Network


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

Hotspot


Dive into the research topics where Jun-ichi Nezu is active.

Publication


Featured researches published by Jun-ichi Nezu.


Journal of Biological Chemistry | 1998

Molecular and Functional Identification of Sodium Ion-dependent, High Affinity Human Carnitine Transporter OCTN2*

Ikumi Tamai; Rikiya Ohashi; Jun-ichi Nezu; Hikaru Yabuuchi; Asuka Oku; Miyuki Shimane; Yoshimichi Sai; Akira Tsuji

Primary carnitine deficiency, because of a defect of the tissue plasma membrane carnitine transporters, causes critical symptoms. However, the transporter has not been molecularly identified. In this study, we screened a human kidney cDNA library and assembled a cDNA-encoding OCTN2 as a homologue of the organic cation transporter OCTN1, and then we examined the function of OCTN2 as a carnitine transporter. OCTN2-cDNA encodes a polypeptide of 557 amino acids with 75.8% similarity to OCTN1. Northern blot analysis showed that OCTN2 is strongly expressed in kidney, skeletal muscle, heart, and placenta in adult humans. When OCTN2 was expressed in HEK293 cells, uptake ofl-[3H]carnitine was strongly enhanced in a sodium-dependent manner with K m value of 4.34 μm, whereas typical substrates for previously known organic cation transporters, tetraethylammonium and guanidine, were not good substitutes. OCTN2-mediatedl-[3H]carnitine transport was inhibited by the d-isomer, acetyl-d,l-carnitine, and γ-butyrobetaine with high affinity and by glycinebetaine with lower affinity, whereas choline, β-hydroxybutyric acid, γ-aminobutyric acid, lysine, and taurine were not inhibitory. Because the observed tissue distribution of OCTN2 is consistent with the reported distribution of carnitine transport activity and the functional characteristics of OCTN2 coincide with those reported for plasma membrane carnitine transport, we conclude that OCTN2 is a physiologically important, high affinity sodium-carnitine cotransporter in humans.


Nature Genetics | 1999

Primary systemic carnitine deficiency is caused by mutations in a gene encoding sodium ion-dependent carnitine transporter

Jun-ichi Nezu; Ikumi Tamai; Asuka Oku; Rikiya Ohashi; Hikaru Yabuuchi; Noriyoshi Hashimoto; Hiroko Nikaido; Yoshimichi Sai; Akio Koizumi; Yutaka Shoji; Goro Takada; Toyojiro Matsuishi; Makoto Yoshino; Hirohisa Kato; Toshihiro Ohura; Gozoh Tsujimoto; Miyuki Shimane; Akira Tsuji

Primary systemic carnitine deficiency (SCD; OMIM 212140) is an autosomal recessive disorder characterized by progressive cardiomyopathy, skeletal myopathy, hypoglycaemia and hyperammonaemia. SCD has also been linked to sudden infant death syndrome. Membrane-physiological studies have suggested a defect of the carnitine transport system in the plasma membrane in SCD patients and in the mouse model, juvenile visceral steatosis (jvs; ref. 6). Although the responsible loci have been mapped in both human and mouse, the underlying gene has not yet been identified. Recently, we cloned and analysed the function of a novel transporter protein termed OCTN2 (ref. 9). Our observation that OCTN2 has the ability to transport carnitine in a sodium-dependent manner prompted us to search for mutations in the gene encoding OCTN2, SLC22A5. Initially, we analysed the mouse gene and found a missense mutation in Slc22a5 in jvs mice. Biochemical analysis revealed that this mutation abrogates carnitine transport. Subsequent analysis of the human gene identified four mutations in three SCD pedigrees. Affected individuals in one family were homozygous for the deletion of a 113-bp region containing the start codon. In the second pedigree, the affected individual was shown to be a compound heterozygote for two mutations that cause a frameshift and a premature stop codon, respectively. In an affected individual belonging to a third family, we found a homozygous splice-site mutation also resulting in a premature stop codon. These mutations provide the first evidence that loss of OCTN2 function causes SCD.


