Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
2.
Biochemistry ; 56(27): 3434-3442, 2017 07 11.
Article in English | MEDLINE | ID: mdl-28627879

ABSTRACT

The heterodimeric human immunodeficiency virus type 1 reverse transcriptase is composed of p66 and p51 subunits. While in the p51 subunit, the connection domain is tucked in the polymerase cleft; it is effectively displaced from the cleft of the catalytically active p66 subunit. How is the connection domain relocated from the polymerase cleft of p66? Does the RNase H domain have any role in this process? To answer this question, we extended the C-terminal region of p51 by stepwise addition of N-terminal motifs of RNase H domain to generate p54, p57, p60, and p63 derivatives. We found all of the C-terminal extended derivatives of p51 assume open conformation, bind to the template-primer, and catalyze the polymerase reaction. Glycerol gradient ultracentrifugation analysis showed that only p54 sedimented as a monomer, while other derivatives were in a homodimeric conformation. We proposed a model to explain the monomeric conformation of catalytically active p54 derivative carrying additional 21-residues long ß1'-ß2' motif from the RNase H domain. Our results indicate that the ß1'-ß2' motif of the RNase H domain may be responsible for displacing the connection domain from the polymerase cleft of putative monomeric p66. The unstable elongated p66 molecule may then readily dimerize with p51 to assume a stable dimeric conformation.


Subject(s)
HIV Reverse Transcriptase/chemistry , HIV-1/enzymology , Models, Molecular , Ribonuclease H, Human Immunodeficiency Virus/chemistry , Amino Acid Motifs , Dimerization , Enzyme Stability , HIV Reverse Transcriptase/genetics , HIV Reverse Transcriptase/metabolism , Hydrophobic and Hydrophilic Interactions , Kinetics , Molecular Docking Simulation , Molecular Weight , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Conformation , Protein Conformation, beta-Strand , Protein Folding , Protein Interaction Domains and Motifs , Protein Multimerization , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Ribonuclease H, Human Immunodeficiency Virus/genetics , Ribonuclease H, Human Immunodeficiency Virus/metabolism
3.
Virology ; 500: 35-49, 2017 01.
Article in English | MEDLINE | ID: mdl-27770702

ABSTRACT

We recently identified a cell-factor, ErbB3 binding protein 1 (Ebp-1), which specifically interacts with the viral RNA genome and modulates HCV replication and translation. Ebp1 has two isoforms, p48, and p42, that result from differential splicing. We found that both isoforms interact with HCV proteins NS5A and NS5B, as well as cell-factor PKR. The p48 isoform, which localizes in the cytoplasm and nuclei, promoted HCV replication, whereas the shorter p42 isoform, which resides exclusively in the cytoplasm, strongly inhibited HCV replication. Transient expression of individual isoforms in Ebp1-knockdown MH14 cells confirmed that the p48 isoform promotes HCV replication, while the p42 isoform inhibits it. We found that Ebp1-p42 significantly enhanced autophosphorylation of PKR, while Ebp1-p48 isoform strongly inhibited it. We propose that modulation of autophosphorylation of PKR by p48 isoform is an important mechanism whereby the HCV virus escapes innate antiviral immune responses by circumventing p42-mediated inhibition of its replication.


Subject(s)
Hepacivirus/physiology , Hepatitis C/virology , Virus Replication , Cell Line, Tumor , DNA Replication , Hepacivirus/genetics , Hepatitis C/genetics , Hepatitis C/metabolism , Humans , Keratin-20/genetics , Keratin-20/metabolism , Phosphorylation , Protein Biosynthesis , Protein Isoforms/genetics , Protein Isoforms/metabolism , eIF-2 Kinase/genetics , eIF-2 Kinase/metabolism
4.
Nucleic Acids Res ; 44(11): 5271-87, 2016 06 20.
Article in English | MEDLINE | ID: mdl-27106056

ABSTRACT

Persistent hepatitis C virus (HCV) infection leads to chronic hepatitis C (CHC), which often progresses to liver cirrhosis (LC) and hepatocellular carcinoma (HCC). The molecular mechanisms that establish CHC and cause its subsequent development into LC and HCC are poorly understood. We have identified a cytoplasmic double-stranded RNA binding protein, Stau1, which is crucial for HCV replication. In this study, Stau1 specifically interacted with the variable-stem-loop region in the 3' NTR and domain IIId of the HCV-IRES in the 5' NTR, and promoted HCV replication and translation. Stau1 coimmunoprecipitates HCV NS5B and a cell factor, protein kinase R (PKR), which is critical for interferon-induced cellular antiviral and antiproliferative responses. Like Stau1, PKR displayed binding specificity to domain IIId of HCV-IRES. Stau1 binds to PKR and strongly inhibits PKR-autophosphorylation. We demonstrated that the transport of HCV RNA on the polysomes is Stau1-dependent, being mainly localized in the monosome fractions when Stau1 is downregulated and exclusively localized in the polysomes when Stau1 is overexpressed. Our findings suggest that HCV may appropriate Stau1 to its advantage to prevent PKR-mediated inhibition of eIF2α, which is required for the synthesis of HCV proteins for translocation of viral RNA genome to the polysomes for efficient translation and replication.


