33 results on '"Murakami, S"'
Search Results
2. The interleukin-8 AP-1 and kappa B-like sites are genetic end targets of FK506-sensitive pathway accompanied by calcium mobilization.
- Author
-
Okamoto, S., primary, Mukaida, N., additional, Yasumoto, K., additional, Rice, N., additional, Ishikawa, Y., additional, Horiguchi, H., additional, Murakami, S., additional, and Matsushima, K., additional
- Published
- 1994
- Full Text
- View/download PDF
3. Structure and function of the intracellular portion of the mouse interleukin 1 receptor (type I). Determining the essential region for transducing signals to activate the interleukin 8 gene
- Author
-
Kuno, K., primary, Okamoto, S., additional, Hirose, K., additional, Murakami, S., additional, and Matsushima, K., additional
- Published
- 1993
- Full Text
- View/download PDF
4. Tumor necrosis factor alpha and interferon gamma synergistically induce interleukin 8 production in a human gastric cancer cell line through acting concurrently on AP-1 and NF-kB-like binding sites of the interleukin 8 gene.
- Author
-
Yasumoto, K, primary, Okamoto, S, additional, Mukaida, N, additional, Murakami, S, additional, Mai, M, additional, and Matsushima, K, additional
- Published
- 1992
- Full Text
- View/download PDF
5. Hepatitis B virus X protein transactivates human interleukin-8 gene through acting on nuclear factor kB and CCAAT/enhancer-binding protein-like cis-elements
- Author
-
Mahé, Y., primary, Mukaida, N., additional, Kuno, K., additional, Akiyama, M., additional, Ikeda, N., additional, Matsushima, K., additional, and Murakami, S., additional
- Published
- 1991
- Full Text
- View/download PDF
6. Potent inhibition of the master chondrogenic factor Sox9 gene by interleukin-1 and tumor necrosis factor-alpha.
- Author
-
Murakami, S, Lefebvre, V, and de Crombrugghe, B
- Abstract
The inflammatory cytokines interleukin-1 (IL-1) and tumor necrosis factor-alpha (TNF-alpha) strongly inhibit the expression of genes for cartilage extracellular matrix proteins. We have recently obtained genetic evidence indicating that the high mobility group domain containing transcription factor Sox9 is required for cartilage formation and for expression of chondrocyte-specific genes including the gene for type II collagen (Col2a1). We show here that IL-1 and TNF-alpha cause a marked and rapid decrease in the levels of Sox9 mRNA and/or protein in chondrocytes. A role for the transcription factor NFkappaB in Sox9 down-regulation was suggested by the ability of pyrrolidine dithiocarbamate, an inhibitor of the NFkappaB pathway, to block the effects of IL-1 and TNF-alpha. This role was further supported by the ability of a dominant-negative mutant of IkappaBalpha to block the IL-1 and TNF-alpha inhibition of Sox9-dependent Col2a1 enhancer elements. Furthermore, forced expression of the NFkappaB subunits p65 or p50 also inhibited Sox9-dependent Col2a1 enhancer. Because Sox9 is essential for chondrogenesis, the marked down-regulation of the Sox9 gene by IL-1 and TNF-alpha in chondrocytes is sufficient to account for the inhibition of the chondrocyte phenotype by these cytokines. The down-regulation of Sox9 may have a crucial role in inhibiting expression of the cartilage phenotype in inflammatory joint diseases.
- Published
- 2000
7. The hepatitis B virus X protein is a co-activator of activated transcription that modulates the transcription machinery and distal binding activators.
- Author
-
Lin, Y, Tang, H, Nomura, T, Dorjsuren, D, Hayashi, N, Wei, W, Ohta, T, Roeder, R, and Murakami, S
- Abstract
Hepatitis B virus X protein (HBx) transactivates viral and cellular genes through a wide variety of cis-elements, but the mechanism has not been well elucidated. Evidence for nuclear events in HBx transactivation has been reported. Here we examine the role of HBx in modulation of transcription with a transient transfection system and an in vitro transcription assay. Reporters bearing Gal4-binding sites were applied to avoid the effects of endogenous transcription factors with or without signaling processes. The Gal4-DNA binding domain fused form of HBx exhibited no effect on Gal4-responsive reporters. However, HBx augmented activated transcription by transcriptional activators, suggesting HBx retains a co-activator but not a transcriptional activator function. The functional domain for co-activation was the same as that for HBx transactivation, and the transcription factor IIB- and RNA polymerase II subunit 5-interacting sites of HBx, which were critical for HBx transactivation, were shown to be crucial for the co-activation function. Importantly, HBx stimulated transcription on templates bearing the X responsive elements in vitro with endogenous activators. These results imply that HBx acts as a co-activator that modulates transcriptional machinery and distal-binding activators, which may explain one of the mechanisms of transactivation by HBx when localized in nuclei.
- Published
- 1998
8. RNA-dependent RNA polymerase activity of the soluble recombinant hepatitis C virus NS5B protein truncated at the C-terminal region.
- Author
-
Yamashita, T, Kaneko, S, Shirota, Y, Qin, W, Nomura, T, Kobayashi, K, and Murakami, S
- Abstract
The hepatitis C virus (HCV) NS5B protein encodes an RNA-dependent RNA polymerase (RdRP), which is the central catalytic enzyme of HCV replicase. We established a new method to purify soluble HCV NS5B in the glutathione S-transferase-fused form NS5Bt from Escherichia coli which lacks the C-terminal 21 amino acid residues encompassing a putative anchoring domain (anino acids 2990-3010). The recombinant soluble protein exhibited RdRP activity in vitro which was dependent upon the template and primer, but it did not exhibit the terminal transferase activity that has been reported to be associated with the recombinant NS5B protein from insect cells. The RdRP activity of purified glutathione S-transferase-NS5Bt and thrombin-cleavaged non-fused NS5Bt shares most of the properties. Substitution mutations of NS5Bt at the GDD motif, which is highly conserved among viral RdRPs, and at the clustered basic residues (amino acids 2919-2924 and 2693-2699) abolished the RdRP activity. The C-terminal region of NS5B, which is dispensable for the RdRP activity, dramatically affected the subcellular localization of NS5B retaining it in perinuclear sites in transiently overexpressed mammalian cells. These results may provide some clues to dissecting the molecular mechanism of the HCV replication and also act as a basis for developing new anti-viral drugs.
- Published
- 1998
9. Novel genes encoding 2-aminophenol 1,6-dioxygenase from Pseudomonas species AP-3 growing on 2-aminophenol and catalytic properties of the purified enzyme.
- Author
-
Takenaka, S, Murakami, S, Shinke, R, Hatakeyama, K, Yukawa, H, and Aoki, K
- Abstract
2-Aminophenol 1,6-dioxygenase was purified from the cell extracts of Pseudomonas sp. AP-3 grown on 2-aminophenol. The product from 2-aminophenol by catalysis of the purified enzyme was identified as 2-aminomuconic 6-semialdehyde by gas chromatographic and mass spectrometric analyses. The molecular mass of the native enzyme was 140 kDa based on gel filtration. It was dissociated into molecular mass subunits of 32 (alpha-subunit) and 40 kDa (beta-subunit) by sodium dodecyl sulfate-polyacrylamide gel electrophoresis, indicating that the dioxygenase was a heterotetramer of alpha2beta2. The genes coding for the alpha- and beta-subunits of the enzyme were cloned and sequenced. Open reading frames of the genes (amnA and amnB) were 816 and 918 base pairs in length, respectively. The amino acid sequences predicted from the open reading frames of amnA and amnB corresponded to the NH2-terminal amino acid sequences of the alpha-subunit (AmnA) and beta-subunit (AmnB), respectively. The deduced amino acid sequences of AmnB showed identities to some extent with HpaD (25.4%) and HpcB (24.4%) that are homoprotocatechuate 2,3-dioxygenases from Escherichia coli W and C, respectively, belonging to class III in the extradiol dioxygenases. On the other hand, AmnA had identity (23.3%) with only AmnB among the enzymes examined.
