25 results on '"Xiong H"'
Search Results
2. Mechanistic studies of the processing of human S-adenosylmethionine decarboxylase proenzyme. Isolation of an ester intermediate.
- Author
-
Xiong, H and Pegg, A E
- Abstract
Human S-adenosylmethionine decarboxylase is synthesized as a proenzyme that undergoes an autocatalytic cleavage reaction generating the alpha and beta subunits and forming the pyruvate prosthetic group, which is derived from an internal Ser residue (Ser-68). The mechanism of this processing reaction was studied using site-directed mutagenesis of conserved residues (His-243 and Ser-229) located close to the cleavage site. Mutant S229A failed to process, and mutant S229C cleaved very slowly, whereas mutant S229T processed normally, suggesting that the hydroxyl group of residue 229 is required for the processing reaction where Ser-229 may act as a proton acceptor. Mutant His-243A cleaved very slowly, forming a small amount of the correctly processed pyruvoyl enzyme but a much larger proportion of the alpha subunit with an amino-terminal Ser. The cleavage to form the latter was greatly enhanced by hydroxylamine. This result suggests that the N-O acyl shift needed for ester formation occurs normally in this mutant but that the next step, which is a beta-elimination reaction leading to the two subunits, does not occur. His-243 may therefore act as the basic residue that extracts the hydrogen of the alpha-carbon of Ser-68 in the ester in order to facilitate this reaction. The availability of the recombinant H243A S-adenosylmethionine decarboxylase proenzyme provides a useful model system to examine the processing reaction in vitro and test the design of specific inactivators aimed at blocking the production of the pyruvoyl prosthetic group.
- Published
- 1999
3. Alanine-scanning mutagenesis of the epsilon subunit of the F1-F0 ATP synthase from Escherichia coli reveals two classes of mutants.
- Author
-
Xiong, H and Vik, S B
- Abstract
Alanine-scanning mutagenesis was applied to the epsilon subunit of the F1-F0 ATP synthase from E. coli. Nineteen amino acid residues were changed to alanine, either singly or in pairs, between residues 10 and 93. All mutants, when expressed in the epsilon deletion strain XH1, were able to grow on succinate minimal medium. Membranes were prepared from all mutants and assayed for ATP-driven proton translocation, ATP hydrolysis +/- lauryldiethylamine oxide, and sensitivity of ATPase activity to N,N'-dicyclohexylcarbodiimide (DCCD). Most of the mutants fell into 2 distinct classes. The first group had inhibited ATPase activity, with near normal levels of membrane-bound F1, but decreased sensitivity to DCCD. The second group had stimulated ATPase activity, with a reduced level of membrane-bound F1, but normal sensitivity to DCCD. Membranes from all mutants were further characterized by immunoblotting using 2 monoclonal antibodies. A model for the secondary structure of epsilon and its role in the function of the ATP synthase has been developed. Some residues are important for the binding of epsilon to F1 and therefore for inhibition. Other residues, from Glu-59 through Glu-70, are important for the release of inhibition by epsilon that is part of the normal enzyme cycle.
- Published
- 1995
4. Phosphorylation of eukaryotic protein synthesis initiation factor 4E by insulin-stimulated protamine kinase.
- Author
-
Makkinje, A, Xiong, H, Li, M, and Damuni, Z
- Abstract
Insulin-stimulated protamine kinase (cPK) and protein kinase C (PKC) phosphorylated eukaryotic protein synthesis initiation factor 4E (eIF-4E) on serine and threonine residues located on an identical tryptic fragment as judged by two-dimensional phosphopeptide mapping. With cPK and PKC, the apparent Km for eIF-4E was about 1.2 and 50 microM, respectively. Relative to recombinant human eIF-4E, cPK exhibited about 100% and < or = 5% activity with eIF-4ES209A and eIF-4ET210A, respectively, and eIF-4ES209A was phosphorylated exclusively on threonines. Bovine kidney eIF-4E enhanced up to 1.8-fold globin synthesis in m7GTP-Sepharose-treated reticulocyte lysates. In contrast, following incubation with cPK, these eIF-4E preparations stimulated globin synthesis up to 6-fold. Compared to the dephosphorylation of the cPK-modified serine on eIF-4E, reticulocyte lysates and highly purified protein phosphatase 2A exhibited marked preference for the cPK-modified threonine. The results indicate that cPK phosphorylates eIF-4E on Ser209 and Thr210, that the hydroxyl group or phosphorylation of Thr210 is necessary for cPK to act on Ser209, and that Ser209 phosphorylation activates reticulocyte globin synthesis. The results suggest that cPK could contribute to the insulin-stimulated phosphorylation of eIF-4E, but that protein phosphatase 2A may confer the site specificity of this response.
- Published
- 1995
5. Disulfide linkage of biotin identifies a 106-kDa Ca2+release channel in sarcoplasmic reticulum*
- Author
-
Zaidi, N F, Lagenaur, C F, Hilkert, R J, Xiong, H, Abramson, J J, and Salama, G
- Abstract
Reactive disulfide reagents (RDSs) with a biotin moiety have been synthesized and found to cause Ca2+release from sarcoplasmic reticulum (SR) vesicles. The RDSs oxidize SH sites on SR proteins via a thiol-disulfide exchange, with the formation of mixed disulfide bonds between SR proteins and biotin. Biotinylated RDSs identified a 106-kDa protein which was purified by biotin-avidin chromatography. Disulfide reducing agents, like dithiothreitol, reverse the effect of RDSs and thus promoted active re-uptake of Ca2+and dissociated biotin from the labeled protein indicating that biotin was covalently linked to the 106-kDa protein via a disulfide bond. Several lines of evidence indicate that this protein is not Ca2+,Mg2+-ATPase and is not a proteolytic fragment or a subunit of the 400-kDa Ca2+-ryanodine receptor complex (RRC). Monoclonal antibodies against the ATPase did not cross-react with the 106-kDa protein, and polyclonal antibodies against the 106-kDa did not cross-react with either the ATPase or the 400-kDa RRC. RDSs did not label the 400-kDa RRC with biotin. Linear sucrose gradients used to purify the RRC show that the 106-kDa protein migrated throughout 5–20% linear sucrose gradients, including the high sucrose density protein fractions containing 400-kDa RRC. Protease inhibitors diisopropylfluorophosphate used to prevent proteolysis of 400-kDa proteins did not alter the migration of 106-kDa in sucrose gradients nor the patterns of biotin labeling of the 106-kDa protein. Incorporation of highly purified 106-kDa protein (free of RRC) in planar bilayers revealed cationic channels with large Na+(gNa+= 375 ± 15 pS) and Ca2+(gCa2+= 107.7 ± 12 pS) conductances which were activated by micromolar [Ca2+]freeor millimolar [ATP] and blocked by micromolar ruthenium red or millimolar [Mg2+]. Thus, the SR contains a sulfhydryl-activated 106-kDa Ca2+channel with apparently similar characteristics to the 400-kDa “feet” proteins.
