84 results on '"Kato S"'
Search Results
2. Positive and negative regulation of retinoid X receptor gene expression by thyroid hormone in the rat. Transcriptional and post-transcriptional controls by thyroid hormone.
- Author
-
Mano, H., primary, Mori, R., additional, Ozawa, T., additional, Takeyama, K., additional, Yoshizawa, Y., additional, Kojima, R., additional, Arao, Y., additional, Masushige, S., additional, and Kato, S., additional
- Published
- 1994
- Full Text
- View/download PDF
3. Human glyoxalase I. cDNA cloning, expression, and sequence similarity to glyoxalase I from Pseudomonas putida
- Author
-
Kim, N.S., primary, Umezawa, Y., additional, Ohmura, S., additional, and Kato, S., additional
- Published
- 1993
- Full Text
- View/download PDF
4. Tissue-specific enhancer of the human multidrug-resistance (MDR1) gene.
- Author
-
Kohno, K, primary, Sato, S, additional, Uchiumi, T, additional, Takano, H, additional, Kato, S, additional, and Kuwano, M, additional
- Published
- 1990
- Full Text
- View/download PDF
5. The tamoxifen-responsive estrogen receptor alpha mutant D351Y shows reduced tamoxifen-dependent interaction with corepressor complexes.
- Author
-
Yamamoto, Y, Wada, O, Suzawa, M, Yogiashi, Y, Yano, T, Kato, S, and Yanagisawa, J
- Abstract
The effects of estrogen and anti-estrogen are mediated through the estrogen receptors ERalpha and beta, which function as ligand-induced transcriptional factors. The nonsteroidal anti-estrogen tamoxifen is the most commonly used endocrine in the treatment of all stages of breast cancer in both pre- and postmenopausal women. Several lines of evidence have indicated that tamoxifen promotes association between ERalpha and corepressors N-CoR or silencing mediator for retinoid and thyroid hormone receptor (SMRT). Our results indicate that N-CoR/SMRT recognize and interact with helices H3 and H5 of the ERalpha ligand-binding domain in a 4-hydroxy tamoxifen-dependent manner. The mutant ERalpha(D351Y), derived from a tamoxifen-stimulated tumor and containing an amino acid substitution at position 351 within H3, showed reduced interaction with N-CoR/SMRT and high tamoxifen-induced activation function-1 (AF-1) activity. While the estradiol-dependent transcriptional activity of ERalpha(D351Y) was almost equal to that of wild-type ERalpha, the mutant exhibited higher levels of transcriptional activity in the presence of both E2 and 4-hydroxy tamoxifen compared with wild-type ERalpha. These results may explain the observation that the growth of tumor cells expressing ERalpha(D351Y) can be stimulated by tamoxifen, E2, or both.
- Published
- 2001
- Full Text
- View/download PDF
6. Suppression subtractive hybridization identifies high glucose levels as a stimulus for expression of connective tissue growth factor and other genes in human mesangial cells.
- Author
-
Murphy, M, Godson, C, Cannon, S, Kato, S, Mackenzie, H S, Martin, F, and Brady, H R
- Abstract
Accumulation of mesangial matrix is a pivotal event in the pathophysiology of diabetic nephropathy. The molecular triggers for matrix production are still being defined. Here, suppression subtractive hybridization identified 15 genes differentially induced when primary human mesangial cells are exposed to high glucose (30 mM versus 5 mM) in vitro. These genes included (a) known regulators of mesangial cell activation in diabetic nephropathy (fibronectin, caldesmon, thrombospondin, and plasminogen activator inhibitor-1), (b) novel genes, and (c) known genes whose induction by high glucose has not been reported. Prominent among the latter were genes encoding cytoskeleton-associated proteins and connective tissue growth factor (CTGF), a modulator of fibroblast matrix production. In parallel experiments, elevated CTGF mRNA levels were demonstrated in glomeruli of rats with streptozotocin-induced diabetic nephropathy. Mannitol provoked less mesangial cell CTGF expression in vitro than high glucose, excluding hyperosmolality as the key stimulus. The addition of recombinant CTGF to cultured mesangial cells enhanced expression of extracellular matrix proteins. High glucose stimulated expression of transforming growth factor beta1 (TGF-beta1), and addition of TGF-beta1 to mesangial cells triggered CTGF expression. CTGF expression induced by high glucose was partially suppressed by anti-TGF-beta1 antibody and by the protein kinase C inhibitor GF 109203X. Together, these data suggest that 1) high glucose stimulates mesangial CTGF expression by TGFbeta1-dependent and protein kinase C dependent pathways, and 2) CTGF may be a mediator of TGFbeta1-driven matrix production within a diabetic milieu.
- Published
- 1999
7. Evidence that a phosphatidylinositol 3,4,5-trisphosphate-binding protein can function in nucleus.
- Author
-
Tanaka, K, Horiguchi, K, Yoshida, T, Takeda, M, Fujisawa, H, Takeuchi, K, Umeda, M, Kato, S, Ihara, S, Nagata, S, and Fukui, Y
- Abstract
PIP3BP is a phosphatidylinositol 3,4,5-trisphosphate-binding protein (PIP3BP) abundant in brain, containing a zinc finger motif and two pleckstrin homology (PH) domains. Staining of rat brain cells with anti-PIP3BP antibody and determination of localization of PIP3BP fused to the green fluorescent protein (GFP-PIP3BP) revealed that PIP3BP was targeted to the nucleus. Targeting was dependent on a putative nuclear localization signal in PIP3BP. Generation of PIP3 in the nucleus was detected in H2O2-treated 293T cells, nerve growth factor (NGF)-treated PC12 cells, and platelet-derived growth factor (PDGF)-treated NIH 3T3 cells. Translocation of phosphatidylinositol 3-kinase (PI 3-kinase) to the nucleus and enhanced activity of PI 3-kinase in the nucleus fraction were observed after H2O2 treatment of 293T cells, suggesting that PI 3-kinase can be activated in the nucleus as well as in the membrane after appropriate stimulation of the cells. Co-expression of the constitutively active PI 3-kinase with PIP3BP resulted in exportation of the protein from the nucleus to the cytoplasm, suggesting that PIP3BP can function as a PIP3-binding protein in the intact cells. These results imply that there may be an unknown function of PI 3-kinase in the nucleus.
- Published
- 1999
8. Association of two nuclear proteins, Npw38 and NpwBP, via the interaction between the WW domain and a novel proline-rich motif containing glycine and arginine.
- Author
-
Komuro, A, Saeki, M, and Kato, S
- Abstract
We have previously reported a nuclear protein possessing a WW domain, Npw38 (Komuro, A., Saeki, M., and Kato, S. (1999) Nucleic Acids Res. 27, 1957-1965). Here we report a Npw38-binding protein, NpwBP, isolated from HeLa cell nuclear extracts and its characterization using a cloned cDNA. NpwBP contains two proline-rich regions that are capable of binding to the WW domain of Npw38. The binding analysis using an oligopeptide-immobilized membrane revealed that the WW domain of Npw38 preferentially recognizes a short proline-rich sequence, PPGPPP, surrounded by an arginine residue, so we named it a PGR motif. Localization analysis using green fluorescent protein fusion protein and immunostaining showed that Npw38 and NpwBP are colocalized in the same subnuclear region. Coimmunoprecipitation experiments confirmed the association between Npw38 and NpwBP, which were expressed as epitope-tagged forms in COS7 cells. Furthermore, the N-terminal region of NpwBP has binding ability for poly(rG) and G-rich single-stranded DNA. These results suggest that NpwBP is a physiological ligand of Npw38 and that the Npw38-NpwBP complex may function as a component of an mRNA factory in the nucleus.
- Published
- 1999
9. Positive and negative modulation of vitamin D receptor function by transforming growth factor-beta signaling through smad proteins.
- Author
-
Yanagi, Y, Suzawa, M, Kawabata, M, Miyazono, K, Yanagisawa, J, and Kato, S
- Abstract
Several lines of experiments demonstrated the interplay between the transforming growth factor-beta (TGF-beta) and vitamin D signaling pathways. Recently, we found that Smad3, a downstream component of the TGF-beta signaling pathway, potentiates ligand-induced transactivation of vitamin D receptor (VDR) as a coactivator of VDR (Yanagisawa, J., Yanagi, Y., Masuhiro, Y., Suzawa, M., Watanabe, M., Kashiwagi, K., Toriyabe, T., Kawabata, M., Miyazono, K., and Kato, S. (1999) Science 283, 1317-1321). Here, we investigated the roles of inhibitory Smads, Smad6 and Smad7, which are negative regulators of the TGF-beta/bone morphogenetic protein signaling pathway, on the Smad3-mediated potentiation of VDR function. We found that Smad7, but not Smad6, abrogates the Smad3-mediated VDR potentiation. Interaction studies in vivo and in vitro showed that Smad7 inhibited the formation of the VDR-Smad3 complex, whereas Smad6 had no effect. Taken together, our results strongly suggest that the interplay between the TGF-beta and vitamin D signaling pathways is, at least in part, mediated by the two classes of Smad proteins, which modulate VDR transactivation function both positively and negatively.
- Published
- 1999
10. Differential interactions of the C terminus and the cytoplasmic I-II loop of neuronal Ca2+ channels with G-protein alpha and beta gamma subunits. I. Molecular determination.
- Author
-
Furukawa, T, Nukada, T, Mori, Y, Wakamori, M, Fujita, Y, Ishida, H, Fukuda, K, Kato, S, and Yoshii, M
- Abstract
Interactions of G-protein alpha (Galpha) and beta gamma subunits (Gbeta gamma) with N- (alpha1B) and P/Q-type (alpha1A) Ca2+ channels were investigated using the Xenopus oocyte expression system. Gi3alpha was found to inhibit both N- and P/Q-type channels by receptor agonists, whereas Gbeta1 gamma2 was responsible for prepulse facilitation of N-type channels. L-type channels (alpha1C) were not regulated by Galpha or Gbeta gamma. For N-type, prepulse facilitation mediated via Gbeta gamma was impaired when the cytoplasmic I-II loop (loop 1) was deleted or replaced with the alpha1C loop 1. Galpha-mediated inhibitions were also impaired by substitution of the alpha1C loop 1, but only when the C terminus was deleted. For P/Q-type, by contrast, deletion of the C terminus alone diminished Galpha-mediated inhibition. Moreover, a chimera of L-type with the alpha1B loop 1 gained Gbeta gamma-dependent facilitation, whereas an L-type chimera with the N- or P/Q-type C terminus gained Galpha-mediated inhibition. These findings provide evidence that loop 1 of N-type channels is a regulatory site for Gbeta gamma and the C termini of P/Q- and N-types for Galpha.
- Published
- 1998
11. 1alpha,25-dehydroxyvitamin D3 synergism toward transforming growth factor-beta1-induced AP-1 transcriptional activity in mouse osteoblastic cells via its nuclear receptor.
