Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Mol Cell ; 59(2): 188-202, 2015 Jul 16.
Article in English | MEDLINE | ID: mdl-26166704

ABSTRACT

Enhancers instruct spatio-temporally specific gene expression in a manner tightly linked to higher-order chromatin architecture. Critical chromatin architectural regulators condensin I and condensin II play non-redundant roles controlling mitotic chromosomes. But the chromosomal locations of condensins and their functional roles in interphase are poorly understood. Here we report that both condensin complexes exhibit an unexpected, dramatic estrogen-induced recruitment to estrogen receptor α (ER-α)-bound eRNA(+) active enhancers in interphase breast cancer cells, exhibiting non-canonical interaction with ER-α via its DNA-binding domain (DBD). Condensins positively regulate ligand-dependent enhancer activation at least in part by recruiting an E3 ubiquitin ligase, HECTD1, to modulate the binding of enhancer-associated coactivators/corepressors, including p300 and RIP140, permitting full eRNA transcription, formation of enhancer:promoter looping, and the resultant coding gene activation. Collectively, our results reveal an important, unanticipated transcriptional role of interphase condensins in modulating estrogen-regulated enhancer activation and coding gene transcriptional program.


Subject(s)
Adenosine Triphosphatases/metabolism , DNA-Binding Proteins/metabolism , Enhancer Elements, Genetic , Estrogen Receptor alpha/metabolism , Multiprotein Complexes/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Adenosine Triphosphatases/antagonists & inhibitors , Adenosine Triphosphatases/genetics , Base Sequence , Binding Sites , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Chromatin/genetics , Chromatin/metabolism , DNA, Neoplasm/genetics , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Estradiol/metabolism , Female , Gene Knockdown Techniques , Humans , Interphase , MCF-7 Cells , Models, Biological , Molecular Sequence Data , Multiprotein Complexes/antagonists & inhibitors , Multiprotein Complexes/genetics , Nuclear Proteins/metabolism , Nuclear Receptor Interacting Protein 1 , Promoter Regions, Genetic , Protein Binding , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
2.
J Virol ; 93(1)2019 01 01.
Article in English | MEDLINE | ID: mdl-30282717

ABSTRACT

Lytic infection of human parvovirus B19 (B19V) takes place exclusively in human erythroid progenitor cells of bone marrow and fetal liver, which disrupts erythropoiesis. During infection, B19V expresses three nonstructural proteins (NS1, 11-kDa, and 7.5-kDa) and two structural proteins (VP1 and VP2). While NS1 is essential for B19V DNA replication, 11-kDa enhances viral DNA replication significantly. In this study, we confirmed the enhancement role of 11-kDa in viral DNA replication and elucidated the underlying mechanism. We found that 11-kDa specially interacts with cellular growth factor receptor-bound protein 2 (Grb2) during virus infection and in vitro We determined a high affinity interaction between 11-kDa and Grb2 that has an equilibrium dissociation constant (KD ) value of 18.13 nM. In vitro, one proline-rich motif was sufficient for 11-kDa to sustain a strong interaction with Grb2. In consistence, in vivo during infection, one proline-rich motif was enough for 11-kDa to significantly reduce phosphorylation of extracellular signal-regulated kinase (ERK). Mutations of all three proline-rich motifs of 11-kDa abolished its capability to reduce ERK activity and, accordingly, decreased viral DNA replication. Transduction of a lentiviral vector encoding a short hairpin RNA (shRNA) targeting Grb2 decreased the expression of Grb2 as well as the level of ERK phosphorylation, which resulted in an increase of B19V replication. These results, in concert, indicate that the B19V 11-kDa protein interacts with cellular Grb2 to downregulate ERK activity, which upregulates viral DNA replication.IMPORTANCE Human parvovirus B19 (B19V) infection causes hematological disorders and is the leading cause of nonimmunological fetal hydrops during pregnancy. During infection, B19V expresses two structural proteins, VP1 and VP2, and three nonstructural proteins, NS1, 11-kDa, and 7.5-kDa. While NS1 is essential, 11-kDa plays an enhancing role in viral DNA replication. Here, we elucidated a mechanism underlying 11-kDa protein-regulated B19V DNA replication. 11-kDa is tightly associated with cellular growth factor receptor-bound protein 2 (Grb2) during infection. In vitro, 11-kDa interacts with Grb2 with high affinity through three proline-rich motifs, of which at least one is indispensable for the regulation of viral DNA replication. 11-kDa and Grb2 interaction disrupts extracellular signal-regulated kinase (ERK) signaling, which mediates upregulation of B19V replication. Thus, our study reveals a novel mechanism of how a parvoviral small nonstructural protein regulates viral DNA replication by interacting with a host protein that is predominately expressed in the cytoplasm.


