Curriculum Vitaes

Yoshikatsu Kanai

  (金井 好克)

Profile Information

Affiliation
Professor, Research Promotion Headquarters, Center for Society-Academia Collaboration , Fujita Health University
Professor, Premium Research Institute for Human Metaverse Medicine, Osaka University
Degree
MD(Mar, 1984)
Ph.D.(Mar, 1988)

Researcher number
60204533
J-GLOBAL ID
200901003262194571
researchmap Member ID
1000305140

External link

Education

 2

Papers

 464
  • Shinichi Sakamoto, Yukio Naya, Junryo Rii, Kazumi Taguchi, Masaaki Fujimura, Yasuhiro Shigeta, Arthit Chairoungdua, Motoi Nishimura, Ken Wakai, Yasutaka Yamada, Xue Zhao, Yusuke Imamura, Sawako Tajima, Natsumi Sato, Chiaki Hosaka, Mizuka Sekine, Takeshi Ueda, Shuzo Hamamoto, Takahiro Yasui, Yoshikatsu Kanai, Koichiro Akakura, Yuzuru Ikehara, Naohiko Anzai, Tomohiko Ichikawa
    Scientific Reports, 15(1), Sep 26, 2025  Peer-reviewed
  • Anzu Nishiwaki, Hiroki Okanishi, Yoshikatsu Kanai, Ryoji Masui
    FEBS Open Bio, Sep 15, 2025  Peer-reviewed
    In Thermus thermophilus, an aerobic Gram‐negative eubacterium used as a model organism, more than half of the phosphorylation sites identified by proteomic analysis are located near the ligand‐binding site, including the active site, of the enzyme in the three‐dimensional structure. We investigated the effect of these phosphorylation events on the activity of six enzymes (three nucleoside monophosphate kinases, isocitrate kinase, malate dehydrogenase and inorganic pyrophosphatase) by introducing phosphomimetic mutations, Glu, into the phosphorylation sites. All phosphomimetic mutants showed severely reduced activity compared with the wild‐type, particularly in the turnover number. The proteins analyzed in this study belong to different families and have various functions. This suggests that there is a widespread mechanism by which phosphorylation of amino acid residues near the active site reduces enzyme activity independent of the protein family and function.
  • Yongchan Lee, Chunhuan Jin, Ryuichi Ohgaki, Minhui Xu, Satoshi Ogasawara, Rangana Warshamanage, Keitaro Yamashita, Garib Murshudov, Osamu Nureki, Takeshi Murata, Yoshikatsu Kanai
    Nature Communications, 16(1), Feb 14, 2025  Peer-reviewedLast authorCorresponding author
    Abstract LAT1 (SLC7A5) transports large neutral amino acids and plays pivotal roles in cancer proliferation, immune response and drug delivery. Despite recent advances in structural understanding of LAT1, how it discriminates substrates and inhibitors including the clinically relevant drugs remains elusive. Here we report six structures of LAT1 across three conformations with bound ligands, elucidating its substrate transport and inhibitory mechanisms. JPH203 (also known as nanvuranlat or KYT-0353), an anticancer drug in clinical trials, traps LAT1 in an outward-facing state with a U-shaped conformer, with its amino-phenylbenzoxazol moiety pushing against transmembrane helix 3 (TM3) and bending TM10. Physiological substrates like ʟ-Phe lack such effects, whereas melphalan poses steric hindrance, explaining its inhibitory activity. The “classical” system L inhibitor BCH induces an occluded state critical for transport, confirming its substrate-like behavior. These findings provide a structural basis for substrate recognition and inhibition of LAT1, guiding future drug design.
  • Yunlong Sui, Namiko Hoshi, Norihiro Okamoto, Yuta Inoue, Takumi Funatsu, Yuna Ku, Makoto Ooi, Daisuke Watanabe, Haruka Miyazaki, Misaki Agawa, Hirotaka Nakamura, Ryuichi Ohgaki, Yoshikatsu Kanai, Hui Yang, Yuzo Kodama
    Biochemical and Biophysical Research Communications, 151446-151446, Feb, 2025  Peer-reviewed
  • Chunhuan Jin, Xinyu Zhou, Minhui Xu, Hiroki Okanishi, Ryuichi Ohgaki, Yoshikatsu Kanai
    Scientific Reports, 15(1), Jan 23, 2025  Peer-reviewedLast authorCorresponding author
  • Yumiao Liu, Ryuichi Ohgaki, Hiroki Okanishi, Minhui Xu, Yoshikatsu Kanai
    Scientific Reports, 14(1), Nov 18, 2024  Peer-reviewedLast authorCorresponding author
  • Kazuko Kaneda-Nakashima, Yoshifumi Shirakami, Kentaro Hisada, Sifan Feng, Yuichiro Kadonaga, Kazuhiro Ooe, Tadashi Watabe, Yoshiyuki Manabe, Atsushi Shimoyama, Masashi Murakami, Atsushi Toyoshima, Hiromitsu Haba, Yoshikatsu Kanai, Koichi Fukase
    International Journal of Molecular Sciences, 25(22) 12386-12386, Nov 18, 2024  Peer-reviewed
    We investigated nuclear medicine therapeutics targeting the L-type amino acid transporter 1 (LAT1). We previously reported that a nuclear medicine therapeutic drug using astatine 211 (211At), an alpha-emitting nuclide that can be produced in an accelerator and targets LAT1 as a molecular target, is effective. The seed compound was 3-[211At] Astato-α-methyl-L-tyrosine (211At-AAMT-OH-L). We used a unique labeling method. By changing the OH group of phenol to a methyl group, retention was successfully increased. It was also found that the amount of the L-isomer taken up by the D-isomer and L-isomer was clearly higher, and the L-isomer was superior as a therapeutic drug. Compounds in which the methyl group was replaced with an ethyl or propyl group were also examined, but their retention did not increase significantly. In fact, we observed increased non-specific accumulation and dynamics, suggesting that labeling may be off. In addition, 211At-AAMT-O-Me-L, which has a simple structure, was clearly superior in terms of uptake speed for several candidate compounds. As a result, we were able to develop a compound that can be easily labeled, has high specific radioactivity, is stable, and has a strong therapeutic effect.
  • Masatoshi Fukuda, Hiroki Okanishi, Daisuke Ino, Kazuya Ono, Takeru Ota, Eri Wakai, Takashi Sato, Yumi Ohta, Yoshiaki Kikkawa, Hidenori Inohara, Yoshikatsu Kanai, Hiroshi Hibino
    iScience, 27(11) 111214-111214, Nov, 2024  Peer-reviewed
  • Ryo Taguchi, Kyoichi Kaira, Yu Miura, Tetsuya Umesaki, Atsuto Mouri, Hisao Imai, Hiroshi Kagamu, Masanori Yasuda, Yoshikatsu Kanai, Hiroyuki Nitanda, Hironori Ishida, Hirozo Sakaguchi
    Heliyon, 10(17) e37414-e37414, Sep, 2024  Peer-reviewed
  • Ryuichi Ohgaki, Yuma Hirase, Minhui Xu, Hiroki Okanishi, Yoshikatsu Kanai
    Scientific reports, 14(1) 19635-19635, Aug 23, 2024  Peer-reviewedLast authorCorresponding author
