Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Biochem Biophys Res Commun ; 463(3): 453-7, 2015 Jul 31.
Article in English | MEDLINE | ID: mdl-26043696

ABSTRACT

Valosin-containing protein (VCP or p97) is required for the proteasomal degradation of polyubiquitinated proteins. However, the molecular mechanism for VCP to process the polyubiquitinated proteins remains unclear. Here, we show that VCP can unfold polyubiquitinated proteins. It preferably unfolds the pentaubiquitin-over monoubiquin-conjugated dihydrofolate reductase (Ub5-DHFR or Ub-DHFR) in a dose dependent manner. In addition, the unfolding activity of VCP does not depend on its ATPase activity, on the contrary, ATP and its non-hydrolysable analogs suppress the unfolding of Ub5-DHFR. The structural and functional analysis showed that either D1 or D2 domain of VCP is sufficient to carry out this unfolding activity. The structure of the substrates also affects its unfolding by VCP. VCP is unable to unfold Ub5-DHFR in a tight structure when it binds with methotrexate, a folate analog with high affinity to DHFR. Thus, these results support that VCP is capable of unfolding polyubiquitinated proteins and suggest that VCP may facilitate the proteasomal degradation of polyubiquitinated proteins through its unfolding activity.


Subject(s)
Adenosine Triphosphatases/metabolism , Adenosine Triphosphate/metabolism , Cell Cycle Proteins/metabolism , Protein Unfolding , Ubiquitination , Adenosine Triphosphatases/chemistry , Cell Cycle Proteins/chemistry , Chymotrypsin/metabolism , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Tetrahydrofolate Dehydrogenase/chemistry , Tetrahydrofolate Dehydrogenase/metabolism , Valosin Containing Protein
2.
Biochim Biophys Acta ; 1853(1): 222-32, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25447673

ABSTRACT

Valosin-containing protein (VCP or p97), a member of the AAA family (ATPases associated with diverse cellular activities), plays a key role in many important cellular activities. A genetic deficiency of VCP can cause inclusion body myopathy associated with Paget's disease of bone and frontotemporal dementia (IBMPFD). Previous studies showed that the VCP N domain is essential for the regulation of nuclear entry of VCP. Here we report that IBMPFD mutations, which are mainly located in the N domain, suppress the nuclear entry of VCP. Moreover, the peptide sequence G780AGPSQ in the C-terminal region regulates the retention of VCP in the nucleus. A mutant lacking this sequence can increase the nuclear distribution of IBMPFD VCP, suggesting that this sequence is a potential molecular target for correcting the deficient nucleocytoplasmic shuttling of IBMPFD VCP proteins.


Subject(s)
Adenosine Triphosphatases/metabolism , Cell Cycle Proteins/metabolism , Cell Nucleus/metabolism , Active Transport, Cell Nucleus , Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/physiology , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/genetics , Frontotemporal Dementia/genetics , HEK293 Cells , Humans , Muscular Dystrophies, Limb-Girdle/genetics , Myositis, Inclusion Body/genetics , Osteitis Deformans/genetics , Protein Structure, Tertiary , Valosin Containing Protein
3.
J Immunol ; 192(11): 5305-13, 2014 Jun 01.
Article in English | MEDLINE | ID: mdl-24778447

ABSTRACT

The cross-regulation of G protein-coupled receptors (GPCRs) plays an important role in the immune response. Studies from several laboratories have suggested that a hierarchy of sensitivities to cross-desensitization exists for the chemoattractant GPCRs. We carried out experiments to study the capacity of the formyl peptide receptor-1 (FPR1) to desensitize chemokine receptors CCR1 and CCR2. Our results show that activation of FPR1 resulted in the desensitization and partial internalization of CCR1, but not CCR2, in both primary human monocytes and HEK293 cells coexpressing CCR1, CCR2, and FPR1 (HR1R2F cells). The desensitization of CCR1 by FPR1 stimulation was not due to the simple depletion of the Ca(2+) stores, but was dependent on activation of protein kinase C. Furthermore, we found that the cross-desensitization of CCR1 by FPR1 was associated with CCR1 phosphorylation and moderate reduction of CCR1 cell-surface expression. In contrast, CCR2 was not phosphorylated or internalized after FPR1 activation. Additional studies showed that optimal cross talk between FPR1 and CCR1 was dependent on the functional activity of protein kinase Cß. These results provide a mechanistic basis for the capacity of certain GPCR ligands to exert rapid and selective cross-inactivation of other chemoattractant receptors, and suggest that FPR1 is able to exert "traffic control" in the migration of inflammatory cells by rapidly inhibiting the cell responses to potentially "low-priority" chemoattractants such as CCR1 agonists without inhibiting the response to "higher priority" CCR2 chemoattractants.