- Author
-
Takeshita, A, Imai, K, Kato, S, Kitano, S, and Hanazawa, S
- Abstract
The present study demonstrates 1alpha,25-dehydroxyvitamin D3 (1alpha-25-(OH)2D3) synergism toward transforming growth factor (TGF)-beta1-induced activation protein-1 (AP-1) activity in mouse osteoblastic MC3T3-E1 cells via the nuclear receptor of the vitamin. 1alpha-25-(OH)2D3 synergistically stimulated TGF-beta1-induced expression of the c-jun gene in the cells but not that of the c-fos gene. We actually showed by a gel mobility shift assay 1alpha-25-(OH)2D3 synergism of TGF-beta1-induced AP-1 binding to the 12-(O-tetradecanoylphorbol-13-acetate response element (TRE). 1alpha-25-(OH)2D3 markedly stimulated the transient activity of TGF-beta1-induced AP-1 in the cells transfected with a TRE-chloramphenicol acetyltransferase (CAT) reporter gene. Also, a synergistic increase in TGF-beta1-induced CAT activity was observed in the cells cotransfected with an expression vector encoding vitamin D3 receptor (VDR) and the reporter gene. However, the synergistic CAT activity was inhibited by pretreatment with VDR antisense oligonucleotides. In addition, in a Northern blot assay, we observed 1alpha-25-(OH)2D3 synergism of TGF-beta1-induced expression of the c-jun gene in the cells transfected with the VDR expression vector and also found that the synergistic action was clearly blocked by VDR antisense oligonucleotide pretreatment. The present study strongly suggests a novel positive regulation by 1alpha-25-(OH)2D3 of TGF-beta1-induced AP-1 activity in osteoblasts via "genomic action."
- Published
- 1998
12. A sulfotransferase-sulfatase system in avian oviduct which catalyzes a conversion of UDP-N-acetylgalactosamine 4-sulfate to the 6-sulfate isomer.
- Author
-
Otsu, K, Inoue, H, Nakanishi, Y, Kato, S, Tsuji, M, and Suzuki, S
- Abstract
Magnum from quail oviduct was subfractionated to yield epithelium and tubular glands. The in vitro enzymatic activities involved in sulfated sugar nucleotide biosynthesis were assayed in these isolated tissues. The results demonstrated that the activities necessary for a series of reactions, UDP-N-acetylgalactosamine—-UDP-N-acetylgalactosamine 4-sulfate—-UDP-N-acetylgalactosamine 4,6- bisulfate —-UDP-N-acetylgalactosamine 6-sulfate, are located predominantly in the tubular gland. Both time course and pulse-chase studies with [35S]sulfate gave results that were consistent with this reaction scheme. A microsomal preparation from the magnum was shown to be capable of labeling all three sulfate sugar nucleotides with [35S]sulfate upon incubation with UDP-N-acetylgalactosamine and 3‘- phosphoadenylyl [35S]sulfate. Again, their relative labeling rates were in the order necessary to allow for a synthesis of sulfated sugar nucleotides in the sequence described above. Furthermore, incubation of the microsomal preparation with UDP-N-[14C]acetylgalactosamine 4-sulfate and 3‘- phosphoadenylyl sulfate resulted in the formation of UDP-N-[14C]acetylgalactosamine 6-sulfate. Also shown was the existence in the microsomal preparation of a sulfatase specific for the sulfate at position 4 of UDP-N-acetylgalactosamine 4,6- bisulfate . The results, together with those obtained in previous investigations, suggest that the tubular gland of quail oviduct contains a microsomal multienzyme system which catalyzes a series of sulfation and desulfation of N-acetylgalactosamine residues at the nonreducing terminal position of either sugar nucleotides or polysaccharide chains.
- Published
- 1984
- Full Text
- View/download PDF
13. Molecular mechanism of human CD38 gene expression by retinoic acid. Identification of retinoic acid response element in the first intron.
- Author
-
Kishimoto, H, Hoshino, S, Ohori, M, Kontani, K, Nishina, H, Suzawa, M, Kato, S, and Katada, T
- Abstract
CD38 is a nonlineage-restricted type II transmembrane glycoprotein possessing ecto-NAD+ glycohydrolase activity. Because of its unique expression pattern in lymphocyte differentiation, it appears to function as an immunoregulatory molecule. We previously reported that CD38 was specifically induced by all-trans-retinoic acid (RA) in human promyelocytic leukemia HL-60 cells. Here we studied the molecular mechanism of the RA-dependent induction of human CD38. The expression of CD38 mRNA by RA appeared to be caused by the transcriptional stimulation of the gene, since it was blocked by an RNA synthesis inhibitor, but not by a protein synthesis inhibitor. In search of the RA response element (RARE) possibly present in human CD38 gene promoter, we isolated and sequenced the genomic DNA covering the 5'-flanking region, exon 1, and partial intron 1. Transient transfection experiments revealed that the responsiveness to RA was conferred through an RARE consisting of two direct repeat TGACCT-like hexamer motifs with a 5-nucleotide spacer, which was located in the first intron rather than the 5'-flanking region of the CD38 gene. This RARE interacted with heterodimer composed of RA receptor and retinoid X receptor in vitro. Thus, the RA-induced expression of the human CD38 gene was demonstrated to be mediated through the RARE located in the first intron.
- Published
- 1998
14. Decreased transfer of oligosaccharide from oligosaccharide-lipid to protein acceptors in regenerating rat liver.
- Author
-
Oda-Tamai, S, Kato, S, Hara, S, and Akamatsu, N
- Abstract
The transfer of [14C]glucose from UDP-[14C]glucose to lipid intermediates and glycoproteins was decreased in regenerating rat liver microsomes 24 h after partial hepatectomy. In regenerating liver microsomes, the concentration of free dolichyl phosphate (Dol-P) was significantly decreased. However, it was only about 10% of total Dol-P, which was not significantly changed. On the addition of exogenous Dol-P, the transfer of [14C]glucose to glycoproteins was still decreased, while the decrease of the transfer to lipid intermediates was no longer observed. These results suggest that the glycoprotein synthesis is not regulated by the amount of Dol-P in regenerating liver microsomes. Oligosaccharide obtained from [14C]glucosyl-oligosaccharide-lipid was not distinguishable between regenerating liver and control by paper chromatography. The oligosaccharide transfer to protein in microsomes was compared by using [14C]glucosyl-oligosaccharide-lipid as oligosaccharide donor. The transfer of oligosaccharide to endogenous proteins decreased to 77% of control in regenerating liver and the transfer to exogenously added denatured alpha-lactalbumin decreased to 59% of control. Therefore, it is unlikely that the acceptor capacity of endogenous protein is decreased in regenerating liver. Neither the change in oligosaccharide-lipid under the condition for oligosaccharide transfer assay nor the stability of oligosaccharide transferase was different between regenerating liver and control. These results strongly suggest that the decrease in the activity of the oligosaccharide transferase in microsomes causes the decrease of glycoprotein synthesis in regenerating liver, which was shown in our previous studies.
- Published
- 1985
- Full Text
- View/download PDF
15. Mechanism of heterologous desensitization of the adenylate cyclase system by glucagon in primary cultures of adult rat hepatocytes.
- Author
-
Noda, C, Shinjyo, F, Tomomura, A, Kato, S, Nakamura, T, and Ichihara, A
- Abstract
During treatment of primary cultured hepatocytes with either glucagon or isoproterenol for several hours, the stimulations of cAMP formation by these hormones decreased time dependently. Glucagon treatment also reduced the response to isoproterenol, but isoproterenol treatment did not decrease the response to glucagon. Treatment with isoproterenol caused more rapid desensitization than treatment with glucagon. Assays of glucagon and beta-adrenergic receptors showed that the receptor number of only the hormone with which the cells were treated decreased and that dissociation constants of the receptors did not change. Moreover, in glucagon-desensitized cells, the effect of GTP on competition of the bindings of antagonist and agonist for the beta-adrenergic receptor did not change. After treatment with isoproterenol, stimulation of adenylate cyclase activity by the agonist was decreased without any decrease in the stimulations of activity by other effectors. In contrast, glucagon treatment greatly decreased the stimulations of activity by glucagon, isoproterenol, and guanyl-5'-yl imidodiphosphate and slightly decreased that by fluoride. However, after glucagon treatment, the cells showed normal responses to cholera toxin of activation of adenylate cyclase and ADP-ribosylation of guanine nucleotide binding regulatory protein (Ns). The maximal response of glucagon-treated cells to forskolin was about two-thirds that of untreated cells and this treatment also impaired the shift toward a low Kact value for forskolin observed in the presence of either glucagon or isoproterenol. These results indicate that isoproterenol caused homologous desensitization consisting of only a “down regulation” of the beta-adrenergic receptor, whereas glucagon caused heterologous desensitization, mainly by alteration of the Ns component, as well as “down regulation” of glucagon receptor. This altered Ns seems to be coupled normally to the beta-adrenergic receptor, but to have impaired coupling to the catalytic component of adenylate cyclase.
- Published
- 1984
- Full Text
- View/download PDF
16. Okadaic acid suppresses neural differentiation-dependent expression of the neurofilament-L gene in P19 embryonal carcinoma cells by post-transcriptional modification.
- Author
-
Sasahara, Y, Kobayashi, T, Onodera, H, Onoda, M, Ohnishi, M, Kato, S, Kusuda, K, Shima, H, Nagao, M, Abe, H, Yanagawa, Y, Hiraga, A, and Tamura, S
- Abstract
Mouse P19 embryonal carcinoma cells in aggregation culture in the presence of 10(-6) M retinoic acid followed by monolayer culture differentiate into nerve and glial cells. In this study, we demonstrated that the neurofilament-L (NF-L) mRNA and protein levels of these cells were enhanced in accordance with their retinoic acid-induced neural differentiation. Okadaic acid (OA) treatment of the cells markedly suppressed this differentiation-dependent NF-L gene expression increase and neurite outgrowth of the cells. Similar results were obtained when tautomycin was used instead of OA, suggesting that inhibition of protein phosphatase(s) is involved in the suppression of neural differentiation. OA treatment did not affect the NF-L gene transcription level, determined by the nuclear run-on transcription assay, but it did reduce the stability of both the 3.5- and 2.3-kilobase NF-L mRNAs. The expression and activity levels of protein phosphatase 2A (PP2A) and 2B (PP2B) but not protein phosphatase 1 (PP1) in P19 cells increased in accordance with the enhanced NF-L gene expression. The presence of OA in the culture medium during the course of the neural differentiation caused a reduced PP2A activity but not PP1 and PP2B activities of the cell extracts. On the other hand, both PP1 and PP2B activities but not PP2A activity of cell extracts were suppressed by the addition of cyclosporin A or FK506 in the culture medium. However, both cyclosporin A and FK506 treatments affected neither NF-L gene expression nor neurite outgrowth. These results demonstrate that the OA treatment inhibits the differentiation-dependent increase in NF-L gene expression by destabilizing its mRNAs and suggest that PP2A plays key roles in the differentiation-dependent enhanced expression of the NF-L gene and is the point of the action of OA.
- Published
- 1996
17. Primary structures of the mRNAs encoding the rat precursors for bradykinin and T-kinin. Structural relationship of kininogens with major acute phase protein and alpha 1-cysteine proteinase inhibitor.
