Network


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

Hotspot


Dive into the research topics where Kyoko Kojima-Aikawa is active.

Publication


Featured researches published by Kyoko Kojima-Aikawa.


Thrombosis and Haemostasis | 2004

Levels of annexin IV and V in the plasma of pregnant and postpartum women

Junko Masuda; Eiji Takayama; Ayano Satoh; Michiru Ida; Tadashi Shinohara; Kyoko Kojima-Aikawa; Fumitaka Ohsuzu; Kuniaki Nakanishi; Koichi Kuroda; Mitsutaka Murakami; Kimihiro Suzuki; Isamu Matsumoto

Annexin (Anx) V is pivotal in the maintenance of pregnancy by preventing the activation of blood coagulation. The homology of the amino acid sequence between Anx IV and Anx V is highest in Anx family proteins. However, little is known about the roles of Anx IV in pregnancy. The aim of this study is to clarify the roles of circulating Anx IV and Anx V in normal pregnancy. Subjects were non-pregnant women (n = 50), 120 pregnant women, and maternal subjects just after delivery (n = 53) or postpartum (n = 67). Anx IV in the plasma of non-pregnant women was at a concentration 20 times that of Anx V. The plasma levels of Anx IV suddenly increase after delivery, but Anx V levels remain low during this period. Anx IV and Anx V exert similar levels of anticoagulant activity. Anx IV protein was expressed on the basal surface of syncytiotrophoblasts; Anx V protein, on the apical surface of syncytiotrophoblasts. These results suggest that Anx IV enters the maternal bloodstream just after delivery and might play a role in preventing disseminated intravascular coagulopathy, and that Anx V helps to prevent clotting in the placenta during pregnancy.


Journal of Neuroscience Research | 2002

CNS myelinogenesis in vitro: Myelin basic protein deficient shiverer oligodendrocytes

Chika Seiwa; Kyoko Kojima-Aikawa; Isamu Matsumoto; Hiroaki Asou

The shiverer mutant mouse is an autosomal recessive mutant characterized by incomplete myelin sheath formation in the central nervous system (CNS). Such mice contain a deletion in the MBP gene, do not produce MBP proteins, and have little or no compact myelin in the CNS. To investigate the myelin sheath formation in shiverer mutant mice resulting from the absence of compact myelin, firstly we developed new methods for generating oligodendrocyte precursor cells (OPCs) from an E17 mouse brain, and examined homozygous shiverer (shi/shi) OPCs with respect to myelinogenesis in vitro. After treatment of shi/shi OPCs in vitro with PDGF or bFGF, proliferation of shi/shi OPCs was enhanced similar to that observed in wild‐type OPCs. The majority of cells from the shiverer mutant mouse, however, remained as A2B5‐immunoreactive early OPCs. To determine which molecular events affect the differentiation of shi/shi OPCs, we determined the signaling pathway that could be responsible for activating myelin sheath‐specific proteins. We found that the developmental schedule of shi/shi OPCs in vitro was accelerated by the addition of cyclic AMP analogs, dibutyryl cAMP (dbcAMP). Treatment of shi/shi OPCs with dbcAMP had significant effect on the differentiation of OPCs that became MAG‐expressing oligodendrocytes. To further determine the possible mechanism involved in the activation of MAG by dbcAMP, we examined the cAMP‐dependent signaling cascades. The activation of JNK was markedly stimulated by treatment with dbcAMP, and the phosphorylation of transcription factor ATF‐2 was also stimulated by dbcAMP. We demonstrated that the MAG‐positive shi/shi oligodendrocytes extend processes around axons and finally covered the axon, this was clearly observed by immunocytochemistry of shi/shi oligodendrocyte‐DRG cocultures. These results suggest that ATF‐2 coupled to specific signal transduction cascades plays an important regulatory role in MAG expression at a specific stage of shi/shi oligodendrocyte differentiation, and OPCs grow to become myelin‐forming cells with numerous cell processes that wraps around an axon to form a thin myelin sheath.


Biochemical and Biophysical Research Communications | 2011

Crystal structures of human secretory proteins ZG16p and ZG16b reveal a Jacalin-related β-prism fold

Mayumi Kanagawa; Tadashi Satoh; Akemi Ikeda; Yukiko Nakano; Hirokazu Yagi; Koichi Kato; Kyoko Kojima-Aikawa; Yoshiki Yamaguchi