- Published
- 1989
- Full Text
- View/download PDF
6. Disulfide linkage of biotin identifies a 106-kDa Ca2+ release channel in sarcoplasmic reticulum
- Author
-
Zaidi, N F, primary, Lagenaur, C F, additional, Hilkert, R J, additional, Xiong, H, additional, Abramson, J J, additional, and Salama, G, additional
- Published
- 1989
- Full Text
- View/download PDF
7. αB-Crystallin Interacts with Nav1.5 and Regulates Ubiquitination and Internalization of Cell Surface Nav1.5.
- Author
-
Huang Y, Wang Z, Liu Y, Xiong H, Zhao Y, Wu L, Yuan C, Wang L, Hou Y, Yu G, Huang Z, Xu C, Chen Q, and Wang QK
- Subjects
- Animals, Cell Line, Cell Membrane metabolism, Endosomal Sorting Complexes Required for Transport chemistry, Endosomal Sorting Complexes Required for Transport genetics, Endosomal Sorting Complexes Required for Transport metabolism, Gene Knockdown Techniques, HEK293 Cells, Humans, Mutant Proteins chemistry, Mutant Proteins genetics, Mutant Proteins metabolism, Myocardium metabolism, NAV1.5 Voltage-Gated Sodium Channel genetics, Nedd4 Ubiquitin Protein Ligases, Protein Binding, Protein Interaction Domains and Motifs, Protein Stability, Protein Transport, Rats, Recombinant Proteins chemistry, Recombinant Proteins genetics, Recombinant Proteins metabolism, Ubiquitin-Protein Ligases chemistry, Ubiquitin-Protein Ligases genetics, Ubiquitin-Protein Ligases metabolism, Ubiquitination, alpha-Crystallin B Chain genetics, NAV1.5 Voltage-Gated Sodium Channel chemistry, NAV1.5 Voltage-Gated Sodium Channel metabolism, alpha-Crystallin B Chain chemistry, alpha-Crystallin B Chain metabolism
- Abstract
Nav1.5, the pore-forming α subunit of the cardiac voltage-gated Na(+) channel complex, is required for the initiation and propagation of the cardiac action potential. Mutations in Nav1.5 cause cardiac arrhythmias and sudden death. The cardiac Na(+) channel functions as a protein complex; however, its complete components remain to be fully elucidated. A yeast two-hybrid screen identified a new candidate Nav1.5-interacting protein, αB-crystallin. GST pull-down, co-immunoprecipitation, and immunostaining analyses validated the interaction between Nav1.5 and αB-crystallin. Whole-cell patch clamping showed that overexpression of αB-crystallin significantly increased peak sodium current (INa) density, and the underlying molecular mechanism is the increased cell surface expression level of Nav1.5 via reduced internalization of cell surface Nav1.5 and ubiquitination of Nav1.5. Knock-out of αB-crystallin expression significantly decreased the cell surface expression level of Nav1.5. Co-immunoprecipitation analysis showed that αB-crystallin interacted with Nedd4-2; however, a catalytically inactive Nedd4-2-C801S mutant impaired the interaction and abolished the up-regulation of INa by αB-crystallin. Nav1.5 mutation V1980A at the interaction site for Nedd4-2 eliminated the effect of αB-crystallin on reduction of Nav1.5 ubiquitination and increases of INa density. Two disease-causing mutations in αB-crystallin, R109H and R151X (nonsense mutation), eliminated the effect of αB-crystallin on INa This study identifies αB-crystallin as a new binding partner for Nav1.5. αB-Crystallin interacts with Nav1.5 and increases INa by modulating the expression level and internalization of cell surface Nav1.5 and ubiquitination of Nav1.5, which requires the protein-protein interactions between αB-crystallin and Nav1.5 and between αB-crystallin and functionally active Nedd4-2., (© 2016 by The American Society for Biochemistry and Molecular Biology, Inc.)
- Published
- 2016
- Full Text
- View/download PDF
8. Thromboxane A2 Receptor Inhibition Suppresses Multiple Myeloma Cell Proliferation by Inducing p38/c-Jun N-terminal Kinase (JNK) Mitogen-activated Protein Kinase (MAPK)-mediated G2/M Progression Delay and Cell Apoptosis.
- Author
-
Liu Q, Tao B, Liu G, Chen G, Zhu Q, Yu Y, Yu Y, and Xiong H
- Subjects
- 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid pharmacology, Bone Marrow Cells cytology, Bridged Bicyclo Compounds, Heterocyclic, CDC2 Protein Kinase, Cell Line, Tumor, Cell Proliferation drug effects, Cells, Cultured, Cyclin B1 agonists, Cyclin B1 antagonists & inhibitors, Cyclin B1 genetics, Cyclin B1 metabolism, Cyclin-Dependent Kinases antagonists & inhibitors, Cyclin-Dependent Kinases chemistry, Cyclin-Dependent Kinases genetics, Cyclin-Dependent Kinases metabolism, Cyclooxygenase Inhibitors pharmacology, Fatty Acids, Unsaturated, Gene Expression Regulation, Neoplastic drug effects, Humans, Hydrazines pharmacology, Leukocytes, Mononuclear cytology, Leukocytes, Mononuclear drug effects, Leukocytes, Mononuclear metabolism, Multiple Myeloma chemically induced, Multiple Myeloma metabolism, Multiple Myeloma pathology, Neoplasm Proteins agonists, Neoplasm Proteins genetics, Neoplasm Proteins metabolism, RNA Interference, Receptors, Thromboxane A2, Prostaglandin H2 agonists, Receptors, Thromboxane A2, Prostaglandin H2 genetics, Receptors, Thromboxane A2, Prostaglandin H2 metabolism, Antineoplastic Agents pharmacology, Apoptosis drug effects, G2 Phase drug effects, MAP Kinase Signaling System drug effects, Multiple Myeloma drug therapy, Neoplasm Proteins antagonists & inhibitors, Receptors, Thromboxane A2, Prostaglandin H2 antagonists & inhibitors
- Abstract
Multiple myeloma (MM) is a plasma cell malignancy without effective therapeutics. Thromboxane A2 (TxA2)/TxA2 receptor (T prostanoid receptor (TP)) modulates the progression of some carcinomas; however, its effects on MM cell proliferation remain unclear. In this study, we evaluated cyclooxygenase (COX) enzymes and downstream prostaglandin profiles in human myeloma cell lines RPMI-8226 and U-266 and analyzed the effects of COX-1/-2 inhibitors SC-560 and NS-398 on MM cell proliferation. Our observations implicate COX-2 as being involved in modulating cell proliferation. We further incubated MM cells with prostaglandin receptor antagonists or agonists and found that only the TP antagonist, SQ29548, suppressed MM cell proliferation. TP silencing and the TP agonist, U46619, further confirmed this finding. Moreover, SQ29548 and TP silencing promoted MM cell G2/M phase delay accompanied by reducing cyclin B1/cyclin-dependent kinase-1 (CDK1) mRNA and protein expression. Notably, cyclin B1 overexpression rescued MM cells from G2/M arrest. We also found that the TP agonist activated JNK and p38 MAPK phosphorylation, and inhibitors of JNK and p38 MAPK depressed U46619-induced proliferation and cyclin B1/CDK1 protein expression. In addition, SQ29548 and TP silencing led to the MM cell apoptotic rate increasing with improving caspase 3 activity. The knockdown of caspase 3 reversed the apoptotic rate. Taken together, our results suggest that TxA2/TP promotes MM cell proliferation by reducing cell delay at G2/M phase via elevating p38 MAPK/JNK-mediated cyclin B1/CDK1 expression and hindering cell apoptosis. The TP inhibitor has potential as a novel agent to target kinase cascades for MM therapy., (© 2016 by The American Society for Biochemistry and Molecular Biology, Inc.)