    L-type amino acid transporter 1 (LAT1) is upregulated in various cancer types and contributes to disease progression. Previous studies have demonstrated or suggested that hypoxia-inducible factors (HIFs), the key transcription factors in hypoxic responses, control the expression of LAT1 gene in several types of cancer cells. However, this regulatory relationship has not been investigated yet in colorectal cancer (CRC), one of the cancer types in which the increased LAT1 expression holds prognostic significance. In this study, we found that neither LAT1 mRNA nor protein is induced under hypoxic condition (1% O2) in CRC HT-29 cells in vitro, regardless of the prominent HIF-1/2α accumulation and HIFs-dependent upregulation of glucose transporter 1. The hypoxic treatment generally did not increase either the mRNA or protein expression of LAT1 in eight CRC cell lines tested, in contrast to the pronounced upregulation by amino acid restriction. Interestingly, knockdown of von Hippel-Lindau ubiquitin ligase to inhibit the proteasomal degradation of HIFs caused an accumulation of HIF-2α and increased the LAT1 expression in certain CRC cell lines. This study contributes to delineating the molecular mechanisms responsible for the pathological expression of LAT1 in CRC cells, emphasizing the ambiguity of HIFs-dependent transcriptional upregulation of LAT1 across cancer cells.
  • Fredrick J Rosario, Kenneth Barentsen, Theresa L Powell, Johann Urschitz, Thomas L Brown, Yoshikatsu Kanai, Thomas Jansson
    PNAS Nexus, 3(6), May 31, 2024  Peer-reviewed
    Abstract Placental System L amino acid transporter activity is decreased in pregnancies complicated by intrauterine growth restriction (IUGR) and increased in fetal overgrowth. However, it is unknown if changes in the expression/activity of placental Large Neutral Amino Acid Transporter Small Subunit 1 (Slc7a5/LAT1) are mechanistically linked to placental function and fetal growth. We hypothesized that trophoblast-specific Slc7a5 overexpression increases placental transport of essential amino acids, activates the placental mechanistic target of rapamycin (mTOR) signaling, and promotes fetal growth in mice. Using lentiviral transduction of blastocysts with a Slc7a5 transgene, we achieved trophoblast-specific overexpression of Slc7a5 (Slc7a5 OX) with increased fetal (+27%) and placental weights (+10%). Trophoblast-specific Slc7a5 overexpression increased trophoblast plasma membrane (TPM) LAT1 protein abundance and TPM System L transporter (+53%) and System A transporter activity (+ 21%). Slc7a5 overexpression also increased transplacental transport of leucine (+ 85%) but not of the System A tracer, 14C-methylamino isobutyric acid, in vivo. Trophoblast-specific overexpression of Slc7a5 activated placental mTORC1, as assessed by increased (+44%) phosphorylation of S6 ribosomal protein (Ser 235/236), and mTORC2 as indicated by phosphorylation of PKCα-Tyr-657 (+47%) and Akt-Ser 473 (+96%). This is the first demonstration that placental transport of essential amino acids is mechanistically linked to fetal growth. The decreased placental System L activity in human IUGR and the increased placental activity of this transporter in some cases of fetal overgrowth may directly contribute to the development of these pregnancy complications.
  • Masatoshi Fukuda, Hiroki Okanishi, Daisuke Ino, Kazuya Ono, Satoru Kawamura, Eri Wakai, Tsuyoshi Miyoshi, Takashi Sato, Yumi Ohta, Takashi Saito, Takaomi C. Saido, Hidenori Inohara, Yoshikatsu Kanai, Hiroshi Hibino
    PLOS ONE, 19(5) e0303375-e0303375, May 10, 2024  Peer-reviewed
    Hearing loss is a pivotal risk factor for dementia. It has recently emerged that a disruption in the intercommunication between the cochlea and brain is a key process in the initiation and progression of this disease. However, whether the cochlear properties can be influenced by pathological signals associated with dementia remains unclear. In this study, using a mouse model of Alzheimer’s disease (AD), we investigated the impacts of the AD-like amyloid β (Aβ) pathology in the brain on the cochlea. Despite little detectable change in the age-related shift of the hearing threshold, we observed quantitative and qualitative alterations in the protein profile in perilymph, an extracellular fluid that fills the path of sound waves in the cochlea. Our findings highlight the potential contribution of Aβ pathology in the brain to the disturbance of cochlear homeostasis.
  • Yu Ma, Suguru Okuda, Hiroki Okanishi, Minhui Xu, Chunhuan Jin, Hitoshi Endou, Ryuichi Ohgaki, Yoshikatsu Kanai
    Journal of Pharmacological Sciences, 155(1) 14-20, May, 2024  Peer-reviewedLast authorCorresponding author
  • Shinpei Saito, Keisuke Ando, Shinichi Sakamoto, Minhui Xu, Yasutaka Yamada, Junryo Rii, Sanji Kanaoka, Jiaxing Wei, Xue Zhao, Sangjon Pae, Manato Kanesaka, Yusuke Goto, Tomokazu Sazuka, Yusuke Imamura, Yoshie Reien, Norie Hamaguchi-Suzuki, Shota Saito, Yuri Hirayama, Hirofumi Hashimoto, Yoshikatsu Kanai, Tomohiko Ichikawa, Naohiko Anzai
    Cancer science, Apr 24, 2024  Peer-reviewed
    L-type amino acid transporter 1 (LAT1) is specifically expressed in many malignancies, contributes to the transport of essential amino acids, such as leucine, and regulates the mammalian target of rapamycin (mTOR) signaling pathway. We investigated the expression profile and functional role of LAT1 in prostate cancer using JPH203, a specific inhibitor of LAT1. LAT1 was highly expressed in castration-resistant prostate cancer (CRPC) cells, including C4-2 and PC-3 cells, but its expression level was low in castration-sensitive LNCaP cells. JPH203 significantly inhibited [14C] leucine uptake in CRPC cells but had no effect in LNCaP cells. JPH203 inhibited the proliferation, migration, and invasion of CRPC cells but not of LNCaP cells. In C4-2 cells, Cluster of differentiation (CD) 24 was identified by RNA sequencing as a novel downstream target of JPH203. CD24 was downregulated in a JPH203 concentration-dependent manner and suppressed activation of the Wnt/β-catenin signaling pathway. Furthermore, an in vivo study showed that JPH203 inhibited the proliferation of C4-2 cells in a castration environment. The results of this study indicate that JPH203 may exert its antitumor effect in CRPC cells via mTOR and CD24.
  • Xinyu Zhou, Ryuichi Ohgaki, Chunhuan Jin, Minhui Xu, Hiroki Okanishi, Hitoshi Endou, Yoshikatsu Kanai
