. It can be speculated that the cleavage of CLEC3A by MMP-7 weakens the stable adhesion of tumor cells to the matrix and promotes their migration in tumor microenvironments. J. Cell. Biochem. 106: 693-702, 2009. (C) 2009 Wiley-Liss, Inc.

    DOI: 10.1002/jcb.22062

    Web of Science

    PubMed

    researchmap

  • Identification of Amino Acid Residues of Matrix Metalloproteinase-7 Essential for Binding to Cholesterol Sulfate Reviewed

    Shouichi Higashi, Miwa Oeda, Kazuhiro Yamamoto, Kaoru Miyazaki

    JOURNAL OF BIOLOGICAL CHEMISTRY   283 ( 51 )   35735 - 35744   2008.12

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:AMER SOC BIOCHEMISTRY MOLECULAR BIOLOGY INC  

    Matrix metalloproteinase-7 (MMP-7; matrilysin) induces homotypic adhesion of colon cancer cells by cleaving cell surface protein(s) and enhances their metastatic potential. Our previous study (Yamamoto, K., Higashi, S., Kioi, M., Tsunezumi, J., Honke, K., and Miyazaki, K. (2006) J. Biol. Chem. 281, 9170-9180) demonstrated that binding of MMP-7 to cell surface cholesterol sulfate (CS) is essential for the cell membrane-associated proteolytic action of the protease. To determine the region of MMP-7 essential for binding to CS, we constructed chimeric proteases consisting of various parts of MMP-7 and those of the catalytic domain of MMP-2; the latter protease does not have an affinity for CS. Studies of these chimeric proteases and other mutants of MMP-7 revealed that Ile(29), Arg(33), Arg(51), and Trp(55), in the internal sequence, and the C-terminal three residues corresponding to residues 171-173 of MMP-7 are essential for binding to CS. An MMP-7 mutant, which had the internal 4 residues at positions 29, 33, 51, and 55 of MMP-7 replaced with the corresponding residues of MMP-2 and the C-terminal 3 residues deleted, had essentially no affinity for CS. This mutant and wild-type MMP-7 showed similar proteolytic activity toward fibronectin, whereas the mutant lacked the ability to induce the colon cancer cell aggregation. In the three-dimensional structure of MMP-7, the residues essential for binding to CS are located on the molecular surface in the opposite side of the catalytic cleft of the protease. Therefore, it is assumed that the active site of MMP-7 bound to cell surface is directed outside. We speculate that the direction of the cell-bound MMP-7 makes it feasible for the protease to cleave its substrates on cell surface.

    DOI: 10.1074/jbc.M806285200

    Web of Science

    PubMed

    researchmap

  • Matrilysin (matrix metalloprotease-7) cleaves membrane-bound annexin II and enhances binding of tissue-type plasminogen activator to cancer cell surfaces Reviewed

    Jun Tsunezumi, Kazuhiro Yamamoto, Shouichi Higashi, Kaoru Miyazaki

    FEBS JOURNAL   275 ( 19 )   4810 - 4823   2008.10

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:WILEY-BLACKWELL  

    Matrilysin (matrix metalloproteinase-7) plays important roles in tumor progression. It was previously found that matrilysin binds to the surface of colon cancer cells to promote their metastatic potential. In this study, we identified annexin II as a novel membrane-bound substrate of matrilysin. Treatment of human colon cancer cell lines with active matrilysin released a 35 kDa annexin II form, which lacked its N-terminal region, into the culture supernatant. The release of the 35 kDa annexin II by matrilysin was significantly enhanced in the presence of serotonin or heparin. Matrilysin hydrolyzed annexin II at the Lys9-Leu10 bond, thus dividing the protein into an N-terminal nonapeptide and the C-terminal 35 kDa fragment. Annexin II is known to serve as a cell surface receptor for tissue-type plasminogen activator (tPA). Although the matrilysin treatment liberated the 35 kDa fragment of annexin II from the cell surface, it significantly increased tPA binding to the cell membrane. A synthetic N-terminal nonapeptide of annexin II bound to tPA more efficiently than intact annexin II. This peptide formed a heterodimer with intact annexin II in test tubes and on cancer cell surfaces. These and other results suggested that the nonapeptide generated by matrilysin treatment might be anchored to the cell membrane, possibly by binding to intact annexin II, and interact with tPA via its C-terminal lysine. It is supposed that the cleavage of cell surface annexin II by matrilysin contributes to tumor invasion and metastasis by enhancing tPA-mediated pericellular proteolysis by cancer cells.

    DOI: 10.1111/j.1742-4658.2008.06620.x

    Web of Science

    PubMed

    researchmap

  • Identification of amino acid residues of the matrix metalloproteinase-2 essential for its selective inhibition by beta-amyloid precursor protein-derived inhibitor Reviewed

    Shouichi Higashi, Kaoru Miyazaki

    JOURNAL OF BIOLOGICAL CHEMISTRY   283 ( 15 )   10068 - 10078   2008.4

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:AMER SOC BIOCHEMISTRY MOLECULAR BIOLOGY INC  

    The extracellular domain of beta-amyloid precursor protein (APP) contains an inhibitor against matrix metalloproteinase-2 (MMP-2, gelatinase A). Our previous study (Higashi, S. and Miyazaki, K. (2003) J Biol Chem 278, 14020 - 14028) demonstrated that the inhibitor is localized within the ISYGN-DALMP sequence of APP, and a synthetic decapeptide containing this sequence (named APP-derived inhibitory peptide, APP-IP) selectively inhibits the activity of MMP-2. To determine the region of interaction that correlates with the selective inhibition, we constructed various MMP-2 mutants. An MMP-2 mutant, which had the hemopexin-like domain and three fibronectin-like type II domains of MMP-2 deleted, and native MMP-2 showed similar affinities for APP-IP, suggesting that only the catalytic domain of MMP-2 is essential for the interaction. Studies of chimeric proteases, consisting of various parts of the MMP-2 catalytic domain and those of MMP-7 (matrilysin) or MMP-9 (gelatinase B), further revealed that Ala88 and Gly94 in the non-prime side and Tyr145 and Thr146 in the prime side of the substrate- binding cleft of MMP-2 contribute separately to the selective inhibition. Replacement of the amino acid residue at position 94 of a chimeric MMP mutant affected its interaction with the C-terminal Pro(10) of APP-IP, whereas that of residues 145 148 affected the interaction with Tyr(3) of the inhibitor, suggesting that the N to C direction of APP-IP relative to the substrate- binding cleft of MMP is analogous to that of propeptide in proMMP, and opposite to that of substrate. When the APP-IP sequence was added to the N terminus of the catalytic domain of MMP-2, the activity of the protease was intramolecularly inhibited. We speculate that the direction of interaction makes the active site-bound APP-IP resistant to cleavage, thereby supporting the inhibitory action of the peptide inhibitor.

    DOI: 10.1074/jbc.M709509200

    Web of Science

    PubMed

    researchmap

  • Matriptase activates stromelysin (MMP-3) and promotes tumor growth and angiogenesis Reviewed

    Xinlian Jin, Motoki Yagi, Nagisa Akiyama, Tomomi Hirosaki, Shouichi Higashi, Chen-Yong Lin, Robert B. Dickson, Hitoshi Kitamura, Kaoru Miyazaki

    CANCER SCIENCE   97 ( 12 )   1327 - 1334   2006.12

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:BLACKWELL PUBLISHING  

    Matriptase/MT-SP1, a type II membrane serine protease widely expressed in normal epithelial cells and human carcinoma cells, is thought to be involved in cancer progression. To clarify this possibility, we overexpressed exogenous matriptase in the human stomach cancer cell line AZ521. In vitro, the matriptase transfectant (Mat-AZ521) and the control transfectant (Mock-AZ521) showed a similar growth rate, although the saturation cell density was significantly higher with the Mat-AZ521. When implanted into nude mice subcutaneously or intraperitoneally, Mat-AZ521 cells grew faster and produced much larger solid tumors than Mock-AZ521 cells. The overexpression of matriptase in AZ521 cells shortened the survival time of tumor-bearing mice. Histological analysis showed that both the number and the size of blood vessels in tumor tissues were significantly higher in the Mat-AZ521 tumors than the Mock-AZ521 ones. Moreover, it was found that purified matriptase activated one of the important matrix metalloproteinases, stromelysin (MMP-3). These results suggest the possibility that the matriptase-dependent activation of MMP-3, as well as the direct activity of matriptase, promotes tumor growth and angiogenesis by enhancing extracellular matrix degradation in tumor cell microenvironments.

    DOI: 10.1111/j.1349-7006.2006.00328.x

    Web of Science

    PubMed

    researchmap

  • Binding of active matrilysin to cell surface cholesterol sulfate is essential for its membrane-associated proteolytic action and induction of homotypic cell adhesion Reviewed

    K Yamamoto, S Higashi, M Kioi, J Tsunezumi, K Honke, K Miyazaki

    JOURNAL OF BIOLOGICAL CHEMISTRY   281 ( 14 )   9170 - 9180   2006.4

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:AMER SOC BIOCHEMISTRY MOLECULAR BIOLOGY INC  

    Regulation of cell surface molecules by matrix metalloproteinases (MMPs), as well as MMPs-catalyzed degradation of extracellular matrix, is important for tumor invasion and metastasis. Our previous study (Kioi, M., Yamamoto, K., Higashi, S., Koshikawa, N., Fujita, K., and Miyazaki, K. (2003) Oncogene 22, 8662-8670) demonstrated that active matrilysin specifically binds to the surface of colon cancer cells and induces notable cell aggregation due to processing of the cell membrane protein(s). Furthermore, these aggregated cells showed a dramatically enhanced metastatic potential. To elucidate the mechanism of matrilysin-induced cell aggregation, we attempted to identify the matrilysin-binding substance on the cell surface. Here, we demonstrate that cholesterol sulfate on the cell surface is a major matrilysin-binding substance. We found that active matrilysin bound to the cell membrane and cholesterol sulfate incorporated into liposomes with similar affinities. Treatment of colon cancer cells with beta-cyclodextrin significantly reduced not only matrilysin binding to the cell surface but also matrilysin-dependent proteolysis and cell aggregation. Interestingly, replenishment of cholesterol sulfate, but not cholesterol, neutralized the effects of beta-cyclodextrin. Taken together, it is likely that binding of matrilysin to cholesterol sulfate facilitates the matrilysin-catalyzed modulation of cell surface proteins, thus inducing the cancer cell aggregation.

    DOI: 10.1074/jbc.M510377200

    Web of Science

    PubMed

    researchmap

  • Identification of membrane-bound serine proteinase matriptase as processing enzyme of insulin-like growth factor binding protein-related protein-1 (IGFBP-rP1/angiomodulin/mac25) Reviewed

    S Ahmed, XL Jin, M Yagi, C Yasuda, Y Sato, S Higashi, CY Lin, RB Dickson, K Miyazaki

    FEBS JOURNAL   273 ( 3 )   615 - 627   2006.2

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:BLACKWELL PUBLISHING  

    Insulin-like growth factor (IGF) binding protein-related protein-1 (IGFBP-rP1) modulates cellular adhesion and growth in an IGF/insulin-dependent or independent manner. It also shows tumor-suppressive activity in vivo. We recently found that a single-chain IGFB-rP1 is proteolytically cleaved to a two-chain form by a trypsin-like, endogenous serine proteinase, changing its biological activities. In this study, we attempted to identify the IGFBP-rP1-processing enzyme. Of nine human cell lines tested, seven cell lines secreted IGFBP-rP1 at high levels, and two of them, ovarian clear cell adenocarcinoma (OVISE) and gastric carcinoma (MKN-45), highly produced the cleaved TGFBP-rP1. Serine protemase inhibitors effectively blocked the IGFBP-rP1 cleavage in the OVISE cell culture. The conditioned medium of OVISE cells did not cleave purified IGFBP-rP1, but their membrane fraction had an IGFBP-rP1-cleaving activity. The membrane fraction contained an 80-kDa gelatinolytic enzyme, which was identified as the membrane-type serine proteinase matriptase (MT-SP1) by immunoblotting. When the membrane fraction was separated by SDS/PAGE, the IGFBP-rP1-cleaving activity comigrated with matriptase. A soluble form of matriptase purified in an inhibitor-free form efficiently cleaved IGFBP-rP1 at the same site as that found in a naturally cleaved IGFBP-rP1. Furthermore, small interfering RNAs for matriptase efficiently blocked both the matriptase expression and the cleavage of IGBP-rP1 in OVISE cells. These results demonstrate that IGFBP-rP1 is processed to the two-chain form by matriptase on the cell surface.