- Published
- 2016
- Full Text
- View/download PDF
9. Alteration of lysosome fusion and low-grade inflammation mediated by super-low-dose endotoxin.
- Author
-
Baker B, Geng S, Chen K, Diao N, Yuan R, Xu X, Dougherty S, Stephenson C, Xiong H, Chu HW, and Li L
- Subjects
- Animals, Endosomes drug effects, Endosomes immunology, Humans, Inflammation chemically induced, Inflammation pathology, Intracellular Signaling Peptides and Proteins genetics, Lysosomes drug effects, Lysosomes pathology, Macrophages drug effects, Macrophages immunology, Mice, Mice, Knockout, Autophagy immunology, Immunity, Innate, Inflammation immunology, Lipopolysaccharides toxicity, Lysosomes immunology
- Abstract
Subclinical super-low-dose endotoxin LPS is a risk factor for the establishment of low-grade inflammation during the pathogenesis and progression of chronic diseases. However, the underlying mechanisms are not well understood. At the cellular level, a disruption of lysosome fusion with endosomes or autophagosomes may contribute to the potentiation of low-grade inflammation. In this study, we identified that subclinical super-low-dose endotoxin LPS can potently inhibit the process of endosome acidification and lysosome fusion with endosomes or autophagosomes in primary macrophages. Super-low-dose LPS induced the inhibitory phosphorylation of VPS34, thus leading to the disruption of endosome-lysosome fusion. This effect may depend upon the clearance and relocation of Tollip in macrophages by super-low-dose LPS. Consistent with this notion, Tollip-deficient macrophages had constitutively elevated levels of VPS34 inhibitory phosphorylation and constitutive disruption of endosome-lysosome fusion. By employing a skin excision wound-healing model, we observed that Tollip-deficient mice had significantly elevated levels of cell stress and reduced wound repair. This study reveals a novel mechanism responsible for the modulation of endosome-lysosome fusion and low-grade inflammation in innate macrophages., (© 2015 by The American Society for Biochemistry and Molecular Biology, Inc.)
- Published
- 2015
- Full Text
- View/download PDF
10. A prototypic lysine methyltransferase 4 from archaea with degenerate sequence specificity methylates chromatin proteins Sul7d and Cren7 in different patterns.
- Author
-
Niu Y, Xia Y, Wang S, Li J, Niu C, Li X, Zhao Y, Xiong H, Li Z, Lou H, and Cao Q
- Subjects
- Amino Acid Motifs, Amino Acid Sequence, Amino Acid Substitution, Archaeal Proteins genetics, Chromatin chemistry, Conserved Sequence, DNA, Archaeal chemistry, Methylation, Molecular Sequence Data, Protein Methyltransferases genetics, Substrate Specificity, Archaeal Proteins chemistry, DNA-Binding Proteins chemistry, Protein Methyltransferases chemistry, Protein Processing, Post-Translational, Sulfolobus enzymology
- Abstract
Background: The origin of eukaryotic histone modification enzymes still remains obscure., Results: Prototypic KMT4/Dot1 from Archaea targets chromatin proteins (Sul7d and Cren7) and shows increased activity on Sul7d, but not Cren7, in the presence of DNA., Conclusion: Promiscuous aKMT4 could be regulated by chromatin environment., Significance: This study supports the prokaryotic origin model of eukaryotic histone methyltransferases and sheds light on chromatin dynamics in Archaea. Histone methylation is one of the major epigenetic modifications even in early diverging unicellular eukaryotes. We show that a widespread lysine methyltransferase from Archaea (aKMT4), bears striking structural and functional resemblance to the core of distantly related eukaryotic KMT4/Dot1. aKMT4 methylates a set of various proteins, including the chromatin proteins Sul7d and Cren7, and RNA exosome components. Csl4- and Rrp4-exosome complexes are methylated in different patterns. aKMT4 can self-methylate intramolecularly and compete with other proteins for the methyl group. Automethylation is inhibited by suitable substrates or DNA in a concentration-dependent manner. The automethylated enzyme shows relatively compromised activity. aKMT4-8A mutant with abrogated automethylation shows a more than 150% increase in methylation of substrates, suggesting a possible mechanism to regulate methyltransferase activity. More interestingly, methylation of Sul7d, but not Cren7, by aKMT4 is significantly enhanced by DNA. MS/MS and kinetic analysis further suggest that aKMT4 methylates Sul7d in the chromatin context. These data provide a clue to the possible regulation of aKMT4 activity by the local chromatin environment, albeit as a promiscuous enzyme required for extensive and variegated lysine methylation in Sulfolobus. This study supports the prokaryotic origin model of eukaryotic histone modification enzymes and sheds light on regulation of archaeal chromatin.
- Published
- 2013
- Full Text
- View/download PDF
11. Roles of STAT3 and ZEB1 proteins in E-cadherin down-regulation and human colorectal cancer epithelial-mesenchymal transition.
- Author
-
Xiong H, Hong J, Du W, Lin YW, Ren LL, Wang YC, Su WY, Wang JL, Cui Y, Wang ZH, and Fang JY
- Subjects
- Apoptosis, Base Sequence, Cell Line, Tumor, Colorectal Neoplasms genetics, Homeodomain Proteins genetics, Humans, Janus Kinases metabolism, Molecular Sequence Data, Neoplasm Invasiveness, Neoplasm Metastasis, Phosphoproteins metabolism, Signal Transduction, Transcription Factors genetics, Vimentin genetics, Vimentin metabolism, Zinc Finger E-box-Binding Homeobox 1, Cadherins genetics, Cadherins metabolism, Colorectal Neoplasms pathology, Down-Regulation, Epithelial-Mesenchymal Transition, Homeodomain Proteins metabolism, STAT3 Transcription Factor metabolism, Transcription Factors metabolism
- Abstract
The progression of colorectal carcinoma (CRC) to invasive and metastatic disease may involve localized occurrences of epithelial-mesenchymal transition (EMT). However, mechanisms of the EMT process in CRC progression are not fully understood. We previously showed that knockdown of signal transducer and activator of transcription 3 (STAT3) up-regulated E-cadherin (a key component in EMT progression) in CRC. In this study, we examined the roles of STAT3 in CRC EMT and ZEB1, an EMT inducer, in STAT3-induced down-regulation of E-cadherin. Knockdown of STAT3 significantly increased E-cadherin and decreased N-cadherin and vimentin expressions in highly invasive LoVo CRC cells. Meanwhile, overexpression of STAT3 significantly reduced E-cadherin and enhanced N-cadherin and vimentin expressions in weakly invasive SW1116 CRC cells. Activation of STAT3 significantly increased CRC cell invasiveness and resistance to apoptosis. Knockdown of STAT3 dramatically enhanced chemosensitivity of CRC cells to fluorouracil. STAT3 regulated ZEB1 expression in CRC cells, and the STAT3-induced decrease in E-cadherin and cell invasion depended on activation of ZEB1 in CRC cells. Additionally, pSTAT3(Tyr-705) and ZEB1 expressions were significantly correlated with TNM (tumor, lymph node, and metastasis stages) (p < 0.01). In conclusion, STAT3 may directly mediate EMT progression and regulate ZEB1 expression in CRC. ZEB1 may participate in STAT3-induced cell invasion and E-cadherin down-regulation in CRC cells. The expressions of pSTAT3(Tyr-705) and ZEB1 may be positively associated with CRC metastasis. Our data may provide potential targets to prevent and/or treat CRC invasion and metastasis.
