79 results on '"Itoh H"'
Search Results
2. Expression and analysis of Gs alpha mutants with decreased ability to activate adenylylcyclase
- Author
-
Itoh, H., primary and Gilman, A.G., additional
- Published
- 1991
- Full Text
- View/download PDF
3. Characterization of the human gene for Gx alpha, a pertussis toxin-insensitive regulatory GTP-binding protein.
- Author
-
Matsuoka, M, primary, Itoh, H, additional, and Kaziro, Y, additional
- Published
- 1990
- Full Text
- View/download PDF
4. Purine but not pyrimidine nucleotides support rotation of F(1)-ATPase.
- Author
-
Noji, H, Bald, D, Yasuda, R, Itoh, H, Yoshida, M, and Kinosita, K
- Abstract
The binding change model for the F(1)-ATPase predicts that its rotation is intimately correlated with the changes in the affinities of the three catalytic sites for nucleotides. If so, subtle differences in the nucleotide structure may have pronounced effects on rotation. Here we show by single-molecule imaging that purine nucleotides ATP, GTP, and ITP support rotation but pyrimidine nucleotides UTP and CTP do not, suggesting that the extra ring in purine is indispensable for proper operation of this molecular motor. Although the three purine nucleotides were bound to the enzyme at different rates, all showed similar rotational characteristics: counterclockwise rotation, 120 degrees steps each driven by hydrolysis of one nucleotide molecule, occasional back steps, rotary torque of approximately 40 piconewtons (pN).nm, and mechanical work done in a step of approximately 80 pN.nm. These latter characteristics are likely to be determined by the rotational mechanism built in the protein structure, which purine nucleotides can energize. With ATP and GTP, rotation was observed even when the free energy of hydrolysis was -80 pN.nm/molecule, indicating approximately 100% efficiency. Reconstituted F(o)F(1)-ATPase actively translocated protons by hydrolyzing ATP, GTP, and ITP, but CTP and UTP were not even hydrolyzed. Isolated F(1) very slowly hydrolyzed UTP (but not CTP), suggesting possible uncoupling from rotation.
- Published
- 2001
- Full Text
- View/download PDF
5. G(i)-dependent activation of c-Jun N-terminal kinase in human embryonal kidney 293 cells.
- Author
-
Yamauchi, J, Kawano, T, Nagao, M, Kaziro, Y, and Itoh, H
- Abstract
Heterotrimeric G proteins stimulate the activities of two stress-activated protein kinases, c-Jun N-terminal kinase (JNK) and p38 mitogen-activated protein kinase in mammalian cells. In this study, we examined whether alpha subunits of G(i) family activate JNK using transient expression system in human embryonal kidney 293 cells. Constitutively activated mutants of Galpha(i1), Galpha(i2), and Galpha(i3) increased JNK activity. In contrast, constitutively activated Galpha(o) and Galpha(z) mutants did not stimulate JNK activity. To examine the mechanism of JNK activation by Galpha(i), kinase-deficient mutants of mitogen-activated protein kinase kinase 4 (MKK4) and 7 (MKK7), which are known to be JNK activators, were transfected into the cells. However, Galpha(i)-induced JNK activation was not blocked effectively by kinase-deficient MKK4 and MKK7. In addition, activated Galpha(i) mutant failed to stimulate MKK4 and MKK7 activities. Furthermore, JNK activation by Galpha(i) was inhibited by dominant-negative Rho and Cdc42 and tyrosine kinase inhibitors, but not dominant-negative Rac and phosphatidylinositol 3-kinase inhibitors. These results indicate that Galpha(i) regulates JNK activity dependent on small GTPases Rho and Cdc42 and on tyrosine kinase but not on MKK4 and MKK7.
- Published
- 2000
6. Requirement for protein-tyrosine phosphatase SHP-2 in insulin-induced activation of c-Jun NH(2)-terminal kinase.
- Author
-
Fukunaga, K, Noguchi, T, Takeda, H, Matozaki, T, Hayashi, Y, Itoh, H, and Kasuga, M
- Abstract
Mitogen-activated protein kinases, including extracellular signal-regulated kinases and c-Jun NH(2)-terminal kinases (JNKs), are activated by insulin. Although the mechanism by which the insulin receptor activates extracellular signal-regulated kinases is relatively well defined, the pathway that leads to JNK activation is poorly understood. Overexpression of a catalytically inactive mutant (SHP-2C/S) of the protein-tyrosine phosphatase SHP-2 in Rat-1 fibroblasts that also express human insulin receptors has now revealed that activation of JNKs by insulin and epidermal growth factor, but not that by anisomycin or sorbitol, requires SHP-2. A dominant negative mutant (RasN17) of Ha-Ras blocked insulin-induced JNK activation, whereas a dominant negative mutant (RacN17) of Rac1 or a specific inhibitor (LY294002) of phosphoinositide 3-kinase did not, indicating a role for Ras, but not for Rac or phosphoinositide 3-kinase, in this effect. SHP-2C/S markedly inhibited Ras activation in response to insulin without affecting insulin-induced tyrosine phosphorylation of cellular substrates or the dissociation of the Crk-p130(Cas) complex. In contrast, SHP-2C/S did not inhibit activation of JNKs induced by a constitutively active mutant (RasV12) of Ha-Ras. Furthermore, expression of myristoylated SOS, which functions as a potent activator of Ras, induced JNK activation even when SHP-2 was inactivated. These results suggest that SHP-2 contributes to JNK activation in response to insulin by positively regulating the Ras signaling pathway at the same level as, or upstream from, SOS.
- Published
- 2000
7. G protein betagamma subunits induce stress fiber formation and focal adhesion assembly in a Rho-dependent manner in HeLa cells.
- Author
-
Ueda, H, Itoh, H, Yamauchi, J, Morishita, R, Kaziro, Y, Kato, K, and Asano, T
- Abstract
In fibroblasts, the G protein alpha subunits Galpha(12) and Galpha(13) stimulate Rho-dependent stress fiber formation and focal adhesion assembly, whereas G protein betagamma subunits instead exert a disruptive influence. We show here that the latter can, however, stimulate the formation of stress fibers and focal adhesions in epithelial-like HeLa cells. Transient expression of beta(1) with gamma(2), gamma(5), gamma(7), and gamma(12) in quiescent HeLa cells induced stress fiber formation and focal adhesion assembly as did expression of the constitutively active Galpha(12). Co-expression of betagamma with Galpha(i2) and the C-terminal fragment of the beta-adrenergic receptor kinase, both of which are known to bind and sequester free betagamma, blocked betagamma-induced stress fiber and focal adhesion formation. Inhibition was also noted with co-expression of a dominant negative mutant of Rho. Botulinum C3 exoenzyme, which ADP-ribosylates and inactivates Rho, and a Rho-associated protein kinase inhibitor, Y-27632, similarly inhibited betagamma-induced stress fiber and focal adhesion assembly. These results indicate that G protein betagamma subunits regulate Rho-dependent actin polymerization in HeLa cells.
- Published
- 2000
8. Critical duration of intracellular Ca2+ response required for continuous translocation and activation of cytosolic phospholipase A2.
- Author
-
Hirabayashi, T, Kume, K, Hirose, K, Yokomizo, T, Iino, M, Itoh, H, and Shimizu, T
- Abstract
When cells are exposed to certain external stimuli, arachidonic acid (AA) is released from the membrane and serves as a precursor of various types of eicosanoids. A Ca2+-regulated cytosolic phospholipase A2 (cPLA2) plays a dominant role in the release of AA. To closely examine the relation between Ca2+ response and AA release by stimulation of G protein-coupled receptors, we established several lines of Chinese hamster ovary cells expressing platelet-activating factor receptor or leukotriene B4 receptor. Measurement of intracellular Ca2+ concentration ([Ca2+]i) demonstrated that cell lines capable of releasing AA elicited a sustained [Ca2+]i increase when stimulated by agonists. The prolonged [Ca2+]i elevation is the result of Ca2+ entry, because this elevation was blocked by EGTA treatment or in the presence of Ca2+ channel blockers (SKF 96365 and methoxyverapamil). cPLA2 fused with a green fluorescent protein (cPLA2-GFP) translocated from the cytosol to the perinuclear region in response to increases in [Ca2+]i. When EGTA was added shortly after [Ca2+]i increase, the cPLA2-GFP returned to the cytosol, without liberating AA. After a prolonged [Ca2+]i increase, even by EGTA treatment, the enzyme was not readily redistributed to the cytosol. Thus, we propose that a critical time length of [Ca2+]i elevation is required for continuous membrane localization and full activation of cPLA2.
- Published
- 1999
9. Phosphorylation of F-actin-associating G protein gamma12 subunit enhances fibroblast motility.
- Author
-
Ueda, H, Yamauchi, J, Itoh, H, Morishita, R, Kaziro, Y, Kato, K, and Asano, T
- Abstract
Eleven isoforms of G protein gamma subunit have been found thus far, but the precise roles of individual gamma subunits are not known. The gamma12 subunit has two unique properties: phosphorylation by protein kinase C and association with F-actin. To elucidate the role of gamma12, we overexpressed gamma12 and other gamma subunits in NIH 3T3 cells together with the beta1 subunit. The overexpressed gamma12 as well as endogenous gamma12, but not gamma2, gamma5, and gamma7 subunits, associated with cytoskeletal components. Expression of gamma12 induced remarkable changes including cell rounding, disruption of stress fibers, and enhancement of cell migration, but expression of other gamma subunits did not induce significant changes. Deletion of the N-terminal region of gamma12 decreased the abilities of gamma12 to associate with cytoskeletal fractions, to induce cell rounding, and to increase cell motility. Replacement by alanine of Ser2 of gamma12 (Ser1 of a mature gamma12 protein), a phosphorylation site for protein kinase C, eliminated these effects of gamma12, whereas a mutant in which Ser2 was replaced with glutamic acid showed effects equivalent to wild-type gamma12. These results indicate that phosphorylation of gamma12 at Ser2 enhances the motility of cells.
- Published
- 1999
10. Mammalian HSP60 is a major target for an immunosuppressant mizoribine.
- Author
-
Itoh, H, Komatsuda, A, Wakui, H, Miura, A B, and Tashima, Y
- Abstract
It has been reported that immunosuppressant cyclosporin A or FK506 binds to immunophilins in the cell and that these immunophilins make a complex with molecular chaperones HSP70 or HSP90. Although mizoribine has been used clinically as an immunosuppressant, immunophilins of the agent have not yet been fully understood. We investigated their specific binding proteins using mizoribine affinity column chromatography and porcine kidney cytosols. By increasing mizoribine in the eluant from the column, two major proteins (with molecular masses of 60 and 43 kDa) were detected by SDS-polyacrylamide gel electrophoresis. Based on the amino acid sequence analysis of these proteins, 60- and 43-kDa mizoribine-binding proteins were identified with HSP60 and cytosolic actin, respectively. A considerable amount of actin was also eluted from the affinity column by nucleotides, but a very low quantity of HSP60 was eluted under the same conditions. On the other hand, HSP60 was eluted as a major protein in the eluant that was eluted preferentially, with nucleotide followed by mizoribine. Actin was also detected in the eluant, but the quantity of the protein was very low. These results indicated that HSP60 has high affinity to mizoribine, and the interaction was also observed on surface plasmon resonance analysis. Although HSP60 or GroE facilitated refolding of citrate synthase in vitro, mizoribine interfered with the chaperone activity of HSP60. On different types of mizoribine affinity columns, HSP60 or actin recognized the NH(2) group of mizoribine, and this group may be a functional group of the agent.
- Published
- 1999
11. Differential regulation of mitogen-activated protein kinase kinase 4 (MKK4) and 7 (MKK7) by signaling from G protein beta gamma subunit in human embryonal kidney 293 cells.