Cancer Research | 2004

Identification of Soluble NH2-Terminal Fragment of Glypican-3 as a Serological Marker for Early-Stage Hepatocellular Carcinoma

Yoshitaka Hippo; Kiyotaka Watanabe; Akira Watanabe; Yutaka Midorikawa; Shogo Yamamoto; Sigeo Ihara; Susumu Tokita; Hiroko Iwanari; Yukio Ito; Kiyotaka Nakano; Jun-ichi Nezu; Hiroyuki Tsunoda; Takeshi Yoshino; Iwao Ohizumi; Masayuki Tsuchiya; Shin Ohnishi; Masatoshi Makuuchi; Takao Hamakubo; Tatsuhiko Kodama; Hiroyuki Aburatani

For detection of hepatocellular carcinoma (HCC) in patients with liver cirrhosis, serum α-fetoprotein has been widely used, but its sensitivity has not been satisfactory, especially in small, well-differentiated HCC, and complementary serum marker has been clinically required. Glypican-3 (GPC3), a heparan sulfate proteoglycan anchored to the plasma membrane, is a good candidate marker of HCC because it is an oncofetal protein overexpressed in HCC at both the mRNA and protein levels. In this study, we demonstrated that its NH2-terminal portion [soluble GPC3 (sGPC3)] is cleaved between Arg358 and Ser359 of GPC3 and that sGPC3 can be specifically detected in the sera of patients with HCC. Serum levels of sGPC3 were 4.84 ± 8.91 ng/ml in HCC, significantly higher than the levels seen in liver cirrhosis (1.09 ± 0.74 ng/ml; P < 0.01) and healthy controls (0.65 ± 0.32 ng/ml; P < 0.001). In well- or moderately-differentiated HCC, sGPC3 was superior to α-fetoprotein in sensitivity, and a combination measurement of both markers improved overall sensitivity from 50% to 72%. These results indicate that sGPC3 is a novel serological marker essential for the early detection of HCC.


Pharmaceutical Research | 2001

Functional characterization of human organic anion transporting polypeptide B (OATP-B) in comparison with liver-specific OATP-C.

Ikumi Tamai; Takashi Nozawa; Minako Koshida; Jun-ichi Nezu; Yoshimichi Sai; Akira Tsuji

AbstractPurpose. To assess the functional characteristics of human organic anion transporter B (OATP-B) in comparison with those of the known, liver-specific OATP-C. Methods. OATP-B or -C was expressed in HEK293 cells or Xenopus oocytes, and uptakes of estradiol-17β-glucuronide and estrone-3-sulfate were measured using radiolabeled compounds. Results. OATP-C transported both estrone-3-sulfate and estradiol-17β-glucuronide, whereas OATP-B transported only the former. OATP-C-mediated uptake of estrone-3-sulfate exhibited biphasic saturation kinetics, whereas transports of estradiol-17β-glucuronide by OATP-C and estrone-3-sulafte by OATP-B followed single-saturation kinetics. Inhibition kinetics showed that only the high-affinity site for estrone-3-sulfate on OATP-C was shared with glucuronide conjugates. Uptake of [3H]estrone-3-sulfate by OATP-B was inhibited by sulfate conjugates but not by glucuronide conjugates, whereas its uptake by OATP-C was inhibited by both types of conjugates. Conclusions. OATP-B accepted sulfate conjugates of steroids but not glucuronide conjugates, whereas OATP-C transported both types of steroid conjugates. Transport of estrone-3-sulfate by OATP-B and -C followed single- and biphasic-saturation kinetics, respectively, and the high-affinity site on OATP-C was the same as that for estradiol-17β-glucuronide. Other OATPs, OATP-A and OATP-8, reportedly exhibit different preferences for steroid conjugates, and the specific recognition of sulfate conjugates seems to be unique to OATP-B.


American Journal of Human Genetics | 1998

Loss of LKB1 Kinase Activity in Peutz-Jeghers Syndrome, and Evidence for Allelic and Locus Heterogeneity

Hamid Mehenni; Corinne Gehrig; Jun-ichi Nezu; Asuka Oku; Miyuki Shimane; Colette Rossier; Nicolas Guex; Jean-Louis Blouin; Hamish S. Scott