Subject(s)
GRB2 Adaptor Protein/metabolism , Parvoviridae Infections/metabolism , Parvovirus B19, Human/physiology , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/metabolism , Amino Acid Motifs , Binding Sites , DNA Replication , Humans , Molecular Weight , Mutation , Parvovirus B19, Human/metabolism , Phosphorylation , Proline/metabolism , Protein Binding , Virus Replication
3.
J Virol ; 92(8)2018 04 15.
Article in English | MEDLINE | ID: mdl-29437973

ABSTRACT

Human parvovirus B19 (B19V) expresses a single precursor mRNA (pre-mRNA), which undergoes alternative splicing and alternative polyadenylation to generate 12 viral mRNA transcripts that encode two structural proteins (VP1 and VP2) and three nonstructural proteins (NS1, 7.5-kDa protein, and 11-kDa protein). Splicing at the second 5' donor site (D2 site) of the B19V pre-mRNA is essential for the expression of VP2 and the 11-kDa protein. We previously identified that cis-acting intronic splicing enhancer 2 (ISE2) that lies immediately after the D2 site facilitates the recognition of the D2 donor for its efficient splicing. In this study, we report that ISE2 is critical for the expression of the 11-kDa viral nonstructural protein. We found that ISE2 harbors a consensus RNA binding motif protein 38 (RBM38) binding sequence, 5'-UGUGUG-3'. RBM38 is expressed during the middle stage of erythropoiesis. We first confirmed that RBM38 binds specifically with the ISE2 element in vitro The knockdown of RBM38 significantly decreases the level of spliced mRNA at D2 that encodes the 11-kDa protein but not that of the D2-spliced mRNA that encodes VP2. Importantly, we found that the 11-kDa protein enhances viral DNA replication and virion release. Accordingly, the knockdown of RBM38 decreases virus replication via downregulating 11-kDa protein expression. Taken together, these results suggest that the 11-kDa protein facilitates B19V DNA replication and that RBM38 is an essential host factor for B19V pre-mRNA splicing and for the expression of the 11-kDa protein.IMPORTANCE B19V is a human pathogen that can cause fifth disease, arthropathy, anemia in immunocompromised patients and sickle cell disease patients, myocarditis, and hydrops fetalis in pregnant women. Human erythroid progenitor cells (EPCs) are most susceptible to B19V infection and fully support viral DNA replication. The exclusive tropism of B19V for erythroid-lineage cells is dependent not only on the expression of viral receptors and coreceptors on the cell surface but also on the intracellular host factors that support B19V replication. Our present study shows that B19V uses a host factor, RNA binding motif protein 38 (RBM38), for the processing of its pre-mRNA during virus replication. Specifically, RBM38 interacts with the intronic splicing enhancer 2 (ISE2) element of B19V pre-mRNA and promotes 11-kDa protein expression, thereby regulating the 11-kDa protein-mediated augmentation of B19V replication. The identification of this novel host-pathogen interaction will provide mechanistic insights into B19V replication and aid in finding new targets for anti-B19V therapeutics.