    DOI: 10.1111/j.1742-4658.2005.05094.x

    Web of Science

    PubMed

    researchmap

  • Production of soluble matriptase by human cancer cell lines and cell surface activation of its zymogen by trypsin Reviewed

    XL Jin, T Hirosaki, CY Lin, RB Dickson, S Higashi, H Kitamura, K Miyazaki

    JOURNAL OF CELLULAR BIOCHEMISTRY   95 ( 3 )   632 - 647   2005.6

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:WILEY-LISS  

    The membrane-bound serine proteinase matriptase, which is often released from the plasma membrane of epithelial and carcinoma cells, has been implicated to play important roles in both physiological and pathological conditions. However, the regulatory mechanism of its activity is poorly understood. In the present study, we examined expression and activation state of soluble matriptase in 24 human cancer cell lines. Soluble matriptase was detected in the conditioned media from all of 5 colon and 4 breast carcinoma cell lines and 8 of 10 stomach carcinoma cell lines tested. Only two of five lung cancer cell lines released the matriptase protein into the culture media. Out of the five matriptase-negative cell lines, two cell lines expressed the matriptase mRNA. Among 24 cancer cell lines tested, 13 cell lines secreted trypsin in an active or latent form and all of them released matriptase. Most of the 24 cell lines released a latent, single-chain matriptase of 75 kDa as a major form, as well as low levels of complex forms of an activated two-chain enzyme with its specific inhibitor HAI-1. Thus, these soluble matriptases appeared to have little proteolytic activity. Treatment of stomach and colon cancer cell lines with epidermal growth factor stimulated the release of matripatase/HAI-1 complexes. In cancercell lines secreting active trypsin, however, matriptase was released mostly as an inhibitor-free, two-chain active form. Trypsin seemed to activate the membrane-bound, latent matriptase on the cell surface. These results suggest that matriptase and trypsin cooperatively function for extracellular proteolysis. J. Cell. Biochem. 95: 632-647, 2005. (c) 2005 Wiley-Liss, Inc.

    DOI: 10.1002/jcb.20418

    Web of Science

    PubMed

    researchmap

  • Matrilysin (MMP-7) induces homotypic adhesion of human colon cancer cells and enhances their metastatic potential in nude mouse model Reviewed

    M Kioi, K Yamamoto, S Higashi, N Koshikawa, K Fujita, K Miyazaki

    ONCOGENE   22 ( 54 )   8662 - 8670   2003.11

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:NATURE PUBLISHING GROUP  

    Matrilysin (MMP-7) is thought to contribute to invasive growth and metastasis of colon carcinoma and many other human cancers. The present study demonstrates that treatment of human colon carcinoma cells with active matrilysin induces cell aggregation in vitro and promotes liver metastasis in nude mice. When two kinds of colon carcinoma cell lines were incubated with active matrilysin, this enzyme efficiently bound to the cell surface and induced loose cell aggregation, which led to E-cadherin-mediated tight cell aggregation. Synthetic MMP inhibitors inhibited both the membrane binding of matrilysin and matrilysin-induced cell aggregation, while TIMP-2 inhibited only the cell aggregation. Two other active MMPs, stromelysin and gelatinase A, neither bound to cell membrane nor induced cell aggregation. Tumor cells in loose cell aggregates could reaggregate even after they were freed from matrilysin and dispersed. When injected into the spleen of nude mice, the tumor cells in the stable aggregates produced much larger metastatic nodules in the livers than control cells and those in the loose aggregates. These results suggest that matrilysin may enhance metastatic potential of tumor cells by processing a cell surface protein(s) and thereby inducing loose and then tight aggregation of tumor cells.

    DOI: 10.1038/sj.onc.1207181

    Web of Science

    PubMed

    researchmap

  • Proteolytic processing of IGFBP-related protein-1 (TAF/angiomodulin/mac25) modulates its biological activity Reviewed

    S Ahmed, K Yamamoto, Y Sato, T Ogawa, A Herrmann, S Higashi, K Miyazaki

    BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS   310 ( 2 )   612 - 618   2003.10

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:ACADEMIC PRESS INC ELSEVIER SCIENCE  

    Insulin-like growth factor (IGF) binding protein-related protein-1 (IGFBP-rP1) was previously identified as tumor-derived adhesion factor (TAF) secreted from human bladder carcinoma cells. It exhibits growth-stimulatory activity in synergy with insulin or IGFs. In the present study, we found that IGFBP-rP1 was proteolytically cleaved to a two-chain form. The cleavage sequence suggested that a trypsin-like serine proteinase may be responsible for the processing. The cleavage of IGFBP-rP1 led to an almost complete loss of both insulin/IGF-1-binding activity and insulin/IGF-1-dependent growth-stimulatory activity. On the other hand, the cell attachment activity of IGFBP-rP1 was markedly increased by the proteolytic processing. Syndecan-1 was thought to be a cell surface receptor for both intact and cleaved IGFBP-rP1 forms. Although the proteolytic cleavage of IGFBP-rP1 decreased its heparin-binding activity, the cleaved form could bind syndecan-1 efficiently. Thus the proteolytic processing of IGFBP-rP1 seems to modulate its insulin/IGF-dependent and -independent biological functions. (C) 2003 Elsevier Inc. All rights reserved.

    DOI: 10.1016/j.bbrc.2003.09.058

    Web of Science

    PubMed

    researchmap

  • Novel processing of beta-amyloid precursor protein catalyzed by membrane type 1 matrix metalloproteinase releases a fragment lacking the inhibitor domain against gelatinase A Reviewed

    S Higashi, K Miyazaki

    BIOCHEMISTRY   42 ( 21 )   6514 - 6526   2003.6

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:AMER CHEMICAL SOC  

    In various mammalian cell lines, beta-amyloid precursor protein (APP) is proteolytically processed to release its NH2-terminal extracellular domain as a soluble APP (sAPP) that contains the inhibitor domain against gelatinase A. To investigate roles of SAPP in the regulation of gelatinase A activity, we examined the correlation between the activation of progelatinase A and processing of APP. We found that stimulation of HT1080 fibrosarcoma cells with concanavalin A led to an activation of endogenous progelatinase A and to a novel processing of APP, which releases a COOH-terminally truncated form of sAPP (sAPPtrc) into the culture medium. Reverse zymographic analysis showed that sAPPtrc lacked an inhibitory activity against gelatinase A. Analyses of production of sAPPtrc in the presence of various metalloproteinase inhibitors showed that membrane type 1 matrix metalloproteinase (MT1-MMP), an activator of progelatinase A, is most likely responsible for the production of sAPPtrc. When the concanavalin A-stimulated HT 1080 cells were cultured in the condition that inhibited MT1-MMP activity, sAPP and APP were associated with the extracellular matrix deposited by the cells, whereas these gelatinase A inhibitors in the matrix were displaced by sAPPtrc after exertion of MT1-MMP activity. Taken together, these data support a model in which MT1-MMP-catalyzed release of sAPPtrc leads to reduction of the extracellular matrix-associated gelatinase A inhibitor, SAPP, thus making it feasible for gelatinase A to exert proteolytic activity only near its activator, MT1-MMP.

    DOI: 10.1021/bi020643m

    Web of Science

    PubMed

    researchmap

  • Identification of a region of beta-amyloid precursor protein essential for its gelatinase a inhibitory activity Reviewed

    S Higashi, K Miyazaki

    JOURNAL OF BIOLOGICAL CHEMISTRY   278 ( 16 )   14020 - 14028   2003.4

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:AMER SOC BIOCHEMISTRY MOLECULAR BIOLOGY INC  

    Because beta-amyloid precursor protein (APP) has the abilities both to interact with extracellular matrix and to inhibit gelatinase A activity, this molecule is assumed to play a regulatory role in the gelatinase A-catalyzed degradation of extracellular matrix. To determine a region of APP essential for the inhibitory activity, we prepared various derivatives of APP. Functional analyses of proteolytic fragments of soluble APP (sAPP) and glutathione S-transferase fusion proteins, which contain various COOH-terminal parts of sAPP, showed that a site containing residues 579-601 of APP(770) is essential for the inhibitory activity. Moreover, a synthetic decapeptide containing the 579-601 sequence corresponding to residues 586-595 of APP(770) had a gelatinase A inhibitory activity slightly higher than that of sAPP. Studies of deletion of the NH2- and COOH-terminal residues and alanine replacement of internal residues of the decapeptide further revealed that Tyr(588), Asp(591), and Leu(593) of APP mainly stabilize the interaction between gelatinase A and the inhibitor. We also found that the residues of Ile(586), Met(594), and Pro(595) modestly contribute to the inhibitory activity. The APP-derived decapeptide efficiently inhibited the activity of gelatinase A (IC50 = 30 nm), whereas its inhibitory activity toward membrane type 1 matrix metalloproteinase was much weaker (IC50 = 2 muM). The decapeptide had poor inhibitory activity toward gelatinase B, matrilysin, and stromelysin (IC50 > 10 muM). The APP-derived inhibitor formed a complex with active gelatinase A but not with progelatinase A, and the complex formation was prevented completely by a hydroxamate-based synthetic inhibitor. Therefore, the decapeptide region of APP is likely an active site-directed inhibitor that has high selectivity toward gelatinase A.

    DOI: 10.1074/jbc.M212264200

    Web of Science

    PubMed

    researchmap

  • Factor VIIa modified in the 170 loop shows enhanced catalytic activity but does not change the zymogen-like property Reviewed

    K Soejima, J Mizuguchi, M Yuguchi, T Nakagaki, S Higashi, S Iwanaga

    JOURNAL OF BIOLOGICAL CHEMISTRY   276 ( 20 )   17229 - 17235   2001.5

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:AMER SOC BIOCHEMISTRY MOLECULAR BIOLOGY INC  

    Factor VIIa (VIIa) is an unusual trypsin-type serine proteinase that appears to exist in an equilibrium between minor active and dominant zymogen-like inactive conformational states. The binding of tissue factor to VIIa is assumed to shift the equilibrium into the active state. The proteinase domain of VIIa contains a unique structure: a loop formed by a disulfide bond between Cys(310) and Cys(329), which is five residues longer than those of other trypsin types. To examine the functional role of the loop region, we prepared two mutants of VIIa, One of the mutants, named VII-11, had five extra corresponding residues 316-320 of VII deleted. The other mutant, VII-31, had all of the residues in its loop replaced! with those of trypsin, Functional analysis of the two mutants showed that VIIa-11 (K-d = 41 nM) and VIIa-31 (K-d = 160 nM) had lower affinities for soluble tissue factor as compared with the wild-type VIIa (K-d = 11 nM). The magnitude of tissue factor-mediated acceleration of amidolytic activities of VIIa-11 (7-fold) and that of VIIa-31 (2-fold) were also smaller than that of wild-type VIIa (30-fold). In the absence of tissue factor, VIIa-31 but not VIIa-11 showed enhanced activity; the catalytic efficiencies of VIIa-31 toward various chromogenic substrates were 2-18-fold greater than those off the wildtype VIIa. Susceptibility of the alpha -amino group of IIe-153 of VIIa-31 to carbamylation was almost the same as that of wild-type VIIa, suggesting that VIIa-31 as well as wildtype VIIa exist predominantly in the zymogen-like state. Therefore, the tested modifications in the loop region had adverse effects on affinity for tissue factor, disturbed the tissue factor-induced conformational transition, and changed the catalytic efficiency of VIIa, but they did not affect the equilibrium between active and zymogen-like conformational states.

    DOI: 10.1074/jbc.M009206200

    Web of Science

    PubMed

    researchmap

  • A novel processing of β-amyloid precursor protein catalyzed by MT1-MMP abrogates its metalloproteinase inhibitor activity Reviewed

    Shouichi Higashi, Kaoru Miyazaki

    Keio Journal of Medicine   50   44 - 53   2001

     More details

    Language:English   Publishing type:Research paper (scientific journal)  

    DOI: 10.2302/kjm.50.supplement3_44

    Scopus

    researchmap

  • Isolation and activity of proteolytic fragment of laminin-5 alpha 3 chain Reviewed

    Y Tsubota, H Mizushima, T Hirosaki, S Higashi, H Yasumitsu, K Miyazaki

    BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS   278 ( 3 )   614 - 620   2000.11

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:ACADEMIC PRESS INC  

    Laminin-5 (alpha3 beta3 gamma2) is an important component of epithelial basement membranes. The 190-kDa alpha3 chain undergoes extracellular cleavage within the carboxyl (C) terminus consisting of five globular domains (G1 to G5), producing the mature laminin-5 with the 160-kDa alpha3 chain. To understand the physiological meaning of this processing, we isolated the C-terminal fragments of the alpha3 chain from the conditioned media of two kinds of human cell lines. The amino-terminal sequence of the fragments suggested that the cleavage occurs at Gln(1337)-Asp(1338) in the spacer region between the G3 and G4 domains. The G4-G5 fragment itself did not show significant activity, but it stimulated cell migration in the presence of a low concentration of the mature laminin-5, suggesting its regulatory role in cell migration. (C) 2000 Academic Press.