- Published
- 2012
- Full Text
- View/download PDF
12. Feedback regulation of endothelial cell surface plasmin generation by PKC-dependent phosphorylation of annexin A2.
- Author
-
He KL, Sui G, Xiong H, Broekman MJ, Huang B, Marcus AJ, and Hajjar KA
- Subjects
- Animals, Mice, Mice, Knockout, Phosphorylation, Protein Multimerization, Protein Transport, Serine metabolism, Annexin A2 metabolism, Endothelial Cells metabolism, Feedback, Physiological, Fibrinolysin biosynthesis, Protein Kinase C metabolism
- Abstract
In response to blood vessel injury, hemostasis is initiated by platelet activation, advanced by thrombin generation, and tempered by fibrinolysis. The primary fibrinolytic protease, plasmin, can be activated either on a fibrin-containing thrombus or on cells. Annexin A2 (A2) heterotetramer (A2·p11)(2) is a key profibrinolytic complex that assembles plasminogen and tissue plasminogen activator and promotes plasmin generation. We now report that, in endothelial cells, plasmin specifically induces activation of conventional PKC, which phosphorylates serine 11 and serine 25 of A2, triggering dissociation of the (A2·p11)(2) tetramer. The resulting free p11 undergoes ubiquitin-mediated proteasomal degradation, thus preventing further translocation of A2 to the cell surface. In vivo, pretreatment of A2(+/+) but not A2(-/-) mice with a conventional PKC inhibitor significantly reduced thrombosis in a carotid artery injury model. These results indicate that augmentation of fibrinolytic vascular surveillance by blockade of serine phosphorylation is A2-dependent. We also demonstrate that plasmin-induced phosphorylation of A2 requires both cleavage of A2 and activation of Toll-like receptor 4 on the cell surface. We propose that plasmin can limit its own generation by triggering a finely tuned "feedback" mechanism whereby A2 becomes serine-phosphorylated, dissociates from p11, and fails to translocate to the cell surface.
- Published
- 2011
- Full Text
- View/download PDF
13. Activation of IL-27 p28 gene transcription by interferon regulatory factor 8 in cooperation with interferon regulatory factor 1.
- Author
-
Zhang J, Qian X, Ning H, Yang J, Xiong H, and Liu J
- Subjects
- Animals, Cell Nucleus metabolism, Cytokines metabolism, Enzyme-Linked Immunosorbent Assay methods, Humans, Inflammation, Interleukin-17 genetics, Lipopolysaccharides metabolism, Macrophages metabolism, Mice, Mice, Transgenic, RNA, Messenger metabolism, Interferon Regulatory Factor-1 metabolism, Interferon Regulatory Factors metabolism, Interleukin-17 metabolism, Transcription, Genetic
- Abstract
Interferon regulatory factor (IRF) family members, especially interferon regulatory factor-1 (IRF-1) and interferon regulatory factor-8 (IRF-8 or ICSBP), play important roles in interferon signaling in a wide range of host responses to infection and tumor growth. Interleukin-27 (IL-27), as a member of the IL-12 cytokine family, not only acts as a proinflammatory cytokine that regulates the differentiation of naive T helper cells but also possesses anti-inflammatory properties. IL-27 consists of EBI3 (Epstein-Barr virus-induced gene 3) and p28 subunits. Our previous work has shown that IRF-1 regulates IL-27 p28 gene transcription by specifically binding to the IRF-1 response element in the p28 promoter. In this study, we found that IRF-8-deficient macrophages were highly defective in the production of IL-27 p28 at both mRNA and protein levels. Circulating IL-27 p28 in serum was also decreased in IRF-8(-/-) mice in a septic shock model. Lipopolysaccharide, as a potent inducer of IL-27 p28 expression, could activate IRF-8 expression in a MyD88-dependent pathway, which in turn induced p28 gene transcription through NF-kappaB and/or IRF-8. Transcriptional analyses revealed that IRF-8 activated p28 gene transcription through binding to a site located at -57 to -48 in the p28 promoter overlapping the IRF-1 binding site. Consistent with this observation, overexpression of both IRF-8 and IRF-1 additively activated IL-27 p28 promoter. This study provides further mechanistic information regarding how signals initiated during innate and adaptive immune responses synergize to yield greater IL-27 production and sustained cellular immunity.
- Published
- 2010
- Full Text
- View/download PDF
14. Platelet-derived growth factor-mediated induction of the synaptic plasticity gene Arc/Arg3.1.
- Author
-
Peng F, Yao H, Bai X, Zhu X, Reiner BC, Beazely M, Funa K, Xiong H, and Buch S
- Subjects
- Animals, Calcium metabolism, Cell Proliferation, Electrophysiology, Enzyme Activation, Female, Hippocampus metabolism, MAP Kinase Signaling System, Mice, Mice, Inbred C57BL, Nerve Growth Factors metabolism, Phosphatidylinositol 3-Kinases, Rats, Rats, Sprague-Dawley, Cytoskeletal Proteins metabolism, Gene Expression Regulation, Nerve Tissue Proteins metabolism, Platelet-Derived Growth Factor metabolism
- Abstract
Platelet-derived growth factor (PDGF) is a pleiotropic protein with critical roles in both developmental as well as pathogenic processes. In the central nervous system specifically, PDGF is critical for neuronal proliferation and differentiation and has also been implicated as a neuroprotective agent. Whether PDGF also plays a role in synaptic plasticity, however, remains poorly understood. In the present study we demonstrated that in the rat hippocampal neurons PDGF regulated the expression of Arc/Arg3.1 gene that has been implicated in both synapse plasticity and long term potentiation. Relevance of these findings was further confirmed in vivo by injecting mice with intracerebral inoculations of PDGF, which resulted in a rapid induction of Arc in the hippocampus of the injected mice. PDGF induced long term potentiation in rat hippocampal slices, which was abolished by PDGF receptor-tyrosine kinase inhibitor STI-571. We also present evidence that PDGF-mediated induction of Arc/Arg3.1 involved activation of the MAPK/ERK (MEK) pathway. Additionally, induction of Arc/Arg3.1 also involved the upstream release of intracellular calcium stores, an effect that could be blocked by thapsigargin but not by EGTA. Pharmacological approach using inhibitors specific for either MAPK/ERK phosphorylation or calcium release demonstrated that the two pathways converged downstream at a common point involving activation of the immediate early gene Egr-1. Chromatin immunoprecipitation assays demonstrated the binding of Egr-1, but not Egr-3, to the Arc promoter. These findings for the first time, thus, suggest an additional role of PDGF, that of induction of Arc.