- Author
-
Furuto-Kato, S, Matsumoto, A, Kitamura, N, and Nakanishi, S
- Abstract
Three types of cloned cDNA sequences for rat low molecular weight prekininogens were isolated and determined by molecular cloning and sequence analysis. The deduced amino acid sequences indicated that one, termed K-prekininogen, represents the counterpart of the known low molecular weight prekininogen present in other mammals, while the other two, called T-prekininogens, contain a novel T-kinin sequence which was recently identified from rat plasma. Although T- and K-prekininogens are highly homologous with each other, both of the T-prekininogens contain methionine, instead of arginine or lysine, as an amino acid preceding T-kinin and exhibit two consecutive amino acid deletions in the preceding region of T-kinin as compared with K-prekininogen. The former finding accounts for the previous observation of strong resistance of T-kininogens to cleavage with trypsin or kallikreins, while the latter finding has been explained by the structural analysis of genomic clones in which T-kinin-coding exon is contracted at its intron junction. A partial nucleotide sequence reported recently for the rat major acute phase protein (alpha 1-MAP) mRNA was found to be extremely related to the corresponding portion of the rat T-prekininogen mRNA. Furthermore, consistent with the previous report of the structural identity of major acute phase protein and alpha 1-cysteine proteinase inhibitor, kininogen closely resembles not only the former but also the latter in the amino acid compositions. The interrelationship among the triad of these proteins has been discussed.
- Published
- 1985
- Full Text
- View/download PDF
18. Location of regions of the opioid receptor involved in selective agonist binding.
- Author
-
Fukuda, K, Kato, S, and Mori, K
- Abstract
To elucidate which portions of the opioid receptor molecules are involved in the ligand selectivity, we have expressed chimeric receptors between the rat delta- and mu-opioid receptors from cDNAs and analysed their ligand binding properties. We demonstrate that the major binding determinant for the delta-selective enkephalin-related peptide, [D-Pen2,D-Pen5]enkephalin, resides within the region comprising the transmembrane segments V-VII and the intervening loop regions. On the other hand, the region spanning from the intracellular loop I to the amino-terminal half of the transmembrane segment III is shown to be involved in determining high-affinity binding of the mu-selective enkephalin-related peptides, [D-Ala2, MePhe4,Gly-ol5]enkephalin and [D-Ala2,MePhe4,Met-ol5]enkephalin, whereas the major determinant for binding of the mu-selective alkaloids, morphine and codeine, is demonstrated to exist in the region spanning the transmembrane segments V-VII. These results indicate that distinct regions of the opioid receptor determine the selectivity for the delta- and the mu-selective enkephalin-related peptides and that the binding determinant for the mu-selective alkaloids is distinct from that for the mu-selective enkephalin-related peptides.
- Published
- 1995
19. A terminal 6-sulfotransferase catalyzing a synthesis of N-acetylgalactosamine 4,6-bissulfate residue at the nonreducing terminal position of chondroitin sulfate.
- Author
-
Nakanishi, Y., primary, Shimizu, M., additional, Otsu, K., additional, Kato, S., additional, Tsuji, M., additional, and Suzuki, S., additional
- Published
- 1981
- Full Text
- View/download PDF
20. Oscillation of Cdc20-APC/C-mediated CAMDI stability is critical for cortical neuron migration.
- Author
-
Okuda S, Sato M, Kato S, Nagashima S, Inatome R, Yanagi S, and Fukuda T
- Subjects
- Animals, Brain metabolism, Brain pathology, Cells, Cultured, Centrosome metabolism, Cerebral Cortex metabolism, Cerebral Cortex pathology, Female, Gene Knockdown Techniques methods, Humans, Mental Disorders genetics, Mental Disorders metabolism, Mice, Models, Animal, Neurons metabolism, Anaphase-Promoting Complex-Cyclosome metabolism, Brain growth & development, Cdc20 Proteins metabolism, Cell Cycle Proteins metabolism, Cell Movement, Mental Disorders pathology, Nerve Tissue Proteins metabolism, Neurons pathology
- Abstract
Radial migration during cortical development is required for formation of the six-layered structure of the mammalian cortex. Defective migration of neurons is linked to several developmental disorders such as autism and schizophrenia. A unique swollen structure called the dilation is formed in migrating neurons and is required for movement of the centrosome and nucleus. However, the detailed molecular mechanism by which this dilation forms is unclear. We report that CAMDI, a gene whose deletion is associated with psychiatric behavior, is degraded by cell division cycle protein 20 (Cdc20)-anaphase-promoting complex/cyclosome (APC/C) cell-cycle machinery after centrosome migration into the dilation in mouse brain development. We also show that CAMDI is restabilized in the dilation until the centrosome enters the dilation, at which point it is once again immediately destabilized. CAMDI degradation is carried out by binding to Cdc20-APC/C via the destruction box degron of CAMDI. CAMDI destruction box mutant overexpression inhibits dilation formation and neuronal cell migration via maintaining the stabilized state of CAMDI. These results indicate that CAMDI is a substrate of the Cdc20-APC/C system and that the oscillatory regulation of CAMDI protein correlates with dilation formation for proper cortical migration., Competing Interests: Conflict of interest The authors declare that they have no conflicts of interest with the contents of this article., (Copyright © 2021 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
21. The mechanosensitive channel YbdG from Escherichia coli has a role in adaptation to osmotic up-shock.
- Author
-
Amemiya S, Toyoda H, Kimura M, Saito H, Kobayashi H, Ihara K, Kamagata K, Kawabata R, Kato S, Nakashimada Y, Furuta T, Hamamoto S, and Uozumi N
- Subjects
- Amino Acid Substitution, Escherichia coli genetics, Escherichia coli Proteins genetics, Ion Channels genetics, Mutation, Missense, Protein Domains, Adaptation, Physiological, Escherichia coli metabolism, Escherichia coli Proteins metabolism, Ion Channels metabolism, Mechanotransduction, Cellular, Osmotic Pressure
- Abstract
Mechanosensitive channels play an important role in the adaptation of cells to hypo-osmotic shock. Among members of this channel family in Escherichia coli , the exact function and physiological role of the mechanosensitive channel homolog YbdG remain unclear. Characterization of YbdG's physiological role has been hampered by its lack of measurable transport activity. Using a nitrosoguanidine mutagenesis-aided screen in combination with next-generation sequencing, here we isolated a mutant with a point mutation in ybdG This mutation (resulting in a I167T change) conferred sensitivity to high osmotic stress, and the mutant cells differed from WT cells in morphology during hyperosmotic stress at alkaline pH. Interestingly, unlike the cells containing the I167T variant, a null- ybdG mutant did not exhibit this sensitivity and phenotype. Although I167T was located near the putative ion-conducting pore in a transmembrane region of YbdG, no change in ion channel activities of YbdG-I167T was detected. Of note, introduction of the WT C-terminal cytosolic region of YbdG into the I167T variant complemented the osmo-sensitive phenotype. Co-precipitation of proteins interacting with the C-terminal YbdG region led to the isolation of HldD and FbaA, whose overexpression in cells containing the YbdG-I167T variant partially rescued the osmo-sensitive phenotype. This study indicates that YbdG functions as a component of a mechanosensing system that transmits signals triggered by external osmotic changes to intracellular factors. The cellular role of YbdG uncovered here goes beyond its predicted function as an ion or solute transport protein., (© 2019 Amemiya et al.)
- Published
- 2019
- Full Text
- View/download PDF
22. Double PHD fingers protein DPF2 recognizes acetylated histones and suppresses the function of estrogen-related receptor α through histone deacetylase 1.
- Author
-
Matusyama R, Takada I, Yokoyama A, Fujiyma-Nakamura S, Tsuji N, Kitagawa H, Fujiki R, Kim M, Kouza-Fujita M, Yano T, and Kato S
- Published
- 2015
- Full Text
- View/download PDF
23. Positive and negative modulation of vitamin D receptor function by transforming growth factor-β signaling through Smad proteins.
- Author
-
Yanagi Y, Suzawa M, Kawabata M, Miyazono K, Yanagisawa J, and Kato S
- Published
- 2013
- Full Text
- View/download PDF
24. Phosphorylation of Williams syndrome transcription factor by MAPK induces a switching between two distinct chromatin remodeling complexes.
- Author
-
Oya H, Yokoyama A, Yamaoka I, Fujiki R, Yonezawa M, Youn MY, Takada I, Kato S, and Kitagawa H
- Published
- 2013
- Full Text
- View/download PDF
25. An hGCN5/TRRAP histone acetyltransferase complex co-activates BRCA1 transactivation function through histone modification.
- Author
-
Oishi H, Kitagawa H, Wada O, Takezawa S, Tora L, Kouzu-Fujita M, Takada I, Yano T, Yanagisawa J, and Kato S
- Published
- 2013
- Full Text
- View/download PDF
26. The tamoxifen-responsive estrogen receptor α mutant D351Y shows reduced tamoxifen-dependent interaction with corepressor complexes.
- Author
-
Yamamoto Y, Wada O, Suzawa M, Yogiashi Y, Yano T, Kato S, and Yanagisawa J
- Published
- 2013
- Full Text
- View/download PDF
27. Ligand type-specific interactions of peroxisome proliferator-activated receptor γ with transcriptional coactivators.
- Author
-
Kodera Y, Takeyama KI, Murayama A, Suzawa M, Masuhiro Y, and Kato S
- Published
- 2013
- Full Text
- View/download PDF
28. Heparin cofactor II, a serine protease inhibitor, promotes angiogenesis via activation of the AMP-activated protein kinase-endothelial nitric-oxide synthase signaling pathway.
- Author
-
Ikeda Y, Aihara K, Yoshida S, Iwase T, Tajima S, Izawa-Ishizawa Y, Kihira Y, Ishizawa K, Tomita S, Tsuchiya K, Sata M, Akaike M, Kato S, Matsumoto T, and Tamaki T
- Subjects
- Animals, Aorta cytology, Aorta metabolism, Cell Movement drug effects, Cell Movement physiology, Endothelial Cells cytology, Endothelial Cells metabolism, Heparin Cofactor II pharmacology, Humans, Male, Mice, Mice, Mutant Strains, Neovascularization, Physiologic drug effects, Signal Transduction drug effects, AMP-Activated Protein Kinases metabolism, Heparin Cofactor II metabolism, Neovascularization, Physiologic physiology, Nitric Oxide Synthase Type III metabolism, Signal Transduction physiology
- Abstract
We previously clarified that heparin cofactor II (HCII), a serine proteinase inhibitor, exerts various protective actions on cardiovascular diseases in both experimental and clinical studies. In the present study, we aimed to clarify whether HCII participates in the regulation of angiogenesis. Male heterozygous HCII-deficient (HCII(+/-)) mice and male littermate wild-type (HCII(+/+)) mice at the age of 12-16 weeks were subjected to unilateral hindlimb ligation surgery. Laser speckle blood flow analysis showed that blood flow recovery in response to hindlimb ischemia was delayed in HCII(+/-) mice compared with that in HCII(+/+) mice. Capillary number, arteriole number, and endothelial nitric-oxide synthase (eNOS), AMP-activated protein kinase (AMPK), and liver kinase B1 (LKB1) phosphorylation in ischemic muscles were decreased in HCII(+/-) mice. Human purified HCII (h-HCII) administration almost restored blood flow recovery, capillary density, and arteriole number as well as phosphorylation levels of eNOS, AMPK, and LKB1 in ischemic muscles of HCII(+/-) mice. Although treatment with h-HCII increased phosphorylation levels of eNOS, AMPK, and LKB1 in human aortic endothelial cells (HAECs), the h-HCII-induced eNOS phosphorylation was abolished by compound C, an AMPK inhibitor, and by AMPK siRNA. In a similar fashion, tube formation, proliferation, and migration of HAECs were also promoted by h-HCII treatment and were abrogated by pretreatment with compound C. HCII potentiates the activation of vascular endothelial cells and the promotion of angiogenesis in response to hindlimb ischemia via an AMPK-eNOS signaling pathway. These findings suggest that HCII is a novel therapeutic target for treatment of patients with peripheral circulation insufficiency.