    DOI: 10.1006/bbrc.2000.3851

    Web of Science

    PubMed

    researchmap

  • Expression of trypsin in human cancer cell lines and cancer tissues and its tight binding to soluble form of Alzheimer amyloid precursor protein in culture Reviewed

    S Miyata, N Koshikawa, S Higashi, Y Miyagi, Y Nagashima, S Yanoma, Y Kato, H Yasumitsu, K Miyazaki

    JOURNAL OF BIOCHEMISTRY   125 ( 6 )   1067 - 1076   1999.6

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:JAPANESE BIOCHEMICAL SOC  

    It was recently found that overexpression of the trypsin gene in tumor cells stimulates their growth in culture and in nude mice. In the present study, expression of trypsin in various human cancer cell lines and tissues was studied by gelatin zymography and immunoblotting before and after enterokinase treatment and by immunohistochemistry. The analyses showed that many stomach, colon, and breast cancer cell lines secreted trypsinogens-1 and/or -2, as well as an unidentified serine proteinase of about 70 kDa, into culture medium. Lung cancer cell lines secreted 18- and 19-kDa unidentified trypsin-like proteins. Stomach cancer cell lines frequently secreted active trypsin, suggesting that they produced an endogenous activator of trypsinogen, most likely enterokinase. Active trypsin formed a complex with a soluble form of Alzheimer amyloid precursor protein (sAPP), a Kunitz-type trypsin inhibitor, which was secreted by all cell lines tested. This indicated that sAPP is a primary inhibitor of secreted trypsin, Immunohistochemical analysis showed that trypsin(ogen) was frequently expressed at high levels in stomach and colon cancers, but scarcely in breast cancers. In the stomach cancers, the trypsin immunoreactivity was higher in the malignant, non-cohesive type than in the cohesive type. These results support the hypothesis that tumor-derived trypsin is involved in the malignant growth of tumor cells, especially stomach cancer cells.

    Web of Science

    PubMed

    researchmap

  • Reactive site-modified tissue inhibitor of metalloproteinases-2 inhibits the cell-mediated activation of progelatinase A Reviewed

    S Higashi, K Miyazaki

    JOURNAL OF BIOLOGICAL CHEMISTRY   274 ( 15 )   10497 - 10504   1999.4

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:AMER SOC BIOCHEMISTRY MOLECULAR BIOLOGY INC  

    Tissue inhibitor of metalloproteinases-a (TIMP-2) is supposed to play a regulatory role in the cell-mediated activation of progelatinase A To investigate the mechanism of the regulation, we prepared and characterized a chemically modified TIMP-2, and examined its effects on the activation of progelatinase A. We found that treatment of TIMP-2 with cyanate ion led to loss of inhibitory activity toward matrilysin or gelatinase A Structural and functional analyses of the modified TIMP-2 showed that carbamylation of the alpha-amino group of the NH2-terminal Cys(1) of TIMP-2 led to complete loss of the inhibitory activity. When the reactive-site modified TIMP-2 was added to culture medium of concanavalin A-stimulated HT1080 cells, the conversion of endogenous progelatinase A to the intermediate form was partially inhibited, whereas that of the intermediate form to the mature one was strongly inhibited. The reactive site-modified TIMP-2 also prevented an accumulation of active gelatinase A on the cell surface. We speculate that occupation of the hemopexin-like domain of gelatinase A by the reactive site-modified TIMP-2 makes it unable for gelatinase A to be retained on the cell surface, thus preventing the autocatalytic conversion of the intermediate fern of gelatinase A to its mature form.

    DOI: 10.1074/jbc.274.15.10497

    Web of Science

    PubMed

    researchmap

  • Role of tissue inhibitor of metalloproteinases-2 (TIMP-2) in regulation of pro-gelatinase A activation catalyzed by membrane-type matrix metalloproteinase-1 (MT1-MRP) in human cancer cells Reviewed

    K Shofuda, K Moriyama, A Nishihashi, S Higashi, H Mizushima, H Yasumitsu, K Miki, H Sato, M Seiki, K Miyazaki

    JOURNAL OF BIOCHEMISTRY   124 ( 2 )   462 - 470   1998.8

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:JAPANESE BIOCHEMICAL SOC  

    To clarify the regulatory mechanism of pro-gelatinase A (proGelA) activation at a cellular level, expression of gelatinase A (GelA), three MT-RIMPs, and TIMP-2 was examined with 11 human cancer cell lines cultured in the presence and absence of stimulants. MT1-MMP mRNA was expressed in 8 cell lines, while MT2-MMP and MT3-MMP mRNAs were expressed in fewer cell lines, The cells with high proGelA activation strongly expressed MTI-MMP mRNA but not MTS-MMP and MT3-MMP mRNAs, suggesting that MT1-MMP was responsible for the proGelA activation in the cancer cells. Treatments with concanavalin A (Con A) and a phorbor ester (TPA) enhanced the MTI-MMP expression, but only Con A stimulated the proGelA activation in many cell lines. In HT1080 fibrosarcoma cells, however, TPA also stimulated the activation. The level of TIMP-8 secreted into culture medium inversely correlated with proGelA activation, For example, 2 squamous cell carcinoma lines (HSC-3 and HSC-4) and 3 HT1080 clones, which efficiently activated proGelA, secreted little TIMP-8 into medium, whereas other cell lines and other HT1080 clones, which hardly activated proGelA, secreted TIMP-8 at high levels, When HSC-3 cells were incubated with TIMP-8 protein or transfected with TIMP-8 cDNA, the proGelA activation was strongly inhibited, These results indicated that. extracellular TIMP-8 was an important negative regulator of proGelA activation. However, the level of extracellular TIMP-8 was not consistent with that of TIMP-2 mRNA in some cell lines. Other experimental results suggested that TIMP-2 might be rapidly metabolized after binding to MT1-MMP, and Con A treatment might stabilize the complex of TIMP-8 and MT1-MMP on cell membranes.

    Web of Science

    PubMed

    researchmap

  • Molecular interaction between factor VII and tissue factor Reviewed

    Shouichi Higashi, Sadaaki Iwanaga

    International Journal of Hematology   67 ( 3 )   229 - 241   1998

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:Springer-Verlag Tokyo  

    Complex formation between serine protease factor VIIa (VIIa) and tissue factor (TF) dramatically enhances the catalytic activity of VIIa, leading to the initiation of extrinsic blood coagulation. The recently determined crystal structure of the complex formed between VIIa and the extracellular domain of TF has revealed in detail the interacting surfaces of the two molecules. Mutational and biochemical studies have further identified regions of VIIa and/or those of TF essential for the high affinity interaction. These studies indicate that the regions extending from the γ-carboxyglutamic acid domain through the first epidermal growth factor-like domain and protease domain of VIIa are involved separately in the interaction with the distinct sites of TF. The interaction of TF with the protease domain of VIIa apparently induces a conformational transition of the VIIa active site. Studies of the chemical modification of VIIa provided a model for the mechanism of TF-mediated acceleration of VIIa activity. In this model, the protease domain of VIIa exists in equilibrium between minor active and dominant zymogen-like inactive conformational states, and preferential binding of TF to the active state leads to a shift in equilibrium into the active state, thereby accelerating VIIa activity. Overall docking of TF with VIIa in the presence of phospholipids further supports a recognition of macromolecular substrates, such as factors IX and X. Therefore, single VIIa- TF interaction contains several distinct mechanisms for enhancing the coagulant activity of VIIa, which may be important for the initiation of coagulation specifically mediated by TF. Studies of the interaction between VIIa and TF provide not only a detailed understanding of the regulated initiation of blood coagulation but also the potential to design novel anticoagulants for the treatment of thrombotic diseases.

    DOI: 10.1016/S0925-5710(98)00018-8

    Scopus

    PubMed

    researchmap

  • Conformation of factor VIIa stabilized by a labile disulfide bond (Cys-310-Cys-329) in the protease domain is essential for interaction with tissue factor Reviewed

    S Higashi, N Matsumoto, S Iwanaga

    JOURNAL OF BIOLOGICAL CHEMISTRY   272 ( 41 )   25724 - 25730   1997.10

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:AMER SOC BIOCHEMISTRY MOLECULAR BIOLOGY INC  

    Unlike other trypsin-type serine proteases, zymogen-to-enzyme transition of conformation of factor VII apparently requires not only conversion of the zymogen to active form factor VIIa (VIIa) but also interaction of VIIa with tissue factor (TF), To determine the region of interaction that correlates with maturation of the Wa active site, we modified intramolecular disulfide bonds in VIIa and examined the interaction of the modified VIIa with soluble TF (sTF). We found that partial reduction and S-carboxamidomethylation of disulfide bonds in VIIa led to losses of amidolytic activity and the binding ability to sTF. To determine the sites of modification that associate with the loss of functions, partially S-carboxamidomethylated Ma was separated on a column of immobilized sTF, Each of the sTF-bound and sTF-unbound fractions and native VIIa was then digested by trypsin, and the digest was analyzed by reversed-phase high performance liquid chromatography, We found that reduction and S-carboxamidomethylation of a disulfide bond between Cys-310 and Cys-329 in the protease domain of VIIa led to loss of the binding ability with sTF, and the modification of a disulfide bond between Cys-340 and Cys-368 of VIIa led to loss of the amidolytic activity. In the three-dimensional structures of trypsinogen and trypsin, the disulfide bonds corresponding to Cys-340-Cys-368 and Cys-310-Cys-329 of Ma are, respectively, in and adjacent to the activation domain, which has flexible conformation in trypsinogen but not in trypsin. Furthermore, the crystal structure of human VIIa TF complex indicates that the region next to Cys-310-Cys-329 is in contact with sTF, We speculate that a regional flexibility, reflected by the labile nature of disulfide bonds of Cys-310-Cys-329 and Cys-340-Cys-368 in the protease domain, contributes to the inability of VIIa to attain the active conformation. Interaction of TF with this flexible region may stabilize the structure in a conformation similar to that of the active state of VIIa.

    DOI: 10.1074/jbc.272.41.25724

    Web of Science

    PubMed

    researchmap

  • Molecular mechanism of tissue factor-mediated acceleration of factor VIIa activity Reviewed

    S Higashi, N Matsumoto, S Iwanaga

    JOURNAL OF BIOLOGICAL CHEMISTRY   271 ( 43 )   26569 - 26574   1996.10

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:AMER SOC BIOCHEMISTRY MOLECULAR BIOLOGY INC  

    The mechanism of the acceleration of the catalytic activity of factor VIIa (VIIa) in the presence of tissue factor (TF) was investigated, To explore the VIIa's site(s) that correlates with TF-mediated acceleration, zymogen VII, VIIa, and active site-modified VIIa were prepared, and dissociation constants (K-d) for their bindings to TF or soluble TF in solution were determined. We found that conversion of zymogen VII to VIIa led to an increase in affinity (Delta Delta G = 4.3-4.4 kJ/mol) for TFs., Dansyl-Glu-Gly-Arg chloromethyl ketone (DNS-EGRck) treatment of VIIa led to a further increase in the affinity (Delta Delta G = 7.3-12 kJ/mol), Neither removal of the Gla domain from VIIa nor truncation of the COOH-terminal membrane and cytoplasmic regions of TF affected the affinity enhanced after DNS-EGRck treatment of VIIa. Treatment of VIIa with (p-amidinophenyl)methanesulfonyl fluoride also enhanced its affinity for soluble TF, whereas treatment with 4-(2-aminoethyl)benzenesulfonyl fluoride, phenylmethylsulfonyl fluoride, or diisopropyl fluorophosphate had a slight effect on the affinity, On the other hand, DNS-EGRck and (p-amidinophenyl)methanesulfonyl fluoride treatments, but not diisopropyl fluorophosphate treatment, of VIIa led to protection of its alpha-amino group of Ile-153 from carbamylation, Protection of the alpha-amino group was consistent with formation of a critical salt bridge between Ile-153 and Asp-343 in the protease domain of VIIa, Therefore, TF may preferentially bind to the active conformational state of VIIa. When one assumes that free VIIa exists in equilibrium between minor active and dominant zymogen-like inactive conformational states, preferential binding of TF to the active state leads to a shift in equilibrium, We speculate that TF traps the active conformational state of VIIa and converts its zymogen-like state into an active state, thereby accelerating the VIIa activity.

    DOI: 10.1074/jbc.271.43.26569

    Web of Science

    PubMed

    researchmap

  • IDENTIFICATION OF REGIONS OF BOVINE FACTOR-VII ESSENTIAL FOR BINDING TO TISSUE FACTOR Reviewed

    S HIGASHI, H NISHIMURA, K AITA, S IWANAGA

    JOURNAL OF BIOLOGICAL CHEMISTRY   269 ( 29 )   18891 - 18898   1994.7

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:AMER SOC BIOCHEMISTRY MOLECULAR BIOLOGY INC  

    Initiation of the extrinsic blood coagulation pathway is mediated by a complex formed between plasma-derived factor VII/VIIa and cell-derived tissue factor (TF). To identify the site(s) of interaction, zymogen VII and VIIa were enzymatically and chemically modified, and their affinities for TF were estimated by measuring their inhibitory effects on the amidolytic activity enhanced after formation of the VIIa-TF complex. We found that the VIIa-light chain (K-i = 3.5 x 10(-7) M) and its fragment consisting of the gamma-carboxyglutamic acid (Gla)-domain and the first epidermal growth factor (EGF)-like domain (Gla-EGF1 peptide; K-i = 1.0 x 10(-6) M) have an affinity for TF. Therefore, one of the binding sites of VII with TF is probably located in the Gla-EGF1 region. On the other hand, a dansyl-Glu-Gly-Arg chloromethyl ketone-treated Gla-domainless VIIa (K-i = 0.7 x 10(-7) M) showed a high affinity for TF: whereas the corresponding Gla-domainless VII similarly treated showed no binding potential, thereby indicating that binding site(s) other than in the Gla-EGF1 region are present in VIIa but not in VII. Acetylation or carbamylation of the alpha-amino group of the NH2-terminal Ile-153 of VIIa resulted in the loss of binding affinity for TF; such modifications convert VIIa into a zymogen-like inactive form by destroying the salt bridge between Ile-153 and Asp-343 in VIIa. The rate of carbamylation of VIIa was reduced in the presence of TF. Protection of the alpha-amino group of Ile-153 from carbamylation after complex formation was consistent with salt bridge formation between Ile-153 and Asp-343 in the VIIa-TF complex. Therefore, binding of TF with the heavy chain of VIIa may induce a conformational change that brings the alpha-amino group of Ile-153 close to the beta-carboxyl group of Asp-343 to make a stable salt bridge.