- Author
-
Yamauchi, J, Kaziro, Y, and Itoh, H
- Abstract
Heterotrimeric G protein beta gamma subunit (Gbeta gamma) mediates signals to two types of stress-activated protein kinases, c-Jun NH2-terminal kinase (JNK) and p38 mitogen-activated protein kinase, in mammalian cells. To investigate the signaling mechanism whereby Gbeta gamma regulates the activity of JNK, we transfected kinase-deficient mutants of two JNK kinases, mitogen-activated protein kinase kinase 4 (MKK4) and 7 (MKK7), into human embryonal kidney 293 cells. Gbeta gamma-induced JNK activation was blocked by kinase-deficient MKK4 and to a lesser extent by kinase-deficient MKK7. Moreover, Gbeta gamma increased MKK4 activity by 6-fold and MKK7 activity by 2-fold. MKK4 activation by Gbeta gamma was blocked by dominant-negative Rho and Cdc42, whereas MKK7 activation was blocked by dominant-negative Rac. In addition, Gbeta gamma-mediated MKK4 activation, but not MKK7 activation, was inhibited completely by specific tyrosine kinase inhibitors PP2 and PP1. These results indicate that Gbeta gamma induces JNK activation mainly through MKK4 activation dependent on Rho, Cdc42, and tyrosine kinase, and to a lesser extent through MKK7 activation dependent on Rac.
- Published
- 1999
12. Presence of three distinct molecular species of Gi protein alpha subunit. Structure of rat cDNAs and human genomic DNAs.
- Author
-
Itoh, H, Toyama, R, Kozasa, T, Tsukamoto, T, Matsuoka, M, and Kaziro, Y
- Abstract
We have cloned a new species of rat Gi alpha (Gi3 alpha) cDNA and genomic DNAs for three distinct human Gi alpha proteins (Gi1 alpha, Gi2 alpha, and Gi3 alpha). Gi3 alpha cDNA codes for a protein of 354 amino acids (Mr 40,522) whose sequence is closely related but distinct from that of the previously isolated rat Gi alpha (Gi2 alpha). By screening the human genomic libraries with the two rat Gi alpha cDNAs as probes, clones encoding human Gi1 alpha, Gi2 alpha, and Gi3 alpha were isolated. The human Gi2 alpha and Gi3 alpha genes are composed of eight coding exons and seven introns and possess a completely identical exon-intron organization. Southern blot analysis indicates that a single copy of each Gi alpha gene is present per haploid human genome.
- Published
- 1988
- Full Text
- View/download PDF
13. Activation and maturation mechanisms of boar acrosin zymogen based on the deduced primary structure
- Author
-
Baba, T, Kashiwabara, S, Watanabe, K, Itoh, H, Michikawa, Y, Kimura, K, Takada, M, Fukamizu, A, and Arai, Y
- Abstract
We have isolated cDNA clones encoding boar acrosin, a serine protease participating in the initial stage of fertilization, from boar testis λgt11 cDNA libraries. Nucleotide sequencing of the overlapping clones indicates that the composite cDNA inserts contain 1,391 base pairs coding for a 5′-untranslated region, an open reading frame, a stop codon, a 3′-untranslated region, and a poly(A)+tail. A polyadenylation signal, AATAAA, is located 33 bases upstream from the start of the poly(A)+tail. The amino acid sequence deduced from the cDNAs shows that boar acrosin is initially synthesized as a prepro-protein with a 16-residue signal peptide at the NH2terminus. This signal sequence is followed by a 399-residue sequence corresponding to the acrosin zymogen. COOH-terminal sequence analysis of boar sperm 55-kDa proacrosin and its processed forms indicates that the mature acrosin molecule contains 322 amino acid residues in two polypeptide chains, a 23-residue light chain and a 299-residue heavy chain, with a combined molecular mass of 35,735 Da, and that the 55-kDa proacrosin molecule has 14-, 18-, and 43-residue segments as COOH-terminal extensions that are removed during proacrosin maturation. The COOH-terminal 43-residue segment is rich in proline residues, including an unusual repeat of 23 consecutive prolines. The deduced amino acid sequence of boar acrosin shows a high degree of identity with major portions of other serine proteases, including the active site region and the location of cysteine residues. We conclude that boar acrosin is synthesized as a single-chain polypeptide with the regions corresponding to the light and heavy chains covalently connected by two disulfide bonds, and that the single-chain molecule is autoactivated by cleavage of the Arg23-Val24bond after removal of the COOH-terminal 14-residue segment, resulting in the formation of the light and heavy chains. This two-chain molecule is then converted to the mature enzyme by removal of the COOH-terminal 18- and 43-residue segments.
- Published
- 1989
- Full Text
- View/download PDF
14. Alpha helix content of G protein alpha subunit is decreased upon activation by receptor mimetics.
- Author
-
Tanaka, T, Kohno, T, Kinoshita, S, Mukai, H, Itoh, H, Ohya, M, Miyazawa, T, Higashijima, T, and Wakamatsu, K
- Abstract
To elucidate the mechanism whereby liganded receptor molecules enhance nucleotide exchange of GTP-binding regulatory proteins (G proteins), changes in the secondary structure of the recombinant Gi1 alpha subunit (Gi1alpha) upon binding with receptor mimetics, compound 48/80 and mastoparan, were analyzed by circular dichroism spectroscopy. Compound 48/80 enhanced the initial rate of GTPgammaS binding to soluble Gi1alpha 2.6-fold with an EC50 of 30 microg/ml. With the same EC50, the mimetic decreased the magnitude of ellipticity, which is ascribed to a reduction in alpha helix content of the Gi1alpha by 7%. Likewise, mastoparan also enhanced the rate of GTPgammaS binding by 3.0-fold and decreased the magnitude of ellipticity of Gi1alpha similar to compound 48/80. In corresponding experiments using a K349P-Gi1alpha, a Gi1alpha counterpart of the unc mutant in Gsalpha in which Pro was substituted for Lys349, enhancement of the GTPgammaS binding rate by both activators was quite small. In addition, compound 48/80 showed a negligible effect on the circular dichroism spectrum of the mutant. On the other hand, a proteolytic fragment of Gi1alpha lacking the N-terminal 29 residues was activated and showed decreased ellipticity upon interaction with the compound, as did the wild-type Gi1alpha. Taken together, our results strongly suggest that the activator-induced unwinding of the alpha helix of the G protein alpha subunit is mechanically coupled to the enhanced release of bound GDP from the alpha subunit.
- Published
- 1998
15. Identification of cis-elements of the human endothelin-A receptor gene and inhibition of the gene expression by the decoy strategy.
- Author
-
Yamashita, J, Yoshimasa, T, Arai, H, Hiraoka, J, Takaya, K, Miyamoto, Y, Ogawa, Y, Itoh, H, and Nakao, K
- Abstract
Previously, we succeeded in molecular cloning of the cDNA and the gene for human endothelin-A receptor (ET-AR). In the present study, we define cis-elements in the 5'-flanking region of the ET-AR gene. Deletion analyses were performed in A7r5 cells, rat vascular smooth muscle cell line, and Chinese hamster ovary cells using ET-AR promoter-luciferase gene constructs including 5 kilobases of the 5'-flanking region. These analyses demonstrated the existence of one negative regulatory element (-2.0 kilobases to -857 bases) and two positive regulatory elements (-137 to -53 and -53 to +251). Gel mobility shift assay revealed a nuclear protein binding to the region (-104 to -78) (R1). DNase I footprinting analysis showed a footprint spanning from -91 to -83 whose sequence is CCCCACCTT (ETA-P1). When a plasmid including R1 fragments (R1 decoy) was co-transfected into A7r5 cells with ET-AR (-137 to +251)-luciferase gene construct, it significantly reduced the luciferase activity in a dose-dependent manner. Moreover, R1 decoy down-regulated the endogenous ET-AR mRNA in A7r5 cells by a maximum of 75%. Thus, we identified cis-elements that regulate basal transcriptional activity of the ET-AR gene and proved the feasibility to suppress the expression of the ET-AR gene by the DNA decoy strategy using the positive regulatory element we identified.
- Published
- 1998
16. Purification, Identification, and Characterization of Two GTP-binding Proteins with Molecular Weights of 25,000 and 21,000 in Human Platelet Cytosol
- Author
-
Nagata, K, Itoh, H, Katada, T, Takenaka, K, Ui, M, Kaziro, Y, and Nozawa, Y
- Abstract
We have purified, characterized, and identified two GTP-binding proteins with Mrof 25,000 (c25KG) and 21,000 (c21KG) from the cytosol fraction of human platelets. These two proteins were not copurified with the βγ subunits of heterotrimeric GTP-binding proteins. Amino acid sequences of tryptic fragments of c21KG completely matched with those of rap1 protein (Pizon, V., Chardin, P., Lerosey, I., Olofsson, B., and Tavitian, A. (1988) Oncogene3, 201–204), smgp21 (Kawata, M., Matsui, Y., Kondo, J., Hishida, T., Teranishi, Y., and Takai, Y. (1988) J. Biol. Chem. 263, 18965–18971), and Krev-1 protein (Kitayama, H., Sugimoto, Y., Matsuzaki, T., Ikawa, Y., and Noda, M. (1989) Cell56, 77–84). The partial amino acid sequence analysis of c25KG revealed that this protein was different from any low MrGTP-binding proteins already reported. c25KG bound about 1 mol of [35S] guanosine 5′-(3-O-thio)triphosphate (GTPγS)/mol of protein, with a Kdvalue of about 45 nM. [35S]GTPγS-binding to c25KG was specifically inhibited by guanine nucleotides, GTP and GDP, but not by adenine nucleotides such as ATP and adenyl-5′-yl, β,γ-imidodiphosphate. The binding activity was not inhibited by pretreatment with N-ethylmaleimide. c25KG hydrolyzed GTP to librate Piwith the specific activity of 1.8 mmol of Pi/mol of protein/min, which are different from the activities of the already purified low MrGTP-binding proteins. We conclude that c25KG is a novel GTP-binding protein and c21KG is a rap1/smgp21/Krev-1 product.
- Published
- 1989
- Full Text
- View/download PDF
17. Transcription factor ZBP-89 regulates the activity of the ornithine decarboxylase promoter.
- Author
-
Law, G L, Itoh, H, Law, D J, Mize, G J, Merchant, J L, and Morris, D R
- Abstract
Appropriate cellular levels of polyamines are required for cell growth and differentiation. Ornithine decarboxylase is a key regulatory enzyme in the biosynthesis of polyamines, and precise regulation of the expression of this enzyme is required, according to cellular growth state. A variety of mitogens increase the level of ornithine decarboxylase activity, and, in most cases, this elevation is due to increased levels of mRNA. A GC box in the proximal promoter of the ornithine decarboxylase gene is required for basal and induced transcriptional activity, and two proteins, Sp1 and NF-ODC1, bind to this region in a mutually exclusive manner. Using a yeast one-hybrid screening method, ZBP-89, a DNA-binding protein, was identified as a candidate for the protein responsible for NF-ODC1 binding activity. Three lines of evidence verified this identification; ZBP-89 copurified with NF-ODC1 binding activity, ZBP-89 antibodies specifically abolished NF-ODC1 binding to the GC box, and binding affinities of 12 different double-stranded oligonucleotides were indistinguishable between NF-ODC1, in nuclear extract, and in vitro translated ZBP-89. ZBP-89 inhibited the activation of the ornithine decarboxylase promoter by Sp1 in Schneider's Drosophila line 2, consistent with properties previously attributed to NF-ODC1.
- Published
- 1998
18. Betagamma subunits of pertussis toxin-sensitive G proteins mediate A1 adenosine receptor agonist-induced activation of phospholipase C in collaboration with thyrotropin. A novel stimulatory mechanism through the cross-talk of two types of receptors.