- Published
- 2012
- Full Text
- View/download PDF
29. Targeting serous epithelial ovarian cancer with designer zinc finger transcription factors.
- Author
-
Lara H, Wang Y, Beltran AS, Juárez-Moreno K, Yuan X, Kato S, Leisewitz AV, Cuello Fredes M, Licea AF, Connolly DC, Huang L, and Blancafort P
- Subjects
- Actin Cytoskeleton genetics, Actin Cytoskeleton metabolism, Animals, Cell Line, Tumor, Female, Mice, Mice, Transgenic, Neoplasm Invasiveness, Neoplasms, Experimental genetics, Neoplasms, Experimental pathology, Neoplasms, Experimental therapy, Oligonucleotide Array Sequence Analysis, Ovarian Neoplasms genetics, Ovarian Neoplasms pathology, Ovarian Neoplasms therapy, RNA, Messenger genetics, RNA, Messenger metabolism, RNA, Messenger pharmacology, Serpins biosynthesis, Tetraspanins genetics, Tetraspanins metabolism, Transcription Factors genetics, Transcriptome, Gene Expression Regulation, Neoplastic, Neoplasms, Experimental metabolism, Ovarian Neoplasms metabolism, Transcription Factors biosynthesis, Zinc Fingers
- Abstract
Ovarian cancer is the leading cause of death among gynecological malignancies. It is detected at late stages when the disease is spread through the abdominal cavity in a condition known as peritoneal carcinomatosis. Thus, there is an urgent need to develop novel therapeutic interventions to target advanced stages of ovarian cancer. Mammary serine protease inhibitor (Maspin) represents an important metastasis suppressor initially identified in breast cancer. Herein we have generated a sequence-specific zinc finger artificial transcription factor (ATF) to up-regulate the Maspin promoter in aggressive ovarian cancer cell lines and to interrogate the therapeutic potential of Maspin in ovarian cancer. We found that although Maspin was expressed in some primary ovarian tumors, the promoter was epigenetically silenced in cell lines derived from ascites. Transduction of the ATF in MOVCAR 5009 cells derived from ascitic cultures of a TgMISIIR-TAg mouse model of ovarian cancer resulted in tumor cell growth inhibition, impaired cell invasion, and severe disruption of actin cytoskeleton. Systemic delivery of lipid-protamine-RNA nanoparticles encapsulating a chemically modified ATF mRNA resulted in inhibition of ovarian cancer cell growth in nude mice accompanied with Maspin re-expression in the treated tumors. Gene expression microarrays of ATF-transduced cells revealed an exceptional specificity for the Maspin promoter. These analyses identified novel targets co-regulated with Maspin in human short-term cultures derived from ascites, such as TSPAN12, that could mediate the anti-metastatic phenotype of the ATF. Our work outlined the first targeted, non-viral delivery of ATFs into tumors with potential clinical applications for metastatic ovarian cancers.
- Published
- 2012
- Full Text
- View/download PDF
30. JMJD5, a Jumonji C (JmjC) domain-containing protein, negatively regulates osteoclastogenesis by facilitating NFATc1 protein degradation.
- Author
-
Youn MY, Yokoyama A, Fujiyama-Nakamura S, Ohtake F, Minehata K, Yasuda H, Suzuki T, Kato S, and Imai Y
- Subjects
- Cell Differentiation physiology, Enzyme Activation physiology, Epigenesis, Genetic physiology, HEK293 Cells, Histone Demethylases genetics, Humans, Hydroxylation physiology, NFATC Transcription Factors genetics, Proteasome Endopeptidase Complex metabolism, Protein Processing, Post-Translational physiology, Transcriptional Activation physiology, Ubiquitin metabolism, Histone Demethylases metabolism, NFATC Transcription Factors metabolism, Osteoclasts cytology, Osteoclasts enzymology
- Abstract
Osteoclastogenesis is a highly regulated process governed by diverse classes of regulators. Among them, nuclear factor of activated T-cells calcineurin-dependent 1 (NFATc1) is the primary osteoclastogenic transcription factor, and its expression is transcriptionally induced during early osteoclastogenesis by receptor activation of nuclear factor κB ligand (RANKL), an osteoclastogenic cytokine. Here, we report the novel enzymatic function of JMJD5, which regulates NFATc1 protein stability. Among the tested Jumonji C (JmjC) domain-containing proteins, decreased mRNA expression levels during osteoclastogenesis were found for JMJD5 in RAW264 cells stimulated by RANKL. To examine the functional role of JMJD5 in osteoclast differentiation, we established stable JMJD5 knockdown cells, and osteoclast formation was assessed. Down-regulated expression of JMJD5 led to accelerated osteoclast formation together with induction of several osteoclast-specific genes such as Ctsk and DC-STAMP, suggesting that JMJD5 is a negative regulator in osteoclast differentiation. Although JMJD5 was recently reported as a histone demethylase for histone H3K36me2, no histone demethylase activity was detected in JMJD5 in vitro or in living cells, even for other methylated histone residues. Instead, JMJD5 co-repressed transcriptional activity by destabilizing NFATc1 protein. Protein hydroxylase activity mediated by the JmjC domain in JMJD5 was required for the observed functions of JMJD5. JMJD5 induced the association of hydroxylated NFATc1 with the E3 ubiquitin ligase Von Hippel-Lindau tumor suppressor (VHL), thereby presumably facilitating proteasomal degradation of NFATc1 via ubiquitination. Taken together, the present study demonstrated that JMJD5 is a post-translational co-repressor for NFATc1 that attenuates osteoclastogenesis.
- Published
- 2012
- Full Text
- View/download PDF
31. Zinc finger protein 467 is a novel regulator of osteoblast and adipocyte commitment.
- Author
-
Quach JM, Walker EC, Allan E, Solano M, Yokoyama A, Kato S, Sims NA, Gillespie MT, and Martin TJ
- Subjects
- Adipocytes pathology, Animals, Antigens, Differentiation biosynthesis, Antigens, Differentiation genetics, Cell Line, DNA-Binding Proteins, Mice, Nuclear Proteins genetics, Osteoblasts pathology, Osteoporosis genetics, Osteoporosis metabolism, Osteoporosis pathology, Retroviridae, Transcription Factors genetics, Transduction, Genetic, Adipocytes metabolism, Cell Differentiation, Nuclear Proteins metabolism, Osteoblasts metabolism, Response Elements, Transcription Factors metabolism, Transcriptional Activation
- Abstract
Osteoblasts and adipocytes are derived from common mesenchymal progenitor cells. The bone loss of osteoporosis is associated with altered progenitor differentiation from an osteoblastic to an adipocytic lineage. cDNA microarrays and quantitative real-time PCR (Q-PCR) were carried out in a differentiating mouse stromal osteoblastic cell line, Kusa 4b10, to identify gene targets of factors that stimulate osteoblast differentiation including parathyroid hormone (PTH) and gp130-binding cytokines, oncostatin M (OSM) and cardiotrophin-1 (CT-1). Zinc finger protein 467 (Zfp467) was rapidly down-regulated by PTH, OSM, and CT-1. Retroviral overexpression and RNA interference for Zfp467 in mouse stromal cells showed that this factor stimulated adipocyte formation and inhibited osteoblast commitment compared with controls. Regulation of adipocyte markers, including peroxisome proliferator-activated receptor (PPAR) γ, C/EBPα, adiponectin, and resistin, and late osteoblast/osteocyte markers (osteocalcin and sclerostin) by Zfp467 was confirmed by Q-PCR. Intra-tibial injection of calvarial cells transduced with retroviral Zfp467 doubled the number of marrow adipocytes in C57Bl/6 mice compared with vector control-transduced cells, providing in vivo confirmation of a pro-adipogenic role of Zfp467. Furthermore, Zfp467 transactivated a PPAR-response element reporter construct and recruited a histone deacetylase complex. Thus Zfp467 is a novel co-factor that promotes adipocyte differentiation and suppresses osteoblast differentiation. This has relevance to therapeutic interventions in osteoporosis, including PTH-based therapies currently available, and may be of relevance for the use of adipose-derived stem cells for tissue engineering.
- Published
- 2011
- Full Text
- View/download PDF
32. Double PHD fingers protein DPF2 recognizes acetylated histones and suppresses the function of estrogen-related receptor alpha through histone deacetylase 1.
- Author
-
Matsuyama R, Takada I, Yokoyama A, Fujiyma-Nakamura S, Tsuji N, Kitagawa H, Fujiki R, Kim M, Kouzu-Fujita M, Yano T, and Kato S
- Subjects
- Acetylation, Animals, Cell Differentiation, Cell Nucleus metabolism, DNA-Binding Proteins genetics, Histone Deacetylase 1 genetics, Histones chemistry, Humans, Mice, Mutation, Protein Structure, Tertiary, RNA, Messenger metabolism, Repressor Proteins genetics, Transcription Factors, Transcription, Genetic, Transcriptional Activation, ERRalpha Estrogen-Related Receptor, DNA-Binding Proteins chemistry, Histone Deacetylase 1 chemistry, Receptors, Estrogen chemistry, Repressor Proteins chemistry
- Abstract
Estrogen-related receptor alpha (ERRalpha) is a member of the nuclear receptor superfamily and regulates many physiological functions, including mitochondrial biogenesis and lipid metabolism. ERRalpha enhances the transactivation function without endogenous ligand by associating with coactivators such as peroxisome proliferator-activated receptor gamma coactivator 1 alpha and beta (PGC-1alpha and -beta) and members of the steroid receptor coactivator family. However, the molecular mechanism by which the transactivation function of ERRalpha is converted from a repressive state to an active state is poorly understood. Here we used biochemical purification techniques to identify ERRalpha-associated proteins in HeLa cells stably expressing ERRalpha. Interestingly, we found that double PHD fingers protein DPF2/BAF45d suppressed PGC-1alpha-dependent transactivation of ERRalpha by recognizing acetylated histone H3 and associating with HDAC1. DPF2 directly bound to ERRalpha and suppressed the transactivation function of nuclear receptors such as androgen receptor. DPF2 was recruited to ERR target gene promoters in myoblast cells, and knockdown of DPF2 derepressed the level of mRNA expressed by target genes of ERRalpha. These results show that DPF2 acts as a nuclear receptor-selective co-repressor for ERRalpha by associating with both acetylated histone H3 and HDAC1.