- Published
- 2010
- Full Text
- View/download PDF
15. AP-1 activated by toll-like receptors regulates expression of IL-23 p19.
- Author
-
Liu W, Ouyang X, Yang J, Liu J, Li Q, Gu Y, Fukata M, Lin T, He JC, Abreu M, Unkeless JC, Mayer L, and Xiong H
- Subjects
- Activating Transcription Factor 2 genetics, Activating Transcription Factor 2 immunology, Activating Transcription Factor 2 metabolism, Animals, Autoimmune Diseases, Cell Line, Dendritic Cells immunology, Dendritic Cells metabolism, Gene Expression Regulation drug effects, Interleukin-10 immunology, Interleukin-10 pharmacology, Interleukin-23 Subunit p19 genetics, Interleukin-23 Subunit p19 immunology, Lipopolysaccharides pharmacology, MAP Kinase Signaling System drug effects, MAP Kinase Signaling System physiology, Macrophages, Peritoneal cytology, Macrophages, Peritoneal immunology, Mice, Mice, Knockout, Mitogen-Activated Protein Kinase Kinases genetics, Mitogen-Activated Protein Kinase Kinases immunology, Mitogen-Activated Protein Kinase Kinases metabolism, Mutation, Myeloid Differentiation Factor 88 genetics, Myeloid Differentiation Factor 88 immunology, NF-kappa B genetics, NF-kappa B immunology, NF-kappa B metabolism, Proto-Oncogene Proteins c-jun genetics, Proto-Oncogene Proteins c-jun immunology, Proto-Oncogene Proteins c-jun metabolism, Response Elements genetics, Response Elements immunology, Toll-Like Receptor 4 genetics, Toll-Like Receptor 4 immunology, Transcription Factor AP-1 genetics, Transcription Factor AP-1 immunology, Gene Expression Regulation physiology, Interleukin-23 Subunit p19 biosynthesis, Macrophages, Peritoneal metabolism, Myeloid Differentiation Factor 88 metabolism, Toll-Like Receptor 4 metabolism, Transcription Factor AP-1 metabolism
- Abstract
Interleukin (IL)-23, a new member of the IL-12 family, plays a central role in the Th17 immune response and in autoimmune diseases. It is clear that activated macrophages and dendritic cells produce IL-23, but the molecular mechanisms whereby inflammatory signals stimulate IL-23 expression are not fully understood. We demonstrate that induction of IL-23 p19 gene expression by LPS depends on the TLR4 and MyD88 pathways. All three MAPK pathways (ERK, JNK, and p38) that are activated by lipopolysaccharide (LPS) stimulation were shown to exert a positive effect on p19 expression. We cloned a 1.3-kb putative p19 promoter and defined its transcription initiation sites by the 5'-rapid amplification of cDNA ends method. By analyzing IL-23 p19 promoter mutants, we have identified a promoter region (-413 to +10) that contains several important elements, including NF-kappaB and AP-1. In addition to NF-kappaB, we have demonstrated that the proximal AP-1 site is important for p19 promoter activation. Mutation of the AP-1 site resulted in the loss of p19 promoter activation. Electrophoretic mobility shift assay (EMSA) analysis showed that c-Jun and c-Fos bind to the AP-1 site, which was confirmed by a chromatin immunoprecipitation assay. Furthermore, co-transfection of c-Jun and ATF2 synergistically induced p19 promoter activation, and c-Jun and ATF2 formed a protein complex, demonstrated by co-immunoprecipitation. Finally, LPS-stimulated peritoneal macrophages from IL-10-deficient mice expressed significantly higher IL-23 p19 than macrophages from wild type mice, and the addition of recombinant IL-10 strongly inhibited LPS-induced p19 expression. Thus, this study suggests that MyD88-dependent Toll-like receptor signaling induces IL-23 p19 gene expression through both MAPKs and NF-kappaB.
- Published
- 2009
- Full Text
- View/download PDF
16. Endothelial cell annexin A2 regulates polyubiquitination and degradation of its binding partner S100A10/p11.
- Author
-
He KL, Deora AB, Xiong H, Ling Q, Weksler BB, Niesvizky R, and Hajjar KA
- Subjects
- Animals, Annexin A2 genetics, Antiphospholipid Syndrome blood, Antiphospholipid Syndrome genetics, Autoantibodies blood, Cell Line, Tumor, Fibrin genetics, Fibrin metabolism, Fibrinolysin genetics, Fibrinolysin metabolism, Fibrinolysis genetics, Hemorrhage blood, Hemorrhage etiology, Hemorrhage genetics, Humans, Leukemia, Promyelocytic, Acute blood, Leukemia, Promyelocytic, Acute complications, Leukemia, Promyelocytic, Acute genetics, Mice, Mice, Knockout, Oncogene Protein pp60(v-src) genetics, Oncogene Protein pp60(v-src) metabolism, Proteasome Endopeptidase Complex genetics, Proteasome Endopeptidase Complex metabolism, Protein Binding genetics, Protein Transport genetics, S100 Proteins genetics, Thrombosis blood, Thrombosis etiology, Tissue Plasminogen Activator genetics, Tissue Plasminogen Activator metabolism, Ubiquitin genetics, Ubiquitin metabolism, Annexin A2 metabolism, Endothelial Cells metabolism, S100 Proteins metabolism, Ubiquitination genetics
- Abstract
The annexin A2 (A2) heterotetramer, consisting of two copies of A2 and two copies of S100A10/p11, promotes fibrinolytic activity on the surface of vascular endothelial cells by assembling plasminogen and tissue plasminogen activator (tPA) and accelerating the generation of plasmin. In humans, overexpression of A2 by acute promyelocytic leukemia cells is associated with excessive fibrinolysis and hemorrhage, whereas anti-A2 autoantibodies appear to accentuate the risk of thrombosis in patients with anti-phospholipid syndrome. Complete deficiency of A2 in mice leads to a lack of tPA cofactor activity, accumulation of intravascular fibrin, and failure to clear arterial thrombi. Within the endothelial cell, p11 is required for Src kinase-mediated tyrosine phosphorylation of A2, which signals translocation of both proteins to the cell surface. Here we show that p11 is expressed at very low levels in the absence of A2 both in vitro and in vivo. We demonstrate further that unpartnered p11 becomes polyubiquitinated and degraded via a proteasome-dependent mechanism. A2 stabilizes intracellular p11 through direct binding, thus masking an autonomous p11 polyubiquitination signal that triggers proteasomal degradation. This interaction requires both the p11-binding N-terminal domain of A2 and the C-terminal domain of p11. This mechanism prevents accumulation of free p11 in the endothelial cell and suggests that regulation of tPA-dependent cell surface fibrinolytic activity is precisely tuned to the intracellular level of p11.
- Published
- 2008
- Full Text
- View/download PDF
17. Structural and functional characterization of a secreted hookworm Macrophage Migration Inhibitory Factor (MIF) that interacts with the human MIF receptor CD74.