- Author
-
Tomura, H, Itoh, H, Sho, K, Sato, K, Nagao, M, Ui, M, Kondo, Y, and Okajima, F
- Abstract
COS-7 cells were transiently transfected with human thyrotropin receptor and dog A1 adenosine receptor cDNAs. An A1 agonist, N6-(L-2-phenylisopropyl) adenosine (PIA), which is ineffective alone, enhanced the thyrotropin (TSH)-induced inositol phosphate production, reflecting phospholipase C (PLC) activation, but inhibited the TSH-induced cAMP accumulation, reflecting adenylyl cyclase inhibition. These PIA-induced actions were completely inhibited by pertussis toxin (PTX) treatment. Moreover, in the cells expressing a PTX-insensitive mutant of Gi2alpha or Gi3alpha, in which a glycine residue was substituted for a cysteine residue to be ADP-ribosylated by PTX, at the fourth position of the C terminus, PIA effectively exerted both stimulatory and inhibitory effects on the TSH-induced actions although the cells were treated with the toxin. Overexpression of the betagamma subunits of the G proteins enhanced the TSH-induced inositol phosphate production without any significant effect on the cAMP response; under these conditions, PIA did not further increase the elevated inositol phosphate response to TSH. On the contrary, overexpression of a constitutively active mutant of Gi2alpha, in which the guanosine triphosphatase activity is lost, inhibited the TSH-induced cAMP accumulation but hardly affected the inositol phosphate response; under these conditions, PIA never exerted further inhibitory effects on the cAMP response to TSH. In contrast to the case of the TSH-induced inositol phosphate response, the response to a constitutively active G11alpha mutant was not appreciably affected, and that to NaF was rather inhibited by PIA and overexpression of the betagamma subunits. Taken together, these results suggest that a single type of PTX-sensitive G protein mediates the A1 adenosine receptor-linked modulation of two signaling pathways in collaboration with an activated thyrotropin receptor; alpha subunits of the PTX-sensitive G proteins mediate the inhibitory action on adenylyl cyclase, and the betagamma subunits mediate the stimulatory action on PLC. In the case of the latter stimulatory action on PLC, the betagamma subunits may not directly activate PLC. The possible mechanism by which betagamma subunits enhance the TSH-induced PLC activation is discussed.
- Published
- 1997
19. C-terminal mutation of G protein beta subunit affects differentially extracellular signal-regulated kinase and c-Jun N-terminal kinase pathways in human embryonal kidney 293 cells.
- Author
-
Yamauchi, J, Kaziro, Y, and Itoh, H
- Abstract
G protein beta and gamma subunits (Gbeta and Ggamma) form a complex that is involved in various signaling pathways. We reported that the C-terminal 10 amino acids of Gbeta are required for association with Ggamma (Yamauchi, J., Kaziro, Y., and Itoh, H. (1995) Biochem. Biophys. Res. Commun., 214, 694-700). To evaluate further the significance of the C-terminal region of Gbeta in the formation of a Gbetagamma complex and its signal transduction, we constructed several C-terminal mutants and expressed them in human embryonal kidney 293 cells. The mutant lacking the C-terminal 2 amino acids (DeltaC2) failed to associate with Ggamma, whereas deletion of the C-terminal amino acid (DeltaC1), replacement of Trp at -2 position by Ala (W339A), and addition of six histidines ((His)6) at the C terminus did not affect the association with Ggamma. We also studied the effect of these mutations on the activation of mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK) and c-Jun N-terminal kinase/stress-activated protein kinase (JNK/SAPK). Co-expression of the DeltaC2 or (His)6 mutant with Ggamma did not activate MAPK/ERK at all, whereas the DeltaC1 or W339A mutant showed the MAPK/ERK activation. The JNK/SAPK activity was stimulated by the W339A, DeltaC2, or (His)6 mutant, but not by the DeltaC1 mutant. These results suggest that the C-terminal region of Gbeta participates differentially in the signaling for MAPK/ERK and JNK/SAPK activations in mammalian cells.
- Published
- 1997
20. Activation of p38 mitogen-activated protein kinase by signaling through G protein-coupled receptors. Involvement of Gbetagamma and Galphaq/11 subunits.
- Author
-
Yamauchi, J, Nagao, M, Kaziro, Y, and Itoh, H
- Abstract
Various extracellular stimuli activate three classes of mitogen-activated protein kinases (MAPKs): extracellular signal-regulated kinase, c-Jun N-terminal kinase, and p38 MAPK. In mammalian cells, p38 MAPK is activated by endotoxins, inflammatory cytokines, and environmental stresses. We show here that p38 MAPK is also activated upon stimulation of G protein-coupled receptors (Gq/G11-coupled m1 and Gi-coupled m2 muscarinic acetylcholine and Gs-coupled beta-adrenergic receptors) in human embryonal kidney 293 cells. The activation of p38 MAPK through the m2 and beta-adrenergic receptors was completely inhibited by coexpression of Galphao, whereas the activation by the m1 receptor was only partially inhibited. Furthermore, we show that overexpression of Gbetagamma or a constitutively activated mutant of Galpha11, but not Galphas and Galphai, can stimulate p38 MAPK. These results suggest that the signal from the m2 and beta-adrenergic receptors to p38 MAPK is mediated by Gbetagamma, whereas the signal from the m1 receptor is mediated by both Gbetagamma and Galphaq/11.
- Published
- 1997
21. Mammalian 60-kDa stress protein (chaperonin homolog). Identification, biochemical properties, and localization.
- Author
-
Itoh, H, Kobayashi, R, Wakui, H, Komatsuda, A, Ohtani, H, Miura, A B, Otaka, M, Masamune, O, Andoh, H, and Koyama, K
- Abstract
Mammalian chaperonin homolog (HSP60) was purified from porcine livers cytosol using a tandem ATP-Sepharose column and Mono Q column chromatography. A partial amino acid sequence (96 amino acid residues) of this protein was determined and coincided with those of human HSP60 with 96.9% homology, which was deduced from the nucleotide sequence of the cDNA. The sequence of the NH2 termini of the purified protein (5 amino acid residues) coincided with the signal sequence of HSP60. These facts led to the identification of the 60-kDa liver protein with the chaperonin homolog. Dihydrofolate reductase was able to form a stable complex with the liver chaperonin homolog. The liver chaperonin homolog was detected by at least five spots around pI = 5.6 on two-dimensional gel electrophoresis. Immunoblotting studies using an antibody against chaperonin homolog showed that the chaperonin homolog was localized in the cytosol, mitochondrial, and nuclear fractions of porcine liver. The chaperonin homolog was localized both in the mitochondria and cytoplasm of rat kidneys at the electron microscopic level. The chaperonin homolog in the cytosol, but not in the other subcellular fractions, was cross-reacted with an antibody against the synthetic peptide corresponding to the signal peptide of HSP60 as well as the purified chaperonin homolog on immunoblotting. These results suggested that the functional chaperonin homolog in the cytosol may be transported into the mitochondria and the protein may be processed to mitochondrial HSP60 in the organella.
- Published
- 1995
22. Conversion of adrenergic mechanism from an alpha- to a beta-type during primary culture of rat hepatocytes. Accompanying decreases in the function of the inhibitory guanine nucleotide regulatory component of adenylate cyclase identified as the substrate of islet-activating protein.
- Author
-
Itoh, H, Okajima, F, and Ui, M
- Abstract
Adrenergic mechanism for phosphorylase activation was gradually converted from an alpha 1- to a beta 2-type during primary culture of rat hepatocytes. beta 2-Receptor-mediated cAMP generation was also much greater in 8-h cultured cells than in fresh cells. Incubation of hepatocyte membranes with [alpha-32P]NAD and the preactivated A-protomer (an active component) of islet-activating protein (IAP), pertussis toxin, resulted in the ADP-ribosylation of a specific IAP substrate protein (Mr = 41,000). This ADP-ribosylation diminished progressively when the membrane-donor hepatocytes had been cultured. The early diminution was interfered with by the addition of nicotinamide or isonicotinamide, a potent inhibitor of ADP-ribosyltransferase, to the culture medium. The decrease of the IAP substrate was well correlated with the potentiation of beta-adrenergic functions under various conditions of culture. beta-Receptor-mediated activation of GTP-dependent membrane adenylate cyclase was, but glucagon-induced activation was not enhanced by either prior culture of hepatocytes or prior exposure of membranes to the A-protomer of IAP. There was no further enhancement, however, when membranes from cultured cells were exposed to the active toxin. Thus, the IAP-susceptible inhibitory guanine nucleotide-regulatory protein is coupled to beta-adrenergic receptors in such a manner as to reduce the degree of activation of cyclase, and the decrease in this IAP substrate may be responsible, at least partly, for development of beta-receptor functions during culture of hepatocytes. Its possible relation to accompanying inhibition of alpha 1-receptor functions is discussed.
- Published
- 1984
- Full Text
- View/download PDF
23. Involvement of protein kinase C and Src family tyrosine kinase in Galphaq/11-induced activation of c-Jun N-terminal kinase and p38 mitogen-activated protein kinase.
- Author
-
Nagao, M, Yamauchi, J, Kaziro, Y, and Itoh, H
- Abstract
Mitogen-activated protein kinases (MAPKs) are activated by various extracellular stimuli. The signaling pathways from G protein-coupled receptors to extracellular signal-regulated kinase have been partially elucidated, whereas the mechanisms by which G protein-coupled receptors stimulate c-Jun N-terminal kinase (JNK) and p38 MAPK activities remain largely unknown. We have recently demonstrated that the signal from Gq/11-coupled m1 muscarinic acetylcholine receptor to p38 MAPK is mediated by both Galphaq/11 and Gbeta gamma in HEK-293 cells (Yamauchi, J., Nagao, M., Kaziro, Y., and Itoh, H. (1997) J. Biol. Chem. 272, 27771-27777). In the present study, we report that a constitutively activated mutant of Galpha11 (Galpha11 Q209L) activated not only p38 MAPK, but also JNK, and the activation of JNK and p38 MAPK by Galpha11 Q209L was partially inhibited by prolonged treatment with phorbol 12-myristate 13-acetate and calphostin C. In addition, the Galpha11 Q209L-stimulated activation of both kinases was blocked by a specific inhibitor of protein tyrosine kinases (PP2) and Csk (C-terminal Src kinase). Furthermore, we demonstrated that Galpha11 Q209L stimulated Src family kinase activity and induced tyrosine phosphorylation of several proteins in HEK-293 cells. These results suggest that Galphaq/11 stimulates JNK and p38 MAPK activities through protein kinase C- and Src family kinase-dependent signaling pathways.
- Published
- 1998
24. ATPase activity and ATP-dependent conformational change in the co-chaperone HSP70/HSP90-organizing protein (HOP).
- Author
-
Yamamoto S, Subedi GP, Hanashima S, Satoh T, Otaka M, Wakui H, Sawada K, Yokota S, Yamaguchi Y, Kubota H, and Itoh H
- Subjects
- Adenosine Triphosphatases genetics, Adenosine Triphosphate genetics, Amino Acid Sequence, HSP70 Heat-Shock Proteins genetics, HSP90 Heat-Shock Proteins genetics, Heat-Shock Proteins genetics, Humans, Hydrolysis, Protein Structure, Tertiary, Sequence Deletion, Adenosine Triphosphatases metabolism, Adenosine Triphosphate metabolism, HSP70 Heat-Shock Proteins metabolism, HSP90 Heat-Shock Proteins metabolism, Heat-Shock Proteins metabolism, Protein Folding
- Abstract
Co-chaperones help to maintain cellular homeostasis by modulating the activities of molecular chaperones involved in protein quality control. The HSP70/HSP90-organizing protein (HOP) is a co-chaperone that cooperates with HSP70 and HSP90 in catalysis of protein folding and maturation in the cytosol. We show here that HOP has ATP-binding activity comparable to that of HSP70/HSP90, and that HOP slowly hydrolyzes ATP. Analysis of deletion mutants revealed that the ATPase domain of HOP is in the N-terminal TPR1-DP1-TPR2A segment. In addition, HOP changes its conformation in the presence of ATP. These results indicate that HOP is a unique co-chaperone that undergoes an ATP-dependent conformational change.