Subject(s)
Monocytes/immunology , Receptors, CCR1/immunology , Receptors, CCR2/immunology , Receptors, Formyl Peptide/immunology , Calcium/immunology , Gene Expression Regulation/immunology , HEK293 Cells , Humans , Monocytes/cytology , Protein Kinase C beta/immunology
4.
J Biol Chem ; 286(23): 20354-65, 2011 Jun 10.
Article in English | MEDLINE | ID: mdl-21454526

ABSTRACT

We have previously shown that the µ-opioid receptor (MOR) is capable of mediating cross-desensitization of several chemokine receptors including CCR5, but the biochemical mechanism of this process has not been fully elucidated. We have carried out a series of functional and biochemical studies and found that the mechanism of MOR-induced cross-desensitization of CCR5 involves the activation of PKCζ. Inhibition of PKCζ by its pseudosubstrate inhibitor, or its siRNA, or dominant negative mutants suppresses the cross-desensitization of CCR5. Our results further indicate that the activation of PKCζ is mediated through a pathway involving phosphoinositol-dependent kinase-1 (PDK1). In addition, activation of MOR elevates the phosphorylation level and kinase activity of PKCζ. The phosphorylation of PKCζ can be suppressed by a dominant negative mutant of PDK1. We observed that following MOR activation, the interaction between PKCζ and PDK1 is immediately increased based on the analysis of fluorescent resonance energy transfer in cells with the expression of PKCζ-YFP and PDK1-CFP. In addition, cells expressing PKCζ kinase motif mutants (Lys-281, Thr-410, Thr-560) fail to exhibit full MOR-induced desensitization of CCR5 activity. Taken together, we propose that upon DAMGO treatment, MOR activates PKCζ through a PDK1-dependent signaling pathway to induce CCR5 phosphorylation and desensitization. Because CCR5 is a highly proinflammatory receptor, and a critical coreceptor for HIV-1, these results may provide a novel approach for the development of specific therapeutic agents to treat patients with certain inflammatory diseases or AIDS.


Subject(s)
Protein Kinase C/metabolism , Receptors, Opioid, mu/metabolism , Signal Transduction/physiology , Acquired Immunodeficiency Syndrome/drug therapy , Acquired Immunodeficiency Syndrome/genetics , Acquired Immunodeficiency Syndrome/metabolism , Amino Acid Motifs , Analgesics, Opioid/pharmacology , Animals , CHO Cells , Cricetinae , Cricetulus , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , HIV-1 , Humans , Mice , Mutation , Peptide Fragments , Phosphorylation/drug effects , Phosphorylation/physiology , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Receptors, CCR5 , Receptors, Opioid, mu/genetics , Signal Transduction/drug effects
5.
Semin Respir Crit Care Med ; 31(3): 257-66, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20496295

ABSTRACT

Chronic obstructive pulmonary disease is characterized by an abnormal persistent inflammatory response to noxious environmental stimuli, most commonly cigarette smoke. Although cigarette smoking elicits airway inflammation in all of those who smoke, persistent inflammation and clinically significant COPD occurs in only a minority of smokers. The pathogenesis of COPD involves the recruitment and regulation of neutrophils, macrophages, and lymphocytes to the lung, as well as the induction of oxidative stress, all of which result in lung parenchymal destruction and airway remodeling. Recent research has generated a greater understanding of the mechanisms responsible for COPD development, including new concepts in T cell biology and the increasing recognition that the processes governing lung cell apoptosis are upregulated. We are also starting to understand the reasons for continued inflammation even after smoking cessation, which accelerates the rate of lung function decline in COPD. Herein we review our current knowledge of the inflammatory pathways involved in COPD pathogenesis, as well as newer concepts that have begun to unfold in recent years.