- Published
- 1997
10. Hepatitis B virus X protein is a transcriptional modulator that communicates with transcription factor IIB and the RNA polymerase II subunit 5.
- Author
-
Lin, Y, Nomura, T, Cheong, J, Dorjsuren, D, Iida, K, and Murakami, S
- Abstract
Hepatitis B virus X protein (HBx) transactivates viral and cellular genes through a wide variety of cis-elements. However, the mechanism is still obscure. Our finding that HBx directly interacts with RNA polymerase II subunit 5 (RPB5), a common subunit of RNA polymerases, implies that HBx directly modulates the function of RNA polymerase (Cheong, J. H., Yi, M., Lin, Y., and Murakami, S. (1995) EMBO J. 14, 142-150). In this context, we examined the possibility that HBx and RPB5 interact with other general transcription factors. HBx and RPB5 specifically bound to transcription factor IIB (TFIIB) in vitro, both of which were detected by either far-Western blotting or the glutathione S-transferase-resin pull-down assay. Delineation of the binding regions of these three proteins revealed that HBx, RPB5, and TFIIB each has two binding regions for the other two proteins. Co-immunoprecipitation using HepG2 cell lysates that express HBx demonstrated trimeric interaction in vivo. Some HBx substitution mutants, which had severely impaired transacting activity, exhibited reduced binding affinity with either TFIIB or RPB5 in a mutually exclusive manner, suggesting that HBx transactivation requires the interactions of both RPB5 and TFIIB. These results indicated that HBx is a novel virus modulator that facilitates transcriptional initiation by stabilizing the association between RNA polymerase and TFIIB through communication with RPB5 and TFIIB.
- Published
- 1997
11. Characterization of Rab32- and Rab38-positive lysosome-related organelles in osteoclasts and macrophages.
- Author
-
Noda K, Lu SL, Chen S, Tokuda K, Li Y, Hao F, Wada Y, Sun-Wada GH, Murakami S, Fukuda M, Itoh T, and Noda T
- Abstract
Both the biogenesis and functions of osteoclasts and macrophages involves dynamic membrane traffic. We screened transcript levels for Rab family small GTPases related to osteoclasts and identified Rab38. Rab38 expression is upregulated during osteoclast differentiation and maturation. In osteoclasts, both Rab38 and its paralog, Rab32, colocalize to lysosome-related organelles (LROs). In macrophages, Rab32 is also found in LROs. LROs are part of the endocytic pathway but are distinct from lysosomes. After receptor activator of NF-κB ligand stimulation, LROs contain cathepsin K and tartrate-resistant acid phosphatase inside and help both proteins to accumulate around bone resorption pits. After osteoclast maturation, these enzymes are hardly found within LROs. In macrophages derived from Rab32 and Rab38 double knockout mice, both acidification and V-ATPase a3 localization were severely compromised. Both the double knockout macrophage and bafilomycin-treated wildtype macrophage show an increase in Lamp1-positive organelles, implying that biogenesis of lysosomes and LROs are related. These results indicate that Rab32 and Rab38 both play a crucial role in LRO biogenesis in macrophages and in osteoclasts., Competing Interests: Conflict of interest The authors declare no conflict of interest with the contents of this article., (Copyright © 2023 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
12. Glucocorticoid receptor signaling represses the antioxidant response by inhibiting histone acetylation mediated by the transcriptional activator NRF2.
- Author
-
Alam MM, Okazaki K, Nguyen LTT, Ota N, Kitamura H, Murakami S, Shima H, Igarashi K, Sekine H, and Motohashi H
- Subjects
- Acetylation drug effects, Animals, Histones genetics, Kelch-Like ECH-Associated Protein 1 genetics, Kelch-Like ECH-Associated Protein 1 metabolism, Membrane Proteins genetics, Membrane Proteins metabolism, Mice, Mice, Knockout, NF-E2-Related Factor 2 genetics, Oxidative Stress drug effects, Phosphoproteins genetics, Phosphoproteins metabolism, Receptors, Glucocorticoid genetics, Dexamethasone pharmacology, Histones metabolism, NF-E2-Related Factor 2 metabolism, Receptors, Glucocorticoid metabolism, Signal Transduction drug effects
- Abstract
NRF2 (nuclear factor erythroid 2-related factor 2) is a key transcriptional activator that mediates the inducible expression of antioxidant genes. NRF2 is normally ubiquitinated by KEAP1 (Kelch-like ECH-associated protein 1) and subsequently degraded by proteasomes. Inactivation of KEAP1 by oxidative stress or electrophilic chemicals allows NRF2 to activate transcription through binding to antioxidant response elements (AREs) and recruiting histone acetyltransferase CBP (CREB-binding protein). Whereas KEAP1-dependent regulation is a major determinant of NRF2 activity, NRF2-mediated transcriptional activation varies from context to context, suggesting that other intracellular signaling cascades may impact NRF2 function. To identify a signaling pathway that modifies NRF2 activity, we immunoprecipitated endogenous NRF2 and its interacting proteins from mouse liver and identified glucocorticoid receptor (GR) as a novel NRF2-binding partner. We found that glucocorticoids, dexamethasone and betamethasone, antagonize diethyl maleate-induced activation of NRF2 target genes in a GR-dependent manner. Dexamethasone treatment enhanced GR recruitment to AREs without affecting chromatin binding of NRF2, resulting in the inhibition of CBP recruitment and histone acetylation at AREs. This repressive effect was canceled by the addition of histone deacetylase inhibitors. Thus, GR signaling decreases NRF2 transcriptional activation through reducing the NRF2-dependent histone acetylation. Consistent with these observations, GR signaling blocked NRF2-mediated cytoprotection from oxidative stress. This study suggests that an impaired antioxidant response by NRF2 and a resulting decrease in cellular antioxidant capacity account for the side effects of glucocorticoids, providing a novel viewpoint for the pathogenesis of hypercorticosteroidism., (© 2017 by The American Society for Biochemistry and Molecular Biology, Inc.)
- Published
- 2017
- Full Text
- View/download PDF
13. DHHC7 Palmitoylates Glucose Transporter 4 (Glut4) and Regulates Glut4 Membrane Translocation.
- Author
-
Du K, Murakami S, Sun Y, Kilpatrick CL, and Luscher B
- Subjects
- 3T3-L1 Cells, Acyltransferases genetics, Adipocytes metabolism, Animals, Cell Membrane metabolism, Glucose Tolerance Test, HEK293 Cells, Humans, Hyperglycemia metabolism, Insulin metabolism, Mice, Mice, Inbred C57BL, Mice, Knockout, Protein Transport, Acyltransferases metabolism, Glucose Transporter Type 4 metabolism, Palmitic Acid metabolism
- Abstract
Insulin-dependent translocation of glucose transporter 4 (Glut4) to the plasma membrane plays a key role in the dynamic regulation of glucose homeostasis. We recently showed that this process is critically dependent on palmitoylation of Glut4 at Cys-223. To gain further insights into the regulation of Glut4 palmitoylation, we set out to identify the palmitoyl acyltransferase (PAT) involved. Here we report that among 23 mammalian DHHC proteins, DHHC7 is the major Glut4 PAT, based on evidence that ectopic expression of DHHC7 increased Glut4 palmitoylation, whereas DHHC7 knockdown in 3T3-L1 adipocytes and DHHC7 KO in adipose tissue and muscle decreased Glut4 palmitoylation. Moreover, inactivation of DHHC7 suppressed insulin-dependent Glut4 membrane translocation in both 3T3-L1 adipocytes and primary adipocytes. Finally, DHHC7 KO mice developed hyperglycemia and glucose intolerance, thereby confirming that DHHC7 represents the principal PAT for Glut4 and that this mechanism is essential for insulin-regulated glucose homeostasis., (© 2017 by The American Society for Biochemistry and Molecular Biology, Inc.)