    Web of Science

    PubMed

    researchmap

  • DEVELOPMENT OF ANTITISSUE FACTOR ANTIBODIES IN PATIENTS AFTER LIVER SURGERY Reviewed

    H TSUDA, S HIGASHI, S IWANAGA, T KUBOTA, T MORITA, K YANAGA

    BLOOD   82 ( 1 )   96 - 102   1993.7

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:W B SAUNDERS CO  

    Web of Science

    PubMed

    researchmap

  • EXPRESSION OF HUMAN SOLUBLE TISSUE FACTOR IN YEAST AND ENZYMATIC-PROPERTIES OF ITS COMPLEX WITH FACTOR-VIIA Reviewed

    Y SHIGEMATSU, T MIYATA, S HIGASHI, T MIKI, JE SADLER, S IWANAGA

    JOURNAL OF BIOLOGICAL CHEMISTRY   267 ( 30 )   21329 - 21337   1992.10

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:AMER SOC BIOCHEMISTRY MOLECULAR BIOLOGY INC  

    The extracellular domain of human tissue factor (TF, amino acids 1-217) was expressed in Saccharomyces cerevisiae, using the inducible yeast acid phosphatase promoter and the yeast invertase signal sequence to direct its secretion into the culture broth. Two active soluble forms sTFalpha (high molecular weight form) and sTFbeta (low molecular weight form) were purified, the yield being approximately 10 and 1 mg/liter of culture supernatant, respectively. sTFalpha had an apparent molecular mass of 150 kDa on SDS-polyacrylamide gel electrophoresis and contained more than 200 residues of mannose/mol of protein. sTFbeta had an apparent molecular mass of 37 kDa and contained 22 residues of mannose/mol of protein. N-Glycosidase F treatments of both rTFs reduced the apparent molecular mass to 35 kDa. The amino-terminal sequences and amino acid compositions of sTFalpha and sTFbeta were consistent with those deduced from the cDNA sequence, thereby indicating that the difference in molecular mass is caused by heterogeneity of oligosaccharide structures. Of these recombinant TFs, sTFbeta enhanced factor VIIa-amidolytic activity 40-fold toward the chromogenic substrate and 147-fold toward the fluorogenic substrate, affecting mainly the k(cat) value. The enhancement was comparable with that of TF purified from human placenta. The TF-mediated enhancement of factor VIIa-amidolytic activity was inhibited by heparin-activated antithrombin III, forming a high molecular weight complex. As treatment of sTFbeta with denaturants such as guanidine hydrochloride or urea led to a biphasic loss of the activity, the extracellular domain of TF probably consists of two discrete domains. This expression system provides a significant amount of the extracellular domain of TF so that studies of interactions with factor VII are feasible.

    Web of Science

    PubMed

    researchmap

  • TISSUE FACTOR POTENTIATES THE FACTOR-VIIA-CATALYZED HYDROLYSIS OF AN ESTER SUBSTRATE Reviewed

    S HIGASHI, H NISHIMURA, S FUJII, K TAKADA, S IWANAGA

    JOURNAL OF BIOLOGICAL CHEMISTRY   267 ( 25 )   17990 - 17996   1992.9

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:AMER SOC BIOCHEMISTRY MOLECULAR BIOLOGY INC  

    We designed a simple and sensitive method to assay the activity of the factor VIIa-tissue factor complex, using as a substrate N(alpha)-benzyloxycarbonyl-L-arginine p-nitrobenzyl ester (Z-Arg-ONb) (Zur, M., and Nemerson, Y. (1978) J. Biol. Chem. 253, 2203-2209). The principle was to measure the amount of p-nitrobenzyl alcohol released during ester hydrolysis using reversed-phase high performance liquid chromatography. Z-Arg-ONb had a broad specificity for plasma serine proteases and factor IXa. Using this method, we examined the effect of tissue factor on the esterase activity of factor VIIa under various conditions. We found that tissue factor greatly potentiates the factor VIIa-catalyzed hydrolysis of Z-Arg-ONb. Phospholipids were not required for the factor VIIa-catalyzed hydrolysis of Z-Arg-ONb, even in the presence of tissue factor. The K(m) value of factor VIIa alone toward the ester substrate was six times higher than that of a VIIa-tissue factor complex (3.2 versus 0.54 mM), whereas the k(cat) value was 12 times lower than that of the VIIa-tissue factor complex (14.3 versus 173 s-1). Thus, tissue factor apparently affects the catalytic site of factor VIIa and enhances hydrolysis of the ester substrate. This enhancing effect of tissue factor disappeared on removal of the gamma-carboxyglutamic acid domain from factor VIIa, whereas the esterase activity in the absence of tissue factor was not affected by this modification. The gamma-carboxyglutamic acid domain is probably required as a potent determinant for interactions with tissue factor, even in the absence of phospholipids in the reaction mixture.

    Web of Science

    PubMed

    researchmap

  • Monoclonal antibody (VII-M31) to bovine factor VII: a specific epitope in the gamma-carboxyglutamic acid domain. Reviewed

    Higashi S, Kawabata S, Nishimura H, Funasaki H, Ohyama S, Miyamoto S, Funatsu A, Iwanaga S

    The Journal of biochemistry   108 ( 4 )   654 - 662   1990.10

     More details

▼display all

MISC

  • MMP-7が誘導するがん細胞凝集機構における膜型セリンプロテアーゼ活性発現機序の解析

    五十畑萌, 東昌市

    日本病態プロテアーゼ学会学術集会プログラム抄録集   28th   2023

  • がん悪性進展に関与するMMPsを分子標的とした高特異性阻害剤の開発

    東昌市

    日本病態プロテアーゼ学会学術集会プログラム抄録集   28th   2023

  • マトリプターゼ前駆体の細胞外輸送には触媒ドメインにおける活性型コンフォメーションの誘導が重要である

    池田小春, 東昌市

    日本病態プロテアーゼ学会学術集会プログラム抄録集   28th   2023

  • 高特異性MMP-9阻害タンパク質によるがん悪性進展阻害効果の解析

    東昌市, 佐藤拓輝

    金沢大学がん進展制御研究所がんの転移・薬剤耐性に関わる先導的共同研究拠点   2022   2023

  • MMP-7が誘導するがん細胞凝集誘導機構におけるエンドサイトーシスの寄与

    冨永明里, 東昌市

    日本生化学会大会(Web)   95th   2022

  • 高特異性MMP-9阻害タンパク質によるがん悪性進展阻害効果の解析

    東昌市, 佐藤拓輝

    金沢大学がん進展制御研究所がんの転移・薬剤耐性に関わる先導的共同研究拠点   2021   2022

  • MMP-7が誘導するがん細胞の細胞凝集に及ぼす化合物スラミンの阻害効果

    杢野弘樹, 東昌市, 常住純

    日本病態プロテアーゼ学会学術集会プログラム抄録集   27th   2022

  • MMP-7によるがん細胞凝集誘導機構におけるエンドサイトーシスおよび細胞内シグナルの寄与

    冨永明里, 東昌市

    日本病態プロテアーゼ学会学術集会プログラム抄録集   27th   2022

  • 化合物スラミンはMMP-7が誘導するがん細胞の細胞凝集を阻害する

    杢野弘樹, 常住淳, 東昌市

    日本生化学会大会(Web)   94th   2021

  • MMP-7が誘導するがん細胞凝集機構におけるマトリプターゼ活性の寄与

    冨永明里, 常住淳, 東昌市

    日本生化学会大会(Web)   94th   2021

  • 新規細胞間接着誘導因子可溶性HAI-1の機能制御によるがん転移抑制法の開発

    東昌市

    金沢大学がん進展制御研究所がんの転移・薬剤耐性に関わる先導的共同研究拠点   2019   2020

  • HAI-1およびMMP-7の機能制御によるがん転移抑制法の開発

    東昌市

    金沢大学がん進展制御研究所がんの転移・薬剤耐性に関わる先導的共同研究拠点   2018   2019

  • HAI-1およびMMP-7の機能制御によるがん転移抑制法の開発

    東昌市

    金沢大学がん進展制御研究所がんの転移・薬剤耐性に関わる先導的研究拠点   2017   2018

  • 「HAI-1分子の部分アミノ酸配列を利用した細胞凝集抑制剤の開発」

    伊藤咲, 石川智弘, 東昌市

    日本病態プロテアーゼ学会学術集会プログラム抄録集   23rd   2018

  • MMP-9選択的インヒビターの分子設計とその腫瘍細胞浸潤抑制効果

    滝澤康, 東昌市

    日本生化学会大会(Web)   91st   2018

  • MMP-7による細胞凝集誘導の機序に基づくがん転移抑制剤の開発

    伊藤咲, 石川智弘, 東昌市

    日本生化学会大会(Web)   91st   2018

  • 「MMP-9選択的インヒビターの腫瘍細胞浸潤抑制効果の検証」

    滝澤康, 東昌市

    日本病態プロテアーゼ学会学術集会プログラム抄録集   23rd   2018

  • 可溶型HAI-1による細胞凝集誘導メカニズムの解析及び細胞凝集阻害剤の開発

    石川智弘, 伊藤咲, 木村弥生, 平野久, 東昌市

    日本がん転移学会学術集会・総会プログラム抄録集   27th   2018

  • HAI-1の切断修飾を介したMMP-7によるがん転移能増強機構の解析

    石川智弘, 木村弥生, 平野久, 東昌市

    日本がん転移学会学術集会・総会プログラム抄録集   26th   2017

  • Acinetobacter baumanniiの好中球を利用した新規細菌メカニズムBacterial immunity taxiについて

    鴨志田剛, 永川茂, 上田たかね, 中野竜一, 中野竜一, 彦坂健児, 彦坂健児, 西田智, 祖母井庸之, 東昌市, 斧康雄

    微生物シンポジウム講演要旨集   29th   2017

  • MMP-7により切断・放出された可溶性HAI-1のがん細胞表層との相互作用の解析

    石川智弘, 木村弥生, 平野久, 東昌市

    日本病態プロテアーゼ学会学術集会プログラム抄録集   22nd   2017

  • 細胞間接着因子としての可溶性HAI-1の機能解析

    石川智弘, 木村弥生, 平野久, 東昌市

    日本生化学会大会(Web)   90th   2017

  • 個々のMMP活性を選択的に制御する分子の創出とがん悪性進展抑制法の開発

    東昌市

    金沢大学がん進展制御研究所がんの転移・薬剤耐性に関わる先導的研究拠点   2016   2017

  • 高特異性MMPインヒビターの創出とがん悪性進展抑制法の開発

    東昌市

    金沢大学がん進展制御研究所がんの転移・薬剤耐性に関わる先導的研究拠点   2015   2016

  • MMP-9に対し高い選択性を持つペプチドインヒビターの分子設計

    佐々木祐太, 東昌市

    日本生化学会大会(Web)   89th   2016

  • 可溶性HAI-1による細胞間接着機構の解析

    石川智弘, 木村弥生, 平野久, 東昌市

    日本病態プロテアーゼ学会学術集会プログラム抄録集   21st   2016

  • 培養がん細胞においてMMP-7がセリンプロテアーゼ様活性を促進する機構の解析

    石川智弘, 木村弥生, 平野久, 東昌市

    日本生化学会大会(Web)   89th   2016

  • MMP-2選択的インヒビターペプチドAPP-IPのアミノ酸改変による新規MMP-2インヒビターの開発

    近藤優希, 東昌市

    日本生化学会大会(Web)   89th   2016

  • MMP-9に対して高い阻害活性と選択性を併せ持つペプチドインヒビターの開発

    佐々木祐太, 東昌市

    日本病態プロテアーゼ学会学術集会プログラム抄録集   21st   2016

  • MMP-2選択的インヒビターペプチドAPP-IPのアミノ酸改変による新規MMP-2インヒビターの開発

    近藤優希, 東昌市

    日本病態プロテアーゼ学会学術集会プログラム抄録集   21st   2016

  • 癌転移を支えるMMP-7に対し,高い阻害選択性を持つインヒビターペプチドの開発

    近藤優希, 東昌市

    日本生化学会大会(Web)   88th   2015

  • Mechanism of MMP-2-selective inhibitory action of β-amyloid precursor protein-derived inhibitor and its application to design specific inhibitors against individual MMPs