- Published
- 2014
- Full Text
- View/download PDF
25. Phosphorylation of doublecortin by protein kinase A orchestrates microtubule and actin dynamics to promote neuronal progenitor cell migration.
- Author
-
Toriyama M, Mizuno N, Fukami T, Iguchi T, Toriyama M, Tago K, and Itoh H
- Subjects
- Animals, COS Cells, Cerebral Cortex cytology, Cerebral Cortex embryology, Cerebral Cortex enzymology, Chlorocebus aethiops, Doublecortin Domain Proteins, Doublecortin Protein, GTP-Binding Protein alpha Subunits, Gs metabolism, HEK293 Cells, Humans, Mice, Neural Stem Cells enzymology, Neurons cytology, Neurons enzymology, Organ Culture Techniques, Phosphorylation physiology, Pituitary Adenylate Cyclase-Activating Polypeptide metabolism, Signal Transduction physiology, rac GTP-Binding Proteins metabolism, rac1 GTP-Binding Protein, Actin Cytoskeleton physiology, Cell Movement physiology, Cyclic AMP-Dependent Protein Kinases metabolism, Microtubule-Associated Proteins metabolism, Microtubules physiology, Neural Stem Cells cytology, Neuropeptides metabolism
- Abstract
Doublecortin (DCX) is a microtubule-associated protein that is specifically expressed in neuronal cells. Genetic mutation of DCX causes lissencephaly disease. Although the abnormal cortical lamination in lissencephaly is thought to be attributable to neuronal cell migration defects, the regulatory mechanisms governing interactions between DCX and cytoskeleton in the migration of neuronal progenitor cells remain obscure. In this study we found that the G(s) and protein kinase A (PKA) signal elicited by pituitary adenylate cyclase-activating polypeptide promotes neuronal progenitor cells migration. Stimulation of G(s)-PKA signaling prevented microtubule bundling and induced the dissociation of DCX from microtubules in cells. PKA phosphorylated DCX at Ser-47, and the phospho-mimicking mutant DCX-S47E promoted cell migration. Activation of PKA and DCX-S47E induced lamellipodium formation. Pituitary adenylate cyclase-activating polypeptide and DCX-S47E stimulated the activation of Rac1, and DCX-S47E interacted with Asef2, a guanine nucleotide exchange factor for Rac1. Our data reveal a dual reciprocal role for DCX phosphorylation in the regulation of microtubule and actin dynamics that is indispensable for proper brain lamination.
- Published
- 2012
- Full Text
- View/download PDF
26. Lowering bile acid pool size with a synthetic farnesoid X receptor (FXR) agonist induces obesity and diabetes through reduced energy expenditure.
- Author
-
Watanabe M, Horai Y, Houten SM, Morimoto K, Sugizaki T, Arita E, Mataki C, Sato H, Tanigawara Y, Schoonjans K, Itoh H, and Auwerx J
- Subjects
- 3T3-L1 Cells, Animals, Body Weight drug effects, Diabetes Mellitus metabolism, Dietary Fats pharmacology, Isoxazoles pharmacology, Metabolic Syndrome drug therapy, Mice, Obesity metabolism, Bile Acids and Salts metabolism, Diabetes Mellitus chemically induced, Dietary Fats adverse effects, Energy Metabolism drug effects, Isoxazoles adverse effects, Obesity chemically induced, Receptors, Cytoplasmic and Nuclear agonists
- Abstract
We evaluated the metabolic impact of farnesoid X receptor (FXR) activation by administering a synthetic FXR agonist (GW4064) to mice in which obesity was induced by a high fat diet. Administration of GW4064 accentuated body weight gain and glucose intolerance induced by the high fat diet and led to a pronounced worsening of the changes in liver and adipose tissue. Mechanistically, treatment with GW4064 decreased bile acid (BA) biosynthesis, BA pool size, and energy expenditure, whereas reconstitution of the BA pool in these GW4064-treated animals by BA administration dose-dependently reverted the metabolic abnormalities. Our data therefore suggest that activation of FXR with synthetic agonists is not useful for long term management of the metabolic syndrome, as it reduces the BA pool size and subsequently decreases energy expenditure, translating as weight gain and insulin resistance. In contrast, expansion of the BA pool size, which can be achieved by BA administration, could be an interesting strategy to manage the metabolic syndrome.
- Published
- 2011
- Full Text
- View/download PDF
27. KappaB-Ras is a nuclear-cytoplasmic small GTPase that inhibits NF-kappaB activation through the suppression of transcriptional activation of p65/RelA.
- Author
-
Tago K, Funakoshi-Tago M, Sakinawa M, Mizuno N, and Itoh H
- Subjects
- Amino Acid Substitution, Animals, Cell Nucleus genetics, Cytoplasm genetics, Guanosine Diphosphate genetics, Guanosine Diphosphate metabolism, Guanosine Triphosphate genetics, Guanosine Triphosphate metabolism, Humans, Mice, Monomeric GTP-Binding Proteins genetics, Mutation, Mutation, Missense, NIH 3T3 Cells, Phosphorylation genetics, Transcription Factor RelA genetics, Transcriptional Activation physiology, Cell Nucleus metabolism, Cytoplasm metabolism, Monomeric GTP-Binding Proteins metabolism, Transcription Factor RelA metabolism, Transcription, Genetic physiology
- Abstract
NF-κB is an important transcription factor involved in various biological responses, including inflammation, cell differentiation, and tumorigenesis. κB-Ras was identified as an IκB-interacting small GTPase and is reported to disturb cytokine-induced NF-κB activation. In this study, we established that κB-Ras is a novel type of nuclear-cytoplasmic small GTPase that mainly binds to GTP, and its localization seemed to be regulated by its GTP/GDP-binding state. Unexpectedly, the GDP-binding form of the κB-Ras mutant exhibited a more potent inhibitory effect on NF-κB activation, and this inhibitory effect seemed to be due to suppression of the transactivation of a p65/RelA NF-κB subunit. κB-Ras suppressed phosphorylation at serine 276 on the p65/RelA subunit, resulting in decreased interaction between p65/RelA and the transcriptional coactivator p300. Interestingly, the GDP-bound κB-Ras mutant exhibited higher interactive affinity with p65/RelA and inhibited the phosphorylation of p65/RelA more potently than wild-type κB-Ras. Taken together, these findings suggest that the GDP-bound form of κB-Ras in cytoplasm suppresses NF-κB activation by inhibiting its transcriptional activation.
- Published
- 2010
- Full Text
- View/download PDF
28. Kidney-specific overexpression of Sirt1 protects against acute kidney injury by retaining peroxisome function.
- Author
-
Hasegawa K, Wakino S, Yoshioka K, Tatematsu S, Hara Y, Minakuchi H, Sueyasu K, Washida N, Tokuyama H, Tzukerman M, Skorecki K, Hayashi K, and Itoh H
- Subjects
- Acute Disease, Animals, Antineoplastic Agents adverse effects, Antineoplastic Agents pharmacology, Apoptosis drug effects, Apoptosis genetics, Catalase biosynthesis, Catalase genetics, Cell Line, Cisplatin adverse effects, Cisplatin pharmacology, Kidney Diseases chemically induced, Kidney Diseases genetics, Kidney Diseases therapy, Kidney Tubules, Proximal pathology, Longevity drug effects, Longevity genetics, Male, Mice, Mice, Transgenic, Mitochondria genetics, Mitochondria metabolism, Organ Specificity, Peroxisomes genetics, Reactive Oxygen Species metabolism, Sirtuin 1 genetics, Sodium-Phosphate Cotransporter Proteins, Type IIa biosynthesis, Sodium-Phosphate Cotransporter Proteins, Type IIa genetics, Up-Regulation drug effects, Up-Regulation genetics, Kidney Diseases metabolism, Kidney Tubules, Proximal injuries, Kidney Tubules, Proximal metabolism, Peroxisomes metabolism, Sirtuin 1 biosynthesis
- Abstract
Sirt1, a NAD-dependent protein deacetylase, is reported to regulate intracellular metabolism and attenuate reactive oxidative species (ROS)-induced apoptosis leading to longevity and acute stress resistance. We created transgenic (TG) mice with kidney-specific overexpression of Sirt1 using the promoter sodium-phosphate cotransporter IIa (Npt2) driven specifically in proximal tubules and investigated the kidney-specific role of Sirt1 in the protection against acute kidney injury (AKI). We also elucidated the role of number or function of peroxisome and mitochondria in mediating the mechanisms for renal protective effects of Sirt1 in AKI. Cisplatin-induced AKI decreased the number and function of peroxisomes as well as mitochondria and led to increased local levels of ROS production and renal tubular apoptotic cells. TG mice treated with cisplatin mitigated AKI, local ROS, and renal tubular apoptotic tubular cells. Consistent with these results, TG mice treated with cisplatin also exhibited recovery of peroxisome number and function, as well as rescued mitochondrial function; however, mitochondrial number was not recovered. Immunoelectron microscopic findings consistently demonstrated that the decrease in peroxisome number by cisplatin in wild type mice was restored in transgenic mice. In HK-2 cells, a cultured proximal tubule cell line, overexpression of Sirt1 rescued the cisplatin-induced cell apoptosis through the restoration of peroxisome number, although the mitochondria number was not restored. These results indicate that Sirt1 overexpression in proximal tubules rescues cisplatin-induced AKI by maintaining peroxisomes number and function, concomitant up-regulation of catalase, and elimination of renal ROS levels. Renal Sirt1 can be a potential therapeutic target for the treatment of AKI.
- Published
- 2010
- Full Text
- View/download PDF
29. Ric-8B stabilizes the alpha subunit of stimulatory G protein by inhibiting its ubiquitination.
- Author
-
Nagai Y, Nishimura A, Tago K, Mizuno N, and Itoh H
- Subjects
- Animals, Cell Line, Cyclic AMP metabolism, Cycloheximide pharmacology, GTP-Binding Protein alpha Subunits, Gs chemistry, Guanine Nucleotide Exchange Factors, Humans, Mice, NIH 3T3 Cells, Nuclear Proteins chemistry, Protein Binding, Protein Structure, Tertiary, Signal Transduction, Ubiquitin chemistry, Ubiquitin-Protein Ligases chemistry, GTP-Binding Protein alpha Subunits, Gs physiology, GTP-Binding Proteins chemistry, Nuclear Proteins physiology, Ubiquitination
- Abstract
The alpha subunit of stimulatory G protein (G alpha(s)) activates adenylyl cyclase, which catalyzes cAMP production, and regulates many physiological aspects, such as cardiac regulation and endocrine systems. Ric-8B (resistance to inhibitors of cholinesterase 8B) has been identified as the G alpha(s)-binding protein; however, its role in G(s) signaling remains obscure. In this study, we present evidence that Ric-8B specifically and positively regulates G(s) signaling by stabilizing the G alpha(s) protein. An in vitro biochemical study suggested that Ric-8B does not possess guanine nucleotide exchange factor activity. However, knockdown of Ric-8B attenuated beta-adrenergic agonist-induced cAMP accumulation, indicating that Ric-8B positively regulates G(s) signaling. Interestingly, overexpression and knockdown of Ric-8B resulted in an increase and a decrease in the G alpha(s) protein, respectively, without affecting the G alpha(s) mRNA level. We found that the G alpha(s) protein is ubiquitinated and that this ubiquitination is inhibited by Ric-8B. This Ric-8B-mediated inhibition of G alpha(s) ubiquitination requires interaction between Ric-8B and G alpha(s) because Ric-8B splicing variants, which are defective for G alpha(s) binding, failed to inhibit the ubiquitination. Taken together, these results suggest that Ric-8B plays a critical and specific role in the control of G alpha(s) protein levels by modulating G alpha(s) ubiquitination and positively regulates G(s) signaling.
- Published
- 2010
- Full Text
- View/download PDF
30. 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
31. Mechanism and role of high density lipoprotein-induced activation of AMP-activated protein kinase in endothelial cells.