- Published
- 2017
- Full Text
- View/download PDF
14. Dissociation of Golgi-associated DHHC-type Zinc Finger Protein (GODZ)- and Sertoli Cell Gene with a Zinc Finger Domain-β (SERZ-β)-mediated Palmitoylation by Loss of Function Analyses in Knock-out Mice.
- Author
-
Kilpatrick CL, Murakami S, Feng M, Wu X, Lal R, Chen G, Du K, and Luscher B
- Subjects
- Animals, Brain cytology, Cells, Cultured, Female, Golgi Apparatus metabolism, Lipoylation, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Neurons cytology, Protein Transport, Synapses, Zinc Fingers, Brain metabolism, Intracellular Signaling Peptides and Proteins physiology, Membrane Proteins physiology, Neurons metabolism, Palmitates metabolism, Protein Processing, Post-Translational, Receptors, GABA-A metabolism
- Abstract
The γ2 subunit of GABA type A receptors (GABA
A Rs) is thought to be subject to palmitoylation by both Golgi-associated DHHC-type zinc finger protein (GODZ; also known as DHHC3) and its paralog Sertoli cell gene with a zinc finger domain-β (SERZ-β; DHHC7) based on overexpression of enzymes and substrates in heterologous cells. Here we have further investigated the substrate specificity of these enzymes by characterization of GODZ and SERZ-β knock-out (KO) mice as well as double KO (DKO) neurons. Palmitoylation of γ2 and a second substrate, growth-associated protein of 43 kDa, that is independently implicated in trafficking of GABAA Rs was significantly reduced in brain of GODZ KO versus wild-type (WT) mice but unaltered in SERZ-β KO mice. Accumulation of GABAA Rs at synapses, GABAergic innervation, and synaptic function were reduced in GODZ KO and DKO neurons to a similar extent, indicating that SERZ-β does not contribute to palmitoylation or trafficking of GABAA Rs even in the absence of GODZ. Notably, these effects were seen only when mutant neurons were grown in competition with WT neurons, thereby mimicking conditions of shRNA-transfected neurons previously used to characterize GODZ. However, GABA-evoked whole-cell currents of DKO neurons and the GABAA R cell surface expression in DKO neurons and GODZ or SERZ-β KO brain slices were unaltered, indicating that GODZ-mediated palmitoylation selectively controls the pool of receptors at synapses. The different substrate specificities of GODZ and SERZ-β in vivo were correlated with their differential localization to cis- versus trans-Golgi compartment, a mechanism that was compromised by overexpression of GODZ., (© 2016 by The American Society for Biochemistry and Molecular Biology, Inc.)- Published
- 2016
- Full Text
- View/download PDF
15. β-Lactam selectivity of multidrug transporters AcrB and AcrD resides in the proximal binding pocket.
- Author
-
Kobayashi N, Tamura N, van Veen HW, Yamaguchi A, and Murakami S
- Subjects
- Anti-Bacterial Agents chemistry, Binding Sites, Carrier Proteins metabolism, Cell Membrane drug effects, Crystallography, X-Ray, Escherichia coli metabolism, Mutagenesis, Site-Directed, Plasmids metabolism, Protein Binding, Protein Conformation, Protein Transport, Substrate Specificity, Drug Resistance, Multiple, Bacterial, Escherichia coli drug effects, Escherichia coli Proteins metabolism, Membrane Transport Proteins metabolism, Multidrug Resistance-Associated Proteins metabolism, beta-Lactams chemistry
- Abstract
β-Lactams are mainstream antibiotics that are indicated for the prophylaxis and treatment of bacterial infections. The AcrA-AcrD-TolC multidrug efflux system confers much stronger resistance on Escherichia coli to clinically relevant anionic β-lactam antibiotics than the homologous AcrA-AcrB-TolC system. Using an extensive combination of chimeric analysis and site-directed mutagenesis, we searched for residues that determine the difference in β-lactam specificity between AcrB and AcrD. We identified three crucial residues at the "proximal" (or access) substrate binding pocket. The simultaneous replacement of these residues in AcrB by those in AcrD (Q569R, I626R, and E673G) transferred the β-lactam specificity of AcrD to AcrB. Our findings indicate for the first time that the difference in β-lactam specificity between AcrB and AcrD relates to interactions of the antibiotic with residues in the proximal binding pocket.
- Published
- 2014
- Full Text
- View/download PDF
16. Abundance of prereplicative complexes (Pre-RCs) facilitates recombinational repair under replication stress in fission yeast.
- Author
-
Maki K, Inoue T, Onaka A, Hashizume H, Somete N, Kobayashi Y, Murakami S, Shigaki C, Takahashi TS, Masukata H, and Nakagawa T
- Subjects
- DNA, Fungal genetics, Gene Expression Regulation, Fungal physiology, Multiprotein Complexes genetics, Mutation, Phosphorylation, Schizosaccharomyces genetics, Schizosaccharomyces pombe Proteins genetics, DNA, Fungal biosynthesis, G1 Phase physiology, Multiprotein Complexes metabolism, S Phase physiology, Schizosaccharomyces metabolism, Schizosaccharomyces pombe Proteins metabolism
- Abstract
Mcm2-7 complexes are loaded onto chromatin with the aid of Cdt1 and Cdc18/Cdc6 and form prereplicative complexes (pre-RCs) at multiple sites on each chromosome. Pre-RCs are essential for DNA replication and surviving replication stress. However, the mechanism by which pre-RCs contribute to surviving replication stress is largely unknown. Here, we isolated the fission yeast mcm6-S1 mutant that was hypersensitive to methyl methanesulfonate (MMS) and camptothecin (CPT), both of which cause forks to collapse. The mcm6-S1 mutation impaired the interaction with Cdt1 and decreased the binding of minichromosome maintenance (MCM) proteins to replication origins. Overexpression of Cdt1 restored MCM binding and suppressed the sensitivity to MMS and CPT, suggesting that the Cdt1-Mcm6 interaction is important for the assembly of pre-RCs and the repair of collapsed forks. MMS-induced Chk1 phosphorylation and Rad22/Rad52 focus formation occurred normally, whereas cells containing Rhp54/Rad54 foci, which are involved in DNA strand exchange and dissociation of the joint molecules, were increased. Remarkably, G(1) phase extension through deletion of an S phase cyclin, Cig2, as well as Cdt1 overexpression restored pre-RC assembly and suppressed Rhp54 accumulation. A cdc18 mutation also caused hypersensitivity to MMS and CPT and accumulation of Rhp54 foci. These data suggest that an abundance of pre-RCs facilitates a late step in the recombinational repair of collapsed forks in the following S phase.
- Published
- 2011
- Full Text
- View/download PDF
17. Role of mechanical stress-induced glutamate signaling-associated molecules in cytodifferentiation of periodontal ligament cells.