    東昌市

    日本血栓止血学会誌   26 ( 6 )   2015

  • MT1-MMPに対して高い阻害活性・選択性を併せ持つペプチドインヒビターの開発

    佐々木祐太, 東昌市

    日本病態プロテアーゼ学会学術集会プログラム抄録集   20th   2015

  • 可溶性HAI-1の細胞間接着活性部位の同定

    石川智弘, 木村弥生, 平野久, 東昌市

    日本病態プロテアーゼ学会学術集会プログラム抄録集   20th   2015

  • 標的MMPsに対する高特異性阻害剤の分子設計とがん悪性進展抑制法の開発

    東昌市

    金沢大学がん進展制御研究所がんの転移・薬剤耐性に関わる先導的研究拠点   2014   2015

  • 癌の浸潤,転移を支えるMMP-7に対し,高い選択性を持つインヒビターペプチドの開発

    近藤優希, 東昌市

    日本病態プロテアーゼ学会学術集会プログラム抄録集   20th   2015

  • MMP-14に対し高い選択性を持つペプチドインヒビターの分子設計

    佐々木祐太, 東昌市

    日本生化学会大会(Web)   88th   2015

  • MMP-7による切断を受けた後,細胞間接着の誘導に関与するHAI-1分子内領域の同定

    石川智弘, 木村弥生, 平野久, 東昌市

    日本生化学会大会(Web)   88th   2015

  • がん細胞表層に結合したMMP-7によって切断され,細胞凝集を惹起するタンパク質の同定

    石川智弘, 木村弥生, 平野久, 東昌市

    日本生化学会大会(Web)   87th   2014

  • ヒト26Sプロテアソームサブユニットのリン酸化修飾状態の解析

    得津奏子, 菅原経継, 井野洋子, 倉田洋一, 木村弥生, 東昌市, 平野久

    生物物理化学(Web)   58 ( 2 )   2014

  • がん細胞浸潤および血管内皮細胞増殖に及ぼす高特異性MMP-2インヒビターの効果

    佐野未奈, 東昌市

    日本血栓止血学会誌   25 ( 2 )   2014

  • MMP-7により切断修飾を受ける細胞表層タンパク質の同定

    石川智弘, 木村弥生, 平野久, 東昌市

    日本病態プロテアーゼ学会学術集会プログラム抄録集   19th   2014

  • 高特異性MMP-2インヒビターのがん細胞浸潤抑制効果と血管新生に及ぼす効果

    佐野未奈, 東昌市

    日本生化学会大会(Web)   87th   2014

  • がん浸潤マーカー・ラミニンγ2鎖の血管内皮下浸潤活性とその機構

    佐藤拓輝, 東昌市

    日本血栓止血学会誌   25 ( 2 )   2014

  • 標的MMPsの活性を特異的に制御する機能性分子の開発

    東昌市

    金沢大学がん進展制御研究所がんの転移・薬剤耐性に関わる先導的研究拠点   2013   2014

  • 細胞外マトリックスタンパク質ラミニン-332による単球の腫瘍関連マクロファージへの分化調節

    鴨志田剛, 鴨志田剛, 小川崇, 小柳潤, 佐藤拓輝, 古宮栄利子, 東昌市, 宮崎香, 斧康雄, 辻勉

    Pharmaco-Hematologyシンポジウム講演要旨集   14th   2013

  • 細胞外マトリックスタンパク質ラミニン-332による単球からのマトリックスメタロプロテイナーゼ-9産生調節

    鴨志田剛, 鴨志田剛, 小川崇, 小柳潤, 佐藤拓輝, 古宮栄利子, 東昌市, 宮崎香, 斧康雄, 辻勉

    日本生化学会大会(Web)   86th   2013

  • 高特異性MMPインヒビターの分子設計による制癌剤の開発

    東昌市, 宮崎香

    金沢大学がん進展制御研究所がんの転移・薬剤耐性に関わる先導的研究拠点   2012   2013

  • 極めて特異性の高いMMP-2インヒビタータンパク質の分子設計とそのin vitroがん細胞浸潤に及ぼす効果の解析

    佐野未奈, 東昌市

    日本病態プロテアーゼ学会学術集会プログラム抄録集   18th   2013

  • 3次元浸潤モデル系を用いたMMP-2特異的インヒビターのがん細胞浸潤抑制効果の解析

    佐野未奈, 小柳潤, 宮崎香, 東昌市

    日本生化学会大会(Web)   86th   2013

  • がん細胞表層に結合したMMP-7を分子標的とするがん転移抑制法の開発

    竹内友香, 宮崎香, 東昌市

    日本生化学会大会(Web)   85th   2012

  • カルシウムイオン結合部位を欠くMMP-7改変体は,がん細胞表層のコレステロール硫酸に強く結合し,MMP-7が誘導する細胞凝集を抑制する

    竹内友香, 宮崎香, 東昌市

    日本病態プロテアーゼ学会学術集会プログラム抄録集   17th   2012

  • Epithelial-mesenchymal transition (EMT) stimulates tumor cells to produce invadopodia-like protrusions in collagen matrix, suppressing cell growth

    Jun Oyanagi, Shouichi Higashi, Kaoru Miyazaki

    CANCER RESEARCH   71   2011.4

     More details

    Language:English   Publishing type:Research paper, summary (international conference)   Publisher:AMER ASSOC CANCER RESEARCH  

    DOI: 10.1158/1538-7445.AM2011-3360

    Web of Science

    researchmap

  • APP由来インヒビターペプチドによるMMP-2選択的阻害機構の解明

    竹内友香, 橋本博, 宮崎香, 佐藤衛, 東昌市

    生化学   2011

  • コレステロール硫酸との相互作用に伴うMMP-7の基質特異性変化と細胞膜近傍タンパク質の選択的分解促進機構

    東昌市, 山本和博, 宮崎香

    生化学   2010

  • IGF結合タンパク質様分子IGFBP-rP1の乳がん組織血管での発現誘導

    伊勢まりい, 古宮栄利子, 古屋桃子, 東昌市, 宮崎香

    生化学   2010

  • IGF結合タンパク質様分子(IGFBP-rP1)の腫瘍増殖抑制における間質との相互作用

    古屋桃子, 古宮栄利子, 東昌市, 宮崎香

    生化学   2010

  • がん浸潤マーカー「ラミニンγ2鎖」と血管内皮細胞の相互作用

    佐藤拓輝, 小柳潤, 藤田幸, 東昌市, 宮崎香

    生化学   2010

  • マトリプターゼ(MT-SP1)によるCTGF(IGFBP-rP2)の切断と腫瘍増殖促進作用

    寺田翔, 迫田明子, 東昌市, 宮崎香

    生化学   2009

  • マトリックスメタロプロテアーゼ-2(MMP-2)に対し,高い特異性を持つインヒビタータンパク質の設計

    廣瀬智一, 東昌市, 宮崎香

    生化学   2009

  • Matriptaseによる腫瘍増殖機構の解析-IGFBPsの切断の意義について-

    迫田明子, 山本和博, 寺田翔, 秋山渚, 東昌市, 宮崎香

    生化学   2008

  • 癌の悪性形質を支えるMMP-2およびMMP-7の細胞表層での活性発現機序とそれを応用した高特異性阻害剤の開発

    東昌市, 山本和博, 宮崎香

    生化学   2008

  • 活性型マトリライシンと癌細胞表層コレステロール硫酸との相互作用:とくに癌細胞表層のタンパク質分解および癌転移能に及ぼす効果について

    東昌市, 山本和博, 久田直輝, 大枝民和, 宮崎香

    日本結合組織学会学術大会抄録集   39th   2007

  • がんの病態における基底膜の意義

    宮崎香, 小川崇, 山本和博, 東昌市

    生化学   2007

  • マトリライシン(MMP-7)による細胞外マトリックスに蓄積されたラミニン332(ラミニン5)の切断

    久田直輝, 久田直輝, 山本和博, 小川崇, 東昌市, 宮崎香

    生化学   2007

  • アミロイド前駆体タンパク質(APP)由来インヒビターによるMMP-2選択的阻害機構の解析

    東昌市, 宮崎香

    生化学   2007

  • 活性型マトリライシン(MMP-7)と細胞表層コレステロール硫酸との分子間相互作用の解析

    大枝民和, 東昌市, 山本和博, 宮崎香

    生化学   2007

  • マトリライシン(MMP-7)による細胞表層アネキシンIIの切断および組織プラスミノーゲン活性化因子の結合性の変化

    常住淳, 森山佳谷乃, 山本和博, 東昌市, 宮崎香

    生化学   2007

  • 活性型マトリライシンは癌細胞表層のコレステロール硫酸に結合し,細胞凝集を誘導する

    山本和博, 東昌市, 来生知, 常住淳, 本家孝一, 宮崎香

    日本癌学会学術総会記事   65th   2006

  • マトリックスメタロプロテアーゼの最新知見-癌増殖・浸潤・転移を中心として-マトリライシン(MMP-7)の大腸癌に対する転移促進機構

    山本和博, 東昌市, 宮崎香

    G.I. Research   14 ( 6 )   2006

  • マトリプターゼによるストロムライシ(MMP-3)の活性化と腫瘍増殖の促進

    宮崎香, 山本和博, 東昌市, 北村均

    日本癌学会学術総会記事   65th   2006

  • 癌細胞表層におけるマトリライシン結合分子の同定

    山本和博, 常住淳, 来生知, 東昌市, 本家孝一, 宮崎香

    日本癌学会学術総会記事   64th   2005

  • マトリプターゼ/MT-SP1による腫よう増殖の促進とangiomodulin/IGFBP-rP1の限定分解

    宮崎香, 佐藤佑一朗, 東昌市, 北村均

    日本癌学会学術総会記事   64th   2005

  • IGF結合蛋白質様因子Angiomodulin/IGFBP-rP1の限定分解による腫よう増殖抑制能の変化

    CHEN Zhaoxin, 佐藤佑一朗, 東昌市, 長嶋洋治, 宮崎香

    日本癌学会学術総会記事   64th   2005

  • 癌細胞表層に存在するマトリライシン結合分子の同定

    山本和博, 常住淳, 来生知, 東昌市, 宮崎香

    日本癌学会総会記事   63rd   2004

  • 膜結合型セリンプロテアーゼmatriptaseのヒトがん細胞における発現と腫よう増殖の促進

    JIN X, 東昌市, 北村均, 宮崎香, 広崎智巳, LIN C Y, DICKSON R B

    日本癌学会総会記事   63rd   2004

  • II.基礎研究の進歩 癌転移に関与する諸因子 その他の因子 マトリライシン(MMP-7)の構造と機能

    山本和博, 東昌市, 宮崎香

    日本臨床   61   2003

  • Identification of a Region of β-Amyloid Precursor Protein Essential for Its Gelatinase A Inhibitory Activity

    HIGASHI Shouichi

    Journal of Biological Chemistry   278   2003

  • Novel Processing of β-Amyloid Precursor Protein Catalyzed by Membrane Type 1 Matrix Metalloproteinase Releases a Fragment Lacking the Inhibitor Domain against Gelatinase A

    HIGASHI Shouichi

    Biochemistry   42   2003

     More details

  • マトリライシンによる細胞凝集促進機構の解析

    常住淳, 山本和博, 来生知, 東昌市, 宮崎香

    日本癌学会総会記事   62nd   2003

  • βアミロイド前駆体蛋白質分子内に存在するMMP-2インヒビター部位の同定

    東昌市, 宮崎香

    日本癌学会総会記事   62nd   2003

  • PC12細胞の神経突起伸展におけるメタロプロテアーゼとNGF受容体p75の作用

    堀内理江, 安田知永, 小阪智伯, 苅谷慶喜, 東昌市, 龍野徹, 宮崎香

    生化学   74 ( 8 )   2002

  • βアミロイド前駆体蛋白質分子内に存在するMMP-2インヒビター部位の同定

    東昌市, 宮崎香

    生化学   74 ( 8 )   2002

  • 癌細胞の転移能発現におけるマトリライシン(MMP-7)の役割

    山本和博, 来生知, 東昌市, 宮崎香

    日本癌学会総会記事   61st   2002

  • Roles of membrane-bound matrilysin (MMP-7) in tumor metastasis

    MIYAZAKI Kaoru, KIOI Mitomu, YAMAMOTO Kazuhiro, HIGASHI Shouichi

    50   28 - 28   2001.10

     More details

    Language:English  

    CiNii Books

    researchmap

  • ラット副腎髄質褐色細胞腫由来PC12細胞の細胞死に及ぼすメルトリンγ(ADAM-9)の効果

    丸茂翠, 宮崎咲江, 和久井世紀, 安光英太郎, 広崎智巳, 東昌市, 瀬原(藤沢)淳子, 宮崎香

    生化学   73 ( 8 )   2001

  • 癌細胞の転移能発現におけるマトリライシンの役割

    来生知, 東昌市, 山本和博, 中谷雅年, 越川直彦, 宮崎香

    日本癌学会総会記事   60th   2001

  • 腫ようの増殖・転移におけるマトリックスメタロプロテアーゼの意義

    来生知, 東昌市, 宮崎香

    月刊血液・腫よう科   42 ( 4 )   2001

  • マトリライシン(MMP-7)の癌細胞への結合と癌転移におよぼす効果

    来生知, 東昌市, 宮崎香

    生化学   73 ( 8 )   2001

  • Isolation and activity of proteolytic fragment of laminin-5 alpha 3 chain

    Y Tsubota, H Mizushima, T Hirosaki, S Higashi, H Yasumitsu, K Miyazaki

    BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS   278 ( 3 )   614 - 620   2000.11

     More details

    Language:English   Publisher:ACADEMIC PRESS INC  

    Laminin-5 (alpha3 beta3 gamma2) is an important component of epithelial basement membranes. The 190-kDa alpha3 chain undergoes extracellular cleavage within the carboxyl (C) terminus consisting of five globular domains (G1 to G5), producing the mature laminin-5 with the 160-kDa alpha3 chain. To understand the physiological meaning of this processing, we isolated the C-terminal fragments of the alpha3 chain from the conditioned media of two kinds of human cell lines. The amino-terminal sequence of the fragments suggested that the cleavage occurs at Gln(1337)-Asp(1338) in the spacer region between the G3 and G4 domains. The G4-G5 fragment itself did not show significant activity, but it stimulated cell migration in the presence of a low concentration of the mature laminin-5, suggesting its regulatory role in cell migration. (C) 2000 Academic Press.