- Author
-
Kimura T, Tomura H, Sato K, Ito M, Matsuoka I, Im DS, Kuwabara A, Mogi C, Itoh H, Kurose H, Murakami M, and Okajima F
- Subjects
- AMP-Activated Protein Kinase Kinases, AMP-Activated Protein Kinases genetics, Animals, Aorta cytology, Aorta metabolism, Blotting, Western, Calcium-Calmodulin-Dependent Protein Kinase Kinase genetics, Calcium-Calmodulin-Dependent Protein Kinase Kinase metabolism, Cell Adhesion genetics, Cell Adhesion physiology, Cell Line, Cell Proliferation, Endothelial Cells cytology, Endothelial Cells enzymology, Enzyme Activation drug effects, Humans, In Vitro Techniques, Male, Mice, Phosphorylation drug effects, Protein Serine-Threonine Kinases genetics, Protein Serine-Threonine Kinases metabolism, RNA, Small Interfering, Receptors, Lysosphingolipid genetics, Receptors, Lysosphingolipid metabolism, Reverse Transcriptase Polymerase Chain Reaction, Scavenger Receptors, Class B genetics, Scavenger Receptors, Class B metabolism, Umbilical Veins cytology, AMP-Activated Protein Kinases metabolism, Endothelial Cells drug effects, Endothelial Cells metabolism, Lipoproteins, HDL pharmacology
- Abstract
The upstream signaling pathway leading to the activation of AMP-activated protein kinase (AMPK) by high density lipoprotein (HDL) and the role of AMPK in HDL-induced antiatherogenic actions were investigated. Experiments using genetic and pharmacological tools showed that HDL-induced activation of AMPK is dependent on both sphingosine 1-phosphate receptors and scavenger receptor class B type I through calcium/calmodulin-dependent protein kinase kinase and, for scavenger receptor class B type I system, additionally serine-threonine kinase LKB1 in human umbilical vein endothelial cells. HDL-induced activation of Akt and endothelial NO synthase, stimulation of migration, and inhibition of monocyte adhesion and adhesion molecule expression were dependent on AMPK activation. The inhibitory role of AMPK in the adhesion molecule expression and monocyte adhesion on endothelium of mouse aorta was confirmed in vivo and ex vivo. On the other hand, stimulation of ERK and proliferation were hardly affected by AMPK knockdown but completely inhibited by an N17Ras, whereas the dominant-negative Ras was ineffective for AMPK activation. In conclusion, dual HDL receptor systems differentially regulate AMPK activity through calcium/calmodulin-dependent protein kinase kinase and/or LKB1. Several HDL-induced antiatherogenic actions are regulated by AMPK, but proliferation-related actions are regulated by Ras rather than AMPK.
- Published
- 2010
- Full Text
- View/download PDF
32. Orphan G protein-coupled receptor GPR56 regulates neural progenitor cell migration via a G alpha 12/13 and Rho pathway.
- Author
-
Iguchi T, Sakata K, Yoshizaki K, Tago K, Mizuno N, and Itoh H
- Subjects
- Actins metabolism, Animals, Antibodies immunology, Antibodies pharmacology, Cell Line, Cell Membrane metabolism, Enzyme Activation drug effects, Humans, Mice, NF-kappa B metabolism, Neurons cytology, Neurons drug effects, Receptors, G-Protein-Coupled agonists, Receptors, G-Protein-Coupled genetics, Receptors, G-Protein-Coupled immunology, Serum Response Element, Stem Cells cytology, Stem Cells drug effects, Cell Movement, GTP-Binding Protein alpha Subunits, G12-G13 metabolism, Neurons metabolism, Receptors, G-Protein-Coupled metabolism, Signal Transduction, Stem Cells metabolism, rhoA GTP-Binding Protein metabolism
- Abstract
In the developing forebrain, the migration and positioning of neural progenitor cells (NPCs) are regulated coordinately by various molecules. Mutation of these molecules, therefore, causes cortical malformation. GPR56 has been reported as a cortical malformation-related gene that is mutated in patients with bilateral frontoparietal polymicrogyria. GPR56 encodes an orphan G protein-coupled receptor, and its mutations reduce the cell surface expression. It has also been reported that the expression level of GPR56 is involved in cancer cell adhesion and metastasis. However, it remains to be clarified how GPR56 functions in brain development and which signaling pathways are activated by GPR56. In this study, we showed that GPR56 is highly expressed in NPCs and has the ability to inhibit NPC migration. We found that GPR56 coupled with Galpha(12/13) and induced Rho-dependent activation of the transcription mediated through a serum-responsive element and NF-kappaB-responsive element and actin fiber reorganization. The transcriptional activation and actin reorganization were inhibited by an RGS domain of the p115 Rho-specific guanine nucleotide exchange factor (p115 RhoGEF RGS) and dominant negative form of Rho. Moreover, we have demonstrated that a functional anti-GPR56 antibody, which has an agonistic activity, inhibited NPC migration. This inhibition was attenuated by p115 RhoGEF RGS, C3 exoenzyme, and GPR56 knockdown. These results indicate that GPR56 participates in the regulation of NPC movement through the Galpha(12/13) and Rho signaling pathway, suggesting its important role in the development of the central nervous system.
- Published
- 2008
- Full Text
- View/download PDF
33. Novel dehydrogenase catalyzes oxidative hydrolysis of carbon-nitrogen double bonds for hydrazone degradation.
- Author
-
Itoh H, Suzuta T, Hoshino T, and Takaya N
- Subjects
- Aldehyde Dehydrogenase genetics, Aldehyde Dehydrogenase metabolism, Amino Acid Sequence, Candida genetics, Fungal Proteins genetics, Fungal Proteins metabolism, Hydrazones metabolism, Hydrolysis, Kinetics, Molecular Sequence Data, Mutation, NADP chemistry, NADP metabolism, Oxidation-Reduction, Protein Structure, Quaternary, Saccharomyces cerevisiae enzymology, Saccharomyces cerevisiae genetics, Substrate Specificity genetics, Aldehyde Dehydrogenase chemistry, Candida enzymology, Fungal Proteins chemistry, Hydrazones chemistry, Soil Microbiology
- Abstract
Hydrazines and their derivatives are versatile artificial and natural compounds that are metabolized by elusive biological systems. Here we identified microorganisms that assimilate hydrazones and isolated the yeast, Candida palmioleophila MK883. When cultured with adipic acid bis(ethylidene hydrazide) as the sole source of carbon, C. palmioleophila MK883 degraded hydrazones and accumulated adipic acid dihydrazide. Cytosolic NAD+- or NADP+-dependent hydrazone dehydrogenase (Hdh) activity was detectable under these conditions. The production of Hdh was inducible by adipic acid bis(ethylidene hydrazide) and the hydrazone, varelic acid ethylidene hydrazide, under the control of carbon catabolite repression. Purified Hdh oxidized and hydrated the C=N double bond of acetaldehyde hydrazones by reducing NAD+ or NADP+ to produce relevant hydrazides and acetate, the latter of which the yeast assimilated. The deduced amino acid sequence revealed that Hdh belongs to the aldehyde dehydrogenase (Aldh) superfamily. Kinetic and mutagenesis studies showed that Hdh formed a ternary complex with the substrates and that conserved Cys is essential for the activity. The mechanism of Hdh is similar to that of Aldh, except that it catalyzed oxidative hydrolysis of hydrazones that requires adding a water molecule to the reaction catalyzed by conventional Aldh. Surprisingly, both Hdh and Aldh from baker's yeast (Ald4p) catalyzed the Hdh reaction as well as aldehyde oxidation. Our findings are unique in that we discovered a biological mechanism for hydrazone utilization and a novel function of proteins in the Aldh family that act on C=N compounds.
- Published
- 2008
- Full Text
- View/download PDF
34. Genetic and pharmacological inhibition of Rho-associated kinase II enhances adipogenesis.
- Author
-
Noguchi M, Hosoda K, Fujikura J, Fujimoto M, Iwakura H, Tomita T, Ishii T, Arai N, Hirata M, Ebihara K, Masuzaki H, Itoh H, Narumiya S, and Nakao K
- Subjects
- 3T3-L1 Cells, Actins metabolism, Adipocytes drug effects, Adipogenesis drug effects, Animals, Cytoskeleton metabolism, Dose-Response Relationship, Drug, Fibroblasts metabolism, Insulin metabolism, Mice, Mice, Knockout, Models, Biological, Models, Genetic, Signal Transduction, rho-Associated Kinases, Adipocytes metabolism, Intracellular Signaling Peptides and Proteins metabolism, Protein Serine-Threonine Kinases metabolism
- Abstract
Rho-associated kinase (ROCK) regulates reorganization of actin cytoskeleton. During adipogenesis, the structure of filamentous actin is converted from long stress fibers to cortical actin, suggesting that the ROCK is involved in adipogenesis. Two ROCK isoforms have been identified: ROCK-I and ROCK-II. However, pharmacological inhibitors of ROCK cannot distinguish two ROCK isoforms. In the present study, we examined the role of ROCK in adipogenesis and actin cytoskeleton using genetic and pharmacological approaches. Y-27632, which inhibits the activity of both ROCK isoforms, enhanced adipogenesis through the up-regulation of adipogenic transcription factors in 3T3-L1 cells. Furthermore, Y-27632 restored inhibition of adipogenesis by lysophosphatidic acid, which activates Rho. Regarding actin cytoskeleton, Y-27632 disrupted stress fibers in 3T3-L1 preadipocytes. Next, we analyzed adipogenesis of mouse embryonic fibroblasts (MEFs) derived from ROCK-I and ROCK-II knock-out mice, respectively. Adipogenesis of ROCK-II (-/-) MEFs was markedly enhanced compared with wild-type MEFs while that of ROCK-I (-/-) MEFs was not. In contrast to pharmacological approaches, no obvious alteration was found in actin cytoskeleton of ROCK-II (-/-) MEFs compared with wild-type MEFs. In 3T3-L1 cells, knockdown of ROCK-II by RNA interference enhanced the expression of adipogenic transcription factors while that of ROCK-I did not. Moreover, Y-27632 inhibited IRS-1 serine phosphorylation and enhanced Akt phosphorylation in 3T3-L1 preadipocytes. Similarly, Akt phosphorylation in ROCK-II (-/-) MEFs was augmented compared with wild-type MEFs. In conclusion, inhibition of ROCK-II, not ROCK-I, enhances adipogenesis accompanied by the up-regulation of adipogenic transcription factors. Augmentation of insulin signaling may contribute to the enhancement of adipogenesis.