- Author
-
Cho Y, Jones BF, Vermeire JJ, Leng L, DiFedele L, Harrison LM, Xiong H, Kwong YK, Chen Y, Bucala R, Lolis E, and Cappello M
- Subjects
- Amino Acid Sequence, Ancylostoma, Animals, Cricetinae, Cytokines metabolism, Humans, Intestinal Mucosa metabolism, Mesocricetus, Molecular Sequence Data, Protein Binding, Rabbits, Recombinant Proteins chemistry, Sequence Homology, Amino Acid, Antigens, Differentiation, B-Lymphocyte chemistry, Antigens, Differentiation, B-Lymphocyte physiology, Histocompatibility Antigens Class II chemistry, Histocompatibility Antigens Class II physiology, Macrophage Migration-Inhibitory Factors metabolism
- Abstract
Hookworms, parasitic nematodes that infect nearly one billion people worldwide, are a major cause of anemia and malnutrition. We hypothesize that hookworms actively manipulate the host immune response through the production of specific molecules designed to facilitate infection by larval stages and adult worm survival within the intestine. A full-length cDNA encoding a secreted orthologue of the human cytokine, Macrophage Migration Inhibitory Factor (MIF) has been cloned from the hookworm Ancylostoma ceylanicum. Elucidation of the three-dimensional crystal structure of recombinant AceMIF (rAceMIF) revealed an overall structural homology with significant differences in the tautomerase sites of the human and hookworm proteins. The relative bioactivities of human and hookworm MIF proteins were compared using in vitro assays of tautomerase activity, macrophage migration, and binding to MIF receptor CD74. The activity of rAceMIF was not inhibited by the ligand ISO-1, which was previously determined to be an inhibitor of the catalytic site of human MIF. These data define unique immunological, structural, and functional characteristics of AceMIF, thereby establishing the potential for selectively inhibiting the hookworm cytokine as a means of reducing parasite survival and disease pathogenesis.
- Published
- 2007
- Full Text
- View/download PDF
18. IRF-8/interferon (IFN) consensus sequence-binding protein is involved in Toll-like receptor (TLR) signaling and contributes to the cross-talk between TLR and IFN-gamma signaling pathways.
- Author
-
Zhao J, Kong HJ, Li H, Huang B, Yang M, Zhu C, Bogunovic M, Zheng F, Mayer L, Ozato K, Unkeless J, and Xiong H
- Subjects
- Cell Line, Cytokines metabolism, Humans, Immune System metabolism, Immunoprecipitation, Interferon Regulatory Factors metabolism, Ligands, Lipopolysaccharides chemistry, Macrophages metabolism, NF-kappa B metabolism, Peptidoglycan chemistry, Peritoneum pathology, Phosphorylation, Plasmids metabolism, Protein Binding, Retroviridae genetics, Retroviridae metabolism, Reverse Transcriptase Polymerase Chain Reaction, Signal Transduction, TNF Receptor-Associated Factor 6 metabolism, Time Factors, Transfection, Tumor Necrosis Factor-alpha metabolism, Interferon Regulatory Factors chemistry, Interferon-gamma metabolism, Interferons chemistry, Toll-Like Receptors metabolism
- Abstract
Toll-like receptor (TLR) and interferon-gamma (IFN-gamma) signaling pathways are important for both innate and adaptive immune responses. However, the cross-talk between these two signaling pathways is incompletely understood. Here we show that IFN-gamma and LPS synergistically induce the expression of proinflammatory factors, including interleukin-1 (IL-1), IL-6, IL-12, NO, and tumor necrosis factor-alpha (TNF-alpha). Comparable synergism was observed between IFN-gamma and peptidoglycan (PGN; a TLR2 ligand) and poly(I:C) (a TLR3 ligand) in the induction of IL-12 promoter activity. IFN-gamma enhanced lipopolysaccharide (LPS)-induced ERK and JNK phosphorylation but had no effect on LPS-induced NF-kappaB activation. Interestingly, we found that IRF-8-/- macrophages were impaired in the activation of LPS-induced ERK and JNK and the production of proinflammatory cytokines induced by LPS or IFN-gamma plus LPS. Retroviral transduction of IRF-8 into IRF-8-/- macrophages rescued ERK and JNK activation. Furthermore, co-immunoprecipitation experiments show that IRF-8 physically interacts with TRAF6 at a binding site between amino acid residues 356 and 305 of IRF-8. Transfection of IRF-8 enhanced TRAF6 ubiquitination, which is consistent with a physical interaction of IRF-8 with TRAF6. Taken together, the results suggest that the interaction of IRF-8 with TRAF6 modulates TLR signaling and may contribute to the cross-talk between IFN-gamma and TLR signal pathways.
- Published
- 2006
- Full Text
- View/download PDF
19. Ubiquitin-dependent degradation of interferon regulatory factor-8 mediated by Cbl down-regulates interleukin-12 expression.
- Author
-
Xiong H, Li H, Kong HJ, Chen Y, Zhao J, Xiong S, Huang B, Gu H, Mayer L, Ozato K, and Unkeless JC
- Subjects
- Animals, Cell Line, Humans, Hydrolysis, Immunoprecipitation, Interferon Regulatory Factors, Macrophages, Peritoneal metabolism, Mice, Mice, Inbred Strains, Proto-Oncogene Proteins c-cbl, Down-Regulation, Interleukin-12 metabolism, Proto-Oncogene Proteins physiology, Repressor Proteins metabolism, Ubiquitin metabolism, Ubiquitin-Protein Ligases physiology
- Abstract
Interferon regulatory factor (IRF)-8/interferon consensus sequence-binding protein is regulated by both transcription and degradation. IRF-8 induced in peritoneal macrophages by interferon-gamma and lipopolysaccharide was degraded rapidly, and degradation of IRF-8 was blocked by MG132, the proteasome inhibitor, but inhibitors of calpain and lysosomal enzymes had no effect. The ubiquitination of IRF-8 was shown by co-immunoprecipitation from RAW264.7 macrophages retrovirally transduced with IRF-8 and hemagglutinin-ubiquitin. The dominant negative ubiquitin mutants K48R and K29R inhibited IRF-8 degradation in 293T cells, confirming the relationship between ubiquitination of IRF-8 and its degradation. IRF-8 carboxyl-terminal truncation mutants were not ubiquitinated and were consequently stable, indicating that the carboxyl-terminal domain of IRF-8 controls ubiquitination. The ubiquitin-protein isopeptide ligase (E3) that ubiquitinated IRF-8 was likely to be Cbl, which formed a complex with IRF-8, demonstrable by both immunoprecipitation and gel filtration. Furthermore, IRF-8 stability was increased by dominant negative Cbl, and IRF-8 ubiquitination was significantly attenuated in Cbl-/- cells. Reflecting increased stability and expression, the IRF-8 carboxyl-terminal deletion mutant induced interleukin (IL)-12 p40 promoter activity much more strongly than IRF-8 did. Furthermore, IRF-8-induced IL-12 p40 synthesis in RAW264.7 cells was enhanced by dominant negative Cbl, and peritoneal macrophages from Cbl-/- mice showed increased IL-12 p40 protein production. Taken together, these results suggest that the proteasomal degradation of IRF-8 mediated by the ubiquitin E3 ligase Cbl down-regulates IL-12 expression.