Subject(s)
DNA Replication/physiology , DNA, Viral/metabolism , Down-Regulation/physiology , Erythema Infectiosum/metabolism , Gene Expression Regulation, Viral/physiology , Parvovirus B19, Human/physiology , RNA-Binding Proteins/metabolism , Viral Nonstructural Proteins/biosynthesis , Virus Replication/physiology , DNA, Viral/genetics , Erythema Infectiosum/genetics , Humans , RNA-Binding Proteins/genetics , Viral Nonstructural Proteins/genetics
4.
J Virol ; 92(5)2018 03 01.
Article in English | MEDLINE | ID: mdl-29237843

ABSTRACT

Human parvovirus B19 (B19V) infection of human erythroid progenitor cells (EPCs) induces a DNA damage response and cell cycle arrest at late S phase, which facilitates viral DNA replication. However, it is not clear exactly which cellular factors are employed by this single-stranded DNA virus. Here, we used microarrays to systematically analyze the dynamic transcriptome of EPCs infected with B19V. We found that DNA metabolism, DNA replication, DNA repair, DNA damage response, cell cycle, and cell cycle arrest pathways were significantly regulated after B19V infection. Confocal microscopy analyses revealed that most cellular DNA replication proteins were recruited to the centers of viral DNA replication, but not the DNA repair DNA polymerases. Our results suggest that DNA replication polymerase ƎĀ“ and polymerase α are responsible for B19V DNA replication by knocking down its expression in EPCs. We further showed that although RPA32 is essential for B19V DNA replication and the phosphorylated forms of RPA32 colocalized with the replicating viral genomes, RPA32 phosphorylation was not necessary for B19V DNA replication. Thus, this report provides evidence that B19V uses the cellular DNA replication machinery for viral DNA replication.IMPORTANCE Human parvovirus B19 (B19V) infection can cause transient aplastic crisis, persistent viremia, and pure red cell aplasia. In fetuses, B19V infection can result in nonimmune hydrops fetalis and fetal death. These clinical manifestations of B19V infection are a direct outcome of the death of human erythroid progenitors that host B19V replication. B19V infection induces a DNA damage response that is important for cell cycle arrest at late S phase. Here, we analyzed dynamic changes in cellular gene expression and found that DNA metabolic processes are tightly regulated during B19V infection. Although genes involved in cellular DNA replication were downregulated overall, the cellular DNA replication machinery was tightly associated with the replicating single-stranded DNA viral genome and played a critical role in viral DNA replication. In contrast, the DNA damage response-induced phosphorylated forms of RPA32 were dispensable for viral DNA replication.


Subject(s)
Cell Division , DNA Replication , Host-Pathogen Interactions , Parvoviridae Infections/virology , Parvovirus B19, Human/genetics , Parvovirus B19, Human/metabolism , Virus Replication , Bromodeoxyuridine/metabolism , CD36 Antigens/analysis , CD36 Antigens/metabolism , Cell Cycle , Cell Cycle Checkpoints , Cell Line , DNA Damage , DNA Polymerase III , DNA Polymerase beta , DNA Repair , DNA, Single-Stranded/metabolism , DNA, Viral/genetics , DNA, Viral/metabolism , Erythroid Precursor Cells/cytology , Erythroid Precursor Cells/virology , Fetal Death , Gene Expression Regulation, Viral/physiology , Genome, Viral , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/physiology , Humans , Parvovirus B19, Human/pathogenicity , Phosphorylation , Protein Interaction Maps , Red-Cell Aplasia, Pure/virology , Replication Protein A/genetics , S Phase , Transcriptome , Viremia/virology
5.
Nature ; 498(7455): 516-20, 2013 Jun 27.
Article in English | MEDLINE | ID: mdl-23728302