    Journal of Pharmacological Sciences, 154(3) 182-191, Mar, 2024  Peer-reviewedLast authorCorresponding author
  • Sihui Chen, Chunhuan Jin, Ryuichi Ohgaki, Minhui Xu, Hiroki Okanishi, Yoshikatsu Kanai
    Scientific Reports, 14(1), Feb 26, 2024  Peer-reviewedLast authorCorresponding author
    Abstract L-type amino acid transporter 1 (LAT1) is a transmembrane protein responsible for transporting large neutral amino acids. While numerous LAT1-targeted compound delivery for the brain and tumors have been investigated, their LAT1 selectivity often remains ambiguous despite high LAT1 affinity. This study assessed the LAT1 selectivity of phenylalanine (Phe) analogs, focusing on their structure–activity characteristics. We discovered that 2-iodo-l-phenylalanine (2-I-Phe), with an iodine substituent at position 2 in the benzene ring, markedly improves LAT1 affinity and selectivity compared to parent amino acid Phe, albeit at the cost of reduced transport velocity. l-Phenylglycine (Phg), one carbon shorter than Phe, was found to be a substrate for LAT1 with a lower affinity, exhibiting a low level of selectivity for LAT1 equivalent to Phe. Notably, (R)-2-amino-1,2,3,4-tetrahydro-2-naphthoic acid (bicyclic-Phe), with an α-methylene moiety akin to the α-methyl group in α-methyl-l-phenylalanine (α-methyl-Phe), a known LAT1-selective compound, showed similar LAT1 transport maximal velocity to α-methyl-Phe, but with higher LAT1 affinity and selectivity. In vivo studies revealed tumor-specific accumulation of bicyclic-Phe, underscoring the importance of LAT1-selectivity in targeted delivery. These findings emphasize the potential of bicyclic-Phe as a promising LAT1-selective component, providing a basis for the development of LAT1-targeting compounds based on its structural framework.
  • Xingming Liu, Kou Nishikubo, Ryuichi Ohgaki, Hiroki Okanishi, Suguru Okuda, Minhui Xu, Yoshikatsu Kanai
    Journal of Pharmacological Sciences, Feb, 2024  Peer-reviewedLast authorCorresponding author
  • Junryo Rii, Shinichi Sakamoto, Atsushi Mizokami, Minhui Xu, Ayumi Fujimoto, Shinpei Saito, Hidekazu Koike, Takaaki Tamura, Takayuki Arai, Yasutaka Yamada, Yusuke Goto, Tomokazu Sazuka, Yusuke Imamura, Kazuhiro Suzuki, Yoshikatsu Kanai, Naohiko Anzai, Tomohiko Ichikawa
    Cancer science, Jan 7, 2024  Peer-reviewed
    L-type amino acid transporter 1 (LAT1, SLC7A5) is an amino acid transporter expressed in various carcinomas, and it is postulated to play an important role in the proliferation of cancer cells through the uptake of essential amino acids. Cabazitaxel is a widely used anticancer drug for treating castration-resistant prostate cancer (CRPC); however, its effectiveness is lost when cancer cells acquire drug resistance. In this study, we investigated the expression of LAT1 and the effects of a LAT1-specific inhibitor, JPH203, in cabazitaxel-resistant prostate cancer cells. LAT1 was more highly expressed in the cabazitaxel-resistant strains than in the normal strains. Administration of JPH203 inhibited the growth, migration, and invasive ability of cabazitaxel-resistant strains in vitro. Phosphoproteomics using liquid chromatography-mass spectrometry to comprehensively investigate changes in phosphorylation due to JPH203 administration revealed that cell cycle-related pathways were affected by JPH203, and that JPH203 significantly reduced the kinase activity of cyclin-dependent kinases 1 and 2. Moreover, JPH203 inhibited the proliferation of cabazitaxel-resistant cells in vivo. Taken together, the present study results suggest that LAT1 might be a valuable therapeutic target in cabazitaxel-resistant prostate cancer.
  • Weidong Zhang, Ayako Miura, Md Moin Abu Saleh, Koichiro Shimizu, Yuichiro Mita, Ryota Tanida, Satoshi Hirako, Seiji Shioda, Valery Gmyr, Julie Kerr-Conte, Francois Pattou, Chunhuan Jin, Yoshikatsu Kanai, Kazuki Sasaki, Naoto Minamino, Hideyuki Sakoda, Masamitsu Nakazato
    Nature communications, 14(1) 8158-8158, Dec 9, 2023  Peer-reviewed
    Insulin secretion from pancreatic β cells is regulated by multiple stimuli, including nutrients, hormones, neuronal inputs, and local signalling. Amino acids modulate insulin secretion via amino acid transporters expressed on β cells. The granin protein VGF has dual roles in β cells: regulating secretory granule formation and functioning as a multiple peptide precursor. A VGF-derived peptide, neuroendocrine regulatory peptide-4 (NERP-4), increases Ca2+ influx in the pancreata of transgenic mice expressing apoaequorin, a Ca2+-induced bioluminescent protein complex. NERP-4 enhances glucose-stimulated insulin secretion from isolated human and mouse islets and β-cell-derived MIN6-K8 cells. NERP-4 administration reverses the impairment of β-cell maintenance and function in db/db mice by enhancing mitochondrial function and reducing metabolic stress. NERP-4 acts on sodium-coupled neutral amino acid transporter 2 (SNAT2), thereby increasing glutamine, alanine, and proline uptake into β cells and stimulating insulin secretion. SNAT2 deletion and inhibition abolish the protective effects of NERP-4 on β-cell maintenance. These findings demonstrate a novel autocrine mechanism of β-cell maintenance and function that is mediated by the peptide-amino acid transporter axis.
  • Stephen P. H. Alexander, Doriano Fabbro, Eamonn Kelly, Alistair A. Mathie, John A. Peters, Emma L. Veale, Jane F. Armstrong, Elena Faccenda, Simon D. Harding, Jamie A. Davies, Laura Amarosi, Catriona M. H. Anderson, Philip M. Beart, Stefan Broer, Paul A. Dawson, Gergely Gyimesi, Bruno Hagenbuch, James R. Hammond, Jules C. Hancox, Michal Hershfinkel, Ken‐ichi Inui, Yoshikatsu Kanai, Stephan Kemp, Edmund R. S. Kunji, Gavin Stewart, Sotiria Tavoulari, David T. Thwaites, Tiziano Verri
    British Journal of Pharmacology, 180(S2), Oct, 2023  Peer-reviewed