Subject(s)
Inflammation/etiology , Oxidative Stress , Pulmonary Disease, Chronic Obstructive/physiopathology , Animals , Apoptosis , Humans , Lung/pathology , Lung Diseases, Obstructive/physiopathology , Smoking/adverse effects , Smoking Cessation , Up-Regulation
6.
Hepatology ; 47(6): 1964-73, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18506888

ABSTRACT

UNLABELLED: Melittin, a water-soluble toxic peptide derived from bee venom of Apis mellifera was reported to have inhibitory effects on hepatocellular carcinoma (HCC). However, its role in antimetastasis and the underlying mechanism remains elusive. By utilizing both HCC cell lines and an animal model based assay system, we found that Rac1, which has been shown to be involved in cancer cell metastasis, is highly expressed in aggressive HCC cell lines and its activity correlated with cell motility and cytoskeleton polymerization. In addition, Rac1-dependent activity and metastatic potential of aggressive HCC cells are remarkably high in both cellular and nude mouse models. We provide evidence here that melittin inhibits the viability and motility of HCC cells in vitro, which correlates with its suppression of Rac1-dependent activity, cell motility, and microfilament depolymerization. Furthermore, melittin suppresses both HCC metastasis and Rac1-dependent activity in nude mouse models. The specificity of the effect of melittin on Rac1 was confirmed in HCC cells both in vitro and in vivo. CONCLUSION: Melittin inhibits tumor cell metastasis by reducing cell motility and migration via the suppression of Rac1-dependent pathway, suggesting that melittin is a potential therapeutic agent for HCC.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/prevention & control , Liver Neoplasms/metabolism , Liver Neoplasms/prevention & control , Melitten/pharmacology , Signal Transduction/physiology , rac1 GTP-Binding Protein/metabolism , Actin Cytoskeleton/drug effects , Animals , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , Disease Models, Animal , Gene Expression Regulation, Neoplastic/drug effects , Humans , Liver Neoplasms/pathology , MAP Kinase Kinase 4/metabolism , Mice , Mice, Nude , Neoplasm Metastasis/prevention & control , Signal Transduction/drug effects , Xenograft Model Antitumor Assays , rac1 GTP-Binding Protein/genetics
7.
Toxicol Appl Pharmacol ; 228(3): 351-63, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18261755

ABSTRACT

Cadmium (Cd2+) is a heavy metal ion known to have a long biological half-life in humans. Accumulating evidence shows that exposure to Cd2+ is associated with neurodegenerative diseases characterized by the retention of ubiquitinated and misfolded proteins in the lesions. Here, we report that Cd2+ directly induces the formation of protein inclusion bodies in cells. The protein inclusion body is an aggresome, a major organelle for collecting ubiquitinated or misfolded proteins. Our results show that aggresomes are enriched in the detergent-insoluble fraction of Cd2+-treated cell lysates. Proteomic analysis identified 145 proteins in the aggresome-enriched fractions. One of the proteins is the highly conserved valosin-containing protein (VCP), which has been shown to colocalize with aggresomes and bind ubiquitinated proteins through its N domain (#1-200). Our subsequent examination of VCP's role in the formation of aggresomes induced by Cd2+ indicates that the C-terminal tail (#780-806) of VCP interacts with histone deacetylase HDAC6, a mediator for aggresome formation, suggesting that VCP participates in transporting ubiquitinated proteins to aggresomes. This function of VCP is impaired by inhibition of the deacetylase activity of HDAC6 or by over-expression of VCP mutants that do not bind ubiquitinated proteins or HDAC6. Our results indicate that Cd2+ induces the formation of protein inclusion bodies by promoting the accumulation of ubiquitinated proteins in aggresomes through VCP and HDAC6. Our delineation of the role of VCP in regulating cell responses to ubiquitinated proteins has important implications for understanding Cd2+ toxicity and associated diseases.