    DOI: 10.1006/bbrc.2000.3851

    Web of Science

    researchmap

  • 血管新生と疾患 9 マトリックスメタロプロテアーゼと血管新生

    宮崎香, 泊泰三, 藤田幸子, 来生知, 東昌市

    Mebio   17 ( 3 )   2000

  • ラミニン-5の細胞運動促進活性におけるα3鎖Gドメインのプロテアーゼ

    坪田芳明, 広崎智己, 水島寛人, 東昌市, 宮崎香

    生化学   72 ( 8 )   2000

  • ラミニン-5の細胞運動促進活性におけるα3鎖Gドメインのプロテアーゼ切断の意義

    坪田芳明, 水島寛人, 東昌市, 宮崎香

    Japanese Journal of Cancer Research   91 ( Supplement (Sept) )   2000

  • マトリライシン(MMP-7)の癌細胞膜表面への結合と転移に及ぼす影響

    来生知, 東昌市, 安光英太郎, 藤田浄秀, 宮崎香

    生化学   72 ( 8 )   2000

  • βアミロイド前駆体蛋白質のプロセッシングに伴うゼラチナーゼA阻害活性の消失

    東昌市, 宮崎香

    生化学   72 ( 8 )   2000

  • マトリライシンの大腸癌細胞への結合と肝転移におよぼすその影響

    来生知, 東昌市, 越川直彦, 安光英太郎, 宮崎香

    日本癌学会総会記事   58th   1999

  • Role of secreted MMPs in tumor cell invasion and metastasis.

    来生知, 東昌市, 宮崎香

    月刊組織培養工学   25 ( 9 )   1999

  • Conformation of Factor VIIa Stabilized by a Labile Disulfide Bond(Cys-310-Cys-329) in the Protease Domain Is Essential for Interaction with Tissue Factor.

    東昌市

    日本血栓止血学会誌   10 ( 1 )   1999

  • Molecular interaction between factor VII and tissue factor

    S Higashi, S Iwanaga

    INTERNATIONAL JOURNAL OF HEMATOLOGY   67 ( 3 )   229 - 241   1998.4

     More details

    Language:English   Publishing type:Book review, literature introduction, etc.   Publisher:ELSEVIER SCI IRELAND LTD  

    Complex formation between serine protease factor VIIa (VIIa) and tissue factor (TF) dramatically enhances the catalytic activity of VIIa, leading to the initiation of extrinsic blood coagulation. The recently determined crystal structure of the complex formed between VIIa and the extracellular domain of TF has revealed in detail the interacting surfaces of the two molecules. Mutational and biochemical studies have further identified regions of VIIa and/or those of TF essential for the high affinity interaction. These studies indicate that the regions extending from the gamma-carboxyglutamic acid domain through the first epidermal growth factor-like domain and protease domain of VIIa are involved separately in the interaction with the distinct sites of TF. The interaction of TF with the protease domain of VIIa apparently induces a conformational transition of the VIIa active site. Studies of the chemical modification of VIIa provided a model for the mechanism of TF-mediated acceleration of VIIa activity. In this model, the protease domain of VIIa exists in equilibrium between minor active and dominant zymogen-like inactive conformational states, and preferential binding of TF to the active state leads to a shift in equilibrium into the active state, thereby accelerating VIIa activity. Overall docking of TF with VIIa in the presence of phospholipids further supports a recognition of macromolecular substrates, such as factors IX and X. Therefore, single VIIa-TF interaction contains several distinct mechanisms for enhancing the coagulant activity of VIIa, which may be important for the initiation of coagulation specifically mediated by TF. Studies of the interaction between VIIa and TF provide not only a detailed understanding of the regulated initiation of blood coagulation but also the potential to design novel anticoagulants for the treatment of thrombotic diseases. (C) 1998 Elsevier Science Ireland Ltd. All rights reserved.

    DOI: 10.1016/S0925-5710(98)00018-8

    Web of Science

    researchmap

  • Hemopathy. State of arts (Ver. 2). Fundamental and clinical research on the pathophysiology. Blood coagulation the VII factor. The structure and functions.

    東昌市

    医学のあゆみ   1998

  • Effect of overexpression of TIMP-2 cDNA on neurite outgrowth in PC12 cells.

    加藤みどり, 安光英太郎, 東昌市, 三沢悟, 宮崎香

    組織培養研究   16 ( 1 )   1997

  • [Matrix metalloproteinases: their structures and functions, with special reference to their roles in tumor invasion and metastasis]. Reviewed

    Miyazaki K, Higashi S

    Seikagaku. The Journal of Japanese Biochemical Society   68 ( 12 )   1791 - 1807   1996.12

     More details

  • Start mechanism of the extrinsic clot system reaction.

    東昌市, 岩永貞昭

    Bio Clinica   11 ( 4 )   1996

  • A new method of identification of the interaction sites between two protein-molecules by chemical modification combined with affinity-chromatography and peptide-mapping.

    東昌市, 松本尚美, 相田和博, 岩永貞昭

    タンパク質構造討論会講演要旨集   45th   1994

  • 外因系血液凝固の開始反応機構: 組織因子によるVIIa因子活性増強作用と分子間相互作用の解析

    東昌市, 相田和博, 岩永貞昭

    タンパク質構造討論会講演要旨集   44th   1993

  • Blood disease - state of arts.Advance on basic and clinical research on pathophysiology.Structure and function of tissue factor.

    相田和博, 東昌市

    医学のあゆみ   ( Oct )   1993

  • Blood disease - state of arts.Advance on basic and clinical research on pathophysiology.Blood coagulation, VII factor.Its structure and function.

    東昌市

    医学のあゆみ   ( Oct )   1993

  • Structure and function of blood coagulation factor VII.

    東昌市, 岩永貞昭

    医学のあゆみ   160 ( 9 )   1992

  • MONOCLONAL-ANTIBODY (VII-M31) TO BOVINE FACTOR-VII - A SPECIFIC EPITOPE IN THE GAMMA-CARBOXYGLUTAMIC ACID DOMAIN

    S HIGASHI, S KAWABATA, H NISHIMURA, H FUNASAKI, S OHYAMA, S MIYAMOTO, A FUNATSU, S IWANAGA

    JOURNAL OF BIOCHEMISTRY   108 ( 4 )   654 - 662   1990.10

     More details

    Language:English   Publisher:JAPAN BIOCHEMICAL SOC  

    Web of Science

    researchmap

▼display all

Industrial property rights

  • β-アミロイド前駆体蛋白質由来マトリックスメタロプロテアーゼ-2インヒビターペプチドと組織メタロプロテアーゼ阻害物質との融合タンパク質

    東 昌市

     More details

    Application no:特願2010-532896  Date applied:2009.10

    Announcement no:WO2010-041617  Date announced:2012.3

    Patent/Registration no:特許第5651474号  Date issued:2014.11

    researchmap

  • TIMP修飾体

    東 昌市

     More details

    Application no:特願特願平11-95142  Date applied:1999.4

    Announcement no:特開2000-290294  Date announced:2000.10

    Patent/Registration no:特許第4342027号  Date issued:2009.10

    researchmap

Awards

  • 平成10年度日本血栓止血学会学術奨励賞

    1998  

     More details

    Country:Japan

    researchmap

  • 平成8年度第13回井上研究奨励賞

    1997  

     More details

    Country:Japan

    researchmap

Research Projects

  • 細胞膜タンパク質HAI-1の切断を介した新規細胞間接着とがん転移促進機構の解明

    Grant number:19K07047  2019.4 - 2024.3

    日本学術振興会  科学研究費助成事業  基盤研究(C)

    東 昌市

      More details

    Authorship:Principal investigator 

    Grant amount:\4290000 ( Direct Cost: \3300000 、 Indirect Cost:\990000 )

    MMP-7は種々のがん組織で高発現しているマトリックスメタロプロテアーゼの一種であり、その発現はがん悪性度と高い相関を示す。私達はMMP-7により、がん転移が促進される機序として以下のことを明らかにしてきた。まず、がん細胞により生合成され、分泌・活性化されたMMP-7が、がん細胞の細胞膜成分として存在するコレステロール硫酸と結合する。次にコレステロール硫酸と結合したMMP-7が近傍の細胞表層タンパク質であるHAI-1(hepatocyte growth factor activator inhibitor type 1)を切断することで、細胞凝集を惹起する。これまで、細胞外へ放出されたHAI-1の細胞外領域が細胞間接着を誘導することが示唆されていた。
    しかし、本研究を進める中で、このHAI-1細胞外領域単独でのがん細胞凝集誘導能は極めて弱く、補助的寄与しか持たないことが判明した。一方、2020年度までの研究で、MMP-7が誘導する細胞凝集メカニズムにセリンプロテアーゼ活性が介在することが示唆された。
    2021年度はMMP-7がエンドサイトーシスにより、がん細胞内に取り込まれた後、細胞内小胞にてセリンプロテアーゼの活性発現に寄与しつつ、細胞内シグナルを惹起する可能性を考え、その検証を行った。
    まず、種々のエンドサイトーシス阻害剤の存在下、がん細胞をMMP-7で処理したところ、ダイナミン阻害剤により細胞凝集が有意に抑制されることを見出した。次に、いくつかのセリンプロテアーゼが細胞内小胞においてプロテアーゼ活性化受容体(PARs)を活性化することから、PARs下流のMAPK経路を調べた結果、MMP-7処理に伴いERKのリン酸化が亢進することが判明した。
    以上より、細胞内に取り込まれたMMP-7はMAPK経路を活性化しつつ、細胞凝集を誘導することが示唆された。

    researchmap

  • Molecular mechanism of MMP-7-enhanced liver metastasis of colon cancer, and its application for liver regenerative medicine

    Grant number:16K12900  2016.4 - 2018.3

    Japan Society for the Promotion of Science  Grants-in-Aid for Scientific Research Grant-in-Aid for Challenging Exploratory Research  Grant-in-Aid for Challenging Exploratory Research

    Higashi Shouichi, KIMURA YAYOI

      More details

    Authorship:Principal investigator  Grant type:Competitive

    Grant amount:\3640000 ( Direct Cost: \2800000 、 Indirect Cost:\840000 )

    (1) We found that hepatocyte growth factor activator inhibitor type 1 (HAI-1), a membrane-bound Kunitz-type serine protease inhibitor, is an MMP-7 substrate; MMP-7 cleaves HAI-1 at the membrane-proximal region, and releases the extracellular region as soluble HAI-1(sHAI-1).
    (2) The released sHAI-1, but not the membrane-bound HAI-1, has an ability to induce cancer cell aggregation, and the HAI-1 region corresponding to amino acids 141-249 is essential for the cell aggregation-inducing activity.
    (3) A cell-surface cholesterol sulfate-independent proteolytic action of MMP-7 is critical for the sHAI-1-mediated induction of cell aggregation, whereas cholesterol sulfate is needed for the MMP-7-catalyzed generation of sHAI-1.

    researchmap

  • Roles of tumor invasion marker laminin gamma2 chain in tumor progression and cancer stem cell functions

    Grant number:26430119  2014.4 - 2017.3

    Japan Society for the Promotion of Science  Grants-in-Aid for Scientific Research Grant-in-Aid for Scientific Research (C)  Grant-in-Aid for Scientific Research (C)

    MIYAZAKI KAORU, MIYAGI Yohei, HIGASHI Shouichi

      More details

    Grant amount:\5200000 ( Direct Cost: \4000000 、 Indirect Cost:\1200000 )

    The present study investigated functions of the tumor invasion marker laminin gamma2 chain (Lmg2) in tumor progression. 1. The most N-terminal, domain V (dV) of Lmg2 promoted vascular permeability and tumor cell invasion into endothelial cell layer. 2. The dV protein stimulated migration of metastatic breast cancer cells by binding the cancer stem cell marker CD44. However, it was not clear whether dV supports the stem cell function. 3. Novel antibodies recognizing dV were prepared. Analyses with these antibodies revealed that small proteolytic fragments containing dV were highly produced in various cultured human cancer cell lines, cancer tissues and sera of cancer patients. Immunohistochemistry showed that one of dV antibodies detects invasive lung cancer cells at high sensitivity. These results suggest that dV fragments are produced in cancer tissues and contribute to tumor invasion. The dV antibodies seem useful for pathological analysis and diagnosis of human cancers.