- Author
-
Fujihara C, Yamada S, Ozaki N, Takeshita N, Kawaki H, Takano-Yamamoto T, and Murakami S
- Subjects
- Animals, Biological Transport, Gene Expression Profiling, Homer Scaffolding Proteins, Humans, Male, Mice, Minerals metabolism, Receptors, N-Methyl-D-Aspartate metabolism, Tooth Movement Techniques, Cell Differentiation, Glutamic Acid metabolism, Periodontal Ligament cytology, Periodontal Ligament metabolism, Signal Transduction, Stress, Mechanical
- Abstract
In this study, we analyzed the effects of tensile mechanical stress on the gene expression profile of in vitro-maintained human periodontal ligament (PDL) cells. A DNA chip analysis identified 17 up-regulated genes in human PDL cells under the mechanical stress, including HOMER1 (homer homolog 1) and GRIN3A (glutamate receptor ionotropic N-methyl-d-aspartate 3A), which are related to glutamate signaling. RT-PCR and real-time PCR analyses revealed that human PDL cells constitutively expressed glutamate signaling-associated genes and that mechanical stress increased the expression of these mRNAs, leading to release of glutamate from human PDL cells and intracellular glutamate signal transduction. Interestingly, exogenous glutamate increased the mRNAs of cytodifferentiation and mineralization-related genes as well as the ALP (alkaline phosphatase) activities during the cytodifferentiation of the PDL cells. On the other hand, the glutamate signaling inhibitors riluzole and (+)-MK801 maleate suppressed the alkaline phosphatase activities and mineralized nodule formation during the cytodifferentiation and mineralization. Riluzole inhibited the mechanical stress-induced glutamate signaling-associated gene expressions in human PDL cells. Moreover, in situ hybridization analyses showed up-regulation of glutamate signaling-associated gene expressions at tension sites in the PDL under orthodontic tooth movement in a mouse model. The present data demonstrate that the glutamate signaling induced by mechanical stress positively regulates the cytodifferentiation and mineralization of PDL cells.
- Published
- 2010
- Full Text
- View/download PDF
18. NF449 is a novel inhibitor of fibroblast growth factor receptor 3 (FGFR3) signaling active in chondrocytes and multiple myeloma cells.
- Author
-
Krejci P, Murakami S, Prochazkova J, Trantirek L, Chlebova K, Ouyang Z, Aklian A, Smutny J, Bryja V, Kozubik A, and Wilcox WR
- Subjects
- Animals, Bone and Bones cytology, Bone and Bones drug effects, CHO Cells, Cell Line, Tumor, Chondrocytes drug effects, Cricetinae, Cricetulus, Female, Humans, Mice, Protein Kinases drug effects, Protein Kinases metabolism, RNA drug effects, RNA genetics, RNA, Neoplasm drug effects, RNA, Neoplasm genetics, Reverse Transcriptase Polymerase Chain Reaction, Signal Transduction drug effects, Signal Transduction physiology, Sulfates metabolism, Urinary Bladder Neoplasms physiopathology, Uterine Cervical Neoplasms physiopathology, Benzenesulfonates pharmacology, Chondrocytes physiology, Multiple Myeloma physiopathology, Receptor, Fibroblast Growth Factor, Type 3 antagonists & inhibitors
- Abstract
The FGFR3 receptor tyrosine kinase represents an attractive target for therapy due to its role in several human disorders, including skeletal dysplasias, multiple myeloma, and cervical and bladder carcinomas. By using molecular library screening, we identified a compound named NF449 with inhibitory activity toward FGFR3 signaling. In cultured chondrocytes and murine limb organ culture, NF449 rescued FGFR3-mediated extracellular matrix loss and growth inhibition, which represent two major cellular phenotypes of aberrant FGFR3 signaling in cartilage. Similarly, NF449 antagonized FGFR3 action in the multiple myeloma cell lines OPM2 and KMS11, as evidenced by NF449-mediated reversal of ERK MAPK activation and transcript accumulation of CCL3 and CCL4 chemokines, both of which are induced by FGFR3 activation. In cell-free kinase assays, NF449 inhibited the kinase activity of both wild type and a disease-associated FGFR3 mutant (K650E) in a fashion that appeared non-competitive with ATP. Our data identify NF449 as a novel antagonist of FGFR3 signaling, useful for FGFR3 inhibition alone or in combination with inhibitors that target the ATP binding site.
- Published
- 2010
- Full Text
- View/download PDF
19. Activation of polycystic kidney disease-2-like 1 (PKD2L1)-PKD1L3 complex by acid in mouse taste cells.
- Author
-
Kawaguchi H, Yamanaka A, Uchida K, Shibasaki K, Sokabe T, Maruyama Y, Yanagawa Y, Murakami S, and Tominaga M
- Subjects
- Animals, Calcium metabolism, Cell Membrane metabolism, Glutamate Decarboxylase metabolism, Humans, Mice, Mice, Inbred C57BL, Mice, Transgenic, Tongue metabolism, Acids metabolism, Calcium Channels metabolism, Ion Channels metabolism, Receptors, Cell Surface metabolism, TRPP Cation Channels metabolism, Taste, Taste Buds metabolism
- Abstract
Five basic tastes (bitter, sweet, umami, salty, and sour) are detected in the four taste areas where taste buds reside. Although molecular mechanisms for detecting bitter, sweet, and umami have been well clarified, those for sour and salty remain poorly understood. Several channels including acid-sensing ion channels have been proposed as candidate sour receptors, but they do not encompass all sour-sensing abilities in vivo. We recently reported a novel candidate for sour sensing, the polycystic kidney disease-2-like 1 (PKD2L1)-PKD1L3 channel complex. This channel is not a traditional ligand-gated channel and is gated open only after removal of an acid stimulus, called an off response. Here we show that off responses upon acid stimulus are clearly observed in native taste cells from circumvallate, but not fungiform papillae, of glutamate decarboxylase 67-green fluorescent protein (GAD67-GFP) knock-in mice, from which Type III taste cells can be visualized, using Ca(2+) imaging and patch clamp methods. Off responses were detected in most cells where PKD2L1 immunoreactivity was observed. Interestingly, the pH threshold for acid-evoked intracellular Ca(2+) increase was around 5.0, a value much higher than that observed in HEK293 cells expressing the PKD2L1-PKD1L3 complex. Thus, PKD2L1-PKD1L3-mediated acid-evoked off responses occurred both in HEK293 cells and in native taste cells, suggesting the involvement of the PKD2L1-PKD1L3 complex in acid sensing in vivo.
- Published
- 2010
- Full Text
- View/download PDF
20. PLAP-1/asporin, a novel negative regulator of periodontal ligament mineralization.
- Author
-
Yamada S, Tomoeda M, Ozawa Y, Yoneda S, Terashima Y, Ikezawa K, Ikegawa S, Saito M, Toyosawa S, and Murakami S
- Subjects
- Animals, Calcification, Physiologic, Cell Differentiation, Cell Line, Cloning, Molecular, Dental Sac metabolism, Immunohistochemistry, In Situ Hybridization, Mice, Mice, Inbred BALB C, Osteoblasts metabolism, Periodontal Ligament growth & development, Tooth growth & development, Tooth metabolism, Extracellular Matrix Proteins physiology, Gene Expression Regulation, Periodontal Ligament metabolism
- Abstract
Periodontal ligament-associated protein-1 (PLAP-1)/asporin is a recently identified novel member of the small leucine-rich repeat proteoglycan family. PLAP-1/asporin is involved in chondrogenesis, and its involvement in the pathogenesis of osteoarthritis has been suggested. We report that PLAP-1/asporin is also expressed specifically and predominantly in the periodontal ligament (PDL) and that it negatively regulates the mineralization of PDL cells. In situ hybridization analysis revealed that PLAP-1/asporin was expressed specifically not only in the PDL of an erupted tooth but also in the dental follicle, which is the progenitor tissue of the PDL during tooth development. Overexpression of PLAP-1/asporin in mouse PDL-derived clone cells interfered with both naturally and bone morphogenetic protein 2 (BMP-2)-induced mineralization of the PDL cells. On the other hand, knockdown of PLAP-1/asporin transcript levels by RNA interference enhanced BMP-2-induced differentiation of PDL cells. Furthermore co-immunoprecipitation assays showed a direct interaction between PLAP-1/asporin and BMP-2 in vitro, and immunohistochemistry staining revealed the co-localization of PLAP-1/asporin and BMP-2 at the cellular level. These results suggest that PLAP-1/asporin plays a specific role(s) in the periodontal ligament as a negative regulator of cytodifferentiation and mineralization probably by regulating BMP-2 activity to prevent the periodontal ligament from developing non-physiological mineralization such as ankylosis.