- Published
- 2007
- Full Text
- View/download PDF
35. 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
36. Analysis of rat insulin II promoter-ghrelin transgenic mice and rat glucagon promoter-ghrelin transgenic mice.
- Author
-
Iwakura H, Hosoda K, Son C, Fujikura J, Tomita T, Noguchi M, Ariyasu H, Takaya K, Masuzaki H, Ogawa Y, Hayashi T, Inoue G, Akamizu T, Hosoda H, Kojima M, Itoh H, Toyokuni S, Kangawa K, and Nakao K
- Subjects
- Animals, Arginine chemistry, Blotting, Northern, Body Weight, DNA, Complementary metabolism, Gene Library, Ghrelin, Glucose metabolism, Homeodomain Proteins metabolism, Immunohistochemistry, Insulin metabolism, Insulin Secretion, Introns, Islets of Langerhans metabolism, Lipid Metabolism, Mice, Mice, Transgenic, Models, Genetic, Pancreas metabolism, Peptide Hormones metabolism, Peptides chemistry, RNA, Messenger metabolism, Radioimmunoassay, Rats, Reverse Transcriptase Polymerase Chain Reaction, Time Factors, Trans-Activators metabolism, Glucagon genetics, Insulin genetics, Peptide Hormones genetics, Promoter Regions, Genetic
- Abstract
We developed and analyzed two types of transgenic mice: rat insulin II promoter-ghrelin transgenic (RIP-G Tg) and rat glucagon promoter-ghrelin transgenic mice (RGP-G Tg). The pancreatic tissue ghrelin concentration measured by C-terminal radioimmunoassay (RIA) and plasma desacyl ghrelin concentration of RIP-G Tg were about 1000 and 3.4 times higher than those of nontransgenic littermates, respectively. The pancreatic tissue n-octanoylated ghrelin concentration measured by N-terminal RIA and plasma n-octanoylated ghrelin concentration of RIP-G Tg were not distinguishable from those of nontransgenic littermates. RIP-G Tg showed suppression of glucose-stimulated insulin secretion. Arginine-stimulated insulin secretion, pancreatic insulin mRNA and peptide levels, beta cell mass, islet architecture, and GLUT2 and PDX-1 immunoreactivity in RIP-G Tg pancreas were not significantly different from those of nontransgenic littermates. Islet batch incubation study did not show suppression of insulin secretion of RIP-G Tg in vitro. The insulin tolerance test showed lower tendency of blood glucose levels in RIP-G Tg. Taking lower tendency of triglyceride level of RIP-G Tg into consideration, these results may indicate that the suppression of insulin secretion is likely due to the effect of desacyl ghrelin on insulin sensitivity. RGP-G Tg, in which the pancreatic tissue ghrelin concentration measured by C-RIA was about 50 times higher than that of nontransgenic littermates, showed no significant changes in insulin secretion, glucose metabolism, islet mass, and islet architecture. The present study raises the possibility that desacyl ghrelin may have influence on glucose metabolism.
- Published
- 2005
- Full Text
- View/download PDF
37. DNA topoisomerase II poison TAS-103 transactivates GC-box-dependent transcription via acetylation of Sp1.
- Author
-
Torigoe T, Izumi H, Wakasugi T, Niina I, Igarashi T, Yoshida T, Shibuya I, Chijiiwa K, Matsuo K, Itoh H, and Kohno K
- Subjects
- Acetylation drug effects, Antineoplastic Agents pharmacology, Cell Line, Tumor, DNA Topoisomerases, Type II metabolism, Genes, Reporter genetics, Humans, Nuclear Proteins metabolism, Promoter Regions, Genetic genetics, Protein Binding, Simian virus 40 genetics, Trans-Activators metabolism, Aminoquinolines pharmacology, GC Rich Sequence genetics, Indenes pharmacology, Response Elements genetics, Sp1 Transcription Factor metabolism, Topoisomerase II Inhibitors, Transcription, Genetic drug effects, Transcriptional Activation drug effects
- Abstract
Drug-induced modifications of transcription factors play important roles in both apoptosis and survival signaling. The data presented here show that the DNA topoisomerase II poison TAS-103 transactivated the SV40 promoter in a GC-box-dependent manner and induced Sp1 acetylation in cells expressing p300. This activity was not observed in cells lacking p300. TAS-103 treatment also enhanced the p300 content of the nucleus and the interaction of p300 with Sp1. Cellular susceptibility to TAS-103 was correlated with p300 expression but not with topoisomerase II expression. Furthermore, the presence of p300 significantly sensitized cancer cells to TAS-103 but not to cisplatin. Taken together, these findings demonstrate novel genomic responses to anticancer agents that modulate Sp1 acetylation and Sp1-dependent transcription in an apoptotic pathway.
- Published
- 2005
- Full Text
- View/download PDF
38. The adaptor protein Nck1 mediates endothelin A receptor-regulated cell migration through the Cdc42-dependent c-Jun N-terminal kinase pathway.
- Author
-
Miyamoto Y, Yamauchi J, Mizuno N, and Itoh H
- Subjects
- Adaptor Proteins, Signal Transducing, Animals, Base Sequence, Cell Line, Cell Membrane metabolism, Cell Movement, Cytosol metabolism, Enzyme Activation, Genes, Dominant, Humans, Immunoblotting, JNK Mitogen-Activated Protein Kinases, MAP Kinase Signaling System, Mice, Models, Biological, Molecular Sequence Data, Oncogene Proteins metabolism, Plasmids metabolism, Precipitin Tests, Protein Structure, Tertiary, RNA, Small Interfering metabolism, Signal Transduction, Transfection, src Homology Domains, src-Family Kinases metabolism, Endothelin-1 metabolism, Mitogen-Activated Protein Kinases metabolism, Oncogene Proteins physiology, Receptor, Endothelin A metabolism, cdc42 GTP-Binding Protein metabolism
- Abstract
Cell migration plays key roles in physiological and pathological phenomena, such as development and oncogenesis. The adaptor proteins Grb2, CrkII, and Nck1 are composed of only a single Src homology 2 domain and some Src homology 3 domains, giving specificity to each signal transduction pathway. However, little is known about the relationships between their adaptor proteins and cell migration, which are regulated by the G protein-coupled receptor. Here we showed that Nck1, but not Grb2 or CrkII, mediated the inhibition of cell migration induced by the endothelin-1 and endothelin A receptor. The small interference RNA and dominant negative mutants of Nck1 diminished the endothelin-1-induced inhibition of cell migration. Although overexpression of wild-type Nck1 was detected in the cytosol and did not affect cell migration, expression of the myristoylation signal sequence-conjugated Nck1 was detected in the membrane and induced activation of Cdc42 and c-Jun N-terminal kinase, inhibiting cell migration. Taken together, these results suggest that the endothelin A receptor transduces the signal of inhibition of cell migration through Cdc42-dependent c-Jun N-terminal kinase activation by using Nck1.
- Published
- 2004
- Full Text
- View/download PDF
39. 73-kDa molecular chaperone HSP73 is a direct target of antibiotic gentamicin.
- Author
-
Miyazaki T, Sagawa R, Honma T, Noguchi S, Harada T, Komatsuda A, Ohtani H, Wakui H, Sawada K, Otaka M, Watanabe S, Jikei M, Ogawa N, Hamada F, and Itoh H
- Subjects
- Acetylglucosaminidase urine, Amino Acid Sequence, Animals, Binding Sites, Brain metabolism, Cattle, Chromatography, Circular Dichroism, Creatinine blood, Cytosol metabolism, Disease Models, Animal, HSC70 Heat-Shock Proteins, Immunoblotting, Immunohistochemistry, Kidney metabolism, Kidney Tubules metabolism, Lysosomes metabolism, Male, Microscopy, Electron, Molecular Chaperones pharmacology, Molecular Sequence Data, Nitrogen blood, Peptides chemistry, Protein Binding, Protein Conformation, Protein Structure, Tertiary, Rats, Rats, Sprague-Dawley, Swine, Thiosulfate Sulfurtransferase chemistry, Time Factors, Ultraviolet Rays, Anti-Bacterial Agents pharmacology, Carrier Proteins metabolism, Carrier Proteins physiology, Gentamicins pharmacology, HSP70 Heat-Shock Proteins
- Abstract
Although gentamicin (GM) has been used widely as an antibiotic, the specific binding protein of the drug has not yet been understood sufficiently. Here we show that GM specifically associates with the 73-kDa molecular chaperone HSP73 and reduces its chaperone activity in vitro. In the present study, we investigated GM-specific binding proteins using a GM-affinity column and porcine kidney cytosol. After washing the column, only the 73-kDa protein was eluted from the column by the addition of 10 mm GM. None of the other proteins were found in the eluant. Upon immunoblotting, the protein was identical to HSP73. Upon CD spectrum analysis, the binding of GM to HSP73 resulted in a conformational change in the protein. Although HSP73 prevents aggregation of unfolded rhodanese in vitro, the chaperone activity of HSP73 was suppressed in the presence of GM. Using limited proteolysis of HSP73 by TPCK-trypsin, the GM binding site is a COOH-terminal for one third of the protein known to be a peptide-binding domain. During immunohistochemistry, HSP73 and GM were co-localized in enlarged lysosomes of rat kidneys with GM-induced acute tubular injury in vivo. Our results suggest that the specific association between HSP73 and GM may reduce the chaperone activity of HSP73 in vitro and/or in vivo, and this may have an interaction with GM toxicity in kidneys with GM-induced acute tubular injury.
- Published
- 2004
- Full Text
- View/download PDF
40. S100A1 is a novel molecular chaperone and a member of the Hsp70/Hsp90 multichaperone complex.
- Author
-
Okada M, Hatakeyama T, Itoh H, Tokuta N, Tokumitsu H, and Kobayashi R
- Subjects
- Amino Acid Sequence, Animals, Brain metabolism, Calcium metabolism, Calcium-Binding Proteins chemistry, Calcium-Binding Proteins genetics, Calmodulin metabolism, Carrier Proteins chemistry, Carrier Proteins genetics, Carrier Proteins metabolism, Cattle, HSP70 Heat-Shock Proteins chemistry, HSP70 Heat-Shock Proteins genetics, HSP90 Heat-Shock Proteins chemistry, HSP90 Heat-Shock Proteins genetics, Humans, In Vitro Techniques, Kinetics, Macromolecular Substances, Molecular Chaperones chemistry, Molecular Chaperones genetics, Molecular Sequence Data, Peptidylprolyl Isomerase chemistry, Peptidylprolyl Isomerase genetics, Peptidylprolyl Isomerase metabolism, Recombinant Fusion Proteins chemistry, Recombinant Fusion Proteins genetics, Recombinant Fusion Proteins metabolism, S100 Proteins, Surface Plasmon Resonance, Tacrolimus Binding Proteins chemistry, Tacrolimus Binding Proteins genetics, Tacrolimus Binding Proteins metabolism, Calcium-Binding Proteins metabolism, Cyclophilins, HSP70 Heat-Shock Proteins metabolism, HSP90 Heat-Shock Proteins metabolism, Molecular Chaperones metabolism
- Abstract
Although calmodulin is known to be a component of the Hsp70/Hsp90 multichaperone complex, the functional role of the protein remains uncertain. In this study, we have identified S100A1, but not calmodulin or other S100 proteins, as a potent molecular chaperone and a new member of the multichaperone complex. Glutathione S-transferase pull-down assays and co-immunoprecipitation experiments indicated the formation of stable complexes between S100A1 and Hsp90, Hsp70, FKBP52, and CyP40 both in vitro and in mammalian cells. S100A1 potently protected citrate synthase, aldolase, glyceraldehyde-3-phosphate dehydrogenase, and rhodanese from heat-induced aggregation and suppressed the aggregation of chemically denatured rhodanese and citrate synthase during the refolding pathway. In addition, S100A1 suppressed the heat-induced inactivation of citrate synthase activity, similar to that for Hsp90 and p23. The chaperone activity of S100A1 was antagonized by calmodulin antagonists, such as fluphenazine and prenylamine, that is, indeed an intrinsic function of the protein. The overexpression of S100A1 in COS-7 cells protected transiently expressed firefly luciferase and Escherichia coli beta-galactosidase from inactivation during heat shock. The results demonstrate a novel physiological function for S100A1 and bring us closer to a comprehensive understanding of the molecular mechanisms of the Hsp70/Hsp90 multichaperone complex.
- Published
- 2004
- Full Text
- View/download PDF
41. Src kinase regulates the activation of a novel FGD-1-related Cdc42 guanine nucleotide exchange factor in the signaling pathway from the endothelin A receptor to JNK.