Subject(s)
Adenosine Triphosphatases/physiology , Cadmium/toxicity , Cell Cycle Proteins/physiology , Inclusion Bodies/drug effects , Protein Folding , Ubiquitin/metabolism , Adenosine Triphosphatases/chemistry , Cell Cycle Proteins/chemistry , Cells, Cultured , Histone Deacetylase 6 , Histone Deacetylases/physiology , Humans , Inclusion Bodies/metabolism , Mass Spectrometry , Protein Structure, Tertiary , Valosin Containing Protein
8.
Ai Zheng ; 26(12): 1315-22, 2007 Dec.
Article in Chinese | MEDLINE | ID: mdl-18076793

ABSTRACT

BACKGROUND & OBJECTIVE: Melittin has antitumor effects on osteosarcoma, leukemia, and cervical cancer in vitro. Our previous experiments showed that melittin could inhibit proliferation and induce apoptosis of human hepatocellular carcinoma BEL-7402 cells. This study was to examine the effects of melittin on the growth and angiogenesis of BEL-7402 cell xenografts in nude mice. METHODS: The xenografts derived from BEL-7402 cells were established in BALB/C nude mice. Inoculated mice were randomly divided into normal saline (NS, 10 ml/kg) group, positive control (thalidomide, TLD, 200 mg/kg) group, low dose melittin (40 microg/kg) group, moderate dose melittin (60 microg/kg) group and high dose melittin (80 microg/kg) group. Tumor volume was measured. Tumor tissue was observed under microscope. Microvessel density (MVD) and the expression of vascular endothelial growth factor (VEGF), basic fibroblast growth factor (bFGF) and nuclear factor kappaB (NF-kappaB) were detected by SABC immunohistochemistry. The mRNA levels of VEGF and bFGF were analyzed by real-time fluorescent quantitative polymerase chain reaction. RESULTS: The relative tumor volume (V/V0) and MVD were significantly lower in low, moderate and high dose melittin groups than in NS group (4.42+/-0.58, 3.47+/-0.97, and 3.06+/-1.23 vs. 9.06+/-1.45, P<0.01; 11.33+/-1.86, 9.17+/-1.17, and 6.67+/-1.21 vs. 16.50+/-2.35, P<0.01). Tumor tissue necrosis was observed in melittin-treated groups and tumor vessels were destroyed by melittin. The positive expression indexes of VEGF (2.59+/-0.27, 2.61+/-0.17, 1.55+/-0.22 vs. 3.80+/-0.60, P<0.01), bFGF (2.45+/-0.78, 2.27+/-0.36, 2.10+/-0.27 vs. 4.43+/-0.34, P<0.01) and NF-kappaB (2.79+/-0.29, 2.71+/-0.66, 2.26+/-0.56 vs. 4.98+/-0.63, P<0.01) were significantly lower in low, moderate and high dose melittin groups than in NS group. The mRNA levels of VEGF and bFGF were also significantly lower in melittin-treated groups than in NS group. CONCLUSIONS: Melittin could inhibit the growth of BEL-7402 cell xenografts in nude mice. The down-regulation of VEGF, b-FGF and NF-kappaB expression and the inhibition of angiogenesis might play key roles in the antitumor effect of melittin.


Subject(s)
Liver Neoplasms/pathology , Melitten/pharmacology , Microvessels/drug effects , Neovascularization, Pathologic/pathology , Tumor Burden/drug effects , Animals , Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Down-Regulation , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/metabolism , Humans , Liver Neoplasms/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Nude , NF-kappa B/metabolism , Neoplasm Transplantation , RNA, Messenger/metabolism , Random Allocation , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
9.
Biochemistry ; 46(51): 14889-98, 2007 Dec 25.
Article in English | MEDLINE | ID: mdl-18044963

ABSTRACT

The 97 kDa valosin-containing protein (VCP) belongs to a highly conserved AAA (ATPases associated with a variety of activities) family and contains two ATPase domains, D1 and D2. VCP participates in numerous cellular activities, such as membrane fusion, postmitotic Golgi reassembly, endoplasmic reticulum-associated degradation, ubiquitin-proteasome-mediated proteolysis, and many others. In performing these activities, VCP presumably acts as a molecular chaperone that prevents protein aggregation and modifies protein conformation. In this study, we characterized the aggregation-prevention activity of VCP and identified the structural requirement for this activity. We used multiple methods to treat aggregation-prone luciferase (Luc) and showed that VCP prevents the aggregation of Luc in vitro. These results are in agreement; in vivo RNA interference analyses showed that a reduction of VCP level results in more aggregation of Luc in cells. Structural and functional analyses further demonstrated that the D1 domain of VCP is sufficient to mediate the aggregation-prevention activity, which does not require ATP binding, ATP hydrolysis, or a hexameric structure of VCP. Together, these results indicate that (1) VCP prevents protein aggregation in vitro and in vivo, (2) this aggregation-prevention activity is mediated mainly through the D1 domain of VCP, and (3) this activity does not require ATPase activity or a hexameric structure of VCP.