- Published
- 2007
- Full Text
- View/download PDF
21. Nucleolin interacts with telomerase.
- Author
-
Khurts S, Masutomi K, Delgermaa L, Arai K, Oishi N, Mizuno H, Hayashi N, Hahn WC, and Murakami S
- Subjects
- Catalytic Domain, Cell Line, Cell Line, Tumor, Cell Nucleolus metabolism, Cell Nucleus metabolism, DNA-Binding Proteins, Fibroblasts metabolism, Glutathione Transferase metabolism, Green Fluorescent Proteins metabolism, Humans, Immunoprecipitation, Microscopy, Confocal, Microscopy, Fluorescence, Phosphoproteins chemistry, Plasmids metabolism, Protein Binding, Protein Structure, Tertiary, RNA chemistry, RNA-Binding Proteins chemistry, Recombinant Fusion Proteins metabolism, Recombinant Proteins chemistry, Telomerase chemistry, Transfection, Nucleolin, Phosphoproteins metabolism, RNA-Binding Proteins metabolism, Telomerase metabolism
- Abstract
Telomerase is a specialized reverse transcriptase composed of core RNA and protein subunits which plays essential roles in maintaining telomeres in actively dividing cells. Recent work indicates that telomerase shuttles between subcellular compartments during assembly and in response to specific stimuli. In particular, telomerase colocalizes with nucleoli in normal human fibroblasts. Here, we show that nucleolin, a major nucleolar phosphoprotein, interacts with telomerase and alters its subcellular localization. Nucleolin binds the human telomerase reverse transcriptase subunit (hTERT) through interactions with its RNA binding domain 4 and carboxyl-terminal RGG domain, and this binding also involves the telomerase RNA subunit hTERC. The protein-protein interaction between nucleolin and hTERT is critical for the nucleolar localization of hTERT. These findings indicate that interaction of hTERT and nucleolin participates in the dynamic intracellular localization of telomerase complex.
- Published
- 2004
- Full Text
- View/download PDF
22. Mutational Analysis of Hepatitis C Virus NS5B in the Subgenomic Replicon Cell Culture.
- Author
-
Ma Y, Shimakami T, Luo H, Hayashi N, and Murakami S
- Subjects
- Amino Acid Sequence, Base Sequence, Cells, Cultured, Molecular Sequence Data, RNA-Dependent RNA Polymerase metabolism, Structure-Activity Relationship, Hepacivirus physiology, Replicon, Viral Nonstructural Proteins chemistry, Virus Replication
- Abstract
The hepatitis C virus (HCV) NS5B is an RNA-dependent RNA polymerase (RdRP), a central catalytic enzyme of HCV RNA replication. We previously identified five novel residues of NS5B in a JK-1 isolate indispensable for RdRP activity in vitro (Qin, W., Yamashita, T., Shirota, Y., Lin, Y., Wei, W., and Murakami, S. (2001) Hepatology 33, 728-737). We addressed the role of these residues in HCV RNA replication using a HCV replicon system derived from an M1LE isolate (Kishine, H., Sugiyama, K., Hijikata, M., Kato, N., Takahashi, H., Noshi, T., Nio, Y., Hosaka, M., Miyanari, Y., and Shimotohno, K. (2002) Biochem. Biophys. Res. Commun. 293, 993-999). The five residues of NS5B in M1LE were found to be critical for HCV replication in vivo and also indispensable for RdRP activity in vitro along with purified bacterial recombinant proteins. We also found a chimeric replicon of JK-1 and M1LE in which only the NS5B sequence derived from JK-1 could not replicate in Huh-7 cells. The residues responsible for the phenomenon were mapped by several chimeric and substituted forms of NS5B M1LE and/or JK-1 isolates in the HCV RNA replicon. Two residues, amino acids 220 and 288, were critical, and two residues, amino acids 213 and 231, were important for efficient HCV replication. Mutant JK-1 NS5B harboring all four residues of M1LE was replication-competent in the chimeric replicon and was as efficient as the original M1LE replicon. By comparing the replication competence in vivo and RdRP activity in vitro with various chimeric and mutated versions of NS5B, the HCV replication ability was found to correlate well with the RdRP activity. However, heat- and dilution-sensitive NS5Bs exhibiting weaker RdRP activity in vitro were found to be replication-incompetent, suggesting that HCV replication requires RdRP activity higher than a certain critical threshold.
- Published
- 2004
- Full Text
- View/download PDF
23. SH2-containing inositol phosphatase 2 predominantly regulates Akt2, and not Akt1, phosphorylation at the plasma membrane in response to insulin in 3T3-L1 adipocytes.
- Author
-
Sasaoka T, Wada T, Fukui K, Murakami S, Ishihara H, Suzuki R, Tobe K, Kadowaki T, and Kobayashi M
- Subjects
- 3T3-L1 Cells, Adenoviridae genetics, Adenoviridae metabolism, Animals, Blotting, Western, Chromones pharmacology, Cytosol metabolism, DNA, Complementary metabolism, Enzyme Inhibitors pharmacology, Fibroblasts metabolism, Gene Expression Regulation, Enzymologic, Humans, Mice, Mice, Knockout, Microsomes metabolism, Models, Genetic, Morpholines pharmacology, Phosphatidylinositol 3-Kinases metabolism, Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases, Phosphoric Monoester Hydrolases metabolism, Phosphorylation, Precipitin Tests, Protein Isoforms, Proto-Oncogene Proteins c-akt, Subcellular Fractions metabolism, Time Factors, src Homology Domains, Cell Membrane metabolism, Insulin metabolism, Phosphoric Monoester Hydrolases physiology, Protein Serine-Threonine Kinases metabolism, Proto-Oncogene Proteins metabolism
- Abstract
SH2-containing inositol phosphatase 2 (SHIP2) is a physiologically important negative regulator of insulin signaling by hydrolyzing the phosphatidylinositol (PI) 3-kinase product PI 3,4,5-trisphosphate in the target tissues of insulin. Targeted disruption of the SHIP2 gene in mice resulted in increased insulin sensitivity without affecting biological systems other than insulin signaling. Therefore, we investigated the molecular mechanisms by which SHIP2 specifically regulates insulin-induced metabolic signaling in 3T3-L1 adipocytes. Insulin-induced phosphorylation of Akt, one of the molecules downstream of PI3-kinase, was inhibited by expression of wild-type SHIP2, whereas it was increased by expression of 5'-phosphatase-defective (DeltaIP) SHIP2 in whole cell lysates. The regulatory effect of SHIP2 was mainly seen in the plasma membrane (PM) and low density microsomes but not in the cytosol. In this regard, following insulin stimulation, a proportion of Akt2, and not Akt1, appeared to redistribute from the cytosol to the PM. Thus, insulin-induced phosphorylation of Akt2 at the PM was predominantly regulated by SHIP2, whereas the phosphorylation of Akt1 was only minimally affected. Interestingly, insulin also elicited a subcellular redistribution of both wild-type and DeltaIP-SHIP2 from the cytosol to the PM. The degree of this redistribution was inhibited in part by pretreatment with PI3-kinase inhibitor. Although the expression of a constitutively active form of PI3-kinase myr-p110 also elicited a subcellular redistribution of SHIP2 to the PM, expression of SHIP2 appeared to affect the myr-p110-induced phosphorylation, and not the translocation, of Akt2. Furthermore, insulin-induced phosphorylation of Akt was effectively regulated by SHIP2 in embryonic fibroblasts derived from knockout mice lacking either insulin receptor substrate-1 or insulin receptor substrate-2. These results indicate that insulin specifically stimulates the redistribution of SHIP2 from the cytosol to the PM independent of 5'-phosphatase activity, thereby regulating the insulin-induced translocation and phosphorylation of Akt2 at the PM.