    The Concise Guide to PHARMACOLOGY 2023/24 is the sixth in this series of biennial publications. The Concise Guide provides concise overviews, mostly in tabular format, of the key properties of approximately 1800 drug targets, and over 6000 interactions with about 3900 ligands. There is an emphasis on selective pharmacology (where available), plus links to the open access knowledgebase source of drug targets and their ligands (https://www.guidetopharmacology.org/), which provides more detailed views of target and ligand properties. Although the Concise Guide constitutes almost 500 pages, the material presented is substantially reduced compared to information and links presented on the website. It provides a permanent, citable, point‐in‐time record that will survive database updates. The full contents of this section can be found at http://onlinelibrary.wiley.com/doi/10.1111/bph.16182. Transporters are one of the six major pharmacological targets into which the Guide is divided, with the others being: G protein‐coupled receptors, ion channels, nuclear hormone receptors, catalytic receptors and enzymes. These are presented with nomenclature guidance and summary information on the best available pharmacological tools, alongside key references and suggestions for further reading. The landscape format of the Concise Guide is designed to facilitate comparison of related targets from material contemporary to mid‐2023, and supersedes data presented in the 2021/22, 2019/20, 2017/18, 2015/16 and 2013/14 Concise Guides and previous Guides to Receptors and Channels. It is produced in close conjunction with the Nomenclature and Standards Committee of the International Union of Basic and Clinical Pharmacology (NC‐IUPHAR), therefore, providing official IUPHAR classification and nomenclature for human drug targets, where appropriate.
  • Tadashi Watabe, Naoko Ose, Sadahiro Naka, Eriko Fukui, Toru Kimura, Takashi Kanou, Soichiro Funaki, Hidetaka Sasaki, Takashi Kamiya, Kenta Kurimoto, Kayako Isohashi, Mitsuaki Tatsumi, Eku Shimosegawa, Hiroki Kato, Ryuichi Ohgaki, Yoshikatsu Kanai, Yasushi Shintani
    Clinical nuclear medicine, 48(10) 853-860, Oct 1, 2023  Peer-reviewed
    PURPOSE OF THE REPORT: L-type amino acid transporter-1 (LAT1) is a tumor-specific transporter expressed in various tumor types, with minimal expression in normal organs. We previously demonstrated 18F-fluoro-borono-phenylalanine (18F-FBPA) as a selective PET probe for LAT1 in a preclinical study. Herein, we evaluated LAT1 expression in preoperative patients with lung or mediastinal tumors using 18F-FBPA PET and immunofluorescence staining. PATIENTS AND METHODS: The study population included patients with histopathological diagnosis (n = 55): primary lung cancers (n = 21), lung metastases (n = 6), mediastinal tumors (n = 15), and benign lesion (n = 13). PET scanning was performed 1 hour after the injection of 18F-FBPA (232 ± 32 MBq). Immunofluorescence staining was performed on the resected tumor sections using LAT1 antibody. LAT1 staining was graded on a 4-grade scale and compared with the SUVmax on 18F-FBPA PET. RESULTS: A positive correlation was observed between the SUVmax of 18F-FBPA PET and LAT1 expression by immunofluorescence staining (r = 0.611, P < 0.001). The SUVmax of 18F-FBPA was 3.92 ± 1.46 in grade 3, 3.21 ± 1.82 in grade 2, 2.33 ± 0.93 in grade 1, and 1.50 ± 0.39 in grade 0 of LAT1 expression. Although 18F-FBPA PET showed variable uptake in lung cancers and mediastinal tumors, benign lesions showed significantly lower SUVmax than those in malignant lesions (P < 0.01). CONCLUSIONS: Uptake on 18F-FBPA PET reflected the expression level of LAT1 in lung and mediastinal tumors. It was suggested that 18F-FBPA PET can be used for the precise characterization of the tumor in pretreatment evaluation.
  • Takayuki Minoji, Hidekazu Takahashi, Tadashi Watabe, Satoshi Ishikawa, Hiroki Kato, Ryuichi Ohgaki, Yoshikatsu Kanai, Yuki Sekido, Tsuyoshi Hata, Atsushi Hamabe, Takayuki Ogino, Norikatsu Miyoshi, Mamoru Uemura, Hirofumi Yamamoto, Yuichiro Doki, Hidetoshi Eguchi
    Anticancer research, 43(9) 3913-3921, Sep, 2023  Peer-reviewed
    BACKGROUND/AIM: 18F-fluoro-deoxy-glucose positron emission tomography (18F-FDG PET) has become indispensable for staging colorectal cancer but has limitations. Thus, PET with a focus on metabolism other than glucose, mainly amino acid metabolism, has been developed. L-type amino acid transporter 1 (LAT1) is known to be a cancer-specific amino acid transporter and, although 4-Borono-2-(18)F-fluoro-phenylalanine (FBPA) has been reported to be useful as a probe for LAT1, the significance of LAT1 expression in colorectal cancer is ambiguous and implementation of 18F-FBPA-PET in colorectal cancer has not yet been reported. MATERIALS AND METHODS: The aims of this study were to investigate the expression of LAT1 in primary lesions and metastatic lesions of colorectal cancer by immunohistochemical analysis and report the initial experience of performing 18F-FBPA-PET on colorectal cancer patients in clinical practice. RESULTS: There was a significant correlation between LAT1 protein expression in primary tumors and liver metastases. Furthermore LAT-1 expression was positively correlated with recurrence (p=0.033). We performed 18F-FBPA-PET on three rectal cancer patients and detected cancer. CONCLUSION: LAT1 protein is expressed not only in the primary colorectal tumor, but also in liver metastases. 18F-FBPA-PET can be safely performed in patients with colorectal cancer.
  • Zitong Shi, Kazuko Kaneda-Nakashima, Ryuichi Ohgaki, Minhui Xu, Hiroki Okanishi, Hitoshi Endou, Shushi Nagamori, Yoshikatsu Kanai
    Scientific Reports, 13(1) 13943-13943, Aug 25, 2023  Peer-reviewedLast authorCorresponding author
    Metastasis is the leading cause of mortality in cancer patients. L-type amino acid transporter 1 (LAT1, SLC7A5) is a Na+-independent neutral amino acid transporter highly expressed in various cancers to support their growth. Although high LAT1 expression is closely associated with cancer metastasis, its role in this process remains unclear. This study aimed to investigate the effect of LAT1 inhibition on cancer metastasis using B16-F10 melanoma mouse models. Our results demonstrated that nanvuranlat (JPH203), a high-affinity LAT1-selective inhibitor, suppressed B16-F10 cell proliferation, migration, and invasion. Similarly, LAT1 knockdown reduced cell proliferation, migration, and invasion. LAT1 inhibitors and LAT1 knockdown diminished B16-F10 lung metastasis in a lung metastasis model. Furthermore, nanvuranlat and LAT1 knockdown suppressed lung, spleen, and lymph node metastasis in an orthotopic metastasis model. We discovered that the LAT1 inhibitor reduced the cell surface expression of integrin αvβ3. Our findings revealed that the downregulation of the mTOR signaling pathway, induced by LAT1 inhibitors, decreased the expression of integrin αvβ3, contributing to the suppression of metastasis. These results highlight the critical role of LAT1 in cancer metastasis and suggest that LAT1 inhibition may serve as a potential target for anti-metastasis cancer therapy.