Subject(s)
Adenosine Triphosphatases/metabolism , Cell Cycle Proteins/metabolism , Adenosine Triphosphatases/genetics , Cell Cycle Proteins/genetics , Cell Line , Genes, Reporter/genetics , Humans , Mutation/genetics , Protein Binding , Protein Denaturation , Temperature , Valosin Containing Protein
10.
J Biol Chem ; 280(49): 40515-23, 2005 Dec 09.
Article in English | MEDLINE | ID: mdl-16216872

ABSTRACT

The 97-kDa molecular chaperone valosin-containing protein (VCP) belongs to a highly conserved AAA family and forms a hexameric structure that is essential for its biological functions. The AAA domain contains highly conserved motifs, the Walker A, Walker B, and the second region of homology (SRH). Although Walker A and B motifs mediate ATP binding and hydrolysis, respectively, the function of the SRH in VCP is not clear. We examined the significance of the SRH in VCP, especially the conserved Arg(359) and Arg(362) in the first AAA domain, D1 and Arg(635) and Arg(638) in the second AAA domain, D2. We show that Arg(359) and Arg(362) in D1 are critical for maintaining the hexameric structure and the ability to bind the polyubiquitin chains. Although the rest of the tested SRH mutants retain the hexameric structure, all of them exhibit severely reduced ATPase activity. Tryptophan fluorescence analysis showed that all of the tested mutants can bind to ATP or ADP. Thus, the reduced ATPase activity likely results from the hampered communications among protomers during hydrolysis. Moreover, when the ATPase-defective mutant R635A or R638A is mixed with the Walker A mutant of D2, the ATPase activity is partially restored, suggesting that Arg(635) and Arg(638) can stimulate the ATPase activity of the neighboring protomer. Interestingly, mutation of Arg(359) and Arg(362) uncouples the inhibitory effect of p47, a VCP co-factor, on the ATPase activity of VCP. Therefore, the Arg residues allow D1 to take on a specific conformation that is required for substrate binding and co-factor communications. Taken together, these results demonstrate that the conserved Arg residues in the SRH of both D1 and D2 play critical roles in communicating the conformational changes required for ATP hydrolysis, and SRH in D1 also contributes to substrate binding and co-factor communications.


Subject(s)
Arginine/physiology , Cell Cycle Proteins/chemistry , Conserved Sequence , Sequence Homology , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Arginine/chemistry , Arginine/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/physiology , Hydrolysis , Molecular Sequence Data , Mutagenesis, Site-Directed , Polyubiquitin/metabolism , Protein Conformation , Recombinant Fusion Proteins , Spectrometry, Fluorescence , Structure-Activity Relationship , Tryptophan/chemistry , Valosin Containing Protein
11.
World J Gastroenterol ; 11(26): 4003-7, 2005 Jul 14.
Article in English | MEDLINE | ID: mdl-15996023

ABSTRACT

AIM: To enhance the radiosensitivity of human colon cancer cells by docetaxel. METHODS: Immunoliposomal docetaxel was prepared by coupling monoclonal antibody against carcinoembryonic antigen to cyanuric chloride at the PEG terminus of liposome. LoVo adenocarcinoma cell line was treated with immunoliposomal docetaxel or/and irradiation. MTT colorimetric assay was used to estimate cytotoxicity of immunoliposomal docetaxel and radiotoxicity. Cell cycle redistribution and apoptosis were determined with flow cytometry. Survivin expression in LoVo cells was verified by immunohistochemistry. D801 morphologic analysis system was used to semi-quantify immunohistochemical staining of survivin. RESULTS: Cytotoxicity was induced by immunoliposomal docetaxel alone in a dose-dependent manner. Immunoli-posomal docetaxel yielded a cytotoxicity effect at a low dose of 2 nmol/L. With a single dose irradiation, the relative surviving fraction of LoVo cells showed a dose-dependent response, but there were no significant changes as radiation delivered from 4 to 8 Gy. Compared with liposomal docetaxel or single dose irradiation, strongly radiopotentiating effects of immunoliposomal docetaxel on LoVo cells were observed. A low dose of immunoliposomal docetaxel could yield sufficient radiosensitivity. Immunoliposomal docetaxel were achieved both specificity of the conjugated antibody and drug radiosensitization. Combined with radiation, immunoliposomal docetaxel significantly increased the percentage of G(2)/M cells and induced apoptosis, but significantly decreased the percentage of cells in G(2)/G(1) and S phase by comparison with liposomal docetaxel. Immunohistochemical analysis showed that the brown stained survivin was mainly in cytoplasm of LoVo cells. Semi-quantitative analysis of the survivin immunostaining showed that the expression of survivin in LoVo cells under irradiation with immunoliposomal docetaxel was significantly decreased. CONCLUSION: Immunoliposomal docetaxel is strongly effective for target radiosensitation in LoVo colon carcinoma cells, and may offer the potential to improve local radiotherapy.