ABSTRACT

The functional importance of gene enhancers in regulated gene expression is well established. In addition to widespread transcription of long non-coding RNAs (lncRNAs) in mammalian cells, bidirectional ncRNAs are transcribed on enhancers, and are thus referred to as enhancer RNAs (eRNAs). However, it has remained unclear whether these eRNAs are functional or merely a reflection of enhancer activation. Here we report that in human breast cancer cells 17Ɵ-oestradiol (E2)-bound oestrogen receptor α (ER-α) causes a global increase in eRNA transcription on enhancers adjacent to E2-upregulated coding genes. These induced eRNAs, as functional transcripts, seem to exert important roles for the observed ligand-dependent induction of target coding genes, increasing the strength of specific enhancer-promoter looping initiated by ER-α binding. Cohesin, present on many ER-α-regulated enhancers even before ligand treatment, apparently contributes to E2-dependent gene activation, at least in part by stabilizing E2/ER-α/eRNA-induced enhancer-promoter looping. Our data indicate that eRNAs are likely to have important functions in many regulated programs of gene transcription.


Subject(s)
Enhancer Elements, Genetic/genetics , Estrogens/pharmacology , RNA, Untranslated/genetics , Transcriptional Activation/drug effects , Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Estradiol/pharmacology , Estrogen Receptor alpha/metabolism , Humans , Ligands , MCF-7 Cells , Nucleic Acid Conformation/drug effects , Promoter Regions, Genetic/genetics , RNA, Untranslated/biosynthesis , RNA, Untranslated/metabolism , Transcription, Genetic/drug effects , Transcription, Genetic/genetics , Transcriptional Activation/genetics , Cohesins
6.
PLoS Pathog ; 8(8): e1002899, 2012.
Article in English | MEDLINE | ID: mdl-22956907

ABSTRACT

Human bocavirus 1 (HBoV1) has been identified as one of the etiological agents of wheezing in young children with acute respiratory-tract infections. In this study, we have obtained the sequence of a full-length HBoV1 genome (including both termini) using viral DNA extracted from a nasopharyngeal aspirate of an infected patient, cloned the full-length HBoV1 genome, and demonstrated DNA replication, encapsidation of the ssDNA genome, and release of the HBoV1 virions from human embryonic kidney 293 cells. The HBoV1 virions generated from this cell line-based production system exhibits a typical icosahedral structure of approximately 26 nm in diameter, and is capable of productively infecting polarized primary human airway epithelia (HAE) from the apical surface. Infected HAE showed hallmarks of lung airway-tract injury, including disruption of the tight junction barrier, loss of cilia and epithelial cell hypertrophy. Notably, polarized HAE cultured from an immortalized airway epithelial cell line, CuFi-8 (originally derived from a cystic fibrosis patient), also supported productive infection of HBoV1. Thus, we have established a reverse genetics system and generated the first cell line-based culture system for the study of HBoV1 infection, which will significantly advance the study of HBoV1 replication and pathogenesis.


Subject(s)
Epithelial Cells/virology , Human bocavirus/physiology , Parvoviridae Infections/virology , Respiratory Tract Infections/virology , Virus Replication , Base Sequence , Cell Line , DNA, Viral/chemistry , DNA, Viral/genetics , Epithelium/virology , Female , Genome, Viral/genetics , Human bocavirus/genetics , Human bocavirus/isolation & purification , Humans , Inverted Repeat Sequences , Molecular Sequence Data , Respiratory System/virology , Reverse Genetics , Sequence Analysis, DNA , Transfection
7.
PLoS Pathog ; 7(6): e1002088, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21698228