ZG16p is a secretory protein that mediates condensation-sorting of pancreatic enzymes to the zymogen granule membrane in pancreatic acinar cells. ZG16p interacts with glycosaminoglycans and the binding is considered to be important for condensation-sorting of pancreatic enzymes. ZG16b/PAUF, a paralog of ZG16p, has recently been found to play a role in gene regulation and cancer metastasis. However, the detailed functions of ZG16p and ZG16b remain to be clarified. Here, in order to obtain insights into structure-function relationships, we conducted crystallographic studies of human ZG16p lectin as well as its paralog, ZG16b, and determined their crystal structures at 1.65 and 2.75 Å resolution, respectively. ZG16p has a Jacalin-related β-prism fold, the first to be reported among mammalian lectins. The putative sugar-binding site of ZG16p is occupied by a glycerol molecule, mimicking the mannose bound to Jacalin-related mannose-binding-type plant lectins such as Banlec. ZG16b also has a β-prism fold, but some amino acid residues of the putative sugar-binding site differ from those of the mannose-type binding site suggesting altered preference. A positively charged patch, which may bind sulfated groups of the glycosaminoglycans, is located around the putative sugar-binding site of ZG16p and ZG16b. Taken together, we suggest that the sugar-binding site and the adjacent basic patch of ZG16p and ZG16b cooperatively form a functional glycosaminoglycan-binding site.


Clinical Cancer Research | 2004

HMMC-1 A Humanized Monoclonal Antibody With Therapeutic Potential Against Müllerian Duct-Related Carcinomas

Shiro Nozawa; Daisuke Aoki; Katsumi Tsukazaki; Nobuyuki Susumu; Motoko Sakayori; Nao Suzuki; Atsushi Suzuki; Rie Wakita; Makio Mukai; Yuko Egami; Kyoko Kojima-Aikawa; Isao Ishida; Frederic Belot; Ole Hindsgaul; Minoru Fukuda; Michiko N. Fukuda

Purpose: The purpose of this research was to generate a human monoclonal antibody specific to gynecological cancers and to evaluate such an antibody as therapy for gynecological cancers. Experimental Design: Transchromosomal KM mice were immunized with the human uterine endometrial cancer cell line SNG-S. Hybridomas were constructed between spleen cells from KM mice and mouse myeloma cells. Reactivity of the antibody was evaluated by immunohistochemistry of pathological specimens of gynecological cancers. Cytotoxicity of HMMC-1 against SNG-S cells was tested by in vitro cytotoxicity assays. The epitope of HMMC-1 was determined by transfection with a panel of glycosyltransferase cDNAs and by inhibition assays with chemically synthesized oligosaccharides. Results: HMMC-1 is a human IgM monoclonal antibody that reacts positively with müllerian duct-related carcinomas with positive rates of 54.6% against uterine endometrial adenocarcinoma, 76.9% against uterine cervical adenocarcinoma, and 75.0% against epithelial ovarian cancer. HMMC-1 does not react with normal endometrium at proliferative or secretory phases, normal uterine cervix, or normal and malignant tissue from other organs, whereas it reacts weakly with the epithelium of the gall bladder and the collecting duct of the kidney. HMMC-1 exhibits antigen-dependent and complement-mediated cytotoxicity. Upon cotransfection with cDNAs encoding two glycosyltransferases required for fucosylated extended core 1 O-glycan, mammalian cells express HMMC-1 antigen. Finally, binding of HMMC-1 to SNG-S cells is inhibited by synthetic Fucα1→2Galβ1→4GlcNAcβ1→3Galβ1→3GalNAcα1-octyl. Conclusions: These results indicate that HMMC-1 specifically recognizes a novel O-glycan structure. The unique specificity and cytotoxicity of HMMC-1 strongly suggest a therapeutic potential of this antibody.


Journal of Biological Chemistry | 2012

Identification of Mono- and Disulfated N-Acetyl-lactosaminyl Oligosaccharide Structures as Epitopes Specifically Recognized by Humanized Monoclonal Antibody HMOCC-1 Raised against Ovarian Cancer

Toshiaki Shibata; Fumiko Matsumura; Ping Wang; ShinYi Yu; Chi Chi Chou; Kay Hooi Khoo; Kazuko Kitayama; Tomoya O. Akama; Kazuhiro Sugihara; Naohiro Kanayama; Kyoko Kojima-Aikawa; Peter H. Seeberger; Minoru Fukuda; Atsushi Suzuki; Daisuke Aoki; Michiko N. Fukuda