- Published
- 2010
- Full Text
- View/download PDF
33. NF-YC functions as a corepressor of agonist-bound mineralocorticoid receptor.
- Author
-
Murai-Takeda A, Shibata H, Kurihara I, Kobayashi S, Yokota K, Suda N, Mitsuishi Y, Jo R, Kitagawa H, Kato S, Saruta T, and Itoh H
- Subjects
- Aldosterone pharmacology, Animals, COS Cells, Cells, Cultured, Chlorocebus aethiops, Dose-Response Relationship, Drug, Epithelial Sodium Channels metabolism, Histone Deacetylases metabolism, Humans, Hydrocortisone pharmacology, Immunohistochemistry, Kidney Tubules, Collecting cytology, Male, Mice, Promoter Regions, Genetic physiology, Protein Structure, Tertiary, Receptors, Androgen metabolism, Receptors, Glucocorticoid metabolism, Receptors, Mineralocorticoid agonists, Receptors, Mineralocorticoid chemistry, Receptors, Progesterone metabolism, Two-Hybrid System Techniques, Aldosterone metabolism, CCAAT-Binding Factor metabolism, Hydrocortisone metabolism, Kidney Tubules, Collecting metabolism, Receptors, Mineralocorticoid metabolism
- Abstract
The role of aldosterone has been implicated in the metabolic syndrome and cardiovascular diseases. The biological actions of aldosterone are mediated through mineralocorticoid receptor (MR). Nuclear receptor-mediated gene expression is regulated by dynamic and coordinated recruitment of coactivators and corepressors. To identify new coregulators of the MR, full-length MR was used as bait in yeast two-hybrid screening. We isolated NF-YC, one of the subunits of heterotrimeric transcription factor NF-Y. Specific interaction between MR and NF-YC was confirmed by yeast two-hybrid, mammalian two-hybrid, coimmunoprecipitation assays, and fluorescence subcellular imaging. Transient transfection experiments in COS-7 cells demonstrated that NF-YC repressed MR transactivation in a hormone-sensitive manner. Moreover, reduction of NF-YC protein levels by small interfering RNA potentiated hormonal activation of endogenous target genes in stably MR-expressing cells, indicating that NF-YC functions as an agonist-dependent MR corepressor. The corepressor function of NF-YC is selective for MR, because overexpression of NF-YC did not affect transcriptional activity mediated by androgen, progesterone, or glucocorticoid receptors. Chromatin immunoprecipitation experiments showed that endogenous MR and steroid receptor coactivator-1 were recruited to an endogenous ENaC gene promoter in a largely aldosterone-dependent manner, and endogenous NF-YC was sequentially recruited to the same element. Immunohistochemistry showed that endogenous MR and NF-YC were colocalized within the mouse kidney. Although aldosterone induces interaction of the N and C termini of MR, NF-YC inhibited the N/C interaction. These findings indicate that NF-YC functions as a new corepressor of agonist-bound MR via alteration of aldosterone-induced MR conformation.
- Published
- 2010
- Full Text
- View/download PDF
34. Androgen receptor-dependent transactivation of growth arrest-specific gene 6 mediates inhibitory effects of testosterone on vascular calcification.
- Author
-
Son BK, Akishita M, Iijima K, Ogawa S, Maemura K, Yu J, Takeyama K, Kato S, Eto M, and Ouchi Y
- Subjects
- Adult, Animals, Apoptosis physiology, Calcinosis pathology, Cell Survival, Female, Genes, Reporter, Humans, Male, Promoter Regions, Genetic, RNA Stability, RNA, Small Interfering genetics, RNA, Small Interfering metabolism, Receptors, Androgen genetics, Vascular Diseases pathology, Calcinosis drug therapy, Intercellular Signaling Peptides and Proteins genetics, Intercellular Signaling Peptides and Proteins metabolism, Muscle, Smooth, Vascular drug effects, Muscle, Smooth, Vascular pathology, Receptors, Androgen metabolism, Testosterone pharmacology, Testosterone therapeutic use, Transcriptional Activation, Vascular Diseases drug therapy
- Abstract
Recent epidemiological studies have found that androgen deficiency is associated with a higher incidence of cardiovascular disease in men. However, little is known about the mechanism underlying the cardioprotective effects of androgens. Here we show the inhibitory effects of testosterone on vascular calcification and a critical role of androgen receptor (AR)-dependent transactivation of growth arrest-specific gene 6 (Gas6), a key regulator of inorganic phosphate (P(i))-induced calcification of vascular smooth muscle cells (VSMC). Testosterone and nonaromatizable androgen dihydrotestosterone inhibited P(i)-induced calcification of human aortic VSMC in a concentration-dependent manner. Androgen inhibited P(i)-induced VSMC apoptosis, an essential process for VSMC calcification. The effects on VSMC calcification were mediated by restoration of P(i)-induced down-regulation of Gas6 expression and a subsequent reduction of Akt phosphorylation. These effects of androgen were blocked by an AR antagonist, flutamide, but not by an estrogen receptor antagonist, ICI 182,780. We then explored the mechanistic role of the AR in Gas6 expression and found an abundant expression of AR predominantly in the nucleus of VSMC and two consensus ARE sequences in the Gas6 promoter region. Dihydrotestosterone stimulated Gas6 promoter activity, and this effect was abrogated by flutamide and by AR siRNA. Site-specific mutation revealed that the proximal ARE was essential for androgen-dependent transactivation of Gas6. Furthermore, chromatin immunoprecipitation assays demonstrated ligand-dependent binding of the AR to the proximal ARE of Gas6. These results indicate that AR signaling directly regulates Gas6 transcription, which leads to inhibition of vascular calcification, and provides a mechanistic insight into the cardioprotective action of androgens.
- Published
- 2010
- Full Text
- View/download PDF
35. Phosphorylation of Williams syndrome transcription factor by MAPK induces a switching between two distinct chromatin remodeling complexes.
- Author
-
Oya H, Yokoyama A, Yamaoka I, Fujiki R, Yonezawa M, Youn MY, Takada I, Kato S, and Kitagawa H
- Subjects
- Animals, Cell Line, Cell Line, Tumor, DNA Damage, Epigenesis, Genetic, Gene Expression Regulation, Humans, Mice, Mice, Transgenic, Mutation, Phosphorylation, Protein Structure, Tertiary, Transcription Factors metabolism, Chromatin chemistry, MAP Kinase Signaling System, Transcription Factors chemistry
- Abstract
Changes in the environment of a cell precipitate extracellular signals and sequential cascades of protein modification and elicit nuclear transcriptional responses. However, the functional links between intracellular signaling-dependent gene regulation and epigenetic regulation by chromatin-modifying proteins within the nucleus are largely unknown. Here, we describe novel epigenetic regulation by MAPK cascades that modulate formation of an ATP-dependent chromatin remodeling complex, WINAC (WSTF Including Nucleosome Assembly Complex), an SWI/SNF-type complex containing Williams syndrome transcription factor (WSTF). WSTF, a specific component of two chromatin remodeling complexes (SWI/SNF-type WINAC and ISWI-type WICH), was phosphorylated by the stimulation of MAPK cascades in vitro and in vivo. Ser-158 residue in the WAC (WSTF/Acf1/cbpq46) domain, located close to the N terminus of WSTF, was identified as a major phosphorylation target. Using biochemical analysis of a WSTF mutant (WSTF-S158A) stably expressing cell line, the phosphorylation of this residue (Ser-158) was found to be essential for maintaining the association between WSTF and core BAF complex components, thereby maintaining the ATPase activity of WINAC. WINAC-dependent transcriptional regulation of vitamin D receptor was consequently impaired by this WSTF mutation, but the recovery from DNA damage mediated by WICH was not impaired. Our results suggest that WSTF serves as a nuclear sensor of the extracellular signals to fine-tune the chromatin remodeling activity of WINAC. WINAC mediates a previously unknown MAPK-dependent step in epigenetic regulation, and this MAPK-dependent switching mechanism between the two functionally distinct WSTF-containing complexes might underlie the diverse functions of WSTF in various nuclear events.
- Published
- 2009
- Full Text
- View/download PDF
36. COP1 functions as a FoxO1 ubiquitin E3 ligase to regulate FoxO1-mediated gene expression.
- Author
-
Kato S, Ding J, Pisck E, Jhala US, and Du K
- Subjects
- Animals, CCAAT-Enhancer-Binding Proteins genetics, CCAAT-Enhancer-Binding Proteins metabolism, Carboxy-Lyases genetics, Cell Line, Tumor, Forkhead Box Protein O1, Forkhead Transcription Factors genetics, Gene Knockdown Techniques, Glucose-6-Phosphatase genetics, Humans, Liver, Nerve Tissue Proteins genetics, Protein Binding physiology, Rats, Ubiquitin genetics, Ubiquitin metabolism, Ubiquitin-Protein Ligases genetics, Carboxy-Lyases biosynthesis, Forkhead Transcription Factors metabolism, Gene Expression Regulation, Enzymologic physiology, Gluconeogenesis physiology, Glucose-6-Phosphatase biosynthesis, Nerve Tissue Proteins metabolism, Ubiquitin-Protein Ligases metabolism, Ubiquitination physiology
- Abstract
COP1 is a Ring-Finger E3 ubiquitin ligase that is involved in plant development, mammalian cell survival, growth, and metabolism. Here we report that COP1, whose expression is enhanced by insulin, regulates FoxO1 protein stability. We found that in Fao hepatoma cells, ectopic expression of COP1 decreased, whereas knockdown of COP1 expression increased the level of endogenous FoxO1 protein without impacting other factors such as C/EBPalpha and CREB (cAMP-response element-binding protein). We further showed that COP1 binds FoxO1, enhances its ubiquitination, and promotes its degradation via the ubiquitin-proteasome pathway. To determine the biological significance of COP1-mediated FoxO1 protein degradation, we have examined the impact of COP1 on FoxO1-mediated gene expression and found that COP1 suppressed FoxO1 reporter gene as well as FoxO1 target genes such as glucose-6-phosphatase and phosphoenolpyruvate carboxykinase, two key targets for FoxO1 in the regulation of gluconeogenesis, with corresponding changes of hepatic glucose production in Fao cells. We suggest that by functioning as a FoxO1 E3 ligase, COP1 may play a role in the regulation of hepatic glucose metabolism.
- Published
- 2008
- Full Text
- View/download PDF
37. STAT6 Inhibits TGF-beta1-mediated Foxp3 induction through direct binding to the Foxp3 promoter, which is reverted by retinoic acid receptor.