Subject(s)
Cell Survival/drug effects , Radiation-Sensitizing Agents/toxicity , Taxoids/toxicity , Cell Line, Tumor , Cell Survival/radiation effects , Colonic Neoplasms , Docetaxel , Dose-Response Relationship, Radiation , Humans , Liposomes
12.
J Struct Biol ; 146(1-2): 44-57, 2004.
Article in English | MEDLINE | ID: mdl-15037236

ABSTRACT

The 97-kDa valosin-containing protein (p97 or VCP) is a type-II AAA ( ATPases associated with a variety of activities) ATPases, which are characterized by possessing two conserved ATPase domains. VCP forms a stable homo-hexameric structure, and this two-tier ring-shaped complex acts as a molecular chaperone that mediates many seemingly unrelated cellular activities. The involvement of VCP in the ubiquitin-proteasome degradation pathway and the identification of VCP cofactors provided us important clues to the understanding of how this molecular chaperone works. In this review, we summarize the reported biological functions of VCP and explore the molecular mechanisms underlying the diverse cellular functions. We discuss the structural and biochemical studies, and elucidate how this sophisticated enzymatic machine converts chemical energy into the mechanical forces required for the chaperone activity.


Subject(s)
Cell Cycle Proteins/chemistry , Cell Cycle Proteins/physiology , Adenosine Triphosphatases , Molecular Chaperones/chemistry , Molecular Chaperones/physiology , Molecular Motor Proteins , Protein Structure, Quaternary , Valosin Containing Protein
13.
World J Gastroenterol ; 10(3): 389-92, 2004 Feb 01.
Article in English | MEDLINE | ID: mdl-14760764

ABSTRACT

AIM: A strategy for viral vaccine design is the use of conserved peptides to overcome the problem of sequence diversity. At present it is still unclear whether conserved peptide is safe as a candidate vaccine. We reported it here for the first time not only to highlight the biohazard issue and safety importance for viral peptide vaccine, but also to explore the effect of a fully conserved peptide on HBV replication within the carboxyl terminus of HBx. METHODS: We synthesized the fully conserved peptide (CP) with nine residues, FVLGGCRHK. HBV-producing 2.2.15 cells were treated with or without 3.5 microM CP for 36 hours. Quantitative detection of viral DNA was performed by real-time PCR. HBV antigens were determined by enzyme-linked immunoadsorbent assay (ELISA). Quantitative analyses of p53 and Bax proteins were based on immunofluorescence. Flow cytometry was performed to detect cell cycle and apoptosis. RESULTS: Both extracellular and intracellular copies of HBV DNA per ml were significantly increased after incubation with 3.5 microM of CP. HBsAg and HBeAg in the cultured medium of CP-treatment cells were as abundant as untreated control cells. CP influenced negatively the extracellular viral gene products, and 3.5 microM CP could significantly inhibit intracellular HBsAg expression. In response to CP, intracellular HBeAg displayed an opposite pattern to that of HBsAg, and 3.5 microM CP could efficiently increase the level of intracellular HBeAg. Flow cytometric analyses exhibited no significant changes on cell cycle, apoptosis, p53 and Bax proteins in 2.2.15 cells with or without CP. CONCLUSION: Together with the results generated from the synthetic peptide, we address that the conserved region, a domain of HBx, may be responsible for modulating HBV replication. As conserved peptides from infectious microbes are used as immunogens to elicit immune responses, their latent biological hazard for human beings should be evaluated.