- Published
- 2005
- Full Text
- View/download PDF
20. Interaction of TRAF6 with MAST205 regulates NF-kappaB activation and MAST205 stability.
- Author
-
Xiong H, Li H, Chen Y, Zhao J, and Unkeless JC
- Subjects
- Animals, Cell Line, Enzyme Stability, Macrophages, Mice, NF-kappa B genetics, Promoter Regions, Genetic, Transfection, Tumor Necrosis Factor-alpha metabolism, Microtubule-Associated Proteins metabolism, NF-kappa B metabolism, Protein Serine-Threonine Kinases metabolism, TNF Receptor-Associated Factor 6 metabolism
- Abstract
The binding of immune complexes to macrophage Fcgamma receptor results in a subsequent inhibition of lipopolysaccharide-stimulated interleukin-12 synthesis without affecting the induction of tumor necrosis factor-alpha. RNA interference targeting MAST205, a 205-kDa serine/threonine kinase, and transfection of dominant negative MAST205 mutants also mimic this type II macrophage phenotype. Our previous epistasis experiments suggested that the position of MAST205 in the TLR4 signal pathway was proximal to the IkappaB kinase complex. We now report that MAST205 forms a complex with TRAF6, resulting in the inhibition of TRAF6 NF-kappaB activation. We have identified a peptide (residues 218-233) from the N terminus of MAST205 that, when coupled to a protein transduction domain, inhibits the lipopolysaccharide-stimulated activation of NF-kappaB, modulates the size of the MAST205.TRAF6 complex, and inhibits ubiquitination of TRAF6. A dominant negative N-terminal MAST205 deletion mutant also inhibits TRAF6 ubiquitination. The domain required for degradation of MAST205 after Fcgamma receptor activation resides within the N-terminal 261 residues, and degradation is triggered by protein kinase C isoform phosphorylation of Ser/Thr residues. These results suggest that MAST205 functions as a scaffolding protein controlling TRAF6 activity and, therefore, plays an important role in regulating inflammatory responses.
- Published
- 2004
- Full Text
- View/download PDF
21. Inhibition of interleukin-12 p40 transcription and NF-kappaB activation by nitric oxide in murine macrophages and dendritic cells.
- Author
-
Xiong H, Zhu C, Li F, Hegazi R, He K, Babyatsky M, Bauer AJ, and Plevy SE
- Subjects
- Animals, Blotting, Western, Bone Marrow Cells metabolism, Cell Line, Dendritic Cells cytology, Enzyme Inhibitors pharmacology, Enzyme-Linked Immunosorbent Assay, Humans, Interleukin-1 Receptor-Associated Kinases, Interleukin-12 metabolism, Interleukin-12 Subunit p40, Mice, Mice, Inbred C57BL, Nitric Oxide Donors pharmacology, Nitric Oxide Synthase metabolism, Nitric Oxide Synthase Type II, Precipitin Tests, Promoter Regions, Genetic, Protein Binding, Protein Kinases metabolism, Proteins metabolism, RNA, Messenger metabolism, Reverse Transcriptase Polymerase Chain Reaction, S-Nitroso-N-Acetylpenicillamine pharmacology, Signal Transduction, Spleen cytology, TNF Receptor-Associated Factor 6, Transfection, Dendritic Cells metabolism, Interleukin-12 antagonists & inhibitors, Macrophages metabolism, NF-kappa B metabolism, Nitric Oxide metabolism, Protein Subunits antagonists & inhibitors, Transcription, Genetic
- Abstract
Nitric oxide (NO), an important effector molecule of the innate immune system, can also regulate adaptive immunity. In this study, the molecular effects of NO on the toll-like receptor signaling pathway were determined using interleukin-12 (IL-12) as an immunologically relevant target gene. The principal conclusion of these experiments is that NO inhibits IL-1 receptor-associated kinase (IRAK) activity and attenuates the molecular interaction between tumor necrosis factor receptor-associated factor-6 and IRAK. As a consequence, the NO donor S-nitroso-N-acetylpenicillamine (SNAP) inhibits lipopolysaccharide (LPS)-induced IL-12 p40 mRNA expression, protein production, and promoter activity in murine macrophages, dendritic cells, and the murine macrophage cell line RAW 264.7. Splenocytes from inducible nitric-oxide synthase-deficient mice demonstrate markedly increased IL-12 p40 protein and mRNA expression compared with wild type splenocytes. The inhibitory action of NO on IL-12 p40 is independent of the cytokine IL-10. The effects of NO can be directly attributed to inhibition of NF-kappaB activation through IRAK-dependent pathways. Accordingly, SNAP strongly reduces LPS-induced NF-kappaB DNA binding to the p40 promoter and inhibits LPS-induced IkappaB phosphorylation. Similarly, NO attenuates IL-1beta-induced NF-kappaB activation. These experiments provide another example of how an innate immune molecule may have a profound effect on adaptive immunity.
- Published
- 2004
- Full Text
- View/download PDF
22. Activation of the murine interleukin-12 p40 promoter by functional interactions between NFAT and ICSBP.
- Author
-
Zhu C, Rao K, Xiong H, Gagnidze K, Li F, Horvath C, and Plevy S
- Subjects
- Animals, Base Sequence, Binding Sites genetics, Cell Line, DNA genetics, DNA metabolism, DNA-Binding Proteins chemistry, DNA-Binding Proteins genetics, Gene Expression Regulation, Humans, Interferon Regulatory Factors, Interleukin-12 Subunit p40, Mice, Mutagenesis, Site-Directed, NFATC Transcription Factors, Protein Binding, RNA, Messenger genetics, RNA, Messenger metabolism, Recombinant Fusion Proteins chemistry, Recombinant Fusion Proteins genetics, Recombinant Fusion Proteins metabolism, Repressor Proteins genetics, Transcription Factors chemistry, Transcription Factors genetics, DNA-Binding Proteins metabolism, Interleukin-12 genetics, Nuclear Proteins, Promoter Regions, Genetic, Protein Subunits genetics, Repressor Proteins metabolism, Transcription Factors metabolism
- Abstract
Interleukin (IL)-12 is a heterodimeric cytokine that is critical for the development of a T-helper-1 immune response and immunity against intracellular pathogens. The IL-12 p40 gene product, expressed specifically in macrophages and dendritic cells, heterodimerizes with p35 to form bioactive IL-12, and heterodimerizes with p19 to comprise the cytokine IL-23. Regulation of the murine IL-12 p40 promoter is complex. Multiple cis-acting elements have been characterized that are involved in activation by bacterial products. However, molecular mechanisms through which interferon (IFN)-gamma and bacterial products synergistically activate IL-12 p40 gene expression are less clear. In this study, a composite NFAT/ICSBP binding site at -68 to -54 is identified that is functionally important for p40 promoter activation by lipopolysaccharide (LPS) and LPS plus IFN-gamma. DNA binding of NFAT and ICSBP is demonstrated on the endogenous promoter by chromatin immunoprecipitation. NFAT is required for ICSBP binding to this region. Overexpression of NFAT and ICSBP synergistically activates the p40 promoter. A dominant negative NFAT molecule attenuates LPS- and IFN-gamma-activated endogenous IL-12 p40 mRNA expression. A physical association between NFAT and ICSBP in the absence of DNA is detected by co-immunoprecipitation of endogenous proteins. Three NFAT domains are required for ICSBP interaction. Finally, in LPS- and IFN-gamma-activated RAW-264.7 cells, the association between NFAT and ICSBP is abrogated by IL-10 priming.