Subject(s)
Cytoskeletal Proteins/metabolism , Hepacivirus/physiology , Protein Biosynthesis , RNA, Viral/genetics , RNA, Viral/metabolism , RNA-Binding Proteins/metabolism , Virus Replication , eIF-2 Kinase/metabolism , Binding Sites , Cell Line , Cytoskeletal Proteins/genetics , Genome, Viral , Hepatitis C/metabolism , Hepatitis C/virology , Humans , Phosphorylation , Polyribosomes/genetics , Polyribosomes/metabolism , Protein Binding , Protein Transport , RNA Transport , RNA-Binding Proteins/genetics , Transcription, Genetic , Untranslated Regions , Viral Nonstructural Proteins/metabolism
5.
J Virol ; 89(15): 7905-21, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25995247

ABSTRACT

UNLABELLED: Hepatitis C virus (HCV) is a leading cause of chronic hepatitis C (CHC), liver cirrhosis, and hepatocellular carcinoma (HCC). Immunohistochemistry of archived HCC tumors showed abundant FBP1 expression in HCC tumors with the CHC background. Oncomine data analysis of normal versus HCC tumors with the CHC background indicated a 4-fold increase in FBP1 expression with a concomitant 2.5-fold decrease in the expression of p53. We found that FBP1 promotes HCV replication by inhibiting p53 and regulating BCCIP and TCTP, which are positive and negative regulators of p53, respectively. The severe inhibition of HCV replication in FBP1-knockdown Huh7.5 cells was restored to a normal level by downregulation of either p53 or BCCIP. Although p53 in Huh7.5 cells is transcriptionally inactive as a result of Y220C mutation, we found that the activation and DNA binding ability of Y220C p53 were strongly suppressed by FBP1 but significantly activated upon knockdown of FBP1. Transient expression of FBP1 in FBP1 knockdown cells fully restored the control phenotype in which the DNA binding ability of p53 was strongly suppressed. Using electrophoretic mobility shift assay (EMSA) and isothermal titration calorimetry (ITC), we found no significant difference in in vitro target DNA binding affinity of recombinant wild-type p53 and its Y220C mutant p53. However, in the presence of recombinant FBP1, the DNA binding ability of p53 is strongly inhibited. We confirmed that FBP1 downregulates BCCIP, p21, and p53 and upregulates TCTP under radiation-induced stress. Since FBP1 is overexpressed in most HCC tumors with an HCV background, it may have a role in promoting persistent virus infection and tumorigenesis. IMPORTANCE: It is our novel finding that FUSE binding protein 1 (FBP1) strongly inhibits the function of tumor suppressor p53 and is an essential host cell factor required for HCV replication. Oncomine data analysis of a large number of samples has revealed that overexpression of FBP1 in most HCC tumors with chronic hepatitis C is significantly linked with the decreased expression level of p53. The most significant finding is that FBP1 not only physically interacts with p53 and interferes with its binding to the target DNA but also functions as a negative regulator of p53 under cellular stress. FBP1 is barely detectable in normal differentiated cells; its overexpression in HCC tumors with the CHC background suggests that FBP1 has an important role in promoting HCV infection and HCC tumors by suppressing p53.


Subject(s)
Carcinoma, Hepatocellular/metabolism , DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Hepacivirus/physiology , Hepatitis C/metabolism , Liver Neoplasms/metabolism , Tumor Suppressor Protein p53/genetics , Virus Replication , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/virology , Cell Line, Tumor , DNA Helicases/genetics , DNA-Binding Proteins/genetics , Down-Regulation , Hepacivirus/genetics , Hepatitis C/genetics , Hepatitis C/virology , Host-Pathogen Interactions , Humans , Liver Neoplasms/genetics , Liver Neoplasms/virology , Protein Binding , RNA-Binding Proteins , Tumor Protein, Translationally-Controlled 1 , Tumor Suppressor Protein p53/metabolism
6.
BMC Cancer ; 14: 925, 2014 Dec 08.
Article in English | MEDLINE | ID: mdl-25487856