Background: We produced a humanized monoclonal antibody, designated HMOCC-1, against cell surface carbohydrates presented by malignant ovarian cancer. Results: Co-transfection experiments predicted HMOCC-1 antigenic oligosaccharides structures, which were then chemically synthesized for testing antibody binding. Conclusion: HMOCC-1 antigen is the glycan structure composed of SO3→3Galβ1→4GlcNAcβ1→3(±SO3→6)Galβ1→4GlcNAcβ1→. Significance: The approach employed in this study will be useful in determining specificity of an undefined monoclonal anti-carbohydrate antibody. A humanized monoclonal antibody raised against human ovarian cancer RMG-I cells and designated as HMOCC-1 (Suzuki, N., Aoki, D., Tamada, Y., Susumu, N., Orikawa, K., Tsukazaki, K., Sakayori, M., Suzuki, A., Fukuchi, T., Mukai, M., Kojima-Aikawa, K., Ishida, I., and Nozawa, S. (2004) Gynecol. Oncol. 95, 290–298) was characterized for its carbohydrate epitope structure. Specifically, a series of co-transfections was performed using mammalian expression vectors encoding specific glycosyltransferases and sulfotransferases. These experiments identified one sulfotransferase, GAL3ST3, and one glycosyltransferase, B3GNT7, as required for HMOCC-1 antigen formation. They also suggested that the sulfotransferase CHST1 regulates the abundance and intensity of HMOCC-1 antigen. When HEK293T cells were co-transfected with GAL3ST3 and B3GNT7 expression vectors, transfected cells weakly expressed HMOCC-1 antigen. When cells were first co-transfected with GAL3ST3 and B3GNT7 and then with CHST1, the resulting cells strongly expressed HMOCC-1 antigen. However, when cells were transfected with a mixture of GAL3ST3 and CHST1 before or after transfection with B3GNT7, the number of antigen-positive cells decreased relative to the number seen with only GAL3ST3 and B3GNT7, suggesting that CHST1 plays a regulatory role in HMOCC-1 antigen formation. Because these results predicted that HMOCC-1 antigens are SO3→3Galβ1→4GlcNAcβ1→3(±SO3→6)Galβ1→4GlcNAc, we chemically synthesized mono- and disulfated and unsulfated oligosaccharides. Immunoassays using these oligosaccharides as inhibitors showed the strongest activity by disulfated tetrasaccharide, weak but positive activity by monosulfated tetrasaccharide at the terminal galactose, and no activity by nonsulfated tetrasaccharides. These results establish the HMOCC-1 epitope, which should serve as a useful reagent to further characterize ovarian cancer.


Journal of Biological Chemistry | 2014

Structural Basis for Multiple Sugar Recognition of Jacalin-related Human ZG16p Lectin

Mayumi Kanagawa; Yan Liu; Shinya Hanashima; Akemi Ikeda; Wengang Chai; Yukiko Nakano; Kyoko Kojima-Aikawa; Ten Feizi; Yoshiki Yamaguchi

Background: ZG16p is a soluble mammalian lectin with a Jacalin-related β-prism-fold. Results: ZG16p binds to short α-mannose-related glycans and glycosaminoglycans via the canonical shallow mannose-binding pocket and an adjacent basic surface area, respectively. Conclusion: ZG16p possesses a unique feature of multiple-ligand binding. Significance: Structural insights of ZG16p ligand binding are crucial for understanding the biological functions of the protein. ZG16p is a soluble mammalian lectin, the first to be described with a Jacalin-related β-prism-fold. ZG16p has been reported to bind both to glycosaminoglycans and mannose. To determine the structural basis of the multiple sugar-binding properties, we conducted glycan microarray analyses of human ZG16p. We observed that ZG16p preferentially binds to α-mannose-terminating short glycans such as Ser/Thr-linked O-mannose, but not to high mannose-type N-glycans. Among sulfated glycosaminoglycan oligomers examined, chondroitin sulfate B and heparin oligosaccharides showed significant binding. Crystallographic studies of human ZG16p lectin in the presence of selected ligands revealed the mechanism of multiple sugar recognition. Manα1–3Man and Glcβ1–3Glc bound in different orientations: the nonreducing end of the former and the reducing end of the latter fitted in the canonical shallow mannose binding pocket. Solution NMR analysis using 15N-labeled ZG16p defined the heparin-binding region, which is on an adjacent flat surface of the protein. On-array competitive binding assays suggest that it is possible for ZG16p to bind simultaneously to both types of ligands. Recognition of a broad spectrum of ligands by ZG16p may account for the multiple functions of this lectin in the formation of zymogen granules via glycosaminoglycan binding, and in the recognition of pathogens in the digestive system through α-mannose-related recognition.


Carbohydrate Research | 2011

Caenorhabditis elegans proteins captured by immobilized Galβ1-4Fuc disaccharide units: assignment of 3 annexins

Tomoharu Takeuchi; Kazusa Nishiyama; Atsushi Yamada; Mayumi Tamura; Hideyo Takahashi; Hideaki Natsugari; Jun-ichi Aikawa; Kyoko Kojima-Aikawa; Yoichiro Arata; Ken-ichi Kasai