ABSTRACT

Human parvovirus B19 (B19V) causes a variety of human diseases. Disease outcomes of bone marrow failure in patients with high turnover of red blood cells and immunocompromised conditions, and fetal hydrops in pregnant women are resulted from the targeting and destruction of specifically erythroid progenitors of the human bone marrow by B19V. Although the ex vivo expanded erythroid progenitor cells recently used for studies of B19V infection are highly permissive, they produce progeny viruses inefficiently. In the current study, we aimed to identify the mechanism that underlies productive B19V infection of erythroid progenitor cells cultured in a physiologically relevant environment. Here, we demonstrate an effective reverse genetic system of B19V, and that B19V infection of ex vivo expanded erythroid progenitor cells at 1% O(2) (hypoxia) produces progeny viruses continuously and efficiently at a level of approximately 10 times higher than that seen in the context of normoxia. With regard to mechanism, we show that hypoxia promotes replication of the B19V genome within the nucleus, and that this is independent of the canonical PHD/HIFα pathway, but dependent on STAT5A and MEK/ERK signaling. We further show that simultaneous upregulation of STAT5A signaling and down-regulation of MEK/ERK signaling boosts the level of B19V infection in erythroid progenitor cells under normoxia to that in cells under hypoxia. We conclude that B19V infection of ex vivo expanded erythroid progenitor cells at hypoxia closely mimics native infection of erythroid progenitors in human bone marrow, maintains erythroid progenitors at a stage conducive to efficient production of progeny viruses, and is regulated by the STAT5A and MEK/ERK pathways.


Subject(s)
Erythema Infectiosum/virology , Erythroid Precursor Cells/virology , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Hypoxia/pathology , MAP Kinase Kinase Kinases/physiology , Parvovirus B19, Human/physiology , STAT5 Transcription Factor/physiology , Tumor Suppressor Proteins/physiology , CD36 Antigens/metabolism , Cell Differentiation/physiology , Cell Proliferation , Cells, Cultured , DNA Replication , Erythema Infectiosum/complications , Erythema Infectiosum/metabolism , Erythroid Precursor Cells/metabolism , Erythroid Precursor Cells/pathology , Erythroid Precursor Cells/physiology , Humans , Hypoxia/complications , Hypoxia/virology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , MAP Kinase Kinase Kinases/metabolism , MAP Kinase Signaling System/genetics , MAP Kinase Signaling System/physiology , Models, Biological , STAT5 Transcription Factor/metabolism , Tumor Suppressor Proteins/metabolism , Virus Replication/genetics , Virus Replication/physiology
8.
J Virol ; 85(1): 133-45, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21047968

ABSTRACT

Minute virus of canines (MVC) is an autonomous parvovirus that replicates efficiently without helper viruses in Walter Reed/3873D (WRD) canine cells. We previously showed that MVC infection induces mitochondrion-mediated apoptosis and G(2)/M-phase arrest in infected WRD cells. However, the mechanism responsible for these effects has not been established. Here, we report that MVC infection triggers a DNA damage response in infected cells, as evident from phosphorylation of H2AX and RPA32. We discovered that both ATM (ataxia telangiectasia-mutated kinase) and ATR (ATM- and Rad3-related kinase) were phosphorylated in MVC-infected WRD cells and confirmed that ATM activation was responsible for the phosphorylation of H2AX, whereas ATR activation was required for the phosphorylation of RPA32. Both pharmacological inhibition of ATM activation and knockdown of ATM in MVC-infected cells led to a significant reduction in cell death, a moderate correction of cell cycle arrest, and most importantly, a reduction in MVC DNA replication and progeny virus production. Parallel experiments with an ATR-targeted small interfering RNA (siRNA) had no effect. Moreover, we identified that this ATM-mediated cell death is p53 dependent. In addition, we localized the Mre11-Rad50-Nbs1 (MRN) complex, the major mediator as well as a substrate of the ATM-mediated DNA damage response pathway to MVC replication centers during infection, and show that Mre11 knockdown led to a reduction in MVC DNA replication. Our findings are the first to support the notion that an autonomous parvovirus is able to hijack the host DNA damage machinery for its own replication and for the induction of cell death.