- Author
-
Takaki H, Ichiyama K, Koga K, Chinen T, Takaesu G, Sugiyama Y, Kato S, Yoshimura A, and Kobayashi T
- Subjects
- Animals, Antibodies pharmacology, Cells, Cultured, Chromatin Assembly and Disassembly immunology, Forkhead Transcription Factors genetics, Forkhead Transcription Factors immunology, Hypersensitivity immunology, Hypersensitivity metabolism, Hypersensitivity pathology, Immune Tolerance genetics, Interleukin-4 immunology, Interleukin-4 metabolism, Interleukin-4 pharmacology, Mice, Mice, Knockout, Receptors, Retinoic Acid genetics, Receptors, Retinoic Acid immunology, STAT6 Transcription Factor genetics, STAT6 Transcription Factor immunology, T-Lymphocytes, Regulatory immunology, T-Lymphocytes, Regulatory pathology, Th2 Cells immunology, Th2 Cells pathology, Transforming Growth Factor beta1 immunology, Forkhead Transcription Factors metabolism, Promoter Regions, Genetic immunology, Receptors, Retinoic Acid metabolism, STAT6 Transcription Factor metabolism, T-Lymphocytes, Regulatory metabolism, Th2 Cells metabolism, Transforming Growth Factor beta1 pharmacology
- Abstract
It has been shown that transforming growth factor beta1 (TGF-beta1) is critical in the generation of CD4(+)CD25(+)Foxp3(+)-inducible regulatory T cells (iTregs) from naïve CD4(+)T cells. However, in contrast to natural Tregs, TGF-beta1-induced iTregs rapidly lose both Foxp3 expression and suppression activity. We found that TGF-beta1-induced Foxp3 levels were maintained by the addition of the anti-interleukin 4 (IL-4) antibody or by STAT6 gene deletion. Thus, IL-4 is an important suppressor of Foxp3 induction, and T helper 2 development is a major cause for the disappearance of iTreg during long culture. Using promoter analysis in EL4 cells and primary T cells, we identified a silencer region containing a STAT6 binding site. STAT6 binding to this site reduced TGF-beta1-mediated Foxp3 promoter activation and chromatin modification. Retinoic acid has also been shown to suppress loss of Foxp3 induced by TGF-beta1. Retinoic acid in the presence of TGF-beta1 reduced STAT6 binding to the Foxp3 promoter and enhanced histone acetylation, thereby reverting the effect of IL-4. We propose that antagonistic agents for neutralizing IL-4 could be a novel strategy to facilitate inducible Treg cell generation and the promotion of tolerance in Th2-dominated diseases such as allergy.
- Published
- 2008
- Full Text
- View/download PDF
38. A2E, a pigment of the lipofuscin of retinal pigment epithelial cells, is an endogenous ligand for retinoic acid receptor.
- Author
-
Iriyama A, Fujiki R, Inoue Y, Takahashi H, Tamaki Y, Takezawa S, Takeyama K, Jang WD, Kato S, and Yanagi Y
- Subjects
- Cell Line, Epithelial Cells drug effects, Gene Expression Regulation drug effects, Humans, Ligands, Neovascularization, Physiologic drug effects, Pigment Epithelium of Eye drug effects, Pyridinium Compounds pharmacology, Retinal Pigments pharmacology, Retinoids pharmacology, Vascular Endothelial Growth Factor A genetics, Epithelial Cells metabolism, Lipofuscin metabolism, Pigment Epithelium of Eye cytology, Pigment Epithelium of Eye metabolism, Pyridinium Compounds metabolism, Receptors, Retinoic Acid metabolism, Retinal Pigments metabolism, Retinoids metabolism
- Abstract
Lipofuscin contains fluorophores, which represent a biomarker for cellular aging. Although it remains unsubstantiated clinically, experimental results support that the accumulation of lipofuscin is related to an increased risk of choroidal neovascularization due to age-related macular degeneration, a leading cause of legal blindness. Here, we report that a major lipofuscin component, A2E, activates the retinoic acid receptor (RAR). In vitro experiments using luciferase reporter assay, competitional binding assay, analysis of target genes, and chromatin immunoprecipitation (ChIP) assay strongly suggest that A2E is a bona fide ligand for RAR and induces sustained activation of RAR target genes. A2E-induced vascular endothelial growth factor (VEGF) expression in a human retinal pigment epithelial cell line (ARPE-19) and RAR antagonist blocked the up-regulation of VEGF. The conditioned medium of A2E-treated ARPE-19 cells induced tube formation in human umbilical vascular endothelial cells, which was blocked by the RAR antagonist and anti-VEGF antibody. These results suggest that A2E accumulation results in the phenotypic alteration of retinal pigment epithelial cells, predisposing the environment to choroidal neovascularization development. This is mediated through the agonistic function of A2E, at least in part. The results of this study provide a novel potential therapeutic target for this incurable condition.
- Published
- 2008
- Full Text
- View/download PDF
39. Role of the epsilon subunit of thermophilic F1-ATPase as a sensor for ATP.
- Author
-
Kato S, Yoshida M, and Kato-Yamada Y
- Subjects
- Adenosine Triphosphatases chemistry, Alanine chemistry, Amino Acid Sequence, Binding Sites, Catalytic Domain, Chromatography, Gel, Copper chemistry, Escherichia coli metabolism, Molecular Conformation, Molecular Sequence Data, Protein Binding, Protein Conformation, Protein Structure, Secondary, Adenosine Triphosphate chemistry, Bacillus enzymology, Proton-Translocating ATPases chemistry
- Abstract
The epsilon subunit of F(1)-ATPase from the thermophilic Bacillus PS3 (TF(1)) has been shown to bind ATP. The precise nature of the regulatory role of ATP binding to the epsilon subunit remains to be determined. To address this question, 11 mutants of the epsilon subunit were prepared, in which one of the basic or acidic residues was substituted with alanine. ATP binding to these mutants was tested by gel-filtration chromatography. Among them, four mutants that showed no ATP binding were selected and reconstituted with the alpha(3)beta(3)gamma complex of TF(1). The ATPase activity of the resulting alpha(3)beta(3)gammaepsilon complexes was measured, and the extent of inhibition by the mutant epsilon subunits was compared in each case. With one exception, weaker binding of ATP correlated with greater inhibition of ATPase activity. These results clearly indicate that ATP binding to the epsilon subunit plays a regulatory role and that ATP binding may stabilize the ATPase-active form of TF(1) by fixing the epsilon subunit into the folded conformation.
- Published
- 2007
- Full Text
- View/download PDF
40. Oxidative modification to cysteine sulfonic acid of Cys111 in human copper-zinc superoxide dismutase.
- Author
-
Fujiwara N, Nakano M, Kato S, Yoshihara D, Ookawara T, Eguchi H, Taniguchi N, and Suzuki K
- Subjects
- Amyotrophic Lateral Sclerosis genetics, Amyotrophic Lateral Sclerosis metabolism, Animals, Blotting, Western, Cysteine chemistry, Cysteine genetics, Cysteine metabolism, Humans, Hydrogen Peroxide chemistry, Lewy Bodies chemistry, Lewy Bodies genetics, Lewy Bodies metabolism, Mass Spectrometry, Mercaptoethanol chemistry, Mice, Mutation, Oxidation-Reduction, Oxidative Stress genetics, Recombinant Proteins chemistry, Recombinant Proteins genetics, Recombinant Proteins metabolism, Superoxide Dismutase genetics, Superoxide Dismutase metabolism, Superoxide Dismutase-1, Cysteine analogs & derivatives, Superoxide Dismutase chemistry
- Abstract
Copper-zinc superoxide dismutase (SOD1) plays a protective role against oxidative stress. On the other hand, recent studies suggest that SOD1 itself is a major target of oxidative damage and has its own pathogenicity in various neurodegenerative diseases, including familial amyotrophic lateral sclerosis. Only human and great ape SOD1s among mammals have the highly reactive free cysteine residue, Cys(111), at the surface of the SOD1 molecule. The purpose of this study was to investigate the role of Cys(111) in the oxidative damage of the SOD1 protein, by comparing the oxidative susceptibility of recombinant human SOD1 modified with 2-mercaptoethanol at Cys(111) (2-ME-SOD1) to wild-type SOD1. Wild-type SOD1 was more sensitive to oxidation by hydrogen peroxide-generating fragments, oligomers, and charge isomers compared with 2-ME-SOD1. Moreover, wild-type SOD1, but not 2-ME-SOD1, generated an upper shifted band in reducing SDS-PAGE even by air oxidation. Using mass spectrometry and limited proteolysis, this upper band was identified as an oxidized subunit of SOD1; the sulfhydryl group (Cys-SH) of Cys(111) was selectively oxidized to cysteine sulfinic acid (Cys-SO(2)H) and to cysteine sulfonic acid (Cys-SO(3)H). The antibody raised against a synthesized peptide containing Cys(111)-SO(3)H reacted with only the Cys(111)-peroxidized SOD1 by Western blot analysis and labeled Lewy body-like hyaline inclusions and vacuole rims in the spinal cord of human SOD1-mutated amyotrophic lateral sclerosis mice by immunohistochemical analysis. These results suggest that Cys(111) is a primary target for oxidative modification and plays an important role in oxidative damage to human SOD1, including familial amyotrophic lateral sclerosis mutants.
- Published
- 2007
- Full Text
- View/download PDF
41. Insulin-like growth factor 1/insulin signaling activates androgen signaling through direct interactions of Foxo1 with androgen receptor.
- Author
-
Fan W, Yanase T, Morinaga H, Okabe T, Nomura M, Daitoku H, Fukamizu A, Kato S, Takayanagi R, and Nawata H
- Subjects
- Animals, COS Cells, Cell Line, Tumor, Cell Proliferation, Chlorocebus aethiops, Dihydrotestosterone pharmacology, Forkhead Box Protein O1, Forkhead Transcription Factors chemistry, Humans, Male, PTEN Phosphohydrolase physiology, Phosphatidylinositol 3-Kinases physiology, Promoter Regions, Genetic, Prostate-Specific Antigen genetics, Prostatic Neoplasms pathology, Repressor Proteins physiology, Transcriptional Activation, Forkhead Transcription Factors physiology, Insulin physiology, Insulin-Like Growth Factor I physiology, Prostatic Neoplasms metabolism, Receptors, Androgen genetics, Signal Transduction physiology
- Abstract
The androgen-androgen receptor (AR) system plays vital roles in a wide array of biological processes, including prostate cancer development and progression. Several growth factors, such as insulin-like growth factor 1 (IGF1), can induce AR activation, whereas insulin resistance and hyperinsulinemia are correlated with an elevated incidence of prostate cancer. Here we report that Foxo1, a downstream molecule that becomes phosphorylated and inactivated by phosphatidylinositol 3-kinase/Akt kinase in response to IGF1 or insulin, suppresses ligand-mediated AR transactivation. Foxo1 reduces androgen-induced AR target gene expressions and suppresses the in vitro growth of prostate cancer cells. These inhibitory effects of Foxo1 are attenuated by IGF1 but are enhanced when it is rendered Akt-nonphosphorylatable. Foxo1 interacts directly with the C terminus of AR in a ligand-dependent manner and disrupts ligand-induced AR subnuclear compartmentalization. Foxo1 is recruited by liganded AR to the chromatin of AR target gene promoters, where it interferes with AR-DNA interactions. IGF1 or insulin abolish the Foxo1 occupancy of these promoters. Of interest, a positive feedback circuit working locally in an autocrine/intracrine manner may exist, because liganded AR up-regulates IGF1 receptor expression in prostate cancer cells, presumably resulting in higher IGF1 signaling tension and further enhancing the functions of the receptor itself. Thus, Foxo1 is a novel corepressor for AR, and IGF1/insulin signaling may confer stimulatory effects on AR by attenuating Foxo1 inhibition. These results highlight the potential involvement of metabolic syndrome and hyperinsulinemia in prostate diseases and further suggest that intervention of IGF1/insulin-phosphatidylinositol 3-kinase-Akt signaling may be of clinical value for prostate diseases.