  • Kou Nishikubo, Ryuichi Ohgaki, Xingming Liu, Hiroki Okanishi, Minhui Xu, Hitoshi Endou, Yoshikatsu Kanai
    Cancer Cell International, 23(1), Jun 15, 2023  Peer-reviewedLast authorCorresponding author
    Abstract Background Cytotoxic anticancer drugs widely used in cancer chemotherapy have some limitations, such as the development of side effects and drug resistance. Furthermore, monotherapy is often less effective against heterogeneous cancer tissues. Combination therapies of cytotoxic anticancer drugs with molecularly targeted drugs have been pursued to solve such fundamental problems. Nanvuranlat (JPH203 or KYT-0353), an inhibitor for L-type amino acid transporter 1 (LAT1; SLC7A5), has novel mechanisms of action to suppress the cancer cell proliferation and tumor growth by inhibiting the transport of large neutral amino acids into cancer cells. This study investigated the potential of the combined use of nanvuranlat and cytotoxic anticancer drugs. Methods The combination effects of cytotoxic anticancer drugs and nanvuranlat on cell growth were examined by a water-soluble tetrazolium salt assay in two-dimensional cultures of pancreatic and biliary tract cancer cell lines. To elucidate the pharmacological mechanisms underlying the combination of gemcitabine and nanvuranlat, we investigated apoptotic cell death and cell cycle by flow cytometry. The phosphorylation levels of amino acid-related signaling pathways were analyzed by Western blot. Furthermore, growth inhibition was examined in cancer cell spheroids. Results All the tested seven types of cytotoxic anticancer drugs combined with nanvuranlat significantly inhibited the cell growth of pancreatic cancer MIA PaCa-2 cells compared to their single treatment. Among them, the combined effects of gemcitabine and nanvuranlat were relatively high and confirmed in multiple pancreatic and biliary tract cell lines in two-dimensional cultures. The growth inhibitory effects were suggested to be additive but not synergistic under the tested conditions. Gemcitabine generally induced cell cycle arrest at the S phase and apoptotic cell death, while nanvuranlat induced cell cycle arrest at the G0/G1 phase and affected amino acid-related mTORC1 and GAAC signaling pathways. In combination, each anticancer drug basically exerted its own pharmacological activities, although gemcitabine more strongly influenced the cell cycle than nanvuranlat. The combination effects of growth inhibition were also verified in cancer cell spheroids. Conclusions Our study demonstrates the potential of first-in-class LAT1 inhibitor nanvuranlat as a concomitant drug with cytotoxic anticancer drugs, especially gemcitabine, on pancreatic and biliary tract cancers.
  • 梨井 隼菱, 坂本 信一, 溝上 敦, 斎藤 心平, 田村 貴明, 新井 隆之, 佐塚 智和, 今村 有佑, 安西 尚彦, 金井 好克, 市川 智彦
    日本泌尿器科学会総会, 110回 PP61-08, Apr, 2023  
  • 梨井 隼菱, 坂本 信一, 溝上 敦, 斎藤 心平, 田村 貴明, 新井 隆之, 佐塚 智和, 今村 有佑, 安西 尚彦, 金井 好克, 市川 智彦
    日本泌尿器科学会総会, 110回 PP61-08, Apr, 2023  
  • Yunlong Sui, Namiko Hoshi, Ryuichi Ohgaki, Lingling Kong, Ryutaro Yoshida, Norihiro Okamoto, Masato Kinoshita, Haruka Miyazaki, Yuna Ku, Eri Tokunaga, Yuki Ito, Daisuke Watanabe, Makoto Ooi, Masakazu Shinohara, Kengo Sasaki, Yoh Zen, Takenori Kotani, Takashi Matozaki, Zibin Tian, Yoshikatsu Kanai, Yuzo Kodama
    Journal of Gastroenterology, Feb 5, 2023  Peer-reviewed
    Abstract Background Amino acid transporters play an important role in supplying nutrition to cells and are associated with cell proliferation. L-type amino acid transporter 1 (LAT1) is highly expressed in many types of cancers and promotes tumor growth; however, how LAT1 affects tumor development is not fully understood. Methods To investigate the role of LAT1 in intestinal tumorigenesis, mice carrying LAT1 floxed alleles that also expressed Cre recombinase from the promoter of gene encoding Villin were crossed to an ApcMin/+ background (LAT1fl/fl; vil-cre; ApcMin/+), which were subject to analysis; organoids derived from those mice were also analyzed. Results This study showed that LAT1 was constitutively expressed in normal crypt base cells, and its conditional deletion in the intestinal epithelium resulted in fewer Paneth cells. LAT1 deletion reduced tumor size and number in the small intestine of ApcMin/+ mice. Organoids derived from LAT1-deleted ApcMin/+ intestinal crypts displayed fewer spherical organoids with reduced Wnt/β-catenin target gene expression, suggesting a low tumor-initiation capacity. Wnt3 expression was decreased in the absence of LAT1 in the intestinal epithelium, suggesting that loss of Paneth cells due to LAT1 deficiency reduced the risk of tumor initiation by decreasing Wnt3 production. Conclusions LAT1 affects intestinal tumor development in a cell-extrinsic manner through reduced Wnt3 expression in Paneth cells. Our findings may partly explain how nutrient availability can affect the risk of tumor development in the intestines.
  • Sangjon Pae, Shinichi Sakamoto, Xue Zhao, Shinpei Saito, Takaaki Tamura, Yusuke Imamura, Tomokazu Sazuka, Yoshie Reien, Yuri Hirayama, Hirofumi Hashimoto, Yoshikatsu Kanai, Tomohiko Ichikawa, Naohiko Anzai
    Journal of pharmacological sciences, 150(4) 251-258, Dec, 2022  Peer-reviewed
    Amino acid transporters are responsible for the uptake of amino acids, critical for cell proliferation. L-type amino acid transporters play a major role in the uptake of essential amino acids. L-type amino acid transporter 1 (LAT1) exerts its functional properties by forming a dimer with 4F2hc. Utilizing this cancer-specificity, research on diagnostic imaging and therapeutic agents for malignant tumors targeting LAT1 progresses in various fields. In hormone-sensitive prostate cancer, the up-regulation of L-type amino acid transporter 3 (LAT3) through the androgen receptor (AR) has been identified. On the other hand, in castration-resistant prostate cancer, the negative regulation of LAT1 through AR has been determined. Furthermore, 4F2hc: a binding partner of LAT1, was identified as the specific downstream target of Androgen Receptor Splice Variant 7: AR-V7. LAT1 has been suggested to contribute to acquiring castration resistance in prostate cancer, making LAT1 a completely different therapeutic target from anti-androgens and taxanes. Increased expression of LAT1 has also been found in renal and bladder cancers, suggesting a contribution to acquiring malignancy and progression. In Japan, clinical trials of LAT1 inhibitors for solid tumors are in progress, and clinical applications are now underway. This article will summarize the relationship between LAT1 and urological malignancies.