- Published
- 2004
- Full Text
- View/download PDF
24. Extramembrane central pore of multidrug exporter AcrB in Escherichia coli plays an important role in drug transport.
- Author
-
Murakami S, Tamura N, Saito A, Hirata T, and Yamaguchi A
- Subjects
- Amino Acid Sequence, Blotting, Western, Carrier Proteins metabolism, Cross-Linking Reagents pharmacology, Cysteine chemistry, Dimerization, Drug Resistance, Microbial, Escherichia coli Proteins metabolism, Ethylmaleimide chemistry, Membrane Proteins metabolism, Models, Chemical, Models, Molecular, Molecular Sequence Data, Multidrug Resistance-Associated Proteins, Mutagenesis, Site-Directed, Mutation, Plasmids metabolism, Protein Binding, Protein Conformation, Pyridinium Compounds chemistry, Sequence Homology, Amino Acid, Time Factors, Carrier Proteins chemistry, Drug Resistance, Multiple, Escherichia coli metabolism, Escherichia coli Proteins chemistry, Membrane Proteins chemistry
- Abstract
We previously reported the crystal structure of the major multidrug exporter AcrB in Escherichia coli (Murakami, S., Nakashima, R., Yamashita, E., and Yamaguchi, A. (2002) Nature 419, 587-593). The extramembrane headpiece of the AcrB trimer contains a central pore composed of three alpha-helices. Each pore helix belongs to a different monomer. In this study, we constructed cysteine-scanning mutants as to the residues comprising the pore helix. Of the 21 mutants (D99C to P119C), 5 (D101C, V105C, N109C, Q112C, and P116C) showed significantly reduced drug resistance and drug-exporting activity. These residues are localized on one side of the pore helix, i.e. on the wall of the pore. These observations strongly indicate the important role of this pore in the drug transport process. A N-ethylmaleimide binding experiment revealed that the pore is in the closed state, and the thickness of the permeability barrier in the middle of the pore corresponds to 2.5 alpha-helical turns. Two mutants (V105C and Q112C), which showed the greatest loss of activity of all of the pore mutants, were detected in the form of disulfide cross-linking dimers under normal conditions, suggesting that a conformational change of the pore is indispensable during the transport process.
- Published
- 2004
- Full Text
- View/download PDF
25. Direct interaction between nucleolin and hepatitis C virus NS5B.
- Author
-
Hirano M, Kaneko S, Yamashita T, Luo H, Qin W, Shirota Y, Nomura T, Kobayashi K, and Murakami S
- Subjects
- Animals, COS Cells, Green Fluorescent Proteins, HeLa Cells, Humans, Luminescent Proteins, Nuclear Proteins genetics, Phosphoproteins genetics, Protein Binding, RNA-Binding Proteins genetics, RNA-Dependent RNA Polymerase genetics, RNA-Dependent RNA Polymerase metabolism, Viral Nonstructural Proteins genetics, Nucleolin, Nuclear Proteins metabolism, Phosphoproteins metabolism, RNA-Binding Proteins metabolism, Viral Nonstructural Proteins metabolism
- Abstract
Hepatitis C virus (HCV) NS5B is an RNA-dependent RNA polymerase (RdRP), a central catalytic enzyme in HCV replication. While studying the subcellular localization of a NS5B mutant lacking the C-terminal membrane-anchoring domain, NS5Bt, we found that expression of the green fluorescent protein (GFP)-fused form was exclusively nucleolar. Interestingly, the distribution of endogenous nucleolin changed greatly in the cells expressing GFP-NS5B, with nucleolin colocalized with GFP-NS5B in perinuclear regions in addition to the nucleolus, suggesting that NS5B retains the ability to bind nucleolin. The interaction between nucleolin and NS5B was demonstrated by GST pull-down assay. GST pull-down assay results indicated that C-terminal region of nucleolin was important for its binding to NS5B. Scanning clustered alanine substitution mutants library of NS5B revealed two sites on NS5B that binds nucleolin. NS5B amino acids 208-214 and 500-506 were both found to be indispensable for the nucleolin binding. We reported that the latter sequence is essential for oligomerization of NS5B, which is a prerequisite for the RdRP activity. C-terminal nucleolin inhibited the NS5B RdRP activity in a dose-dependent manner. Taken together, this indicates the binding ability of nucleolin may be involved in NS5B functions.
- Published
- 2003
- Full Text
- View/download PDF
26. The extracellular toll-like receptor 2 domain directly binds peptidoglycan derived from Staphylococcus aureus.
- Author
-
Iwaki D, Mitsuzawa H, Murakami S, Sano H, Konishi M, Akino T, and Kuroki Y
- Subjects
- Base Sequence, Binding Sites, DNA Primers, Humans, Immunoglobulin G, Kinetics, Lipopolysaccharide Receptors chemistry, Lipopolysaccharide Receptors metabolism, Membrane Glycoproteins chemistry, Membrane Glycoproteins immunology, Polymerase Chain Reaction, Receptors, Cell Surface chemistry, Receptors, Cell Surface immunology, Recombinant Proteins metabolism, Toll-Like Receptor 2, Toll-Like Receptors, U937 Cells, Drosophila Proteins, Membrane Glycoproteins metabolism, Peptidoglycan metabolism, Receptors, Cell Surface metabolism, Staphylococcus aureus physiology
- Abstract
Toll-like receptor 2 (TLR2) has been recognized to mediate cell signaling in response to peptidoglycan (PGN), a major cell wall component of Gram-positive bacteria. The mechanism by which TLR2 recognizes PGN is unknown. It is not even clear whether TLR2 directly binds to PGN. In this study, we generated a soluble form of recombinant TLR2 (sTLR2) possessing only its putative extracellular domain by using the baculovirus expression system to examine the direct interaction between sTLR2 and PGN. sTLR2 bound avidly to insoluble PGN (iPGN) from Staphylococcus aureus coated onto microtiter wells in a concentration-dependent manner. In contrast, sTLR2 exhibited a very weak binding to lipopolysaccharide. iPGN cosedimented sTLR2 after the mixture of iPGN and sTLR2 had been incubated and centrifuged. sTLR2 partially attenuated the iPGN-induced NF-kappaB activation in TLR2-transfected HEK 293 cells and the iPGN-induced IL-8 secretion in U937 cells. One of anti-human TLR2 monoclonal antibodies, which blocked iPGN-induced NF-kappaB activation in TLR2-transfected cells, inhibited the binding of sTLR2 to iPGN. In addition, we found that sCD14 interacted with sTLR2 and increased the binding of sTLR2 to iPGN. From these results, we conclude that the extracellular TLR2 domain directly binds to PGN.
- Published
- 2002
- Full Text
- View/download PDF
27. Hepatitis C virus (HCV) NS5A binds RNA-dependent RNA polymerase (RdRP) NS5B and modulates RNA-dependent RNA polymerase activity.