ABSTRACT

BACKGROUND: FUSE binding protein1 (FBP1) is a transactivator of transcription of human c-myc proto-oncogene and expressed mainly in undifferentiated cells. It is also present in differentiated normal cells albeit with very low background. FBP1 is abundantly expressed in the majority of hepatocellular carcinoma tumors and has been implicated in tumor development. Although it down-regulates the expression of proapoptotic p21 protein, it is not known whether FBP1 also interacts and antagonizes the function of tumor suppressor protein p53. METHODS: Western blotting was carried out to detect the expression level of FBP1, p21 and p53, and also p53 regulatory factors, BCCIP and TCTP; real-time quantitative PCR was done to determine the fold change in mRNA levels of target proteins; immunoprecipitation was carried out to determine the interaction of FBP1 with p53, BCCIP and TCTP. Cells stably knockdown for either FBP1; p53 or BCCIP were examined for p53 reporter activity under normal and radiation-induced stress. RESULTS: FBP1 physically interacted with p53, impairing its transcription activity and reducing p53-mediated sensitivity to cellular stress. Knockdown of FBP1 expression activated p53-mediated response to cellular stress while transient expression of FBP1 in FBP-knockdown cells restored the inhibition of p53 activity. FBP1 not only interacted with both BCCIP and TCTP, which, respectively, function as positive and negative regulators of p53, but also regulated their expression under cellular stress. In FBP knockdown cells, TCTP expression was down-regulated under radiation-induced stress whereas expression of BCCIP and p21 were significantly up-regulated suggesting FBP1 as a potential regulator of these proteins. We hypothesize that the FBP1-mediated suppression of p53 activity may occur via preventing the interaction of p53 with BCCIP as well as by FBP1-mediated regulation of p53 regulatory proteins, TCTP and BCCIP. Since FBP1 suppresses p53 activity and is overexpressed in most HCC tumors, it may have a possible role in tumorigenesis. CONCLUSION: FBP1 physically interacts with p53, functions as a regulator of p53-regulatory proteins (TCTP and BCCIP), and suppresses p53 transactivation activity under radiation-induced cellular stress. Since it is abundantly expressed in most HCC tumors, it may have implication in tumorigenesis and thus may be a possible target for drug development.


Subject(s)
Biomarkers, Tumor/metabolism , Calcium-Binding Proteins/metabolism , Carcinoma, Hepatocellular/metabolism , Cell Cycle Proteins/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Liver Neoplasms/metabolism , Nuclear Proteins/metabolism , Transcription, Genetic , Tumor Suppressor Protein p53/metabolism , Calcium-Binding Proteins/genetics , Carcinoma, Hepatocellular/genetics , Cell Cycle Proteins/genetics , Cell Line, Tumor , DNA Helicases/genetics , DNA-Binding Proteins/genetics , Gene Knockdown Techniques , Humans , Liver Neoplasms/genetics , Nuclear Proteins/genetics , Proto-Oncogene Mas , RNA-Binding Proteins , Stress, Physiological , Tumor Protein, Translationally-Controlled 1 , Tumor Suppressor Protein p53/genetics
7.
Mol Cell Proteomics ; 12(6): 1539-52, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23429521

ABSTRACT

Hepatitis C virus (HCV) infection leading to chronic hepatitis is a major factor in the causation of liver cirrhosis, hepatocellular carcinoma, and liver failure. This process may involve the interplay of various host cell factors, as well as the interaction of these factors with viral RNA and proteins. We report a novel strategy using a sequence-specific biotinylated peptide nucleic acid (PNA)-neamine conjugate targeted to HCV RNA for the in situ capture of subgenomic HCV (+) RNA, along with cellular and viral factors associated with it in MH14 host cells. Using this affinity capture system in conjunction with LC/MS/MS, we have identified 83 cellular factors and three viral proteins (NS5B, NS5A, and NS3-4a protease-helicase) associated with the viral genome. The capture was highly specific. These proteins were not scored with cured MH14 cells devoid of HCV replicons because of the absence of the target sequence in cells for the PNA-neamine probe and also because, unlike oligomeric DNA, cellular proteins have no affinity for PNA. The identified cellular factors belong to different functional groups, including signaling, oncogenic, chaperonin, transcriptional regulators, and RNA helicases as well as DEAD box proteins, ribosomal proteins, translational regulators/factors, and metabolic enzymes, that represent a diverse set of cellular factors associated with the HCV RNA genome. Small interfering RNA-mediated silencing of a diverse class of selected proteins in an HCV replicon cell line either enhanced or inhibited HCV replication/translation, suggesting that these cellular factors have regulatory roles in HCV replication.


Subject(s)
Chromatography, Affinity/methods , Gene Expression Regulation, Viral , Genome, Viral , Hepacivirus/genetics , Hepatocytes/virology , RNA, Viral/genetics , Biotinylation , Cell Line, Tumor , Chromatography, Liquid , Framycetin/chemistry , Hepacivirus/metabolism , Hepatocytes/cytology , Hepatocytes/metabolism , Host-Pathogen Interactions , Humans , Peptide Nucleic Acids/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Viral/metabolism , Replicon , Tandem Mass Spectrometry , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism , Virus Replication
8.
Biochemistry ; 52(2): 432-44, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23268692

ABSTRACT

We have used an ATP analogue 5'-[p-(fluorosulfonyl)benzoyl]adenosine (FSBA) to modify HCV replicase in order to identify the ATP binding site in the enzyme. FSBA inactivates HCV replicase activity in a concentration-dependent manner with a binding stoichiometry of 2 moles of FSBA per mole of enzyme. The enzyme activity is protected from FSBA in the presence of rNTP substrates or double-stranded RNA template primers that do not support ATP as the incoming nucleotide but not in the presence of polyrU.rA(26). HPLC analysis of tryptic peptides of FSBA-modified enzyme revealed the presence of two distinct peptides eluted at 23 and 36 min; these were absent in the control. Further we noted that both peptides were protected from FSBA modification in the presence of Mg·ATP. The LC/MS/MS analysis of the affinity-labeled tryptic peptides purified from HPLC, identified two major modification sites at positions 382 (Tyr), and 491 (Lys) and a minor site at position 38 (Tyr). To validate the functional significance of Tyr38, Tyr382, and Lys491 in catalysis, we individually substituted these residues by alanine and examined their ability to catalyze RdRp activity. We found that both Y382A and K491A mutants were significantly affected in their ability to catalyze RdRp activity while Y38A remained unaffected. We further observed that both Y382A and K491A mutants were not affected in their ability to bind template primer but were significantly affected in their ability to photo-cross-link ATP in the absence or presence of template primer.


Subject(s)
Adenosine Triphosphate/metabolism , Adenosine/analogs & derivatives , Affinity Labels/chemistry , Hepacivirus/enzymology , RNA-Dependent RNA Polymerase/chemistry , RNA-Dependent RNA Polymerase/metabolism , Adenosine/chemistry , Adenosine/metabolism , Affinity Labels/metabolism , Amino Acid Sequence , Binding Sites , Hepacivirus/chemistry , Hepacivirus/genetics , Hepatitis C/virology , Humans , Molecular Docking Simulation , Mutagenesis , Nucleotides/metabolism , Point Mutation , Protein Binding , RNA-Dependent RNA Polymerase/genetics , Sequence Alignment , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
9.
J Ethnopharmacol ; 144(3): 545-54, 2012 Dec 18.
Article in English | MEDLINE | ID: mdl-23026306

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Eclipta alba, traditionally known as bhringraj, has been used in Ayurvedic medicine for more than 1000 years in India. It is used for the treatment of infective hepatitis, liver cirrhosis, liver enlargement and other ailments of liver and gall bladder in India. The aim of this study was to evaluate anti-hepatitis C virus activity present in the Eclipta alba extract, perform bioassay based fractionation and identify anti-HCV phytochemicals from the active fractions. MATERIALS AND METHODS: Identification of active compounds was performed by bio-activity guided fractionation approach. Active isolates were separated by the combination of silica gel chromatography and preparative scale reverse phase HPLC. Eclipta alba extract and its isolates were examined for their ability to inhibit HCV replicase (HCV NS5B) activity in vitro and HCV replication in a cell culture system carrying replicating HCV subgenomic RNA replicon. The purified isolates were also examined for their binding affinity to HCV replicase by fluorescence quenching and their cytotoxicity by MTT assay. RESULTS: Eclipta alba extract strongly inhibited RNA dependent RNA polymerase (RdRp) activity of HCV replicase in vitro. In cell culture system, it effectively inhibited HCV replication which resulted in reduced HCV RNA titer and translation level of viral proteins. Bioassay-based fractionations of the extracts and purification of anti-HCV phytochemicals present in the active fractions have identified three compounds, wedelolactone, luteolin, and apigenin. These compounds exhibited dose dependent inhibition of HCV replicase in vitro, and anti-HCV replication activity in the cell culture system CONCLUSION: Eclipta alba extract and phytochemicals isolated from active fractions display anti-HCV activity in vitro and in cell culture system. The standardized Eclipta alba extract or its isolates can be used as an effective alternative and complementary treatment against HCV.


Subject(s)
Antiviral Agents/pharmacology , Eclipta , Hepacivirus/drug effects , Plant Extracts/pharmacology , Antiviral Agents/isolation & purification , Cell Line, Tumor , Hepacivirus/physiology , Humans , RNA, Viral/genetics , RNA-Dependent RNA Polymerase/metabolism , Virus Replication/drug effects
10.
J Nucleic Acids ; 2012: 591025, 2012.
Article in English | MEDLINE | ID: mdl-23029603

ABSTRACT

Human immunodeficiency virus-1 (HIV-1) replication and gene expression entails specific interaction of the viral protein Tat with its transactivation responsive element (TAR), to form a highly stable stem-bulge-loop structure. Previously, we described triphenylphosphonium (TPP) cation-based vectors that efficiently deliver nucleotide analogs (PNAs) into the cytoplasm of cells. In particular, we showed that the TPP conjugate of a linear 16-mer PNA targeting the apical stem-loop region of TAR impedes Tat-mediated transactivation of the HIV-1 LTR in vitro and also in cell culture systems. In this communication, we conjugated TPP to cyclic and hairpin PNAs targeting the loop region of HIV-1 TAR and evaluated their antiviral efficacy in a cell culture system. We found that TPP-cyclic PNAs containing only 8 residues, showed higher antiviral potency compared to hairpin PNAs of 12 or 16 residues. We further noted that the TPP-conjugates of the 8-mer cyclic PNA as well as the 16-mer linear PNA displayed similar antiviral efficacy. However, cyclic PNAs were shown to be highly specific to their target sequences. This communication emphasizes on the importance of small constrained cyclic PNAs over both linear and hairpin structures for targeting biologically relevant RNA hairpins.

11.
J Med Chem ; 55(13): 6021-32, 2012 Jul 12.
Article in English | MEDLINE | ID: mdl-22698070

ABSTRACT

The 6-aminoglucosamine ring of the aminoglycoside antibiotic neomycin B (ring II) was conjugated to a 16-mer peptide nucleic acid (PNA) targeting HIV-1 TAR RNA. For this purpose, we prepared the aminoglucosamine monomer 15 and attached it to the protected PNA prior to its cleavage from the solid support. We found that the resulting PNA-aminoglucosamine conjugate is stable under acidic conditions, efficiently taken up by the human cells and fairly distributed in both cytosol and nucleus without endosomal entrapment because cotreatment with endosome-disrupting agent had no effect on its cellular distribution. The conjugate displayed very high target specificity in vitro and strongly inhibited Tat mediated transactivation of HIV-1 LTR transcription in a cell culture system. The unique properties of this new class of PNA conjugate suggest it to be a potential candidate for therapeutic application.


Subject(s)
Acetylglucosamine/pharmacokinetics , Anti-HIV Agents/pharmacokinetics , Framycetin/analogs & derivatives , HIV-1/drug effects , Peptide Nucleic Acids/pharmacokinetics , Transcriptional Activation/drug effects , tat Gene Products, Human Immunodeficiency Virus/antagonists & inhibitors , Acetylglucosamine/analogs & derivatives , Acetylglucosamine/chemical synthesis , Anti-HIV Agents/chemical synthesis , Anti-HIV Agents/chemistry , Antiviral Agents/chemistry , Binding Sites/genetics , Biological Availability , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Nucleus/ultrastructure , Cytosol/drug effects , Cytosol/metabolism , Cytosol/ultrastructure , Endosomes/drug effects , Endosomes/metabolism , Endosomes/ultrastructure , Fluorescein-5-isothiocyanate/chemistry , Genome, Viral/drug effects , HIV Long Terminal Repeat/drug effects , HIV Long Terminal Repeat/genetics , HIV-1/genetics , Humans , Peptide Nucleic Acids/chemistry , Peptide Nucleic Acids/pharmacology , RNA, Viral/antagonists & inhibitors , RNA, Viral/genetics , Response Elements/drug effects , Spectrometry, Fluorescence , Transcription, Genetic/drug effects , tat Gene Products, Human Immunodeficiency Virus/genetics
12.
Biochemistry ; 50(37): 8067-77, 2011 Sep 20.
Article in English | MEDLINE | ID: mdl-21800837

ABSTRACT

Earlier, we postulated that Gln91 of human immunodeficiency virus type 1 reverse transcriptase (HIV-1 RT) stabilizes the side chain of Tyr183 via hydrogen bonding interaction between O(H) of Tyr183 and CO of Q91 [Harris, D., et al. (1998) Biochemistry 37, 9630-9640]. To test this hypothesis, we generated mutant derivatives of Gln91 and analyzed their biochemical properties. The efficiency of reverse transcription was severely impaired by nonconservative substitution of Gln with Ala, while conservative substitution of Gln with Asn resulted in an approximately 70% loss of activity, a value similar to that observed with the Y183F mutation. The loss of polymerase activity from both Q91A and Q91N was significantly improved by a Met to Val substitution at position 184. Curiously, the Q91N mutant exhibited stringency in discriminating between correct and incorrect nucleotides, suggesting its possible interaction with residues influencing the flexibility of the dNTP binding pocket. In contrast, both double mutants, Q91A/M184V and Q91N/M184V, are found to be as error prone as the wild-type enzyme. We propose a model that suggests that subtle structural changes in the region due to mutation at position 91 may influence the stability of the side chain of Tyr183 in the catalytic YMDD motif of the enzyme, thus altering the active site geometry that may interfere in substrate recognition.


Subject(s)
Deoxyribonucleotides/chemistry , Deoxyribonucleotides/metabolism , Glutamine/chemistry , Glutamine/metabolism , HIV Reverse Transcriptase/chemistry , HIV Reverse Transcriptase/metabolism , Amino Acid Motifs/genetics , Amino Acid Substitution/genetics , Binding Sites/genetics , Catalysis , Catalytic Domain/genetics , Crystallography, X-Ray/methods , Deoxyribonucleotides/genetics , Glutamine/genetics , HIV Reverse Transcriptase/genetics , Humans , Protein Stability , Protein Structure, Secondary , Substrate Specificity/genetics , Tyrosine/metabolism
13.
Mol Cell Biochem ; 338(1-2): 19-33, 2010 May.
Article in English | MEDLINE | ID: mdl-19921401

ABSTRACT

Fingers domain of HIV-1 RT is one of the constituents of the dNTP-binding pocket that is involved in binding of both dNTP and the template-primer. In the ternary complex of HIV-1 RT, two residues Trp-24 and Phe-61 located on the beta1 and beta3, respectively, are seen interacting with N + 1 to N + 3 nucleotides in the template overhang. We generated nonconservative and conservative mutant derivatives of these residues and examined their impact on the template-primer binding and polymerase function of the enzyme. We noted that W24A, F61A, and F61Y and the double mutant (W24A/F61A) were significantly affected in their ability to bind template-primer and also to catalyze the polymerase reaction while W24F remained unaffected. Using a specially designed template-primer with photoactivatable bromo-dU base in the duplex region at the penultimate position to the primer terminus, we demonstrated that F61A, W24A, F61Y as well as the double mutant were also affected in their cross-linking ability with the duplex region of the template-primer. We also isolated the E-TP covalent complexes of these mutants and examined their ability to catalyze single dNTP incorporation onto the immobilized primer terminus. The E-TP covalent complexes from W24F mutant displayed wild-type activity while those from W24A, F61A, F61Y, and the double mutant (W24A/F61A) were significantly impaired in their ability to catalyze dNTP incorporation onto the immobilized primer terminus. This unusual observation indicated that amino acid residues involved in the positioning of the template overhang may also influence the binding and orientation of the duplex region of the template-primer. Molecular modeling studies based on our biochemical results suggested that conformation of both W24 and F61 are interdependent on their interactions with each other, which together are required for proper positioning of the +1 template nucleotide in the binary and ternary complexes.


Subject(s)
DNA Primers , HIV Reverse Transcriptase/metabolism , HIV-1/enzymology , Templates, Genetic , Base Sequence , Circular Dichroism , Cross-Linking Reagents/metabolism , DNA Primers/chemistry , DNA Primers/genetics , DNA Primers/metabolism , DNA-Directed DNA Polymerase/chemistry , DNA-Directed DNA Polymerase/metabolism , Enzyme Stability , HIV Reverse Transcriptase/chemistry , HIV Reverse Transcriptase/genetics , Humans , Macromolecular Substances/chemistry , Macromolecular Substances/metabolism , Models, Molecular , Molecular Conformation , Molecular Sequence Data , Mutagenesis, Site-Directed
14.
J Cell Biochem ; 109(3): 598-605, 2010 Feb 15.
Article in English | MEDLINE | ID: mdl-20013797

ABSTRACT

The human immunodeficiency virus type 1 reverse transcriptase (HIV-1 RT) is a heterodimeric enzyme composed of p66 and p51 subunits. Earlier, we showed that the beta7-beta8 loop of p51 is crucial for polymerase activity of HIV-1 RT as either deletion or Ala substitution of amino acids in the beta7-beta8 loop spanning residues 136-139 in the p51 subunit impaired dimerization and, in turn, polymerase function of the enzyme (Pandey et al. 2001 Biochemistry 40: 9505-9512). In the present study, we generated subunit-specific single-deletion mutants at positions 134, 135, 136, or 137 and examined their effects on the heterodimerization, binary complex formation, and polymerase functions of the enzyme. We found that among these four residues, Ser134, Ile135, and Asn136 in the beta7-beta8 loop of the p51 subunit are crucial residues for dimerization and polymerase function of the enzyme, but have no impact when specifically deleted from the p66 subunit. These results demonstrate the beta7-beta8 loop of the p51 subunit in the formation of stable, functional heterodimeric enzyme which could be an attractive target for anti-HIV-1 drug development.


Subject(s)
HIV Reverse Transcriptase/chemistry , HIV Reverse Transcriptase/genetics , Amino Acid Sequence , Binding Sites , Dimerization , Models, Molecular , Molecular Sequence Data , Protein Structure, Secondary , Protein Subunits/chemistry , Protein Subunits/genetics , Sequence Deletion , Structure-Activity Relationship
15.
Virol J ; 6: 159, 2009 Oct 08.
Article in English | MEDLINE | ID: mdl-19814799

ABSTRACT

Reverse transcriptases from HIV-1 and MuLV respectively prefer Mg2+ and Mn2+ for their polymerase activity, with variable fidelity, on both RNA and DNA templates. The function of the RNase H domain with respect to these parameters is not yet understood. To evaluate this function, two chimeric enzymes were constructed by swapping the RNase H domains between HIV-1 RT and MuLV RT. Chimeric HIV-1 RT, having the RNase H domain of MuLV RT, inherited the divalent cation preference characteristic of MuLV RT on the DNA template with no significant change on the RNA template. Chimeric MuLV RT, likewise partially inherited the metal ion preference of HIV-1 RT. Unlike the wild-type MuLV RT, chimeric MuLV RT is able to use both Mn.dNTP and Mg.dNTP on the RNA template with similar efficiency, while a 30-fold higher preference for Mn.dNTP was seen on the DNA template. The metal preferences for the RNase H activity of chimeric HIV-1 RT and chimeric MuLV RT were, respectively, Mn2+ and Mg2+, a property acquired through their swapped RNase H domains. Chimeric HIV-1 RT displayed higher fidelity and discrimination against rNTPs than against dNTPs substrates, a property inherited from MuLV RT. The overall fidelity of the chimeric MuLV RT was decreased in comparison to the parental MuLV RT, suggesting that the RNase H domain profoundly influences the function of the polymerase domain.


Subject(s)
Cations/pharmacology , Coenzymes/pharmacology , HIV-1/enzymology , Leukemia Virus, Murine/enzymology , Metals/pharmacology , RNA-Directed DNA Polymerase/metabolism , Ribonuclease H/metabolism , Genetic Engineering , HIV-1/genetics , Leukemia Virus, Murine/genetics , RNA-Directed DNA Polymerase/genetics , Recombination, Genetic , Ribonuclease H/genetics , Substrate Specificity
16.
Expert Opin Biol Ther ; 9(8): 975-89, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19534584

ABSTRACT

Since the discovery and synthesis of a novel DNA mimic, peptide nucleic acid (PNA) in 1991, PNAs have attracted tremendous interest and have shown great promise as potential antisense drugs. They have been used extensively as tools for specific modulation of gene expression by targeting translation or transcription processes. This review discusses the present and future therapeutic potential of this class of compound as anti-HIV-1 drugs.


Subject(s)
Genetic Therapy/methods , HIV Infections/therapy , Peptide Nucleic Acids/therapeutic use , Animals , Anti-HIV Agents/therapeutic use , Biological Therapy/methods , CD4-Positive T-Lymphocytes/immunology , Cations , Gene Expression Regulation , HIV Long Terminal Repeat , Humans , Macrophages/metabolism , Mice , Protein Biosynthesis , Transcription, Genetic
17.
Oligonucleotides ; 18(4): 329-35, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19006449

ABSTRACT

Anti-human immunodeficiency virus-1 (HIV-1) polyamide (peptide) nucleic acids (PNAs) conjugated with cell-penetrating peptides (CPPs) targeted to the viral genome are potent virucidal and antiviral agents. Earlier, we have shown that the anti-HIV-1 PNA(TAR)-penetratin conjugate is rapidly taken up by cells and is nontoxic to mice when administered at repeat doses of as high as 100 mg/kg body weight. In the present studies we demonstrate that naked PNA(TAR) is immunologically inert as judged by the proliferation responses of splenocytes and lymph node cells from PNA(TAR)-immunized mice challenged with the immunizing antigen. In contrast, PNA(TAR)-penetratin conjugate is moderately immunogenic mainly due to its penetratin peptide component. Cytokine secretion profiles of the lymph node cells from the conjugate-immunized mice showed marginally elevated levels of proinflammatory cytokines, which are known to promote proliferation of T lymphocytes. Since the candidate compound, PNA(TAR)-penetratin conjugate displays potent virucidal and antiviral activities against HIV-1, the favorable immunological response together with negligible toxicity suggest a strong therapeutic potential for this class of compounds.


Subject(s)
Anti-HIV Agents/immunology , Carrier Proteins/immunology , HIV Long Terminal Repeat/immunology , HIV-1/drug effects , Peptide Nucleic Acids/immunology , RNA, Viral/immunology , Animals , Anti-HIV Agents/administration & dosage , Carrier Proteins/administration & dosage , Cell Proliferation/drug effects , Cell-Penetrating Peptides , Cytokines/biosynthesis , Cytokines/immunology , Female , Genes, Viral , HIV-1/immunology , Lymph Nodes/drug effects , Lymph Nodes/immunology , Mice , Mice, Inbred BALB C , Peptide Nucleic Acids/administration & dosage , RNA, Viral/genetics , Spleen/drug effects , Spleen/immunology , T-Lymphocytes/immunology
18.
Oligonucleotides ; 18(3): 245-56, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18707540

ABSTRACT

With the aim of developing a general and straightforward procedure for the intracellular delivery of naked peptide nucleic acids (PNAs), we designed an intracellularly biodegradable triphenylphosphonium (TPP) cation based transporter system. In this system, TPP is linked, via a biolabile disulfide bridge, to an activated mercaptoethoxycarbonyl moiety, allowing its direct coupling to the N-terminal extremity of a free PNA through a carbamate bond. We found that such TPP-PNA-carbamate conjugates were highly stable in a cell culture medium containing fetal calf serum. In a glutathione-containing medium mimicking the cytosol, the conjugates were rapidly degraded into an unstable intermediate, which spontaneously decomposed, releasing the free PNA. Using a fluorescence-labeled PNA-TPP conjugate, we demonstrated that conjugates were taken up by cells. Efficient cellular uptake and release of the PNA into the cytosol was further confirmed by the anti-HIV activity measured for the TPP-conjugate of a 16-mer PNA targeting the TAR region of the HIV-1 genome. This conjugate exhibited an IC(50) value of 1 microM, while the free 16-mer PNA did not inhibit replication of HIV in the same cellular test.