- Author
-
Miyamoto Y, Yamauchi J, and Itoh H
- Subjects
- Amino Acid Sequence, Catalytic Domain, Cell Line, Cell Movement, Cells, Cultured, DNA, Complementary metabolism, Endothelin-1 metabolism, Endothelins metabolism, Enzyme Activation, Genes, Dominant, Glutathione Transferase metabolism, Humans, JNK Mitogen-Activated Protein Kinases, MAP Kinase Kinase 4, Mitogen-Activated Protein Kinases metabolism, Models, Biological, Molecular Sequence Data, Mutation, Phosphorylation, Proline chemistry, Protein Binding, Receptor, Endothelin A, Recombinant Fusion Proteins metabolism, Recombinant Proteins metabolism, Sequence Homology, Amino Acid, Time Factors, Transfection, Tyrosine metabolism, rac1 GTP-Binding Protein metabolism, rho GTP-Binding Proteins metabolism, rhoA GTP-Binding Protein metabolism, Guanine Nucleotide Exchange Factors chemistry, Guanine Nucleotide Exchange Factors physiology, Mitogen-Activated Protein Kinase Kinases metabolism, Proteins metabolism, Receptors, Endothelin metabolism, Signal Transduction, cdc42 GTP-Binding Protein metabolism, src-Family Kinases physiology
- Abstract
Small GTPases act as binary switches by cycling between an inactive (GDP-bound) and an active (GTP-bound) state. Upon stimulation with extracellular signals, guanine-nucleotide exchange factors (GEFs) stimulate the exchange of GDP to GTP to shift toward the active forms of small GTPases, recognizing the downstream targets. Here we show that KIAA0793, containing substantial sequence homology with the catalytic Dbl homology domain of the faciogenital dysplasia gene product (FGD1), is a specific GEF for Cdc42. We, therefore, tentatively named it FRG (FGD1-related Cdc42-GEF). Src kinase directly phosphorylates and activates FRG, as Vav family GEFs. Additionally, FRG is involved in the signaling pathway from the endothelin A receptor to c-Jun N-terminal kinase, resulting in the inhibition of cell motility. These results suggest that FRG is a member of Cdc42-GEF and plays an important role in the signaling pathway downstream of G protein-coupled receptors.
- Published
- 2003
- Full Text
- View/download PDF
42. A combinatorial G protein-coupled receptor reconstitution system on budded baculovirus. Evidence for Galpha and Galphao coupling to a human leukotriene B4 receptor.
- Author
-
Masuda K, Itoh H, Sakihama T, Akiyama C, Takahashi K, Fukuda R, Yokomizo T, Shimizu T, Kodama T, and Hamakubo T
- Subjects
- Baculoviridae, GTP-Binding Protein alpha Subunits, Gi-Go genetics, Humans, Ligands, Models, Biological, Protein Binding, Protein Conformation, Receptors, Leukotriene B4 genetics, Recombinant Proteins genetics, Virion, GTP-Binding Protein alpha Subunits, Gi-Go analysis, Receptors, Leukotriene B4 analysis, Recombinant Proteins analysis, Signal Transduction
- Abstract
To investigate the coupling selectivity of G proteins and G protein-coupled receptors (GPCRs), we developed a reconstitution system made up of GPCR and heterotrimeric G proteins on extracellular baculovirus particles (budded virus (BV)). BV released from Sf9 cells infected with a recombinant baculovirus coding for human leukotriene B4 receptor (BLT1) cDNA exhibited a high level of BLT1 expression (27.3 pmol/mg of protein) and specific [3H]leukotriene B4 binding activity (Kd = 3.67 nm). The apparent low affinity of the expressed BLT1 is thought to be due to relative non-availability of the Galphai isoform, which couples to BLT1, in BV. Co-infection of heterotrimeric G protein recombinant viruses led to co-expression of BLT1 and G protein subunits on BV. A guanosine-5'-(beta,gamma-imido)triphosphate-sensitive, high affinity ligand binding was observed in the BLT1 BV co-expressing Galphai1beta1gamma2 (Kd = 0.17 nm). A relatively large amount of high affinity receptor protein was recovered in the co-expressing BV fraction (6.81 pmol/mg of protein). A combination of BLT1 and Galphai1 without Gbeta1gamma2 did not exhibit high affinity ligand binding on BV, indicating the low background environment for the GPCR-G protein coupling in this BV reconstitution system. To test other G proteins for coupling, various Galpha subunits were combinatorially expressed in BV with BLT1 and Gbeta1gamma2. The BLT1 BV co-expressing GalphaoAbeta1gamma2 exhibited a comparably high affinity ligand binding as well as ligand-stimulated guanosine 5'-3-O-(thio)triphosphate binding to Galphai1beta1gamma2. Co-expression of other Galpha isoforms such as Galphas, Galpha11, Galpha14, Galpha16, Galpha12, or Galpha13 did not exhibit any significant effects on ligand binding affinity in this system. These results reveal that BLT1 and coupled trimeric G proteins were functionally reconstituted on BV and that Galphao as well as Galphai couples to BLT1. This expression system should prove highly useful for pharmacological characterization, biosensor chip applications, and also drug discovery directed at highly important targets of the membrane receptor proteins.
- Published
- 2003
- Full Text
- View/download PDF
43. Bikunin target genes in ovarian cancer cells identified by microarray analysis.
- Author
-
Suzuki M, Kobayashi H, Tanaka Y, Hirashima Y, Kanayama N, Takei Y, Saga Y, Suzuki M, Itoh H, and Terao T
- Subjects
- Cell Movement drug effects, Female, Gene Expression Profiling, Gene Expression Regulation, Humans, Membrane Glycoproteins pharmacology, Oligonucleotide Array Sequence Analysis, Ovarian Neoplasms genetics, Pregnancy-Associated Plasma Protein-A antagonists & inhibitors, Serine Endopeptidases drug effects, Serine Proteinase Inhibitors genetics, Serine Proteinase Inhibitors pharmacology, Serine Proteinase Inhibitors physiology, Transfection, Trypsin drug effects, Tumor Cells, Cultured, Membrane Glycoproteins physiology, Ovarian Neoplasms pathology, Trypsin Inhibitor, Kunitz Soybean
- Abstract
Bikunin, a Kunitz-type protease inhibitor, could potentially suppress tumor cell invasion and metastasis. Our previous study revealed that overexpression of bikunin in a human ovarian cancer cell line, HRA, resulted in a down-regulation in uPA and uPAR gene expression. For identifying the full repertoire of bikunin-regulated genes, a cDNA microarray hybridization screening was conducted using mRNA from bikunin-treated or bikunin-transfected HRA cells. A number of bikunin-regulated genes were identified, and their regulation was confirmed by Northern blot analysis. Our screen identified 11 bikunin-stimulated genes and 29 bikunin-repressed genes. The identified genes can indeed be classified into distinct subsets. These include transcriptional regulators, oncogenes/tumor suppressor genes, signaling molecules, growth/cell cycle, invasion/metastasis, cytokines, apoptosis, ion channels, extracellular matrix proteins, as well as some proteases. This screen identified suppression of several genes such as CDC-like kinase, LIM domain binding, Ets domain transcription factor, Rho GTPase-activating protein, tyrosine phosphorylation-regulated kinase, hyaluronan-binding protein, matriptase, and pregnancy-associated plasma protein-A (PAPP-A), which have previously been implicated in enhancing tumor promotion. Northern blot analysis confirmed that several genes including matriptase and PAPP-A were down-regulated by bikunin by approximately 9-fold. Further, genetic inhibition of matriptase or PAPP-A could lead to diminished invasion. These results show that bikunin alters the pattern of gene expression in HRA cells leading to a block in cell invasion.
- Published
- 2003
- Full Text
- View/download PDF
44. Enhanced expression of the human vacuolar H+-ATPase c subunit gene (ATP6L) in response to anticancer agents.
- Author
-
Torigoe T, Izumi H, Ishiguchi H, Uramoto H, Murakami T, Ise T, Yoshida Y, Tanabe M, Nomoto M, Itoh H, and Kohno K
- Subjects
- Anti-Bacterial Agents pharmacology, Apoptosis, Base Sequence, Binding Sites, Blotting, Northern, Caspase 3, Caspases metabolism, Cell Membrane metabolism, Cell Nucleus metabolism, Chromatin metabolism, Cisplatin pharmacology, DNA metabolism, DNA Damage, DNA Fragmentation, DNA-Binding Proteins metabolism, Enzyme Inhibitors pharmacology, Gene Deletion, Genes, Reporter, Humans, Luciferases metabolism, Mitochondrial Proton-Translocating ATPases, Models, Genetic, Molecular Sequence Data, Mutation, Organic Cation Transporter 1 metabolism, Plasmids metabolism, Precipitin Tests, Promoter Regions, Genetic, RNA, Messenger metabolism, Sp1 Transcription Factor metabolism, Sp3 Transcription Factor, Time Factors, Topoisomerase II Inhibitors, Transcription Factors metabolism, Transcription, Genetic, Transfection, Tumor Cells, Cultured, Up-Regulation, Vacuolar Proton-Translocating ATPases metabolism, Adenosine Triphosphatases biosynthesis, Adenosine Triphosphatases genetics, Antineoplastic Agents pharmacology, Macrolides
- Abstract
We have isolated two overlapping genomic clones that contain the 5'-terminal portion of the human vacuolar H(+)-ATPase c subunit (ATP6L) gene. The sequence preceding the transcription initiation site, which is GC-rich, contains four GC boxes and one Oct1-binding site, but there is no TATA box or CCAAT box. In vivo footprint analysis in human cancer cells shows that two GC boxes and the Oct1-binding site are occupied by Sp1 and Oct1, respectively. We show here that treatment with anticancer agents enhances ATP6L expression. Although cisplatin did not induce ATP6L promoter activity, it altered ATP6L mRNA stability. On the other hand, the DNA topoisomerase II inhibitor, TAS-103, strongly induced promoter activity, and this effect was completely eradicated when a mutation was introduced into the Oct1-binding site. Treatment with TAS-103 increased the levels of both Sp1/Sp3 and Oct1 in nuclear extracts. Cooperative binding of Sp1 and Oct1 to the promoter is required for promoter activation by TAS-103. Incubation of a labeled oligonucleotide probe encompassing the -73/-68 GC box and -64/-57 Oct1-binding site with a nuclear extract from drug-treated KB cells yielded higher levels of the specific DNA-protein complex than an extract of untreated cells. Thus, the two transcription factors, Sp1 and Oct1 interact, in an adaptive response to DNA damage, by up-regulating expression of the vacuolar H(+)-ATPase genes. Furthermore, combination of the vacuolar H(+)-ATPase (V-ATPase) inhibitor, bafilomycin A1, with TAS-103 enhanced apoptosis of KB cells with an associated increase in caspase-3 activity. Our data suggest that the induction of V-ATPase expression is an anti-apoptotic defense, and V-ATPase inhibitors in combination with low-dose anticancer agents may provide a new therapeutic approach.
- Published
- 2002
- Full Text
- View/download PDF
45. 5'-,3'-inverted thymidine-modified antisense oligodeoxynucleotide targeting midkine. Its design and application for cancer therapy.
- Author
-
Takei Y, Kadomatsu K, Itoh H, Sato W, Nakazawa K, Kubota S, and Muramatsu T
- Subjects
- Animals, Apoptosis, Drug Design, Drug Stability, Male, Mice, Midkine, Neoplasms, Experimental drug therapy, Oligodeoxyribonucleotides, Antisense pharmacokinetics, Oligodeoxyribonucleotides, Antisense therapeutic use, Tumor Cells, Cultured, Antineoplastic Agents pharmacology, Carrier Proteins antagonists & inhibitors, Cytokines, Oligodeoxyribonucleotides, Antisense pharmacology
- Abstract
Oligodeoxynucleotides modified at both 5'- and 3'-ends with inverted thymidine (5'-,3'-inverted T) were introduced as new reagents for antisense strategies. These modifications were performed to make the oligodeoxynucleotides resistant to nucleases. The effectiveness of these oligodeoxynucleotides was evaluated in terms of inhibition of synthesis of midkine (MK), a heparin-binding growth factor, and consequent inhibition of growth of CMT-93 mouse rectal carcinoma cells. 5'-,3'-Inverted T antisense MK suppressed synthesis of MK by CMT-93 cells and their growth in culture. Furthermore, 5'-,3'-inverted T oligodeoxynucleotides exhibited less cytotoxicity and better stability than phosphorothioate oligodeoxynucleotides. When 5'-,3'-inverted T antisense MK was mixed with atelocollagen, and injected into CMT-93 tumors pregrown in nude mice, tumor growth was markedly suppressed as compared with tumors injected with sense controls. The suppressive effect of 5'-,3'-inverted T antisense MK on tumor growth was stronger than that of phosphorothioate antisense MK. These findings indicated the usefulness of inverted thymidine-modified antisense oligodeoxynucleotides as a new reagent instead of phosphorothioate-modified oligodeoxynucleotides.