Subject(s)
Hepatitis B virus/physiology , Trans-Activators/chemistry , Trans-Activators/pharmacology , Virus Replication/drug effects , Cell Line, Tumor , Conserved Sequence , Hepatitis B virus/genetics , Humans , Peptide Fragments/chemical synthesis , Peptide Fragments/genetics , Trans-Activators/genetics , Viral Regulatory and Accessory Proteins
14.
World J Gastroenterol ; 9(7): 1521-4, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12854155

ABSTRACT

AIM: To investigate whether the formation of aggregated HBx has a potential linking with its cellular responses. METHODS: Recombinant HBx was expressed in Escherichia coli and purified by Ni-NTA metal-affinity chromatography. Anti-HBx monoclonal antibody was developed for immunocytochemical detection. Bicistronic expression vector harboring full-length DNA of HBx was employed for transfection of human HepG2 cells. Immunocytochemical staining was used to examine the intracellular HBx aggregates in cells. The effects of HBx aggregation on cell cycle and apoptosis were assessed by flow cytometry. RESULTS: Immunocytochemical staining revealed most of the HBx was formed intracellular aggregate in cytoplasm and frequently accumulated in large granules. Flow cytometry analysis showed that HepG2 cells transfected with vector harboring HBx significantly increased apoptosis and largely accumulated in the G0-G1 phase by maintenance in serum medium for 36 hours. Control cells without HBx aggregates in the presence of serum entered S phase and proliferated more rapidly at the same time. EGFP fluorescence in HBx expression cells was significantly decreased. CONCLUSION: Our observations show that cells with HBx aggregate undergo growth arrest and apoptosis, whereas control cells without HBx remain in growth and progression into S phase. Our data may provide helpful information to understand the biological effects of HBx aggregates on cells.


Subject(s)
Apoptosis/physiology , Trans-Activators/metabolism , Antibodies, Monoclonal , Carcinoma, Hepatocellular , Cell Cycle/physiology , Cell Line, Tumor/cytology , Cell Line, Tumor/metabolism , Cell Line, Tumor/virology , Cytoplasm/metabolism , Cytoplasm/virology , Cytoplasmic Granules/metabolism , Cytoplasmic Granules/virology , Flow Cytometry , Genetic Vectors , Green Fluorescent Proteins , Humans , Immunohistochemistry , Indicators and Reagents/metabolism , Liver Neoplasms , Luminescent Proteins/genetics , Trans-Activators/genetics , Trans-Activators/immunology , Transfection , Viral Regulatory and Accessory Proteins
15.
J Biol Chem ; 278(35): 32784-93, 2003 Aug 29.
Article in English | MEDLINE | ID: mdl-12807884

ABSTRACT

The 97-kDa valosin-containing protein (p97-VCP) belongs to the AAA (ATPases associated with various cellular activities) family and acts as a molecular chaperone in diverse cellular events, including ubiquitinproteasome-mediated degradation. We previously showed that VCP contains a substrate-binding domain, N, and two conserved ATPase domains, D1 and D2, of which D2 is responsible for the major enzyme activity. VCP has a barrel-like structure containing two stacked homo-hexameric rings made of the D1 and D2 domains, and this structure is essential for its biological functions. During ATPase cycles, VCP undergoes conformational changes that presumably apply tensions to the bound substrate, leading to the disassembly of protein complexes or unfolding of the substrate. How ATPase activity is coupled with the conformational changes in VCP complex and the D1 and D2 rings is not clear. In this report, we took biochemical approaches to study the structure of VCP in different nucleotide conditions to depict the conformational changes in the ATPase cycles. In contrast to many AAA chaperones that require ATP/ADP to form oligomers, both wild type VCP and ATP-binding site mutants can form hexamers without the addition of nucleotide. This nucleotide-independent hexamerization requires an intact D1 and the down-stream linker sequence of VCP. Tryptophan fluorescence and trypsin digestion analyses showed that ATP/ADP binding induces dramatic conformational changes in VCP. These changes do not require the presence of an intact ATP-binding site in D1 and is thus mainly attributed to the D2 domain. We propose a model whereby D1, although undergoing minor conformational changes, remains as a relatively trypsin-resistant hexameric ring throughout the ATPase cycle, whereas D2 only does so when it binds to ATP or ADP. After ADP is released at the end of the ATP hydrolysis, D2 ring is destabilized and adopts a relatively flexible and open structure.