    researchmap

  • Design of highly selective MMP inhibitors by coupling selective peptide inhibitors with physiological MMP-binding proteins

    Grant number:25440033  2013.4 - 2016.3

    Japan Society for the Promotion of Science  Grants-in-Aid for Scientific Research Grant-in-Aid for Scientific Research (C)  Grant-in-Aid for Scientific Research (C)

    Higashi Shouichi, SATO MAMORU, HASHIMOTO HIROSHI

      More details

    Authorship:Principal investigator  Grant type:Competitive

    Grant amount:\5200000 ( Direct Cost: \4000000 、 Indirect Cost:\1200000 )

    (1) We developed peptide inhibitors with enhanced selectivity toward MMP-7, MMP-9 and MMP-14, respectively, by modifying the amino acid sequence of APP-IP, an MMP-2-selective decapeptide inhibitor. (2) We further designed a highly selective and strong protein inhibitor against MMP-9 and MMP-2 by combining the variant of APP-IP with enhanced MMP-9 selectivity and TIMP-1, which binds specifically to the non-catalytic domains of MMP-9 and MMP-2. (3) We identified hepatocyte growth factor activator inhibitor type I (HAI-1), a type I membrane protein, as a specific substrate of MMP-7 on cancer cell surface, and found that the extracellular domain of HAI-1 released by the MMP-7-catalyzed cleavage acts as a cell-adhesion protein.

    researchmap

  • Disorder of cell adhesion system in tumor progression

    Grant number:23300351  2011.4 - 2014.3

    Japan Society for the Promotion of Science  Grants-in-Aid for Scientific Research Grant-in-Aid for Scientific Research (B)  Grant-in-Aid for Scientific Research (B)

    MIYAZAKI KAORU, HIGASHI Shouichi

      More details

    Authorship:Collaborating Investigator(s) (not designated on Grant-in-Aid)  Grant type:Competitive

    Grant amount:\22100000 ( Direct Cost: \17000000 、 Indirect Cost:\5100000 )

    Non-invasive tumor cells are fixed on basement membranes (BMs) by stably binding to cell adhesion proteins such as laminin-332 (Lm332). However, they start to invade the BMs and then stroma during tumor progression. In this study, we investigated the mechanism of tumor invasion, mainly focusing on the interaction of tumor cells with cell adhesion proteins. Major results are as follows.Laminin gamma2 (Lm-g2), a subunit of Lm332, is often overexpressed in invasive human cancers and hence regarded as a tumor invasion marker. We found that (1) Lm-g2 is induced in association with epithelial-mesenchymal transition of tumor cells, and (2) amino-terminal fragments of Lm-g2 directly interacts with the cancer stem cell marker CD44 in metastatic cancer cells, leading to the stimulation of their migration. These results suggest the cooperation of the two important tumor markers for cancer progression. We also found that Lm-g2 acts on vascular endothelial cells, increasing vascular permeability.

    researchmap

  • Development of highly selective inhibitors against individual MMPs

    Grant number:21570118  2009 - 2011

    Japan Society for the Promotion of Science  Grants-in-Aid for Scientific Research Grant-in-Aid for Scientific Research (C)  Grant-in-Aid for Scientific Research (C)

    HIGASHI Shouichi

      More details

    Authorship:Principal investigator  Grant type:Competitive

    Grant amount:\4940000 ( Direct Cost: \3800000 、 Indirect Cost:\1140000 )

    (1) We determined the crystal structure of the catalytic domain of MMP-2 in complex with decapeptide inhibitor APP-IP, and clarified its mode of the MMP-2-selective inhibition.(2) We designed a highly selective and strong inhibitor against MMP-2 by combining the MMP-2-selective peptide inhibitor APP-IP and physiological MMPs inhibitor TIMP-2.(3) We clarified the mechanism of the cholesterol sulfate-mediated alteration of the substrate preference of MMP-7 ; clarification of the mechanism provides the clue to develop MMP-7-selective inhibitors.

    researchmap

  • Interaction of cancer cells with basement membrane proteins

    Grant number:17014077  2005 - 2010

    Japan Society for the Promotion of Science  Grants-in-Aid for Scientific Research Grant-in-Aid for Scientific Research on Priority Areas  Grant-in-Aid for Scientific Research on Priority Areas

    MIYAZAKI Kaoru, HIGASHI Shouichi, OGAWA Takashi

      More details

    Authorship:Collaborating Investigator(s) (not designated on Grant-in-Aid)  Grant type:Competitive

    Grant amount:\46800000 ( Direct Cost: \46800000 )

    Tumor microenvironments play important roles in tumor progression. The present study demonstrated that the cell adhesion protein laminin-332 (Lm332 ; α3β3γ2) in the basement membrane was converted to a soluble factor capable of strongly promoting tumor cell migration by a proteolytic cleavage of its γ2 chain (Lmγ2). We previously showed that Lmγ2 is solely overexpressed when carcinoma cells invade into stroma. Here, we found that the Lmγ2 expression was induced by TGF-β or some other factors and promoted invasive growth of tumor cells in vivo. Furthermore, we found a novel vascular laminin, laminin-3B11, that was down-regulated in tumor vasculature. The matrix metalloproteinase matrilysin (MMP7) is frequently overexpressed in many types of human cancer and involved in tumor progression and metastasis. Our results suggested that MMP7 might enhance tumor metastasis by binding to cholesterol sulfate of cell membrane and then cleaving cell surface proteins and Lm332. Lmγ2 and MMP7 seem to be good targets for anti-cancer treatments.

    researchmap

  • Application of a new cell-adhesive and cell-migatory molecule, laminin-5B, to regenerative medicine

    Grant number:17300157  2005 - 2006

    Japan Society for the Promotion of Science  Grants-in-Aid for Scientific Research Grant-in-Aid for Scientific Research (B)  Grant-in-Aid for Scientific Research (B)

    MIYAZAKI Kaoru, HIHASHI Shouichi, NAGASHIMA Yoji

      More details

    Grant amount:\14000000 ( Direct Cost: \14000000 )

    Laminins are a family of ECM proteins that are localized mainly in basement membranes and regulate various cellular functions such as adhesion, motility, proliferation, apoptosis and differentiation. More than 15 laminin isoforms consisting of different combinations of the three chains have been identified thus far. Among them, laminin-5 (α3A/B, β3, γ2 ; laminin-332), strongly promotes cellular adhesion and migration. Recently we established efficient expression systems of human recombinant laminin-5A (αA, β3, γ2) and laminin-5B (α3B, β3, γ2). In this study, we investigated the possibility to use these laminins for regenerative medicine. We also attempted to establish a system to produce a novel laminin isoform, laminin-6B (α3B, β1, γ1). Main results are as follows. 1) Mesenchymal stem cell (MSC) is thought to be an important cell type in the regenerative medicine, because it can differentiate into osteoblasts, chondrocytes, neural cells, muscle cells and some others. We found that laminin-5A and laminin-5B efficiently stimulated the growth of human MSCs in culture, while suppressing their chondrogenic differentiation. This suggest the possible use of laminin-5A/B as a growth-stimulatory supplement for the efficient in vitro growth of human MSCs. 2) The presence of laminin-6B is predicted, but it has never been identified. We found that this laminin existed in the basement membranes of micro-blood vessels in various human tissues. We also established an efficient expression system of this novel laminin by introducing cDNAs for human laminin α3B, β1, γ1 chains into HEK293 cells. (α3A, β3, γ2). This laminin may play an important role in angiogenesis and maintenance of vascular structures and functions.

    researchmap

  • 癌細胞表層に存在するMMP-7機能変換分子の同定とその癌転移促進機構の解析

    Grant number:15770070  2003 - 2005

    日本学術振興会  科学研究費助成事業 若手研究(B)  若手研究(B)

    東 昌市

      More details

    Authorship:Principal investigator  Grant type:Competitive

    Grant amount:\3100000 ( Direct Cost: \3100000 )

    これまでに、大腸癌細胞をマトリライシン(MMP-7,MAT)で処理すると、その転移能が著しく上昇することが明らかになっている。また、in vitroではMAT処理により大腸癌細胞の細胞凝集が促進されるが、その凝集が転移能上昇に相関する。一方、種々の癌細胞をMAT処理するとMATが癌細胞表層の特殊ドメインであるラフト・カベオラに結合することから、MATはこの特殊ドメイン内の分子に結合し、かつ、近傍の膜表在性のタンパク質を切断しつつ、細胞凝集を引き起こすことが予想される。昨年度の研究からMAT結合分子の活性はメタノール・クロロホルム混合液で抽出可能であることが判明し、脂質成分であることが示唆された。そこで、癌細胞から抽出した総脂質を陰イオン交換樹脂で分画したところ、吸着画分にのみMAT結合活性が存在することが判明し、この画分に含まれる硫酸化ガラクトシルセラミド、硫酸化ラクトシルセラミド、硫酸化コレステロールおよびカルジオリピンなどの酸性脂質がMAT結合分子の候補として挙げられた。しかしながら、これらの脂質をリボソームに取り込ませた後、MAT結合活性を調べた結果、いずれの脂質もMATと結合したことから、これらの候補を一つに絞り込むことが困難であった。今回、β-シクロデキストリン(β-CD)で癌細胞を処理するとMATの細胞表層への結合が阻害されるとともに、MATが誘導する細胞凝集が顕著に抑制されることを見出した。また、このβ-CDの効果は細胞へのコレステロールの再供給では回復せず、コレステロール硫酸を再供給した場合のみ回復した。β-CDは細胞膜からコレステロールやコレステロール硫酸を抽出するのに対し、硫酸化ガラクトシルセラミドや硫酸化ラクトシルセラミド、カルジオリピンは抽出しないことから、癌細胞表層の主要なMAT結合分子がコレステロール硫酸であることが明らかになった。

    researchmap

  • マトリックスプロテアーゼによるがん細胞の機能制御

    Grant number:13216084  2001 - 2004

    日本学術振興会  科学研究費助成事業 特定領域研究  特定領域研究

    宮崎 香, 東 昌市

      More details

    Authorship:Collaborating Investigator(s) (not designated on Grant-in-Aid)  Grant type:Competitive

    Grant amount:\37600000 ( Direct Cost: \37600000 )

    マトリックスプロテアーゼによる細胞外マトリックス(ECM)分子の機能修飾、およびがん細胞の浸潤・転移への関与について研究を行った。上皮基底膜の主要成分であるラミニン5(LN5:α3/β3/γ2)は細胞の接着と運動を効率的に促進する。BMP-1様酵素によってLN5のα3鎖C末端に存在するG4-5ドメインが除去されることによりLN5が低活性型(前駆体型)から高活性型(成熟型)へと変換されること、また除去されたG4-5ドメインはヘパラン硫酸結合能を介して前駆体LN5をECMに沈着させる役割を果たすことが明らかになった。プロテアーゼの作用にによるこのようなLN5の機能変換は基底膜および間質へのがん細胞の浸潤に寄与すると考えられる。一方、腫瘍血管基底膜に蓄積するIGF結合蛋白質様分子であるアンジオモジュリン/IGFBP-rP1(AGM)が膜結合型セリンプロテアーゼの一種であるマトリプターゼ(Mat)により限定分解されること、これに伴いAGMの細胞接着活性が増大し、IGF/insulin結合活性が消失することなどが判明した。さらに、Matが多様なヒトがん細胞で発現し、可溶性酵素として分泌されることや、Matをヒト胃がん細胞に強制発現させるとヌードマウスでの腫瘍増殖と腫瘍血管新生が著明に促進されることが判明した。この効果はMatによるAGMの分解と関係する可能性が考えられる。以上の結果は細胞外蛋白質に対するプロテアーゼの作用ががん細胞の増殖や浸潤に大きな影響を与えることを示唆する。

    researchmap

  • アミロイド前駆体タンパク質によるMMP活性調節機構の解析

    Grant number:13780485  2001 - 2002

    日本学術振興会  科学研究費助成事業 若手研究(B)  若手研究(B)

    東 昌市

      More details

    Authorship:Principal investigator  Grant type:Competitive

    Grant amount:\2500000 ( Direct Cost: \2500000 )

    アルツハイマー痴呆症は、脳内のβアミロイド蛋白質(βAP)の蓄積が主要な原因であると考えられている。βAPの前駆体蛋白質(APP)は膜結合型蛋白質として生合成された後、大部分がβAP領域内で切断され、その細胞外領域(sAPP,110kDa)が構成的に放出されつつ、βAPを産生しない経路で分解される。しかしながら、APPや放出されるsAPPの生理的役割については殆どわかっていない。私は昨年度の研究から、ゼラチナーゼA (MMP-2)に対するインヒビター領域がAPP770内586-595のアミノ酸配列内に存在することを見い出して来た。さらに、ゼラチナーゼA前駆体の活性化因子である膜型MMP(MT1-MMP)がAPPに作用すると、このインヒビター領域を欠く短い断片」 (80kDa)が細胞外に切り出されることが判明したことから、sAPPtrcを生じる新規プロセッシングには活性化されたゼラチナーゼAの機能を調節するという意義があるのではないかと考えている。本年度は、このAPPの新規プロセッシングを介したゼラチナーゼA活性調節機構を詳細に調べた。sAPPおよびsAPPtrcはともに細胞外マトリックス(ECM)成分であるコラーゲンやラミニンあるいはヘパラン硫酸鎖を持つプロテオグリカンなどと親和性を持つことから、ECM内に存在することが考えられた。実際、HT1080繊維肉腫細胞をMT1-MMP活性を阻害した条件下で培養するとsAPPがECM内に蓄積した。一方、MT1-MMPの阻害剤を除去して培養すると、ECM内のsAPPは速やかにsAPPtrcに置換されることが判明した。したがって、MT1-MMP近傍のECMではインヒビター(sAPP)が除去され、ゼラチナーゼAによる分解を受け易くなることが予想された。こうしたプロテアーゼの活性調節機構は移動しつつある細胞が部位特異的にECM分解を行う上で非常に重要であると考えられる。

    researchmap

  • Structure and Function of Laminin-5, a Cell Adhesion Protein of Basement Membrane