- Published
- 2002
- Full Text
- View/download PDF
46. Galpha11 induces caspase-mediated proteolytic activation of Rho-associated kinase, ROCK-I, in HeLa cells.
- Author
-
Ueda H, Morishita R, Itoh H, Narumiya S, Mikoshiba K, Kato K, and Asano T
- Subjects
- Calcium metabolism, Enzyme Activation, HeLa Cells, Humans, Intracellular Signaling Peptides and Proteins, Protein Kinase C physiology, rho-Associated Kinases, Caspases physiology, Heterotrimeric GTP-Binding Proteins physiology, Protein Serine-Threonine Kinases metabolism
- Abstract
Expression of the constitutively active mutant of Galpha(11) (Galpha(11)QL) induces the formation of vinculin-containing focal adhesion-like structures in HeLa cells. This was found to be inhibited by Y-27632, a specific inhibitor of Rho-associated kinases (ROCK), but not by co-expression with a dominant negative mutant of RhoA, suggesting Rho-independent activation of ROCK by Galpha(11)QL. Investigation of trypan blue exclusion and immunocytochemistry with an antibody against cleaved caspase revealed the cellular phenotype of Galpha(11)QL-expressing cells to be identical to that displayed by cells undergoing apoptosis, and the caspase inhibitor zVAD-fmk blocked all morphological changes induced by Galpha(11)QL. Transfection of Galpha(11)QL induced cleavage of ROCK-I, and this proteolysis was also prevented by zVAD-fmk. ROCK-I C-terminally truncated at its authentic caspase sites also induced the formation of vinculin-containing focal adhesion-like structures. In addition, cleavage of ROCK-I was observed when cells overexpressing m1 muscarinic acetylcholine receptors were stimulated with carbachol. These results suggest that Galpha(11) induces proteolytic activation of ROCK-I by caspase and thereby regulates the actin cytoskeleton during apoptosis.
- Published
- 2001
- Full Text
- View/download PDF
47. Parallel regulation of mitogen-activated protein kinase kinase 3 (MKK3) and MKK6 in Gq-signaling cascade.
- Author
-
Yamauchi J, Tsujimoto G, Kaziro Y, and Itoh H
- Subjects
- Calcium-Calmodulin-Dependent Protein Kinases physiology, Cell Line, GTP-Binding Protein alpha Subunits, Gq-G11, Humans, MAP Kinase Kinase 3, MAP Kinase Kinase 6, Mitogen-Activated Protein Kinase Kinases physiology, Models, Biological, Phosphatidylinositol 3-Kinases physiology, Protein-Tyrosine Kinases physiology, Proto-Oncogene Proteins p21(ras) physiology, Proto-Oncogene Proteins pp60(c-src) physiology, Receptor, Muscarinic M1, Receptors, Muscarinic metabolism, Type C Phospholipases physiology, p38 Mitogen-Activated Protein Kinases, rho GTP-Binding Proteins physiology, Calcium-Calmodulin-Dependent Protein Kinases metabolism, GTP-Binding Protein beta Subunits, GTP-Binding Protein gamma Subunits, Heterotrimeric GTP-Binding Proteins metabolism, MAP Kinase Kinase 4, MAP Kinase Signaling System, Mitogen-Activated Protein Kinase Kinases metabolism, Mitogen-Activated Protein Kinases metabolism, Protein-Tyrosine Kinases metabolism
- Abstract
Heterotrimeric G protein G(q) stimulates the activity of p38 mitogen-activated protein kinase (MAPK) in mammalian cells. To investigate the signaling mechanism whereby alpha and betagamma subunits of G(q) activate p38 MAPK, we introduced kinase-deficient mutants of mitogen-activated protein kinase kinase 3 (MKK3), MKK4, and MKK6 into human embryonal kidney 293 cells. The activation of p38 MAPK by Galpha(q) and Gbetagamma was blocked by kinase-deficient MKK3 and MKK6 but not by kinase-deficient MKK4. In addition, Galpha(q) and Gbetagamma stimulated MKK3 and MKK6 activities. The MKK3 and MKK6 activations by Galpha(q), but not by Gbetagamma, were dependent on phospholipase C and c-Src. Galpha(q) stimulated MKK3 in a Rac- and Cdc42-dependent manner and MKK6 in a Rho-dependent manner. On the other hand, Gbetagamma activated MKK3 in a Rac- and Cdc42-dependent manner and MKK6 in a Rho-, Rac-, and Cdc42-dependent manner. Gbetagamma-induced MKK3 and MKK6 activations were dependent on a tyrosine kinase other than c-Src. These results suggest that Galpha(q) and Gbetagamma stimulate the activity of p38 MAPK by regulating MKK3 and MKK6 through parallel signaling pathways.
- Published
- 2001
- Full Text
- View/download PDF
48. Regulation of Rac and Cdc42 pathways by G(i) during lysophosphatidic acid-induced cell spreading.
- Author
-
Ueda H, Morishita R, Yamauchi J, Itoh H, Kato K, and Asano T
- Subjects
- 3T3 Cells, Animals, Fibroblasts drug effects, Fibroblasts physiology, Guanosine Triphosphate metabolism, Mice, Pertussis Toxin, Phosphorylation, Protein Subunits, Virulence Factors, Bordetella pharmacology, GTP-Binding Protein alpha Subunits, Gi-Go physiology, Lysophospholipids pharmacology, cdc42 GTP-Binding Protein physiology, rac GTP-Binding Proteins physiology
- Abstract
The pertussis toxin-sensitive G protein, G(i), has been implicated in lysophosphatidic acid-induced cell mitogenesis and migration, but the mechanisms remain to be detailed. In the present study, we found that pertussis toxin blocks lysophosphatidic acid-induced cell spreading of NIH 3T3 fibroblasts on fibronectin. This prevention of cell spreading was eliminated by the expression of constitutively active mutants of Rho family small GTP-binding proteins, Rac and Cdc42, but not by Rho. In addition, activation of the endogenous forms was suppressed by pertussis toxin, indicating that G(i)-induced cell spreading is mediated through the Rac and Cdc42 pathway. Transfection of constitutively active mutants of G alpha(i) and G alpha(11) and G beta gamma subunits enhanced spreading of pertussis toxin-treated cells. G beta(1) with G gamma(12), a major G gamma form in fibroblasts, was more effective for increasing cell spreading than G beta(1)gamma(2) or G beta(1) plus G gamma(12)S2A, a mutant in which Ser-2, a phosphorylation site for protein kinase C, is replaced with alanine. In addition, a protein kinase C inhibitor diminished G beta(1)gamma(12)-induced cell spreading, suggesting a role for phosphorylation of the protein. These findings indicate that both G alpha(i) and G beta gamma stimulate Rac and Cdc42 pathways with lysophosphatidic acid-induced cell spreading on fibronectin.
- Published
- 2001
- Full Text
- View/download PDF
49. Hepatocyte growth factor activator inhibitor type 1 is a specific cell surface binding protein of hepatocyte growth factor activator (HGFA) and regulates HGFA activity in the pericellular microenvironment.
- Author
-
Kataoka H, Shimomura T, Kawaguchi T, Hamasuna R, Itoh H, Kitamura N, Miyazawa K, and Koono M
- Subjects
- Animals, Base Sequence, CHO Cells, Cricetinae, DNA Primers, Epithelial Cells metabolism, Humans, Immunohistochemistry, Proteinase Inhibitory Proteins, Secretory, Tetradecanoylphorbol Acetate pharmacology, Tumor Cells, Cultured, Up-Regulation, Membrane Glycoproteins metabolism, Membrane Proteins metabolism, Serine Endopeptidases metabolism
- Abstract
Hepatocyte growth factor activator (HGFA) is responsible for proteolytic activation of the precursor form of hepatocyte growth factor in injured tissues. To date, two specific inhibitors of HGFA have been identified, namely HGFA inhibitor type 1 (HAI-1) and type 2 (HAI-2)/placental bikunin (PB). Both inhibitors are first synthesized as integral membrane proteins having two Kunitz domains and a transmembrane domain, and are subsequently released from cell surface by shedding. Here we show that an active form of HGFA is specifically complexed with membrane-form HAI-1, but not with HAI-2/PB, on the surface of epithelial cells expressing both inhibitors. This binding required the enzyme activity of HGFA. The selective binding of HGFA to the cell surface HAI-1 was further confirmed in an engineered system using Chinese hamster ovary cells, in which only the cells expressing HAI-1 retained exogenous HGFA. The binding of HGFA to HAI-1 was reversible, and no irreversible modifications affecting the enzyme activity occurred during the binding. Importantly, HAI-1 and the HGFA.HAI-1 complex were quickly released from the cell surface by treatment with phorbol 12-myristate 13-acetate or interleukin 1beta accompanying the generation of 58-kDa fragments of HAI-1, which are less potent against HGFA, as well as significant recovery of HGFA activity in the culture supernatant. This regulated shedding was completely inhibited by BB3103, a synthetic zinc-metalloproteinase inhibitor. We conclude that HAI-1 is not only an inhibitor but also a specific acceptor of active HGFA, acting as a reservoir of this enzyme on the cell surface. The latter property appears to ensure the concentrated pericellular HGFA activity in certain cellular conditions, such as tissue injury and inflammation, via the up-regulated shedding of HGFA.HAI-1 complex. These findings shed light on a novel function of the integral membrane Kunitz-type inhibitor in the regulation of pericellular proteinase activity.
- Published
- 2000
- Full Text
- View/download PDF
50. Ras oncoprotein induces CD44 cleavage through phosphoinositide 3-OH kinase and the rho family of small G proteins.
- Author
-
Kawano Y, Okamoto I, Murakami D, Itoh H, Yoshida M, Ueda S, and Saya H
- Subjects
- 3T3 Cells, Animals, CHO Cells, Cricetinae, Mice, Hyaluronan Receptors physiology, Phosphatidylinositol 3-Kinases physiology, Signal Transduction, ras Proteins physiology, rho GTP-Binding Proteins physiology
- Abstract
CD44 is a cell surface adhesion molecule for several extracellular matrix components. We previously showed that CD44 expressed in cancer cells is proteolytically cleaved at the ectodomain through membrane-anchored metalloproteases and that CD44 cleavage plays a critical role in cancer cell migration. Therefore, cellular signals that promote the migration and metastatic activity of cancer cells may regulate the CD44 ectodomain cleavage. Here, we demonstrate that the expression of the dominant active mutant of Ha-Ras (Ha-Ras(Val-12)) induces redistribution of CD44 to the newly generated membrane ruffling area and CD44 ectodomain cleavage. The migration assay revealed that the CD44 cleavage contributes to the Ha-Ras(Val-12)-induced migration of NIH3T3 cells on hyaluronate substrate. Treatment with LY294002, an inhibitor for phosphoinositide 3-OH kinase (PI3K), significantly inhibits Ha-Ras(Val-12)-induced CD44 cleavage, whereas that with PD98059, an inhibitor for MEK, does not. The active mutant p110 subunit of PI3K has also been shown to enhance the CD44 cleavage, suggesting that PI3K mediates the Ras-induced CD44 cleavage. Moreover, the expression of dominant negative mutants of Cdc42 and Rac1 inhibits the Ha-Ras(Val-12)-induced CD44 cleavage. These results suggest that Ras > PI3K > Cdc42/Rac1 pathway plays an important role in CD44 cleavage and may provide a novel molecular basis to explain how the activated Ras facilitates cancer cell migration.
- Published
- 2000
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.