Peutz-Jeghers syndrome (PJS) is an autosomal dominant disease characterized by mucocutaneous pigmentation and hamartomatous polyps. There is an increased risk of benign and malignant tumors in the gastrointestinal tract and in extraintestinal tissues. One PJS locus has been mapped to chromosome 19p13.3; a second locus is suspected on chromosome 19q13.4 in a minority of families. The PJS gene on 19p13.3 has recently been cloned, and it encodes the serine/threonine kinase LKB1. The gene, which is ubiquitously expressed, is composed of 10 exons spanning 23 kb. Several LKB1 mutations have been reported in heterozygosity in PJS patients. In this study, we screened for LKB1 mutations in nine PJS families of American, Spanish, Portuguese, French, Turkish, and Indian origin and detected seven novel mutations. These included two frameshift mutations, one four-amino-acid deletion, two amino-acid substitutions, and two splicing errors. Expression of mutant LKB1 proteins (K78I, D176N, W308C, and L67P) and assessment of their autophosphorylation activity revealed a loss of the kinase activity in all of these mutants. These results provide direct evidence that the elimination of the kinase activity of LKB1 is probably responsible for the development of the PJS phenotypes. In two Indian families, we failed to detect any LKB1 mutation; in one of these families, we previously had detected linkage to markers on 19q13.3-4, which suggests locus heterogeneity of PJS. The elucidation of the molecular etiology of PJS and the positional cloning of the second potential PJS gene will further elucidate the involvement of kinases/phosphatases in the development of cancer-predisposing syndromes.


Journal of Neurochemistry | 2008

Functional relevance of carnitine transporter OCTN2 to brain distribution of l‐carnitine and acetyl‐l‐carnitine across the blood–brain barrier

Yasuto Kido; Ikumi Tamai; Aki Ohnari; Yoshimichi Sai; Toru Kagami; Jun-ichi Nezu; Hiroko Nikaido; Noriyoshi Hashimoto; Masahide Asano; Akira Tsuji

Transport of l‐[3H]carnitine and acetyl‐l‐[3H]carnitine at the blood–brain barrier (BBB) was examined by using in vivo and in vitro models. In vivo brain uptake of acetyl‐l‐[3H]carnitine, determined by a rat brain perfusion technique, was decreased in the presence of unlabeled acetyl‐l‐carnitine and in the absence of sodium ions. Similar transport properties for l‐[3H]carnitine and/or acetyl‐l‐[3H]carnitine were observed in primary cultured brain capillary endothelial cells (BCECs) of rat, mouse, human, porcine and bovine, and immortalized rat BCECs, RBEC1. Uptakes of l‐[3H]carnitine and acetyl‐l‐[3H]carnitine by RBEC1 were sodium ion‐dependent, saturable with Km values of 33.1 ± 11.4 µm and 31.3 ± 11.6 µm, respectively, and inhibited by carnitine analogs. These transport properties are consistent with those of carnitine transport by OCTN2. OCTN2 was confirmed to be expressed in rat and human BCECs by an RT‐PCR method. Furthermore, the uptake of acetyl‐l‐[3H]carnitine by the BCECs of juvenile visceral steatosis (jvs) mouse, in which OCTN2 is functionally defective owing to a genetical missense mutation of one amino acid residue, was reduced. The brain distributions of l‐[3H]carnitine and acetyl‐l‐[3H]carnitine in jvs mice were slightly lower than those of wild‐type mice at 4 h after intravenous administration. These results suggest that OCTN2 is involved in transport of l‐carnitine and acetyl‐l‐carnitine from the circulating blood to the brain across the BBB.


Biochimica et Biophysica Acta | 2001

Na+-coupled transport of l-carnitine via high-affinity carnitine transporter OCTN2 and its subcellular localization in kidney

Ikumi Tamai; Kayoko China; Yoshimichi Sai; Daisuke Kobayashi; Jun-ichi Nezu; Ei Kawahara; Akira Tsuji

The mechanism of Na(+)-dependent transport of L-carnitine via the carnitine/organic cation transporter OCTN2 and the subcellular localization of OCTN2 in kidney were studied. Using plasma membrane vesicles prepared from HEK293 cells that were stably transfected with human OCTN2, transport of L-carnitine via human OCTN2 was characterized. Uptake of L-[(3)H]carnitine by the OCTN2-expressing membrane vesicles was significantly increased in the presence of an inwardly directed Na(+) gradient, with an overshoot, while such transient uphill transport was not observed in membrane vesicles from cells that were mock transfected with expression vector pcDNA3 alone. The uptake of L-[(3)H]carnitine was specifically dependent on Na(+) and the osmolarity effect showed that Na(+) significantly influenced the transport rather than the binding. Changes of inorganic anions in the extravesicular medium and of membrane potential by valinomycin altered the initial uptake activity of L-carnitine by OCTN2. In addition, the fluxes of L-carnitine and Na(+) were coupled with 1:1 stoichiometry. Accordingly, it was clarified that Na(+) is coupled with flux of L-carnitine and the flux is an electrogenic process. Furthermore, OCTN2 was localized on the apical membrane of renal tubular epithelial cells. These results clarified that OCTN2 is important for the concentrative reabsorption of L-carnitine after glomerular filtration in the kidney.