- Published
- 2003
- Full Text
- View/download PDF
23. Complex formation of the interferon (IFN) consensus sequence-binding protein with IRF-1 is essential for murine macrophage IFN-gamma-induced iNOS gene expression.
- Author
-
Xiong H, Zhu C, Li H, Chen F, Mayer L, Ozato K, Unkeless JC, and Plevy SE
- Subjects
- Animals, Binding Sites, Blotting, Western, Cell Line, DNA metabolism, Green Fluorescent Proteins, Interferon Regulatory Factor-1, Interferon Regulatory Factors, Interleukin-4 metabolism, Luminescent Proteins metabolism, Mice, Nitric Oxide metabolism, Nitric Oxide Synthase Type II, Plasmids metabolism, Precipitin Tests, Promoter Regions, Genetic, Protein Binding, Retroviridae genetics, Reverse Transcriptase Polymerase Chain Reaction, Signal Transduction, Time Factors, Transduction, Genetic, Transfection, DNA-Binding Proteins metabolism, Interferon-gamma metabolism, Interferons metabolism, Macrophages metabolism, Nitric Oxide Synthase metabolism, Phosphoproteins metabolism, Repressor Proteins metabolism, Repressor Proteins physiology
- Abstract
This study describes the role of the interferon (IFN) consensus sequence-binding protein (ICSBP or IRF-8) in iNOS gene expression by murine macrophages. An ICSBP binding site in the iNOS promoter region (-923 to -913) was identified using an electrophoretic mobility shift assay and chromatin co-immunoprecipitation. Overexpression of ICSBP greatly enhanced IFN-gamma-induced iNOS promoter activation in RAW264.7 cells, and IFN-gamma-induced iNOS promoter activation was abolished in ICSBP-/- macrophages. Furthermore, transduction of retrovirus-ICSBP in ICSBP-/- macrophages rescued IFN-gamma-induced iNOS gene expression. However, transduction of retrovirus-ICSBP in the absence of IFN-gamma activation did not induce iNOS expression in either RAW264.7 cells or ICSBP-/- macrophages. Interestingly, ICSBP alone transduced into ICSBP-/- macrophages did not bind to IFN-stimulated response element site (-923 to -913) of the iNOS promoter region, although following activation with IFN-gamma, a DNA.protein complex was formed that contains ICSBP and IRF-1. Co-transduction of ICSBP with IRF-1 clearly induces nitric oxide production. In addition, interleukin-4 inhibits IFN-gamma-induced iNOS gene expression by attenuating the physical interaction of ICSBP with IRF-1. Complex formation of ICSBP with IRF-1 is essential for iNOS expression, and interleukin-4 attenuates the physical interaction of ICSBP with IRF-1 resulting in the inhibition of INOS gene expression.
- Published
- 2003
- Full Text
- View/download PDF
24. Conformational remodeling of proteasomal substrates by PA700, the 19 S regulatory complex of the 26 S proteasome.
- Author
-
Liu CW, Millen L, Roman TB, Xiong H, Gilbert HF, Noiva R, DeMartino GN, and Thomas PJ
- Subjects
- Animals, Catalytic Domain, Cattle, Chymotrypsin pharmacology, Disulfides chemistry, Dose-Response Relationship, Drug, Enzyme Inhibitors pharmacology, Ligands, Methotrexate pharmacology, Oxygen metabolism, Peptides chemistry, Proteasome Endopeptidase Complex, Protein Conformation, Protein Folding, Ribonuclease, Pancreatic chemistry, Time Factors, Adenosine Triphosphatases chemistry, Endopeptidases chemistry
- Abstract
PA700, the 19 S regulatory complex of the 26 S proteasome, plays a central role in the recognition and efficient degradation of misfolded proteins. PA700 promotes degradation by recruiting proteasomal substrates utilizing polyubiquitin chains and chaperone-like binding activities and by opening the access to the core of the 20 S proteasome to promote degradation. Here we provide evidence that PA700 in addition to binding misfolded protein substrates also acts to remodel their conformation prior to proteolysis. Scrambled RNase A (scRNase A), a misfolded protein, only slowly refolds spontaneously into an active form because of the rate-limiting unfolding of misfolded disulfide isomers. Notably, PA700 accelerates the rate of reactivation of scRNase A, consistent with its ability to increase the exposure of these disulfide bonds to the solvent. In this regard, PA700 also exposes otherwise buried sites to digestion by exogenous chymotrypsin in a polyubiquitinated enzymatically active substrate, pentaubiquitinated dihydrofolate reductase, Ub(5)DHFR. The dihydrofolate reductase ligand methotrexate counters the ability of PA700 to promote digestion by chymotrypsin. Together, these results indicate that in addition to increasing substrate affinity and opening the access channel to the catalytic sites, PA700 activates proteasomal degradation by remodeling the conformation of protein substrates.
- Published
- 2002
- Full Text
- View/download PDF
25. Processing of mammalian and plant S-adenosylmethionine decarboxylase proenzymes.
- Author
-
Xiong H, Stanley BA, Tekwani BL, and Pegg AE
- Subjects
- Amino Acid Sequence, Amino Acid Substitution, Cysteine metabolism, Humans, Molecular Sequence Data, Mutagenesis, Site-Directed, Putrescine metabolism, Pyruvic Acid metabolism, Serine metabolism, Solanum tuberosum enzymology, Structure-Activity Relationship, Threonine metabolism, Adenosylmethionine Decarboxylase metabolism, Enzyme Precursors metabolism, Protein Processing, Post-Translational
- Abstract
S-Adenosylmethionine decarboxylase (AdoMetDC) is a pyruvoyl enzyme, and the pyruvate is formed in an intramolecular reaction that cleaves a proenzyme precursor and converts a serine residue into pyruvate. The wild type potato AdoMetDC proenzyme processed much faster than the human proenzyme and did not require putrescine for an optimal rate of processing despite the presence of three acidic residues (equivalent to Glu11, Glu178, and Glu256) that were demonstrated in previous studies to be required for the putrescine activation of human AdoMetDC proenzyme processing (Stanley, B. A., Shantz, L. M., and Pegg, A. E. (1994) J. Biol. Chem. 269, 7901-7907). A fourth residue that is also needed for the putrescine stimulation of human AdoMetDC proenzyme processing was identified in the present studies, and this residue (Asp174) is not present in the potato sequence. The site of potato AdoMetDC proenzyme processing was found to be Ser73 in the conserved sequence, YVLSESS, which is the equivalent of Ser68 in the human sequence. Replacement of the serine precursor with threonine or cysteine by site-directed mutagenesis in either the potato or the human AdoMetDC proenzyme did not prevent processing but caused a significant reduction in the rate. Although the COOH-terminal regions of the known eukaryotic AdoMetDCs are not conserved, only relatively small truncations of 8 residues from the human protein and 25 residues from the potato proenzyme were compatible with processing. The maximally truncated proteins show no similarity in COOH-terminal amino acid sequence but each contained 46 amino acid residues after the last conserved sequence, suggesting that the length of this section of the protein is essential for maintaining the proenzyme conformation needed for autocatalytic processing.
- Published
- 1997
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.