    Grant number:11490030  1999 - 2001

    Japan Society for the Promotion of Science  Grants-in-Aid for Scientific Research Grant-in-Aid for Scientific Research (B)  Grant-in-Aid for Scientific Research (B)

    MIYAZAKI Kaoru, HIGASHI Shouichi, YASUMITSU Hidetaro

      More details

    Grant amount:\5200000 ( Direct Cost: \5200000 )

    The basement protein laminin-5(LN5), a heterotrimer of laminin α3, β3 and γ2 chains, potently promotes cellular adhesion and motility. In this study, we investigated the structure and function relationship of LN5 and its pathological roles in tumor invasion. 1) The α3 chain of LN5 contains five globular domains (G1 to G5) in its carboxyl end. To clarify the function of each G domain, we transfected cDNAs for the full length and five deletion derivatives of the α3 chain into human HT1080 cells, which express only the laminin β3 andγ2 chains, and prepared recombinant LN5s lacking each G domain. The deletion of G5 or G4-5 did not affect the biological activities of LN5, but the LN5 without G3-5 showed little activities of cell adhesion and migration. It was also found that G3 contained two different regions to stimulate cell adhesion and migration. Furthermore, we found that the α3 chain was cleaved between G3 and G4 after secretion and determined the cleavage site. 2) The 105-kDa γ2 chain of LN5 is cleaved at an amino-terminal region to produce the 105-kDa form. We found that MT1-MMP catalyzed this processing, and that this processing increased the cell motility activity of LN5 but decreased its cell adhesion activity. These results demonstrate that the amino-terminal region of γ2 chain has an important effect on the biological activity of LN5. 3) Immunohistochemical analysis showed that the laminin γ2 chain monomer was overexpressed in invading human carcinoma cells. When a cDNA for the amino-terminal regions of the γ2 chain was transfected into a human carcinoma cell line, the invasive potential of this cell line in vivo was greatly enhanced. These results suggest that the γ2 chain itself plays some important roles in tumor invasion.

    researchmap

  • アミロイド前駆体タンパク質の細胞外放出機構の解析およびその生理的意義の解明

    Grant number:09780579  1997 - 1998

    日本学術振興会  科学研究費助成事業 奨励研究(A)  奨励研究(A)

    東 昌市

      More details

    Authorship:Principal investigator  Grant type:Competitive

    Grant amount:\2000000 ( Direct Cost: \2000000 )

    これまでの研究成果から、マトリックスメタロプロテアーゼ(MMPs)のひとつであるゼラチナーゼAが、あるいはMT-MMPが細胞膜表層でアミロイド前駆体蛋白質(APP)に作用すると、通常、細胞外に放出されているAPP細胞外領域(sAPP,110 kDa)より短い断片sAPPtrc(80 kDa)が切り出されることを見い出した。本年度は化学修飾によって、機能を変換したtissue inhibitor of metalloproteinases-2(TIMP-2)を用いてプロセシング酵素を特定するとともに、sAPPtrcの機能を解析し、このプロセシングのもつ生理的意義について考察した。まず、TIMP-2をシアン酸イオンで処理すると、TIMP-2のNH_2-末端Cys-1のα-アミノ基がカルバミル化されるとともにインヒビター活性を失うことを見い出した。TIMP-2のもう一つの機能であるゼラチナーゼA前駆体との相互作用はこの化学修飾による影響を受けなかった。このカルバミル化TIMP-2を培養細胞に添加すると細胞膜上のMT-MMP,TIMP-2およびゼラチナーゼA前駆体からなる3分子複合体の形成を拮抗的に阻害し、結果としてゼラチナーゼA前駆体の活性化を抑制した。したがって、カルバミル化TIMP-2はMT-MMPの触媒活性を阻害することなく、活性型ゼラチナーゼの産生を抑えることが判明した。一方、sAPPtrcの産生はTIMP-2によって阻害されるものの、カルバミル化TIMP-2では抑えられず、sAPPtrcはMT-MMPの触媒により産生されることが示唆された。さらに、sAPPtrcはSAPPのもつゼラチナーゼAインヒビター領域をの活性を失っていることが判明し、MT-MMPが触媒するAPPのプロセシングには以下のような意義があるのではないかと考察した。すなわち、MT-MMPの近傍ではsAPPtrcが産生されているため、活性化されたゼラチナーゼAがマトリックス分解能を持つが、それ以外の部分では、ゼラチナーゼA活性がsAPPによって阻害されるため、APPはマトリックス分解を空間的に調節する分子として働くのではないかと考えた。

    researchmap

  • ゼラチナーゼAの活性発現・制御機構の解析

    Grant number:08780564  1996

    日本学術振興会  科学研究費助成事業 奨励研究(A)  奨励研究(A)

    東 昌市

      More details

    Authorship:Principal investigator  Grant type:Competitive

    Grant amount:\1000000 ( Direct Cost: \1000000 )

    アルツハイマー痴呆症は、脳内に蓄積したβアミロイド蛋白質(βAP)により、神経細胞が変性死することが主要な原因と考えられている。βAPは細胞内でより分子量の大きな細胞膜結合型の前駆体蛋白質(APP)として合成された後、細胞内プロテアーゼにより分解される過程で切り出される。一方、APPが細胞表層において未知のプロテアーゼ(α-secretase)で切断されるとAPPの細胞外領域(sAPP)が切り出され、βAPを産生することはない。私達はこれまでマトリックスメタロプロテアーゼ(MMP)の一つであるゼラチナーゼAがα-secretaseとして働く仮説を提唱してきた。本年度はこの仮説を検証するために、培養細胞を用いてsAPPの分泌に及ぼすMMPインヒビターの影響を調べた。1)まず、ヒト繊維肉腫由来HT1080細胞をコンカナバリンA(ConA)で処理すると細胞培養液中のゼラチナーゼA前駆体が活性化されるとともに、sAPP(120kDa)より分子量の低いAPPの分解断片(80〜90kDa)が生じることを見い出した。2)このAPP分離断片は細胞培養液に天然のMMPインヒビターであるTIMP-2や合成MMP阻害剤を添加すると消失した。3)モノクローナル抗体を用いて解析した結果、APP分解断片はsAPPのCOOH末端領域が欠如した分子であることが判明した。4)溶液中でsAPPにゼラチナーゼAを作用させても分解断片を生じないことから、APPの分解は細胞膜表層で起きていると推定された。5)sAPPの分泌はTIMP-2で抑制されないことから、細胞におけるα-secretase活性はMMP以外の酵素が担っていることが示唆された。以上の結果から、当初の予想とは異なり、MMPがα-secretaseとして働く可能性が低いことが示唆された。一方、1)〜4)の結果はα-secretaseに依存しない新しいAPP細胞外領域の放出機構にゼラチナーゼA、あるいはゼラチナーゼA前駆体の活性化因子であるMT-MMPが関わることを示唆している。このMMPによるAPPの細胞外分解経路ではβAP領域内での切断が起きないことから、βAPの産生・蓄積に関与する可能性があり、興味深い。

    researchmap

  • マトリックスメタロプロテアーゼのアルツハイマー病および動脈硬化における関与

    Grant number:08278229  1996

    日本学術振興会  科学研究費助成事業 重点領域研究  重点領域研究

    宮崎 香, 東 昌市, 安光 英太郎

      More details

    Grant amount:\2500000 ( Direct Cost: \2500000 )

    アルツハイマー痴呆症は、脳内に約40アミノ酸からなるβアミロイド蛋白質(βAP)が蓄積することが主要な原因と考えられている。βAPは細胞内でより分子量の大きな細胞膜結合型の前駆体蛋白質(APP)として合成された後、プロテアーゼによりプロセシングされる過程で切り出される。私達はこれまでマトリックスメタロプロテアーゼ(MMP)の一つであるゼラチネ-ゼAがAPPの細胞外領域(sAPP)を切り出す"α-secretase"として働き、βAPを産生しない経路でAPPを分解するという仮説を提唱してきた。本年度はこの仮説を検証するために、培養細胞を用いてsAPPの分泌に及ぼすMMPやそのインヒビターの影響を調べた。1)まず、ヒト繊維肉腫由来HT1080細胞をコンカナバリンA(ConA)で処理すると細胞培養液中のゼラチナーゼA前駆体が活性化されるとともに、sAPP(120kDa)より分子量の低いAPPの分解断片(80〜90kDa)が生じることを見い出した。2)このAPP分解断片は細胞倍溶液に天然のMMPインヒビターであるTIMP-2や合成MMP阻害剤を添加すると消失した。3)モノクローナル抗体を用いて解析した結果、APP分解断片はsAPPのCOOH末端領域が欠如した分子であることが判明した。4)溶液中のsAPPにゼラチナーゼAを作用させても分解断片を生じないことから、APPの分解は細胞膜表層で起きていると推定された。5)sAPPの分泌はTIMP-2が過剰量存在しても、抑えられないことから、細胞におけるα-secretase活性はMMP以外の酵素が担っていることが示唆された。
    以上の結果から、当初の予想とは異なり、MPPがα-secretaseとして働く可能性は低いことが示唆された。一方、1)〜4)の結果はα-secretaseに依存しない新しいAPP細胞外領域の放出機構にゼラチナーゼA、あるいはゼラチナーゼA前駆体の活性化因子であるMT-MMPが関わることを示唆している。今回見い出されたMMPによるAPPの細胞外分解経路ではβAP領域内での切断が起きないことから、この経路がβAPの産生に関与する可能性があり、この経路をさらに詳しく調べることはβAP蓄積の機序を解明する上で重要であると考えられる。

    researchmap

  • Molecular Mechanism of Extrinsic Blood Coagulation Pathway

    Grant number:03044113  1991 - 1993

    Japan Society for the Promotion of Science  Grants-in-Aid for Scientific Research Grant-in-Aid for international Scientific Research  Grant-in-Aid for international Scientific Research

    IWANAGA Sadaaki, HIGASHI Shouichi, NAKAGAWA Kazunori, MUTA Tatsushi, KAWABATA Shun-ichiro, SUEISHI Katsuo, ICHINOSE Akitada, FUJIKAWA Kazuo, DAVIE Earl W

      More details

    Authorship:Collaborating Investigator(s) (not designated on Grant-in-Aid)  Grant type:Competitive

    Grant amount:\9000000 ( Direct Cost: \9000000 )

    Initiation of the extrinsic blood coagulation pathway is mediated by a complex formed between plasma-derived factor VII/VIIa and cell-derived tissue factor (TF). To identify the site(s) of interaction, zymogen VII and VIIa were enzymatically and chemically modified, and their affinities with TF were estimated by measuring their inhibitory effects on the amidolytic activity enhanced after formation of the VIIa-TF complex. We found that the VIIa-light chain(Ki=3.5 X 10^<-7>) and its fragment consisting of the Gla-domain and the first epidermal growth factor (EGF)-like domain (Gla-EGF1 peptide ; Ki=1.0 X 10^<-6>) have an affinity with TF, but their binding capacity disappeared, respectively, by conventional chymotryptic cleavage.Therefore, one of the binding sites of VII with TF probably locates in the Gla-EGF1 region. On the other hand, a dansyl-Glu-Gly-Arg chloromethyl ketone-treated Gla-domainless VIIa(Ki=0.7 X 10^<-7>) showed a high affinity with TF, whereas the corresponding Gla-domainless VII similarly treated showed no binding potential, thereby indicating that binding site(s) other than in the Gla-EGF1 region is present in VIIa but not in VII. Acetylation or carbamylation of alpha-amino group of NH_2-terminal Ile-153 of VIIa resulted in the loss of binding affinity with TF ; such modifications convert VIIa into a zymogen like inactive form by destroying the salt bridge between Ile-153 and Asp343 in VIIa. The carbamylation rate of VIIa in the presence of TF was low, as compared with that in the absence of TF. The protection of alpha-amino group of Ile-153 from carbamylation after complex formation seemed to be due to a salt bridge formation between Ile-153 and Asp-343 in VIIa-TF complex. Therefore, it is concluded that the binding of TF with the heavy chain of VIIa induces a specific conformational change that brings alpha-amino group of Ile-153 close to beta-carboxyl group of Asp-343 to make a stable salt bridge. This salt bridge formed only in the presence of TF is essential for the formation of the catalytic triad of VIIa.

    researchmap

▼display all