  • Hiroki Okanishi, Ryuichi Ohgaki, Minhui Xu, Hitoshi Endou, Yoshikatsu Kanai
    Cancer & Metabolism, 10(1) 10:18, Nov 10, 2022  Peer-reviewedLast authorCorresponding author
    Abstract Background Cancer-upregulated l-type amino acid transporter 1 (LAT1; SLC7A5) supplies essential amino acids to cancer cells. LAT1 substrates are not only needed for cancer rapid growth, but involved in cellular signaling. LAT1 has been proposed as a potential target for cancer treatment—its inhibitor, JPH203, is currently in clinical trials and targets biliary tract cancer (BTC). Here, we revealed to what extent LAT1 inhibitor affects intracellular amino acid content and what kind of cellular signals are directly triggered by LAT1 inhibition. Methods Liquid chromatography assay combined with o-phthalaldehyde- and 9-fluorenyl-methylchloroformate-based derivatization revealed changes in intracellular amino acid levels induced by LAT1 inhibition with JPH203 treatment in three BTC cell lines. Tandem mass tag-based quantitative phosphoproteomics characterized the effect of JPH203 treatment on BTC cells, and suggested key regulators in LAT1-inhibited cells. We further studied one of the key regulators, CK2 protein kinase, by using Western blot, enzymatic activity assay, and co-immunoprecipitation. We evaluated anticancer effects of combination of JPH203 with CK2 inhibitor using cell growth and would healing assay. Results JPH203 treatment decreased intracellular levels of LAT1 substrates including essential amino acids of three BTC cell lines, immediately and drastically. We also found levels of some of these amino acids were partially recovered after longer-time treatment. Therefore, we performed phosphoproteomics with short-time JPH203 treatment prior to the cellular compensatory response, and revealed hundreds of differentially phosphorylated sites. Commonly downregulated phosphorylation sites were found on proteins involved in the cell cycle and RNA splicing. Our phosphoproteomics also suggested key regulators immediately responding to LAT1 inhibition. Focusing on one of these regulators, protein kinase CK2, we revealed LAT1 inhibition decreased phosphorylation of CK2 substrate without changing CK2 enzymatic activity. Furthermore, LAT1 inhibition abolished interaction between CK2 and its regulatory protein NOLC1, which suggests regulatory mechanism of CK2 substrate protein specificity controlled by LAT1 inhibition. Moreover, we revealed that the combination of JPH203 with CK2 inhibitor resulted in the enhanced inhibition of proliferation and migration of BTC cells. Conclusion This study provides new perspectives on LAT1-dependent cellular processes and a rationale for therapeutics targeting reprogrammed cancer metabolism.
  • 佐々木 すみれ, 塩崎 雄治, 小池 萌, 谷藤 和也, 宇賀 穂, Wiriyasermkul Pattama, 長谷川 智香, 網塚 憲生, 宮本 賢一, 永森 收志, 金井 好克, 瀬川 博子
    日本生化学会大会プログラム・講演要旨集, 95回 1T08a-08, Nov, 2022  
  • Maya Nakatani, Shun-Ya Nakahara, Kenji Fukui, Momoka Urano, Yuki Fujii, Takeshi Murakawa, Seiki Baba, Takashi Kumasaka, Hiroki Okanishi, Yoshikatsu Kanai, Takato Yano, Ryoji Masui
    J Struct Biol, 214(4) 107904-107904, Oct 11, 2022  Peer-reviewed
  • Kou Nishikubo, Ryuichi Ohgaki, Hiroki Okanishi, Suguru Okuda, Minhui Xu, Hitoshi Endou, Yoshikatsu Kanai
    Journal of Cellular and Molecular Medicine, Sep 7, 2022  Peer-reviewedLast authorCorresponding author
  • 坂本 信一, 齋藤 心平, 安藤 敬介, 梨井 隼菱, 徐 旻恵, 金井 好克, 安西 尚彦, 市川 智彦
    日本癌学会総会記事, 81回 E-3078, Sep, 2022  
  • 兼田 加珠子, 真鍋 良幸, 下山 敦史, 樺山 一哉, 金井 好克, 豊嶋 厚史, 篠原 厚, 深瀬 浩一
    日本癌学会総会記事, 81回 J-1008, Sep, 2022  
  • 兼田 加珠子, 真鍋 良幸, 下山 敦史, 樺山 一哉, 金井 好克, 豊嶋 厚史, 篠原 厚, 深瀬 浩一
    日本癌学会総会記事, 81回 J-1008, Sep, 2022  
  • Naoko Ohtsu, Ryuichi Ohgaki, Chunhuan Jin, Minhui Xu, Hiroki Okanishi, Ryo Takahashi, Akiko Matsui, Wataru Kishimoto, Naoki Ishiguro, Yoshikatsu Kanai
    Journal of Pharmacological Sciences, 150(1) 41-48, Sep, 2022  Peer-reviewedLast authorCorresponding author
  • Takafumi Kato, Tsukasa Kusakizako, Chunhuan Jin, Xinyu Zhou, Ryuichi Ohgaki, LiLi Quan, Minhui Xu, Suguru Okuda, Kan Kobayashi, Keitaro Yamashita, Tomohiro Nishizawa, Yoshikatsu Kanai, Osamu Nureki
    Nature Communications, 13(1), Aug 11, 2022  Peer-reviewedCorresponding author
    Abstract Glutamate is a pivotal excitatory neurotransmitter in mammalian brains, but excessive glutamate causes numerous neural disorders. Almost all extracellular glutamate is retrieved by the glial transporter, Excitatory Amino Acid Transporter 2 (EAAT2), belonging to the SLC1A family. However, in some cancers, EAAT2 expression is enhanced and causes resistance to therapies by metabolic disturbance. Despite its crucial roles, the detailed structural information about EAAT2 has not been available. Here, we report cryo-EM structures of human EAAT2 in substrate-free and selective inhibitor WAY213613-bound states at 3.2 Å and 2.8 Å, respectively. EAAT2 forms a trimer, with each protomer consisting of transport and scaffold domains. Along with a glutamate-binding site, the transport domain possesses a cavity that could be disrupted during the transport cycle. WAY213613 occupies both the glutamate-binding site and cavity of EAAT2 to interfere with its alternating access, where the sensitivity is defined by the inner environment of the cavity. We provide the characterization of the molecular features of EAAT2 and its selective inhibition mechanism that may facilitate structure-based drug design for EAAT2.
  • Sumire Sasaki, Yuji Shiozaki, Ai Hanazaki, Megumi Koike, Kazuya Tanifuji, Minori Uga, Kota Kawahara, Ichiro Kaneko, Yasuharu Kawamoto, Pattama Wiriyasermkul, Tomoka Hasegawa, Norio Amizuka, Ken-ichi Miyamoto, Shushi Nagamori, Yoshikatsu Kanai, Hiroko Segawa