- Author
-
Shirota Y, Luo H, Qin W, Kaneko S, Yamashita T, Kobayashi K, and Murakami S
- Subjects
- Animals, Base Sequence, COS Cells, DNA Primers, Plasmids, Protein Binding, Recombinant Proteins metabolism, RNA-Dependent RNA Polymerase metabolism, Viral Nonstructural Proteins metabolism
- Abstract
Hepatitis C virus (HCV) NS5B is RNA-dependent RNA polymerase (RdRP), the essential catalytic enzyme for HCV replication. Recently, NS5A has been reported to be important for the establishment of HCV replication in vitro by the adaptive mutations, although its role in viral replication remains uncertain. Here we report that purified bacterial recombinant NS5A and NS5B directly interact with each other in vitro, detected by glutathione S-transferase (GST) pull-down assay. Furthermore, complex formation of these proteins transiently coexpressed in mammalian cells was detected by coprecipitation. Using terminally and internally truncated NS5A, two discontinuous regions of NS5A (amino acids 105-162 and 277-334) outside of the adaptive mutations were identified to be independently essential for the binding both in vivo and in vitro (Yamashita, T., Kaneko, S., Shirota, Y., Qin, W., Nomura, T., Kobayashi, K., and Mkyrakami, S. (1998) J. Biol. Chem. 273, 15479-15486). We previously examined the effect of His-NS5A on RdRP activity of the soluble recombinant NS5Bt in vitro (see Yamashita et al. above). Wild NS5A weakly stimulated at first (when less than 0.1 molar ratio to NS5B) and then inhibited the NS5Bt RdRP activity in a dose-dependent manner. The internal deletion mutants defective in NS5B binding exhibited no inhibitory effect, indicating that the NS5B binding is necessary for the inhibition. Taken together, our results support the idea that NS5A modulates HCV replication as a component of replication complex.
- Published
- 2002
- Full Text
- View/download PDF
28. Two independent regions of human telomerase reverse transcriptase are important for its oligomerization and telomerase activity.
- Author
-
Arai K, Masutomi K, Khurts S, Kaneko S, Kobayashi K, and Murakami S
- Subjects
- Amino Acid Motifs, Animals, Baculoviridae metabolism, Blotting, Western, COS Cells, Cell Division, Cell Line, DNA, Complementary metabolism, DNA-Binding Proteins, Electrophoresis, Polyacrylamide Gel, Enzyme-Linked Immunosorbent Assay, Genetic Vectors, Glutathione Transferase metabolism, Humans, Mutation, Plasmids metabolism, Precipitin Tests, Protein Binding, Protein Structure, Tertiary, RNA metabolism, Recombinant Fusion Proteins metabolism, Recombinant Proteins metabolism, Transfection, Telomerase chemistry, Telomerase metabolism
- Abstract
Human telomerase reverse transcriptase (hTERT), the catalytic subunit of telomerase, contains motifs conserved among reverse transcriptases. Several nucleic acid-dependent polymerases that share a "fingers, palm, and thumb substructure" were shown to oligomerize. Here we demonstrate that hTERT also has this ability using partially purified recombinant hTERTs and mammalian cells co-expressing differently tagged hTERTs. Human template RNA (hTR), by contrast, has no effect on the structural oligomerization of hTERTs. Therefore, hTERT has an intrinsic ability of oligomerization in the absence of hTR. We identified two separate regions as essential for the oligomerization. The regions, amino acids 301-538 (amino-terminal region) and amino acids 914-928 (carboxyl-terminal region), are outside the fingers and palm substructure covering motif T to D and interact with each other in vivo. A substituted mutant of hTERT, hTERT-D712A-V713I, which was reported as a dominant negative form of hTERT, bound to the wild-type hTERT and inhibited its telomerase activity transiently expressed in telomerase-negative finite normal human fibroblast. The truncated forms of hTERT containing the binding region to the wild-type hTERT partially inhibited the telomerase activity, probably by preventing the wild-type hTERT from forming an oligomer. Taken together, the oligomerization of hTERT is an important step for telomerase activity.
- Published
- 2002
- Full Text
- View/download PDF
29. Surfactant protein A inhibits peptidoglycan-induced tumor necrosis factor-alpha secretion in U937 cells and alveolar macrophages by direct interaction with toll-like receptor 2.
- Author
-
Murakami S, Iwaki D, Mitsuzawa H, Sano H, Takahashi H, Voelker DR, Akino T, and Kuroki Y
- Subjects
- Animals, Antibodies, Monoclonal chemistry, Cell Line, Cell Wall metabolism, Cells, Cultured, DNA, Complementary metabolism, Dose-Response Relationship, Drug, Edetic Acid pharmacology, Endotoxins metabolism, Genes, Reporter, Humans, Ligands, Mice, NF-kappa B metabolism, Peptidoglycan metabolism, Protein Binding, Protein Structure, Tertiary, Pulmonary Surfactant-Associated Protein A, Pulmonary Surfactant-Associated Proteins, Rats, Rats, Sprague-Dawley, Recombinant Proteins metabolism, Signal Transduction, Toll-Like Receptor 2, Toll-Like Receptors, Transfection, U937 Cells, Drosophila Proteins, Macrophages, Alveolar metabolism, Membrane Glycoproteins metabolism, Peptidoglycan chemistry, Proteolipids chemistry, Proteolipids metabolism, Pulmonary Surfactants chemistry, Pulmonary Surfactants metabolism, Receptors, Cell Surface metabolism, Tumor Necrosis Factor-alpha metabolism
- Abstract
Pulmonary surfactant protein A (SP-A) plays an important role in modulation of the innate immune system of the lung. Peptidoglycan (PGN), a cell wall component of Gram-positive bacteria, is known to elicit excessive proinflammatory cytokine production from immune cells. In this study we investigated whether SP-A interacts with PGN and alters PGN-elicited cellular responses. Binding studies demonstrate that PGN is not a ligand for SP-A. However, SP-A significantly reduced PGN-elicited tumor necrosis factor alpha (TNF-alpha) secretion by U937 cells and rat alveolar macrophages. The inhibitory effect on TNF-alpha secretion was dependent upon SP-A concentrations in physiological range. Coincubation of SP-A and PGN with human embryonic kidney 293 cells that had been transiently transfected with the cDNA of Toll-like receptor 2 (TLR2), a cell signaling receptor for PGN, significantly attenuated PGN-induced nuclear factor-kappaB activity. SP-A directly bound to a soluble form of the recombinant extracellular TLR2 domain (sTLR2). Coincubation of sTLR2 with SP-A significantly reduced the binding of sTLR2 to PGN. These results indicate that the direct interaction of SP-A with TLR2 alters PGN-induced cell signaling. We propose that SP-A modulates inflammatory responses against the bacterial components by interactions with pattern-recognition receptors.
- Published
- 2002
- Full Text
- View/download PDF
30. Oligomeric interaction of hepatitis C virus NS5B is critical for catalytic activity of RNA-dependent RNA polymerase.
- Author
-
Qin W, Luo H, Nomura T, Hayashi N, Yamashita T, and Murakami S
- Subjects
- Amino Acids metabolism, Biopolymers chemistry, Catalysis, Models, Molecular, Mutagenesis, Site-Directed, Protein Conformation, Solutions, Viral Nonstructural Proteins chemistry, Viral Nonstructural Proteins genetics, Biopolymers metabolism, RNA-Dependent RNA Polymerase metabolism, Viral Nonstructural Proteins metabolism
- Abstract
HCV NS5B is an RNA-dependent RNA polymerase (RdRP), a central catalytic enzyme for HCV replication, which has the "palm and fingers" substructure. We recently identified five novel residues critical for RdRP activity (Qin, W., Yamashita, T., Shirota, Y., Lin, Y., Wei, W., and Murakami, S. (2001) Hepatology 33, 728-737). Among them, GLU-18 and His-502, far from the catalytic center, may be involved in conformational change(s) for RdRP activity as addressed in some palm and fingers enzymes. We examined the possibility that NS5B is oligomerized, and we could detect the interaction between two different tagged NS5B proteins in vitro and transiently expressed in mammalian cells. By scanning 27 clustered and then point alanine substitutions in vivo and in vitro, Glu-18 and His-502 were found to be critical for the homomeric interaction in vivo and in vitro, strongly suggesting a close relationship between the oligomerization and RdRP activity of NS5B. All mutants with substitutions at these two residues failed to bind wild type NS5B, however E18H interacted with H502E in vitro and in vivo. Interestingly, the NS5B protein with E18H or H502E did not exhibit RdRP activity, but a mixture of the two mutant proteins did. These results clearly indicate that two residues of HCV NS5B are critical for the oligomerization that is prerequisite to RdRP activity.