Subject(s)
Adenosine Triphosphatases/chemistry , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/physiology , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Binding Sites , Blotting, Western , Cell Cycle Proteins/genetics , Chromatography, Gel , Chromatography, High Pressure Liquid , Cloning, Molecular , Electrophoresis, Polyacrylamide Gel , Hydrolysis , Models, Biological , Models, Genetic , Molecular Chaperones/metabolism , Mutagenesis, Site-Directed , Mutation , Nucleotides/genetics , Protein Conformation , Protein Structure, Tertiary , Recombinant Fusion Proteins/metabolism , Spectrometry, Fluorescence , Time Factors , Tryptophan/chemistry , Valosin Containing Protein
16.
J Biol Chem ; 278(6): 3648-55, 2003 Feb 07.
Article in English | MEDLINE | ID: mdl-12446676

ABSTRACT

The 97-kDa valosin-containing protein (p97-VCP) plays a role in a wide variety of cellular activities, many of which are regulated by the ubiquitin-proteasome (Ub-Pr)-mediated degradation pathway. We previously demonstrated that VCP binds to multi-ubiquitin chains and may act as a molecular chaperone that targets the ubiquitinated substrates to the proteasome for degradation. In this report, we show that although the ubiquitin chain-binding activity, carried out by the N-terminal 200 residues (N domain), is necessary for the degradation of proteasome substrates, it is not sufficient. Using in vitro degradation assays, we demonstrated that the entire VCP molecule, consisting of the N domain and two ATPase domains D1 and D2, is required for mediating the Ub-Pr degradation. The ATPase activity of VCP requires Mg(2+), and is stimulated by high temperature. Under optimal conditions, VCP hydrolyzes ATP with a K(m) of approximately 0.33 mm and a V(max) of approximately 0.52 nmol P(i) min(-1) microg(-1). At a physiological temperature, mutation in D2 significantly inhibits the ATPase activity, while that in D1 has little effect. Interestingly, mutations in D1, but not D2, abolish the heat-stimulated ATPase activity. Thus, we provide the first demonstration that the ATPase activity of VCP is required for mediating the Ub-Pr degradation, that D2 accounts for the major ATPase activity, and that D1 contributes to the heat-induced activity.


Subject(s)
Adenosine Triphosphatases/metabolism , Cell Cycle Proteins/metabolism , Hot Temperature , Base Sequence , Cell Cycle Proteins/isolation & purification , DNA Primers , Enzyme Activation , Hydrolysis , Mutagenesis, Site-Directed , Valosin Containing Protein
17.
Biochem Biophys Res Commun ; 300(2): 253-60, 2003 Jan 10.
Article in English | MEDLINE | ID: mdl-12504076

ABSTRACT

The 97-kDa valosin-containing protein (p97-VCP or VCP), a hexameric AAA ATPase, plays an important role in diverse cell activities, including ubiquitin-proteasome mediated protein degradation. In this report, we studied dissociation-reassembly kinetics to analyze the structure-function relationship in VCP. Urea-dissociated VCP can reassemble by itself, but addition of ATP, ADP, or ATP-gamma S accelerates the reassembly. Mutation in the ATP-binding site of D1, but not D2, domain abolishes the ATP acceleration effect and further delays the reassembly. Using hybrid hexamers of the wild type and ATP-binding site mutant, we show that hexameric structure and proper communication among the subunits are required for the ATPase activity and ubiquitin-proteasome mediated degradation. Thus, ATP-binding site in D1 plays a major role in VCP hexamerization, of which proper inter-subunit interaction is essential for the activities.


Subject(s)
Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/physiology , Adenosine Triphosphate/metabolism , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/physiology , Adenosine Triphosphatases/metabolism , Animals , Binding Sites , Cell Cycle Proteins/metabolism , Cysteine Endopeptidases/metabolism , Kinetics , Multienzyme Complexes/metabolism , Mutagenesis, Site-Directed , Proteasome Endopeptidase Complex , Protein Denaturation , Protein Structure, Tertiary , Protein Subunits , Structure-Activity Relationship , Ubiquitin/metabolism , Urea/pharmacology , Valosin Containing Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...