    Scientific Reports, 12(1) 6353-6353, May, 2022  Peer-reviewedCorresponding author
    Abstract Renal type II sodium-dependent inorganic phosphate (Pi) transporters NaPi2a and NaPi2c cooperate with other organs to strictly regulate the plasma Pi concentration. A high Pi load induces expression and secretion of the phosphaturic hormones parathyroid hormone (PTH) and fibroblast growth factor 23 (FGF23) that enhance urinary Pi excretion and prevent the onset of hyperphosphatemia. How FGF23 secretion from bone is increased by a high Pi load and the setpoint of the plasma Pi concentration, however, are unclear. Here, we investigated the role of Transmembrane protein 174 (Tmem174) and observed evidence for gene co-expression networks in NaPi2a and NaPi2c function. Tmem174 is localized in the renal proximal tubules and interacts with NaPi2a, but not NaPi2c. In Tmem174-knockout (KO) mice, the serum FGF23 concentration was markedly increased but increased Pi excretion and hypophosphatemia were not observed. In addition, Tmem174-KO mice exhibit reduced NaPi2a responsiveness to FGF23 and PTH administration. Furthermore, a dietary Pi load causes marked hyperphosphatemia and abnormal NaPi2a regulation in Tmem174-KO mice. Thus, Tmem174 is thought to be associated with FGF23 induction in bones and the regulation of NaPi2a to prevent an increase in the plasma Pi concentration due to a high Pi load and kidney injury.
  • Sasagu Kurozumi, Kyoichi Kaira, Hiroshi Matsumoto, Masafumi Kurosumi, Takehiko Yokobori, Yoshikatsu Kanai, Chikako Sekine, Chikako Honda, Ayaka Katayama, Mio Furuya, Sho Shiino, Takaya Makiguchi, Nigel P Mongan, Emad A Rakha, Tetsunari Oyama, Takaaki Fujii, Ken Shirabe, Jun Horiguchi
    Scientific reports, 12(1) 2742-2742, Feb 17, 2022  Peer-reviewed
    L-type amino acid transporter 1 (LAT1), also referred to as SLC7A5, is believed to regulate tumor metabolism and be associated with tumor proliferation. In invasive breast cancer, we clinicopathologically investigated the utility of LAT1 expression. LAT1 expression was evaluated via immunohistochemistry analyses in 250 breast cancer patients undergoing long-term follow-up. We assessed the relationships between LAT1 expression and patient outcomes and clinicopathological factors. Breast cancer-specific survival stratified by LAT1 expression was assessed. Human epidermal growth factor receptor 2 (HER2)-positive patients with metastasis received trastuzumab therapy. The density of tumor-infiltrating lymphocytes (TILs) was evaluated according to the International Working Group guidelines. In the current study, high LAT1 expression was significantly correlated with estrogen receptor (ER) negativity, progesterone receptor negativity, high histological grade, increased TILs, and programmed death ligand 1 positivity. Among the ER-positive and HER2-negative patients, high LAT1 was an independent indicator of poor outcomes (hazard ratio (HR) = 2.97; 95% confidence interval (CI), 1.16-7.62; p = 0.023). Moreover, high LAT1 expression was an independent poor prognostic factor in luminal B-like breast cancer with aggressive features (HR = 3.39; 95% CI 1.35-8.52; p = 0.0094). In conclusion, high LAT1 expression could be used to identify a subgroup of invasive breast cancer characterized by aggressive behavior and high tumor immunoreaction. Our findings suggest that LAT1 might be a candidate therapeutic target for breast cancer patients, particularly those with luminal B-like type breast cancer.
  • Kana Hasegawa, Shunya Ikeda, Moto Yaga, Kouki Watanabe, Rika Urakawa, Akie Iehara, Mai Iwai, Seishin Hashiguchi, Soyoko Morimoto, Fumihiro Fujiki, Hiroko Nakajima, Jun Nakata, Sumiyuki Nishida, Akihiro Tsuboi, Yoshihiro Oka, Satoshi Yoshihara, Masahiro Manabe, Hiroyoshi Ichihara, Atsuko Mugitani, Yasutaka Aoyama, Takafumi Nakao, Asao Hirose, Masayuki Hino, Shiho Ueda, Katsuto Takenaka, Takashi Masuko, Koichi Akashi, Takahiro Maruno, Susumu Uchiyama, Shinji Takamatsu, Naoki Wada, Eiichi Morii, Shushi Nagamori, Daisuke Motooka, Yoshikatsu Kanai, Yusuke Oji, Tomoyoshi Nakagawa, Noriyuki Kijima, Haruhiko Kishima, Atsuyo Ikeda, Takayuki Ogino, Yasushi Shintani, Tateki Kubo, Emiko Mihara, Kosuke Yusa, Haruo Sugiyama, Junichi Takagi, Eiji Miyoshi, Atsushi Kumanogoh, Naoki Hosen
    Science Translational Medicine, 14(632), Feb 16, 2022  Peer-reviewed
  • Yoshikatsu Kanai
    Pharmacology & Therapeutics, 230 107964-107964, Feb, 2022  Peer-reviewedLead authorCorresponding author
  • Nishikubo Kou, Ohgaki Ryuichi, Okanishi Hiroki, Okuda Suguru, Xu Minhui, Endou Hitoshi, Kanai Yoshikatsu
    Proceedings for Annual Meeting of The Japanese Pharmacological Society, 95 1-SS-50, 2022  Peer-reviewedLast authorCorresponding author
    Cancer cells require more nutrients than normal cells to maintain rapid growth and proliferation. L-type amino acid transporter 1 (LAT1: SLC7A5) is highly upregulated in various cancers, and is regarded as a molecular target for cancer therapy. LAT1 preferentially transports large neutral amino acids (Leu, Ile, Val, Met, Phe, Tyr, Trp, and His) including many essential amino acids in a Na+-independent manner. JPH203, an LAT1-specific high-affinity inhibitor, strongly suppresses cancer cell proliferation and tumor growth. However, the contribution of LAT1 to the amino acid uptake in cancer cells and the effect of JPH203 on cellular protein synthesis have not been established. Here, we revealed that JPH203 drastically suppresses the uptake of large neutral amino acids into pancreatic cancer cell lines, regardless of the presence or absence of Na+. This indicates that LAT1 has the main contribution to cellular amino acid uptake among amino acid transporters including Na+-dependent transporters. Furthermore, we revealed, by polysome profiling analysis, that JPH203 treatment suppresses cellular protein synthesis. These results indicate that LAT1 constitutes the main uptake pathway for large neutral amino acids in cancer cells.  Inhibition of LAT1 efficiently suppresses their uptake and reduces the protein synthesis in cancer cells.
  • Pornparn Kongpracha, Pattama Wiriyasermkul, Noriyoshi Isozumi, Satomi Moriyama, Yoshikatsu Kanai, Shushi Nagamori
    Molecular & Cellular Proteomics, 100206-100206, Jan, 2022  Peer-reviewed