Biochemical and Biophysical Research Communications | 2009

Anti-glypican 3 antibodies cause ADCC against human hepatocellular carcinoma cells

Kiyotaka Nakano; Tetsuro Orita; Jun-ichi Nezu; Takeshi Yoshino; Iwao Ohizumi; Masamichi Sugimoto; Koh Furugaki; Yasuko Kinoshita; Takahiro Ishiguro; Takao Hamakubo; Tatsuhiko Kodama; Hiroyuki Aburatani; Hisafumi Yamada-Okabe; Masayuki Tsuchiya

Glypican 3 (GPC3), a GPI-anchored heparan sulfate proteoglycan, is expressed in the majority of hepatocellular carcinoma (HCC) tissues. Using MRL/lpr mice, we successfully generated a series of anti-GPC3 monoclonal antibodies (mAbs). GPC3 was partially cleaved between Arg358 and Ser359, generating a C-terminal 30-kDa fragment and an N-terminal 40-kDa fragment. All mAbs that induced antibody-dependent cellular cytotoxicity (ADCC) and/or complement-dependent cytotoxicity (CDC) against cells expressing GPC3 recognized the 30-kDa fragment, indicating that the C-terminal region of GPC3 serves as an epitope for mAb with ADCC and/or CDC inducing activities. Chimeric mAbs with Fc replaced by human IgG1 were created from GC33, one of the mAbs that reacted with the C-terminal 30-kDa fragment. Chimeric GC33 induced not only ADCC against GPC3-positive human HCC cells but also was efficacious against the Huh-7 human HCC xenograft. Thus, mAbs against the C-terminal 30-kDa fragment such as GC33 are useful in therapy targeting HCC.


Human Mutation | 2000

Two novel missense mutations of the OCTN2 gene (W283R and V446F) in a patient with primary systemic carnitine deficiency

Ertan Mayatepek; Jun-ichi Nezu; Ikumi Tamai; Asuka Oku; Masaki Katsura; Miyuki Shimane; Akira Tsuji

Primary systemic carnitine deficiency (SCD) is an autosomal recessive disorder of fatty acid oxidation caused by defective cellular carnitine transport. The disease is characterized by metabolic derangement simulating Reyes syndrome, hypoglcaemia, progressive cardiomyopathy and skeletal myopathy. Recently, it was shown that SCD is caused by mutations in the organic cation/carnitine transporter OCTN2 (SLC22A5). We report two novel mutations, W283R and V446F, which are both missense mutations in an affected infant. In vitro expression studies demonstrated that both are actually function‐loss mutations with virtually no uptake activity. This is the first report of compound heterozygosity for two missense mutations in a patient with SCD. Hum Mutat 15:118, 2000.


Journal of Pharmacy and Pharmacology | 2003

Contribution of organic anion transporting polypeptide OATP-C to hepatic elimination of the opioid pentapeptide analogue [D-Ala2, D-Leu5]-enkephalin.

Takashi Nozawa; Ikumi Tamai; Yoshimichi Sai; Jun-ichi Nezu; Akira Tsuji

The objective of this study was to examine the transport activity of the human organic anion transporter OATP‐C (SLC21A6) for oligopeptides that are eliminated rapidly from the systemic circulation. We focused on an opioid peptide analogue, [d‐Ala2, d‐Leu5]‐enkephalin (DADLE), a linear pentapeptide modified to be stable. [3H]DADLE was taken up by rat isolated hepatocytes in a saturable manner and highly accumulated in the liver after intravenous administration to rats. The uptake of [3H]DADLE by the isolated hepatocytes was inhibited by several organic anions and pentapeptides, but not by tetra‐ or tripeptides. When OATP‐C was expressed in Xenopus laevis oocytes, a significant increase in uptake of [3H]DADLE was observed. Moreover, the inhibitory effects of various compounds, including some peptides, on [3H]estrone‐3‐sulfate uptake by OATP‐C were similar to those observed in [3H]DADLE uptake by rat isolated hepatocytes. In conclusion, it was demonstrated that OATP‐C contributes to the rapid hepatic excretion of peptides and peptide‐mimetic drugs.

Collaboration


Dive into the Jun-ichi Nezu'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

Tomoyuki Igawa

Chugai Pharmaceutical Co.

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Rikiya Ohashi

Mitsubishi Tanabe Pharma

View shared research outputs
Researchain Logo
Decentralizing Knowledge