- Published
- 2007
- Full Text
- View/download PDF
42. Coactivation of the N-terminal transactivation of mineralocorticoid receptor by Ubc9.
- Author
-
Yokota K, Shibata H, Kurihara I, Kobayashi S, Suda N, Murai-Takeda A, Saito I, Kitagawa H, Kato S, Saruta T, and Itoh H
- Subjects
- Aldosterone metabolism, Animals, COS Cells, Cell Line, Chlorocebus aethiops, Glutathione Transferase metabolism, Humans, Kidney metabolism, Mice, Protein Binding, Protein Structure, Tertiary, Two-Hybrid System Techniques, Receptors, Mineralocorticoid metabolism, Transcriptional Activation, Ubiquitin-Conjugating Enzymes chemistry, Ubiquitin-Conjugating Enzymes physiology
- Abstract
Molecular mechanisms underlying mineralocorticoid receptor (MR)-mediated gene expression are not fully understood. Various transcription factors are post-translationally modified by small ubiquitin-related modifier-1 (SUMO-1). We investigated the role of the SUMO-1-conjugating enzyme Ubc9 in MR transactivation. Yeast two-hybrid, GST-pulldown, and coimmunoprecipitation assays showed that Ubc9 interacted with N-terminal MR-(1-670). Endogenous Ubc9 is associated with stably expressing MR in 293-MR cells. Transient transfection assays in COS-1 cells showed that Ubc9 increased MR transactivation of reporter constructs containing MRE, ENaC, or MMTV promoter in a hormone-sensitive manner. Moreover, reduction of Ubc9 protein levels by small interfering RNA attenuated hormonal activation of a reporter construct as well as an endogenous target gene by MR. A sumoylation-inactive mutant Ubc9(C93S) similarly interacted with MR and potentiated aldosterone-dependent MR transactivation. An MR mutant in which four lysine residues within sumoylation motifs were mutated into arginine (K89R/K399R/K494R/K953R) failed to be sumoylated, but Ubc9 similarly enhanced transactivation by the mutant MR, indicating that sumoylation activity is dispensable for coactivation capacity of Ubc9. Coexpression of Ubc9 and steroid receptor coactivator-1 (SRC-1) synergistically enhanced MR-mediated transactivation in transient transfection assays. Indeed, chromatin immunoprecipitation assays demonstrated that endogenous MR, Ubc9, and SRC-1 were recruited to an endogenous ENaC gene promoter in a largely aldosterone-dependent manner. Coimmunoprecipitation assays showed a complex of MR, Ubc9, and SRC-1 in mammalian cells, and the endogenous proteins were colocalized in the nuclei of the mouse collecting duct cells. These findings support a physiological role of Ubc9 as a transcriptional MR coactivator, beyond the known SUMO E2-conjugating enzyme.
- Published
- 2007
- Full Text
- View/download PDF
43. An hGCN5/TRRAP histone acetyltransferase complex co-activates BRCA1 transactivation function through histone modification.
- Author
-
Oishi H, Kitagawa H, Wada O, Takezawa S, Tora L, Kouzu-Fujita M, Takada I, Yano T, Yanagisawa J, and Kato S
- Subjects
- Adaptor Proteins, Signal Transducing, BRCA1 Protein genetics, Breast Neoplasms genetics, Cell Cycle Proteins genetics, Cell Line, Tumor, Female, Gene Expression Regulation, Neoplastic, Histone Acetyltransferases genetics, Humans, Multienzyme Complexes metabolism, Nuclear Proteins genetics, Transcription Factors genetics, Transcription, Genetic physiology, p300-CBP Transcription Factors, BRCA1 Protein metabolism, Breast Neoplasms metabolism, Cell Cycle Proteins metabolism, Histone Acetyltransferases metabolism, Histones metabolism, Nuclear Proteins metabolism, Transcription Factors metabolism
- Abstract
It is well established that genetic mutations that impair BRCA1 function predispose women to early onset of breast and ovarian cancer. However, the co-regulatory factors that support normal BRCA1 functions remain to be identified. Using a biochemical approach to search for such co-regulatory factors, we identified hGCN5, TRRAP, and hMSH2/6 as BRCA1-interacting proteins. Genetic mutations in the C-terminal transactivation domain of BRCA1, as found in breast cancer patients (Chapman, M. S., and Verma, I. M. (1996) Nature 382, 678-679), caused the loss of physical interaction between BRCA1 and TRRAP and significantly reduced the co-activation of BRCA1 transactivation function by hGCN5/TRRAP. The reported transcriptional squelching between BRCA1 and estrogen receptor alpha (Fan, S., Wang, J., Yuan, R., Ma, Y., Meng, Q., Erdos, M. R., Pestell, R. G., Yuan, F., Auborn, K. J., Goldberg, I. D., and Rosen, E. M. (1999) Science 284, 1354-1356) was rescued by the overexpression of TRRAP or hGCN5. Histone acetyltransferase hGCN5 activity appeared to be indispensable for coregulator complex function in both BRCA1-mediated gene regulation and DNA repair. Biochemical purification of the hGCN5/TRRAP-containing complex suggested that hGCN5/TRRAP formed a complex with hMSH2/hMSH6, presumably as a novel subclass of hGCN5/TRRAP-containing known TFTC (TBP-free TAF-containing)-type histone acetyltransferase complex (hTFTC, hPCAF, and hSTAGA) (Yanagisawa, J., Kitagawa, H., Yanagida, M., Wada, O., Ogawa, S., Nakagomi, M., Oishi, H., Yamamoto, Y., Nagasawa, H., McMahon, S. B., Cole, M. D., Tora, L., Takahashi, N., and Kato, S. (2002) Mol. Cell 9, 553-562). Unlike other subclasses, the isolated complex harbored a previously unknown combination of components including hMSH2 and hMSH6, major components of the BRCA1 genome surveillance repair complex (BASC). Thus, our results suggested that the multiple BRCA1 functions require a novel hGCN5/TRRAP histone acetyltransferase complex subclass.
- Published
- 2006
- Full Text
- View/download PDF
44. Identification of the amino acid residue of CYP27B1 responsible for binding of 25-hydroxyvitamin D3 whose mutation causes vitamin D-dependent rickets type 1.
- Author
-
Yamamoto K, Uchida E, Urushino N, Sakaki T, Kagawa N, Sawada N, Kamakura M, Kato S, Inouye K, and Yamada S
- Subjects
- Amino Acid Sequence, Animals, Blotting, Western, Cholecalciferol chemistry, Escherichia coli metabolism, Glutamine chemistry, Humans, Hydrogen Bonding, Kinetics, Mice, Models, Chemical, Models, Molecular, Molecular Sequence Data, Mutation, Oligonucleotides chemistry, Plasmids metabolism, Point Mutation, Polymerase Chain Reaction, Protein Binding, Protein Conformation, Protein Folding, Protein Structure, Tertiary, Recombinant Proteins chemistry, Sequence Homology, Amino Acid, Spectrophotometry, Substrate Specificity, Threonine chemistry, Time Factors, 25-Hydroxyvitamin D3 1-alpha-Hydroxylase chemistry, Calcifediol metabolism, Rickets genetics, Vitamin D metabolism
- Abstract
We previously reported the three-dimensional structure of human CYP27B1 (25-hydroxyvitamin D3 1alpha-hydroxylase) constructed by homology modeling. Using the three-dimensional model we studied the docking of the substrate, 25-hydroxyvitamin D3, into the substrate binding pocket of CYP27B1. In this study, we focused on the amino acid residues whose point mutations cause vitamin D-dependent rickets type 1, especially unconserved residues among mitochondrial CYPs such as Gln65 and Thr409. Recently, we successfully overexpressed mouse CYP27B1 by using a GroEL/ES co-expression system. In a mutation study of mouse CYP27B1 that included spectroscopic analysis, we concluded that in a 1alpha-hydroxylation process, Ser408 of mouse CYP27B1 corresponding to Thr409 of human CYP27B1 forms a hydrogen bond with the 25-hydroxyl group of 25-hydroxyvitamin D3. This is the first report that shows a critical amino acid residue recognizing the 25-hydroxyl group of the vitamin D3.
- Published
- 2005
- Full Text
- View/download PDF
45. Androgen receptor gene knockout male mice exhibit impaired cardiac growth and exacerbation of angiotensin II-induced cardiac fibrosis.
- Author
-
Ikeda Y, Aihara K, Sato T, Akaike M, Yoshizumi M, Suzaki Y, Izawa Y, Fujimura M, Hashizume S, Kato M, Yagi S, Tamaki T, Kawano H, Matsumoto T, Azuma H, Kato S, and Matsumoto T
- Subjects
- Animals, Blood Pressure, DNA-Binding Proteins metabolism, Fibrosis, Heart Rate, Male, Mice, Mice, Inbred C57BL, Mice, Inbred CBA, Mice, Knockout, Mitogen-Activated Protein Kinase 1 metabolism, Mitogen-Activated Protein Kinase 3 metabolism, Mitogen-Activated Protein Kinase 7 metabolism, RNA, Messenger analysis, Receptors, Androgen genetics, Smad2 Protein, Trans-Activators metabolism, Transforming Growth Factor beta genetics, Transforming Growth Factor beta1, Ventricular Remodeling, Angiotensin II pharmacology, Cardiomegaly etiology, Myocardium pathology, Receptors, Androgen physiology
- Abstract
Androgen has anabolic effects on cardiac myocytes and has been shown to enhance left ventricular enlargement and function. However, the physiological and patho-physiological roles of androgen in cardiac growth and cardiac stress-induced remodeling remains unclear. We aimed to clarify whether the androgen-nuclear androgen receptor (AR) system contributes to the cardiac growth and angiotensin II (Ang II)-stimulated cardiac remodeling by using systemic AR-null male mice. AR knock-out (ARKO) male mice, at 25 weeks of age, and age-matched wild-type (WT) male mice were treated with or without Ang II stimulation (2.0 mg/kg/day) for 2 weeks. ARKO mice with or without Ang II stimulation showed a significant reduction in the heart-to-body weight ratio compared with those of WT mice. In addition, echocardiographic analysis demonstrated impairments of both the concentric hypertrophic response and left ventricular function in Ang II-stimulated ARKO mice. Western blot analysis of the myocardium revealed that activation of extracellular signal-regulated kinases (ERK) 1/2 and ERK5 by Ang II stimulation were lower in ARKO mice than those of WT mice. Ang II stimulation caused more prominent cardiac fibrosis in ARKO mice than in WT mice with enhanced expression of types I and III collagen and transforming growth factor-beta1 genes and with increased Smad2 activation. These results suggest that, in male mice, the androgen-AR system participates in normal cardiac growth and modulates cardiac adaptive hypertrophy and fibrosis during the process of cardiac remodeling under hypertrophic stress.