Galβ1-4Fuc is a key structural motif in Caenorhabditis elegans glycans and is responsible for interaction with C. elegans galectins. In animals of the clade Protostomia, this unit seems to have important roles in glycan-protein interactions and corresponds to the Galβ1-4GlcNAc unit in vertebrates. Therefore, we prepared an affinity adsorbent having immobilized Galβ1-4Fuc in order to capture carbohydrate-binding proteins of C. elegans, which interact with this disaccharide unit. Adsorbed C. elegans proteins were eluted with ethylenediaminetetraacetic acid (EDTA) and followed by lactose (Galβ1-4Glc), digested with trypsin, and were then subjected to proteomic analysis using LC-MS/MS. Three annexins, namely NEX-1, -2, and -3, were assigned in the EDTA-eluted fraction. Whereas, galectins, namely LEC-1, -2, -4, -6, -9, -10, and DC2.3a, were assigned in the lactose-eluted fraction. The affinity of annexins for Galβ1-4Fuc was further confirmed by adsorption of recombinant NEX-1, -2, and -3 proteins to the Galβ1-4Fuc column in the presence of Ca(2+). Furthermore, frontal affinity chromatography analysis using an immobilized NEX-1 column showed that NEX-1 has an affinity for Galβ1-4Fuc, but no affinity toward Galβ1-3Fuc and Galβ1-4GlcNAc. We would hypothesize that the recognition of the Galβ1-4Fuc disaccharide unit is involved in some biological processes in C. elegans and other species of the Protostomia clade.


ChemBioChem | 2015

Defining the Interaction of Human Soluble Lectin ZG16p and Mycobacterial Phosphatidylinositol Mannosides

Shinya Hanashima; Sebastian Götze; Yan Liu; Akemi Ikeda; Kyoko Kojima-Aikawa; Naoyuki Taniguchi; Daniel Varon Silva; Ten Feizi; Peter H. Seeberger; Yoshiki Yamaguchi

ZG16p is a soluble mammalian lectin that interacts with mannose and heparan sulfate. Here we describe detailed analysis of the interaction of human ZG16p with mycobacterial phosphatidylinositol mannosides (PIMs) by glycan microarray and NMR. Pathogen‐related glycan microarray analysis identified phosphatidylinositol mono‐ and di‐mannosides (PIM1 and PIM2) as novel ligand candidates of ZG16p. Saturation transfer difference (STD) NMR and transferred NOE experiments with chemically synthesized PIM glycans indicate that PIMs preferentially interact with ZG16p by using the mannose residues. The binding site of PIM was identified by chemical‐shift perturbation experiments with uniformly 15N‐labeled ZG16p. NMR results with docking simulations suggest a binding mode of ZG16p and PIM glycan; this will help to elucidate the physiological role of ZG16p.


ChemBioChem | 2012

Universal caging group for the in-cell detection of glutathione transferase applied to 19F NMR and bioluminogenic probes.

Mika Ito; Aya Shibata; Jie Zhang; Michio Hiroshima; Yasushi Sako; Yukiko Nakano; Kyoko Kojima-Aikawa; Bengt Mannervik; Satoshi Shuto; Yoshihiro Ito; Ralf Morgenstern; Hiroshi Abe

Universal Caging Group for the in-Cell Detection of Glutathione Transferase Applied to 19F NMR and Bioluminogenic Probes


Glycobiology | 2018

Lectin ZG16p inhibits proliferation of human colorectal cancer cells via its carbohydrate-binding sites

Akiko Mito; Yukiko Nakano; Takako Saitoh; Sabine S. Gouraud; Yoshiki Yamaguchi; Toshiro Sato; Nobuo Sasaki; Kyoko Kojima-Aikawa

Zymogen granule protein 16 (ZG16p) is a soluble lectin that binds to both mannose and heparin/heparan sulfate. It is highly expressed in the human digestive tract and is secreted into the mucus. In this study, we investigated the effect of ZG16p on the proliferation of human colorectal cancer cells. Overexpression of ZG16p in Caco-2 cells decreased cell growth. Recombinant ZG16p markedly inhibited proliferation of Caco-2, LS174T, HCT116 and HCT15 cells. Caco-2 cell growth was not inhibited by two mutated ZG16p proteins, D151A and M5 (K36A, R37A, R53A, R55A and R79A) lacking mannose- and heparin-binding activities, respectively. Immunofluorescent cell staining revealed that ZG16p-D151A maintained its binding to the Caco-2 cell surface, whereas ZG16p-M5 failed to bind to the cells. These results suggest that ZG16p interacts with the cell surface via basic amino acids substituted in ZG16p-M5 and inhibits Caco-2 cell proliferation via Asp151. In addition, growth of patient-derived colorectal tumor organoids in a 3D intestinal stem cell system was suppressed by ZG16p but not by ZG16p-M5. Taken together, our findings indicate that ZG16p inhibits the growth of colorectal cancer cells via its carbohydrate-binding sites in vitro and ex vivo. In this study, a novel pathway in cancer cell growth regulation through cell surface carbohydrate chains is suggested.

Collaboration


Dive into the Kyoko Kojima-Aikawa's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kimihiro Suzuki

National Defense Medical College

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge