Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2022 Dec 12.
Article in English | MEDLINE | ID: mdl-36561180

ABSTRACT

SARS-CoV-2 infection has caused a major global burden. Despite intensive research, the mechanism and dynamics of early viral replication are not completely understood including the kinetics of formation of plus stranded genomic and subgenomic RNAs (gRNA and sgRNA) starting from the RNA from the first virus that enters the cell. We employed single-molecule RNA-fluorescence in situ hybridization (smRNA-FISH) to simultaneously detect viral gRNA and sgRNA in infected cells and carried out a time course analysis to determine the kinetics of their replication. We visualized the single molecules of gRNA within the cytoplasm of infected cells 30 minutes post-infection and detected the co-expression of gRNA and sgRNA within two hours post-infection. Furthermore, we observed the formation of a replication organelle (RO) from a single RNA, which led to the formation of multiple ROs within the same cells. Single molecule analysis indicated that while gRNA resided in the center of these ROs, the sgRNAs were found to radiate and migrate out of these structures. Our results also indicated that after the initial delay, there was a rapid but asynchronous replication, and the gRNA and sgRNAs dispersed throughout the cell within 4-5 hours post-infection forming multiple ROs that filled the entire cytoplasm. These results provide insight into the kinetics of early post-entry events of SARS-CoV-2 and the formation of RO, which will help to understand the molecular events associated with viral infection and facilitate the identification of new therapeutic targets that can curb the virus at a very early stage of replication to combat COVID-19. Author Summary: SARS-CoV-2 infection continues to be a global burden. Soon after the entry, SARS-CoV-2 replicates by an elaborate process, producing genomic and subgenomic RNAs (gRNA and sgRNAs) within specialized structures called replication organelles (RO). Many questions including the timing of multiplication of gRNA and sgRNA, the generation, subcellular localization, and function of the ROs, and the mechanism of vRNA synthesis within ROs is not completely understood. Here, we have developed probes and methods to simultaneously detect the viral gRNA and a sgRNA at single cell single molecule resolution and have employed a method to scan thousands of cells to visualize the early kinetics of gRNA and sgRNA synthesis soon after the viral entry into the cell. Our results reveal that the replication is asynchronous and ROs are rapidly formed from a single RNA that enters the cell within 2 hours, which multiply to fill the entire cell cytoplasm within ~4 hours after infection. Furthermore, our studies provide a first glimpse of the gRNA and sgRNA synthesis within ROs at single molecule resolution. Our studies may facilitate the development of drugs that inhibit the virus at the earliest possible stages of replication to minimize the pathogenic impact of viral infection.

2.
Nat Commun ; 12(1): 2743, 2021 05 12.
Article in English | MEDLINE | ID: mdl-33980829

ABSTRACT

INI1/SMARCB1 binds to HIV-1 integrase (IN) through its Rpt1 domain and exhibits multifaceted role in HIV-1 replication. Determining the NMR structure of INI1-Rpt1 and modeling its interaction with the IN-C-terminal domain (IN-CTD) reveal that INI1-Rpt1/IN-CTD interface residues overlap with those required for IN/RNA interaction. Mutational analyses validate our model and indicate that the same IN residues are involved in both INI1 and RNA binding. INI1-Rpt1 and TAR RNA compete with each other for IN binding with similar IC50 values. INI1-interaction-defective IN mutant viruses are impaired for incorporation of INI1 into virions and for particle morphogenesis. Computational modeling of IN-CTD/TAR complex indicates that the TAR interface phosphates overlap with negatively charged surface residues of INI1-Rpt1 in three-dimensional space, suggesting that INI1-Rpt1 domain structurally mimics TAR. This possible mimicry between INI1-Rpt1 and TAR explains the mechanism by which INI1/SMARCB1 influences HIV-1 late events and suggests additional strategies to inhibit HIV-1 replication.


Subject(s)
HIV Integrase/metabolism , HIV-1/physiology , RNA, Viral/metabolism , SMARCB1 Protein/metabolism , Virus Replication , Genome, Viral , HIV Integrase/chemistry , HIV Integrase/genetics , Host-Pathogen Interactions , Humans , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Docking Simulation , Protein Binding , Protein Domains , RNA, Viral/chemistry , SMARCB1 Protein/chemistry , SMARCB1 Protein/genetics , Virion/growth & development , Virion/metabolism
4.
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
5.
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
6.
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
7.
Oncotarget ; 6(35): 37792-807, 2015 Nov 10.
Article in English | MEDLINE | ID: mdl-26473374

ABSTRACT

Crk is the prototypical member of a class of Src homology 2 (SH2) and Src homology 3 (SH3) domain-containing adaptor proteins that positively regulate cell motility via the activation of Rac1 and, in certain tumor types such as GBM, can promote cell invasion and metastasis by mechanisms that are not well understood. Here we demonstrate that Crk, via its phosphorylation at Tyr251, promotes invasive behavior of tumor cells, is a prominent feature in GBM, and correlating with aggressive glioma grade IV staging and overall poor survival outcomes. At the molecular level, Tyr251 phosphorylation of Crk is negatively regulated by Abi1, which competes for Crk binding to Abl and attenuates Abl transactivation. Together, these results show that Crk and Abi1 have reciprocal biological effects and act as a molecular rheostat to control Abl activation and cell invasion. Finally, these data suggest that Crk Tyr251 phosphorylation regulate invasive cell phenotypes and may serve as a biomarker for aggressive GBM.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Biomarkers, Tumor/metabolism , Cell Movement , Cytoskeletal Proteins/metabolism , Glioblastoma/metabolism , Glioblastoma/pathology , Proto-Oncogene Proteins c-abl/metabolism , Proto-Oncogene Proteins c-crk/metabolism , Apoptosis , Binding Sites , Blotting, Western , Cell Proliferation , Female , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Glioblastoma/mortality , Humans , Immunoenzyme Techniques , Immunoprecipitation , Male , Middle Aged , Neoplasm Grading , Neoplasm Invasiveness , Phenotype , Phosphorylation , Prognosis , Signal Transduction , Survival Rate , Tissue Array Analysis , Tumor Cells, Cultured , Wound Healing
8.
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
9.
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
10.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...