  • Maihulan Maimaiti, Shinichi Sakamoto, Masahiro Sugiura, Manato Kanesaka, Ayumi Fujimoto, Keisuke Matsusaka, Minhui Xu, Keisuke Ando, Shinpei Saito, Ken Wakai, Yusuke Imamura, Keiichi Nakayama, Yoshikatsu Kanai, Atsushi Kaneda, Yuzuru Ikehara, Jun-Ichiro Ikeda, Naohiko Anzai, Tomohiko Ichikawa
    Scientific Reports, 11(1), Dec, 2021  Peer-reviewed
    <title>Abstract</title>The 4F2 cell-surface antigen heavy chain (4F2hc) forms a heterodimeric complex with L-type amino acid transporter 1 (LAT1) and transports large neutral essential amino acids. However, in contrast to the traditional role of LAT1 in various cancers, the role of 4F2hc has largely remained unknown. The role of 4F2hc in prostate cancer was studied. Treatment of C4-2 cells with si4F2hc was found to suppress cellular growth, migratory and invasive abilities, with this effect occurring through the cell cycle, with a significant decrease in S phase and a significant increase in G0/G1 phase, suggesting cell cycle arrest. In addition, it was proven by RNA seq that the key to 4F2hc’s impact on cancer is SKP2. si4F2hc upregulates the protein expression of cyclin-dependent kinase inhibitors (P21cip1, P27kip1) through the downstream target SKP2. Furthermore, the expression of 4F2hc and LAT1 in prostate cancer cells suggests the importance of 4F2hc. Multivariate analysis showed that high 4F2hc expression was an independent prognostic factor for progression-free survival (HR 11.54, <italic>p</italic> = 0.0357). High 4F2hc was related to the clinical tumour stage (<italic>p</italic> = 0.0255) and Gleason score (<italic>p</italic> = 0.0035). Collectively, 4F2hc contributed significantly to prostate cancer (PC) progression. 4F2hc may be a novel marker and therapeutic target in PC.
  • Weijun Liu, Ling Wei, Yoshikatsu Kanai, Xin He
    Evidence-Based Complementary and Alternative Medicine, 2021 1-11, Oct 5, 2021  Peer-reviewed
    Bulbus fritillariae and Radix aconiti praeparata are an incompatible herbal pair in the traditional Chinese medicine theory “eighteen incompatible medicaments,” and they should not be used simultaneously in clinical treatment for safety. This study aimed to investigate the incompatibility mechanism between Bulbus fritillariae and Radix aconiti praeparata based on their interaction with P-glycoprotein (P-gp). The interaction between Bulbus fritillariae and Radix aconiti praeparata during in vitro decocting as well as in vivo absorption was investigated by determining the dry extract yield and by rat single-pass intestinal perfusion (SPIP) model. Inhibition of different species of Bulbus fritillariae on P-gp function was examined using the SPIP model. The mRNA and protein expression of P-gp was determined by PCR and western blotting. The active ingredients of Bulbus fritillariae were predicted and screened for inhibiting P-gp activity by Schrodinger’s molecular docking and MDR1-MDCK cell transport study, respectively. Mediation of monoester alkaloids in Radix aconiti praeparata by P-gp was predicted and examined using Schrodinger’s molecular docking and SPIP experiment, respectively. In the results, when Radix aconiti praeparata was combined with Bulbus fritillariae, the toxic ingredient benzoylmesaconine in Radix aconiti praeparata displayed higher intestinal permeability, whereas the toxic ingredients showed no significant difference during the in vitro decoction process. Bulbus fritillariae thunbergii inhibited both the P-gp function and expression; in contrast, Bulbus fritillariae cirrhosae inhibited the function only. Alkaloids including peimine, peimisine, and imperialine were the active ingredients for inhibiting P-gp activity. Benzoylmesaconine in Radix aconiti praeparata was the substrate of P-gp.
  • 金井 泰和, 渡辺 利光, 大田 洋一郎, 井口 佳哉, 堺 俊博, 仲 定宏, 服部 能英, 上原 幸樹, 片岡 昌治, 金井 好克, 切畑 光統
    核医学, 58(Suppl.) S232-S232, Oct, 2021  
  • Stephen PH Alexander, Eamonn Kelly, Alistair Mathie, John A Peters, Emma L Veale, Jane F Armstrong, Elena Faccenda, Simon D Harding, Adam J Pawson, Christopher Southan, Jamie A Davies, Laura Amarosi, Catriona M. H. Anderson, Philip Mark Beart, Stefan Broer, Paul A. Dawson, Bruno Hagenbuch, James R. Hammond, Ken‐ichi Inui, Yoshikatsu Kanai, Stephan Kemp, Gavin Stewart, David T. Thwaites, Tiziano Verri
    British Journal of Pharmacology, 178(S1), Oct, 2021  Peer-reviewed
  • 兼田 加珠子, 真鍋 良幸, 下山 敦史, 樺山 一哉, 金井 好克, 豊嶋 厚史, 深瀬 浩一, 篠原 厚
    日本癌学会総会記事, 80回 [J19-3], Sep, 2021  Peer-reviewed
  • Junryo Rii, Shinichi Sakamoto, Masahiro Sugiura, Manato Kanesaka, Ayumu Fujimoto, Yasutaka Yamada, Maihulan Maimaiti, Keisuke Ando, Ken Wakai, Minhui Xu, Yusuke Imamura, Norihisa Shindo, Toru Hirota, Atsushi Kaneda, Yoshikatsu Kanai, Yuzuru Ikehara, Naohiko Anzai, Tomohiko Ichikawa
    Cancer Science, Jul 8, 2021  Peer-reviewed
  • Ohgaki Ryuichi, Quan Lili, Okuda Suguru, Okanishi Hiroki, Nagamori Shushi, Endou Hitoshi, Kanai Yoshikatsu
    Proceedings for Annual Meeting of The Japanese Pharmacological Society, 94 3-O-F5-3, 2021  
    Angiogenesis is essential for development and progression of tumors, and regarded as a rational target in anti-cancer treatment. We recently reported that L-type amino acid transporter 1 (LAT1) is upregulated in tumor-associated vascular endothelium of human pancreatic cancer and in vivo animal models. We also reported that tumor growth in animal model was significantly impaired through the inhibition of angiogenesis by targeting endothelial LAT1. To reveal the molecular mechanisms underlying the contribution of LAT1 in tumor angiogenesis, we investigated the effects of abrogating endothelial LAT1 on proliferation and intracellular signaling pathways in human umbilical vein endothelial cells. Pharmacological and genetic inhibition LAT1 drastically suppressed cell proliferation, causing a down-regulation of signaling pathways, GCAA- and mTORC1 pathways that are regulating the translation initiation. Furthermore, LAT1 was fundamental to promote migration, invasion, and tube formation induced by pro-angiogenic factor VEGF-A through the activation of mTORC1 in a manner independent to the tyrosine kinase receptor VEGFR2. These findings highlight a cross-talk between pro-angiogenic signaling and nutrient-sensing in tumor-associated endothelial cells, and may provide novel rational strategies for anti-angiogenic therapy.

Misc.

 431

Books and Other Publications

 41

Presentations

 38

Research Projects

 51

Industrial Property Rights

 68