- Published
- 2002
- Full Text
- View/download PDF
31. Extracellular Toll-like receptor 2 region containing Ser40-Ile64 but not Cys30-Ser39 is critical for the recognition of Staphylococcus aureus peptidoglycan.
- Author
-
Mitsuzawa H, Wada I, Sano H, Iwaki D, Murakami S, Himi T, Matsushima N, and Kuroki Y
- Subjects
- Amino Acid Sequence, Base Sequence, Cell Line, DNA Primers, Humans, Lipopolysaccharide Receptors genetics, Membrane Glycoproteins chemistry, Membrane Glycoproteins genetics, Molecular Sequence Data, Mutagenesis, NF-kappa B metabolism, Receptors, Cell Surface chemistry, Receptors, Cell Surface genetics, Recombinant Fusion Proteins chemistry, Recombinant Fusion Proteins genetics, Recombinant Fusion Proteins metabolism, Reverse Transcriptase Polymerase Chain Reaction, Signal Transduction, Toll-Like Receptor 2, Toll-Like Receptors, Cysteine metabolism, Drosophila Proteins, Isoleucine metabolism, Membrane Glycoproteins metabolism, Peptidoglycan metabolism, Receptors, Cell Surface metabolism, Serine metabolism, Staphylococcus aureus metabolism
- Abstract
Toll-like receptor 2 (TLR2) and CD14 function as pattern recognition receptors for bacterial peptidoglycan (PGN). TLRs and CD14 possess repeats of the leucine-rich motif. To address the role of the extracellular domain of TLR2 in PGN signaling, we constructed CD14/TLR2 chimeras, in which residues 1-356 or 1-323 of CD14 were substituted for the extracellular domain of TLR2, and five deletion mutants of TLR2, in which the progressively longer regions of extracellular TLR2 regions were deleted. PGN induced NF-kappaB activation in HEK293 cells expressing TLR2 but not in cells expressing CD14/TLR2 chimeras. The cells transfected with a deletion mutant TLR2(DeltaCys30-Ile64) as well as TLR2(DeltaCys30-Asp160) and TLR2(DeltaCys30-Asp305) failed to respond to PGN, indicating the importance of the TLR2 region Cys(30)-Ile(64). Although TLR2(DeltaCys30-Ser39) conferred cell responsiveness to PGN, the cells expressing TLR2(DeltaSer40-Ile64) failed to induce NF-kappaB activation. In addition, NF-kappaB activity elicited by PGN was significantly attenuated in the presence of synthetic peptide corresponding to the TLR2 region Ser(40)-Ile(64). From these results, we conclude that; 1) CD14 cannot functionally replace the extracellular domain of TLR2 in PGN signaling; 2) the TLR2 region Cys(30)-Ser(39) is not required for PGN recognition; 3) the TLR2 region containing Ser(40)-Ile(64) is critical for PGN recognition.
- Published
- 2001
- Full Text
- View/download PDF
32. Direct interaction between the subunit RAP30 of transcription factor IIF (TFIIF) and RNA polymerase subunit 5, which contributes to the association between TFIIF and RNA polymerase II.
- Author
-
Wei W, Dorjsuren D, Lin Y, Qin W, Nomura T, Hayashi N, and Murakami S
- Subjects
- Animals, Base Sequence, Binding Sites, COS Cells, DNA Primers, Protein Binding, Transcription Factors chemistry, RNA Polymerase II metabolism, Transcription Factors metabolism, Transcription Factors, TFII
- Abstract
The general transcription factor IIF (TFIIF) assembled in the initiation complex, and RAP30 of TFIIF, have been shown to associate with RNA polymerase II (pol II), although it remains unclear which pol II subunit is responsible for the interaction. We examined whether TFIIF interacts with RNA polymerase II subunit 5 (RPB5), the exposed domain of which binds transcriptional regulatory factors such as hepatitis B virus X protein and a novel regulatory protein, RPB5-mediating protein. The results demonstrated that RPB5 directly binds RAP30 in vitro using purified recombinant proteins and in vivo in COS1 cells transiently expressing recombinant RAP30 and RPB5. The RAP30-binding region was mapped to the central region (amino acids (aa) 47-120) of RPB5, which partly overlaps the hepatitis B virus X protein-binding region. Although the middle part (aa 101-170) and the N-terminus (aa 1-100) of RAP30 independently bound RPB5, the latter was not involved in the RPB5 binding when RAP30 was present in TFIIF complex. Scanning of the middle part of RAP30 by clustered alanine substitutions and then point alanine substitutions pinpointed two residues critical for the RPB5 binding in in vitro and in vivo assays. Wild type but not mutants Y124A and Q131A of RAP30 coexpressed with FLAG-RAP74 efficiently recovered endogenous RPB5 to the FLAG-RAP74-bound anti-FLAG M2 resin. The recovered endogenous RPB5 is assembled in pol II as demonstrated immunologically. Interestingly, coexpression of the central region of RPB5 and wild type RAP30 inhibited recovery of endogenous pol II to the FLAG-RAP74-bound M2 resin, strongly suggesting that the RAP30-binding region of RPB5 inhibited the association of TFIIF and pol II. The exposed domain of RPB5 interacts with RAP30 of TFIIF and is important for the association between pol II and TFIIF.
- Published
- 2001
- Full Text
- View/download PDF
33. Telomerase activity reconstituted in vitro with purified human telomerase reverse transcriptase and human telomerase RNA component.
- Author
-
Masutomi K, Kaneko S, Hayashi N, Yamashita T, Shirota Y, Kobayashi K, and Murakami S
- Subjects
- Benzoquinones, DNA-Binding Proteins, Enzyme Activation drug effects, Enzyme Inhibitors pharmacology, Gene Expression Regulation, Enzymologic, Humans, Lactams, Macrocyclic, Mutation, Quinones pharmacology, RNA genetics, RNA metabolism, Telomerase genetics, Telomerase metabolism
- Abstract
Telomerase is a specialized reverse transcriptase that catalyzes elongation of the telomeric tandem repeat, TTAGGG, by addition of this sequence to the ends of existing telomeres. Human telomerase reverse transcriptase (hTERT) has been identified as a catalytic enzyme involved in telomere elongation that requires telomerase RNA, human telomerase RNA component (hTR), as an RNA template. We established a new method to express and purify soluble insect-expressed recombinant hTERT. The partially purified FLAG-hTERT retained the catalytic activity of telomerase in a complementation assay in vitro to exhibit telomerase activity in telomerase-negative TIG3 cell extract and in a reconstitution assay with FLAG-hTERT and purified hTR in vitro. FLAG-hTERT (D712A) with a mutation in the VDV motif exhibited no telomerase activity, confirming the authentic catalytic activity of FLAG-hTERT. The reconstituted complex of FLAG-hTERT and hTR in vitro was detected by electrophoretic mobility shift assay, and its activity was stimulated by more than 30-fold by TIG3 cell extract. This suggested that some cellular component(s) in the extract facilitated the reconstituted telomerase activity in vitro. Geldanamycin had no effect on the reconstituted activity but partially reduced the stimulated activity of the reconstituted telomerase by the TIG3 cell extract, suggesting that Hsp90 may contribute to the stimulatory effect of the cellular components.
- 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.