- Published
- 2005
- Full Text
- View/download PDF
46. Circulating FGF-23 is regulated by 1alpha,25-dihydroxyvitamin D3 and phosphorus in vivo.
- Author
-
Saito H, Maeda A, Ohtomo S, Hirata M, Kusano K, Kato S, Ogata E, Segawa H, Miyamoto K, and Fukushima N
- Subjects
- Animal Feed, Animals, Calcium blood, Creatinine blood, Dose-Response Relationship, Drug, Enzyme-Linked Immunosorbent Assay, Feedback, Physiological, Fibroblast Growth Factor-23, Humans, Male, Mice, Mice, Knockout, Mice, Transgenic, Phosphates metabolism, Phosphorus blood, Rats, Rats, Sprague-Dawley, Thyroid Gland metabolism, Calcitriol metabolism, Fibroblast Growth Factors blood, Gene Expression Regulation, Phosphorus metabolism
- Abstract
Fibroblast growth factor-23 (FGF-23), a novel phosphate-regulating factor, was elevated in hypophosphatemic patients with X-linked hypophosphatemic rickets/osteomalacia and also in patients with chronic kidney disease. These observations suggested the pathophysiological importance of FGF-23 on phosphate homeostasis. However, regulation of FGF-23 production is still unclear. We investigated effects of both dietary phosphorus and 1alpha,25-dihydroxyvitamin D(3) (1alpha,25(OH)(2)D(3)) on circulating FGF-23 in vivo Administration of. 1alpha,25(OH)(2)D(3) dose-dependently increased serum FGF-23 in thyroparathyroidectomized rats without correlating with serum inorganic phosphorus or serum parathyroid hormone. On the other hand, vitamin D receptor null mice had very low serum FGF-23 and did not respond to the 1alpha,25(OH)(2)D(3) administration. These observations suggested 1alpha,25(OH)(2)D(3) directly or indirectly regulates circulating FGF-23. Serum FGF-23 had a strong correlation with serum inorganic phosphorus controlled by dietary phosphorus in 5/6 nephrectomized rats. High phosphate diet elicited a 5-fold increase in serum FGF-23 compared with sham-operated rats, whereas serum FGF-23 did not correlate with serum calcium or serum creatinine in 5/6 nephrectomized rats. Administration of 1alpha,25-dihydroxyvitamin D(3) also elicited a severalfold increase in serum FGF-23 in the uremic rats. Taken together, this shows that both serum phosphorus and 1alpha,25(OH)(2)D(3) regulate circulating FGF-23 independent of each other. Therefore, we proposed there was a feedback loop existing among serum phosphorus, 1alpha,25(OH)(2)D(3), and FGF-23, in which the novel phosphate-regulating bone-kidney axis integrated with the parathyroid hormone-vitamin D(3) axis in regulating phosphate homeostasis.
- Published
- 2005
- Full Text
- View/download PDF
47. Catalase reaction by myoglobin mutants and native catalase: mechanistic investigation by kinetic isotope effect.
- Author
-
Kato S, Ueno T, Fukuzumi S, and Watanabe Y
- Subjects
- Animals, Binding Sites, Cattle, Deuterium Oxide, In Vitro Techniques, Kinetics, Micrococcus enzymology, Models, Biological, Models, Molecular, Mutation, Myoglobin chemistry, Oxygen Isotopes, Recombinant Proteins chemistry, Recombinant Proteins genetics, Recombinant Proteins metabolism, Spectrophotometry, Thermodynamics, Whales, Catalase metabolism, Myoglobin genetics, Myoglobin metabolism
- Abstract
The catalase reaction has been studied in detail by using myoglobin (Mb) mutants. Compound I of Mb mutants (Mb-I), a ferryl species (Fe(IV)=O) paired with a porphyrin radical cation, is readily prepared by the reaction with a nearly stoichiometric amount of m-chloroperbenzoic acid. Upon the addition of H2O2 to an Mb-I solution, Mb-I is reduced back to the ferric state without forming any intermediates. This indicates that Mb-I is capable of performing two-electron oxidation of H2O2 (catalatic reaction). Gas chromatography-mass spectroscopy analysis of the evolved O2 from a 50:50 mixture of H2(18)O2/H2(16)O2 solution containing H64D or F43H/H64L Mb showed the formation of 18O2 (m/e = 36) and 16O2 (m/e = 32) but not 16O18O (m/e = 34). This implies that O2 is formed by two-electron oxidation of H2O2 without breaking the O-O bond. Deuterium isotope effects on the catalatic reactions of Mb mutants and catalase suggest that the catalatic reactions of Micrococcus lysodeikticus catalase and F43H/H64L Mb proceed via an ionic mechanism with a small isotope effect of less than 4.0, since the distal histidine residue is located at a proper position to act as a general acid-base catalyst for the ionic reaction. In contrast, other Mb mutants such as H64X (X is Ala, Ser, and Asp) and L29H/H64L Mb oxidize H2O2 via a radical mechanism in which a hydrogen atom is abstracted by Mb-I with a large isotope effect in a range of 10-29, due to a lack of the general acid-base catalyst.
- Published
- 2004
- Full Text
- View/download PDF
48. Histone deacetylase 7 associates with hypoxia-inducible factor 1alpha and increases transcriptional activity.
- Author
-
Kato H, Tamamizu-Kato S, and Shibasaki F
- Subjects
- Acetyltransferases metabolism, Active Transport, Cell Nucleus, Animals, Cell Cycle Proteins metabolism, Glucose Transporter Type 1, Histone Acetyltransferases, Histone Deacetylases physiology, Humans, Hypoxia-Inducible Factor 1, alpha Subunit, Monosaccharide Transport Proteins genetics, Protein Binding, Rats, Repressor Proteins, Transcription, Genetic, Up-Regulation, Vascular Endothelial Growth Factor A genetics, p300-CBP Transcription Factors, Histone Deacetylases metabolism, Hypoxia genetics, Transcription Factors metabolism, Transcriptional Activation
- Abstract
Hypoxia-inducible factor (HIF)-1alpha is a transcription factor that controls expression of genes responsive to low oxygen tension, including vascular endothelial growth factor (VEGF), erythropoietin, and glycolytic enzymes. The activity of HIF-1alpha is regulated by binding to the transcriptional co-activator cAMP-response element-binding protein-binding protein (CBP)/p300. Using the yeast two-hybrid screening system, we found that the inhibitory domain of HIF-1alpha strongly interacted with the C-terminal domain of histone deacetylase (HDAC) 7. The o-nitrophenyl beta-d-galactopyranoside assay revealed that regions containing amino acids 735-785 of HIF-1alpha and amino acids 669-952 of HDAC7 were minimum contact sites of the interaction. The binding of HDAC7 with HIF-1alpha was reproduced in HEK293 cells grown under normoxic and hypoxic conditions (2% O(2)). HDAC7 bound solely to HIF-1alpha among other HIF-alpha family members, including HIF-2alpha and HIF-3alpha, whereas HIF-1alpha only interacted with HDAC7 in the class II HDAC family. Although HDAC7 was localized dominantly in the cytoplasm at normal oxygen concentrations, HDAC7 co-translocated to the nucleus with HIF-1alpha under hypoxic conditions. In the nucleus, HDAC7 increased transcriptional activity of HIF-1alpha through the formation of a complex with HIF-1alpha, HDAC7, and p300. Taken together, these results indicate that HDAC7 is a novel transcriptional activator of HIF-1alpha
- Published
- 2004
- Full Text
- View/download PDF
49. Wnt/beta-catenin and estrogen signaling converge in vivo.
- Author
-
Kouzmenko AP, Takeyama K, Ito S, Furutani T, Sawatsubashi S, Maki A, Suzuki E, Kawasaki Y, Akiyama T, Tabata T, and Kato S
- Subjects
- Animals, Apoptosis, Cell Line, Tumor, Chromatin metabolism, Cytoskeletal Proteins genetics, Drosophila, Estrogen Receptor alpha, Genes, Reporter, Humans, Immunoblotting, In Situ Nick-End Labeling, Mutation, Precipitin Tests, Promoter Regions, Genetic, Protein Binding, Receptors, Estrogen metabolism, Signal Transduction, Trans-Activators genetics, Transcription, Genetic, Transcriptional Activation, Transfection, Transgenes, beta Catenin, Cytoskeletal Proteins metabolism, Estrogens metabolism, Trans-Activators metabolism
- Abstract
Wnt and estrogen signaling represent important regulatory pathways, each controlling a wide range of biological processes. While an increasing number of observations suggest potential convergence between these pathways, no direct evidence of their functional interaction has been reported. Using human colon and breast cancer cells, we found that estrogen receptor (ER) alpha- and beta-catenin precipitated within the same immunocomplexes, reciprocally enhanced the transactivation of cognate reporter genes, and were reciprocally recruited to cognate response elements in the promoters of endogenous target genes. Using transgenic Drosophila that ectopically expressed human ERalpha alone or together with metabolically stable beta-catenin/Armadillo mutants, we demonstrated genetic interaction between these signal transducers in vivo. Thus, we present here the first direct evidence of cross-talk between Wnt and estrogen signaling pathways via functional interaction between beta-catenin and ERalpha., (Copyright 2004 American Society for Biochemistry and Molecular Biology, Inc.)
- Published
- 2004
- Full Text
- View/download PDF
50. Disruption of nuclear vitamin D receptor gene causes enhanced thrombogenicity in mice.
- Author
-
Aihara K, Azuma H, Akaike M, Ikeda Y, Yamashita M, Sudo T, Hayashi H, Yamada Y, Endoh F, Fujimura M, Yoshida T, Yamaguchi H, Hashizume S, Kato M, Yoshimura K, Yamamoto Y, Kato S, and Matsumoto T
- Subjects
- Animals, Blood Coagulation physiology, Calcium administration & dosage, Calcium metabolism, Diet, Down-Regulation, Gene Deletion, Gene Expression Regulation, Mice, Mice, Knockout, Nitric Oxide Synthase biosynthesis, Nitric Oxide Synthase Type II, Nitric Oxide Synthase Type III, Organ Specificity, Receptors, Calcitriol metabolism, Thrombin biosynthesis, Thrombomodulin biosynthesis, Blood Coagulation genetics, Receptors, Calcitriol genetics
- Abstract
Vitamin D metabolites influence the expression of various genes involved in calcium homeostasis, cell differentiation, and regulation of the immune system. Expression of these genes is mediated by the activation of the nuclear vitamin D receptor (VDR). Previous studies have shown that a hormonally active form of vitamin D, 1alpha,25-dihydroxyvitamin D3, exerts anticoagulant effects in cultured monocytic cells. To clarify whether activation of VDR plays any antithrombotic actions in vivo, hemostatic/thrombogenic systems were examined in normocalcemic VDR knock-out (KO) mice on a high calcium diet and compared with wild type and hypocalcemic VDRKO mice that were fed a regular diet. Platelet aggregation was enhanced significantly in normocalcemic VDRKO mice compared with wild type and hypocalcemic VDRKO mice. Aortic endothelial nitric-oxide (NO) synthase expression and urinary NOx excretions were reduced in hypocalcemic VDRKO mice, but not in normocalcemic VDRKO mice. Northern blot and RT-PCR analyses revealed that the gene expression of antithrombin in the liver as well as that of thrombomodulin in the aorta, liver and kidney was down-regulated in hypo- and normocalcemic VDRKO mice. Whereas tissue factor mRNA expression in the liver and kidney was up-regulated in VDRKO mice regardless of plasma calcium level. Furthermore, VDRKO mice manifested an exacerbated multi-organ thrombus formation after exogenous lipopolysaccharide injection regardless of the calcemic conditions. These results demonstrate that activation of nuclear VDR elicits antithrombotic effects in vivo, and suggest that the VDR system may play a physiological role in the maintenance of antithrombotic homeostasis.
- Published
- 2004
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.