Subject(s)
Antisense Elements (Genetics) , Drug Delivery Systems , Nylons/metabolism , Peptide Nucleic Acids/metabolism , Anti-HIV Agents/metabolism , Anti-HIV Agents/pharmacology , Cell Line , Culture Media , HIV Long Terminal Repeat/drug effects , HIV-1/drug effects , HIV-1/genetics , HIV-1/physiology , Humans , Nylons/chemistry , Peptide Nucleic Acids/chemical synthesis , Peptide Nucleic Acids/pharmacology , Virus Replication/drug effects
19.
Mol Cancer Ther ; 7(6): 1543-50, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18566225

ABSTRACT

Signal transducers and activators of transcription (STAT) were originally discovered as components of signal transduction pathways. Persistent aberrant activation of STAT3 is a feature of many malignancies including prostate cancer and pancreatic cancer. One consequence of persistently activated STAT3 in malignant cells is that they depend on it for survival; thus, STAT3 is an excellent molecular target for therapy. Previously, we reported that single-stranded oligonucleotides containing consensus STAT3 binding sequences (13410 and 13411) were more effective for inducing apoptosis in prostate cancer cells than antisense STAT3 oligonucleotides. Control oligonucleotides (scrambled sequences) had no effect. Here, we report that authentic STAT3 binding sequences, identified from published literature, were more effective for inducing apoptosis in prostate cancer cells and pancreatic cancer cells than was oligonucleotide 13410. Moreover, the authentic STAT3 binding sequences showed differing efficacies in the malignant cell lines depending on whether the canonical STAT3 binding sequence was truncated at the 5' or the 3' end. Finally, expression of one STAT3-regulated gene was decreased following treatment, suggesting that STAT3 may regulate the same set of genes in the two types of cancer. We conclude that truncating the 5' end left intact enough of the canonical STAT3 binding site for effective hybridization to the genome, whereas truncation of the 3' end, which is outside the canonical binding site, may have affected binding of required cofactors essential for STAT3 activity, thereby reducing the capacity of this modified oligonucleotide to induce apoptosis. Additional experiments to answer this hypothesis are under way.


Subject(s)
Oligonucleotides/chemistry , Oligonucleotides/pharmacology , Pancreatic Neoplasms/metabolism , Prostatic Neoplasms/metabolism , STAT3 Transcription Factor/antagonists & inhibitors , Apoptosis/drug effects , Base Sequence , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Membrane Cofactor Protein/genetics , Membrane Cofactor Protein/metabolism , Molecular Sequence Data , Oligonucleotides/genetics , Oxidation-Reduction/drug effects , Pancreatic Neoplasms/genetics , Prostatic Neoplasms/genetics , Protein Binding/drug effects
20.
J Virol ; 82(12): 5761-73, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18400844

ABSTRACT

Hepatitis C virus (HCV) infection is the leading cause of liver cirrhosis and hepatocellular carcinoma and one of the primary indications for liver transplantation. The molecular mechanisms underlying the actions of host factors in HCV replication remain poorly defined. FUSE (far upstream element of the c-myc proto-oncogene) binding protein (FBP) is a cellular factor that we have identified as a binder of HCV 3' nontranslated region (3'NTR). Mapping of the binding site showed that FBP specifically interacts with the poly(U) tract within the poly(U/UC) region of the 3'NTR. Silencing of FBP expression by small interfering RNA in cells carrying HCV subgenomic replicons severely reduced viral replication, while overexpression of FBP significantly enhanced viral replication. We confirmed these observations by an in vitro HCV replication assay in the cell-free replicative lysate, which suggested that there is a direct correlation between the cellular FBP level and HCV replication. FBP immunoprecipitation coprecipitated HCV nonstructural protein 5A (NS5A), indicating that FBP interacts with HCV NS5A, which is known to function as a link between HCV translation and replication. Although FBP is mainly localized in the nucleus, we found that in MH14 cells a significant level of this protein is colocalized with NS5A in the cytosol, a site of HCV replication. While the mechanism of FBP involvement in HCV replication is yet to be delineated, our findings suggest that it may be an important regulatory component that is essential for efficient replication of HCV.


Subject(s)
Carrier Proteins/physiology , Hepacivirus/physiology , Viral Nonstructural Proteins/metabolism , Virus Replication , Binding Sites , Carrier Proteins/metabolism , Cell Line , Cell Nucleus/metabolism , Cell-Free System , Cross-Linking Reagents/metabolism , Cytosol/metabolism , Fluorescent Antibody Technique, Direct , Gene Expression Regulation, Viral , Genes, Reporter , Hepacivirus/genetics , Humans , Luciferases/analysis , Luciferases/metabolism , Oligonucleotides/metabolism , Plasmids , Protein Binding , Proto-Oncogene Mas , RNA, Small Interfering/metabolism , RNA, Viral/analysis , Replicon , Transfection , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/isolation & purification
SELECTION OF CITATIONS
SEARCH DETAIL
...