Subject(s)
Bocavirus/pathogenicity , Cell Death , DNA Damage , DNA Replication , Animals , Ataxia Telangiectasia Mutated Proteins , Bocavirus/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dogs , Histones/genetics , Histones/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Replication Protein A/genetics , Replication Protein A/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Virus Replication
9.
Blood ; 115(5): 1070-80, 2010 Feb 04.
Article in English | MEDLINE | ID: mdl-19861680

ABSTRACT

Human parvovirus B19 (B19V) infection shows a strong erythroid tropism and drastically destroys erythroid progenitor cells, thus leading to most of the disease outcomes associated with B19V infection. In this study, we systematically examined the 3 B19V nonstructural proteins, 7.5 kDa, 11 kDa, and NS1, for their function in inducing apoptosis in transfection of primary ex vivo-expanded erythroid progenitor cells, in comparison with apoptosis induced during B19V infection. Our results show that 11 kDa is a more significant inducer of apoptosis than NS1, whereas 7.5 kDa does not induce apoptosis. Furthermore, we determined that caspase-10, an initiator caspase in death receptor signaling, is the most active caspase in apoptotic erythroid progenitors induced by 11 kDa and NS1 as well as during B19V infection. More importantly, cytoplasm-localized 11 kDa is expressed at least 100 times more than nucleus-localized NS1 at the protein level in primary erythroid progenitor cells infected with B19V; and inhibition of 11 kDa expression using antisense oligos targeting specifically to the 11 kDa-encoding mRNAs reduces apoptosis significantly during B19V infection of erythroid progenitor cells. Taken together, these results demonstrate that the 11 kDa protein contributes to erythroid progenitor cell death during B19V infection.


Subject(s)
Apoptosis , Erythroid Precursor Cells/metabolism , Parvovirus B19, Human/genetics , Viral Nonstructural Proteins/genetics , Amino Acid Chloromethyl Ketones/pharmacology , Caspase 10/metabolism , Caspase Inhibitors , Cell Line , Cells, Cultured , Cysteine Proteinase Inhibitors/pharmacology , Erythroid Precursor Cells/cytology , Erythroid Precursor Cells/virology , Flow Cytometry , HeLa Cells , Host-Pathogen Interactions , Humans , Immunoblotting , In Situ Nick-End Labeling , K562 Cells , Molecular Weight , Parvovirus B19, Human/metabolism , Parvovirus B19, Human/physiology , Quinolines/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/physiology
10.
J Virol ; 84(11): 5615-26, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20335259

ABSTRACT

Bocavirus is a newly classified genus of the family Parvovirinae. Infection with Bocavirus minute virus of canines (MVC) produces a strong cytopathic effect in permissive Walter Reed/3873D (WRD) canine cells. We have systematically characterized the MVC infection-produced cytopathic effect in WRD cells, namely, the cell death and cell cycle arrest, and carefully examined how MVC infection induces the cytopathic effect. We found that MVC infection induces an apoptotic cell death characterized by Bax translocalization to the mitochondrial outer membrane, disruption of the mitochondrial outer membrane potential, and caspase activation. Moreover, we observed that the activation of caspases occurred only when the MVC genome was replicating, suggesting that replication of the MVC genome induces apoptosis. MVC infection also induced a gradual cell cycle arrest from the S phase in early infection to the G(2)/M phase at a later stage, which was confirmed by the upregulation of cyclin B1 and phosphorylation of cdc2. Cell cycle arrest at the G(2)/M phase was reproduced by transfection of a nonreplicative NS1 knockout mutant of the MVC infectious clone, as well as by inoculation of UV-irradiated MVC. In contrast with other parvoviruses, only expression of the MVC proteins by transfection did not induce apoptosis or cell cycle arrest. Taken together, our results demonstrate that MVC infection induces a mitochondrion-mediated apoptosis that is dependent on the replication of the viral genome, and the MVC genome per se is able to arrest the cell cycle at the G(2)/M phase. Our results may shed light on the molecular pathogenesis of Bocavirus infection in general.


Subject(s)
Apoptosis , Bocavirus , Cell Cycle , Cell Division , Parvoviridae Infections/pathology , Animals , Cell Line , Dogs , Genome, Viral , Mitochondria/pathology , Virus Replication
11.
J Virol ; 84(6): 2687-96, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20042496

ABSTRACT

Aleutian mink disease virus (AMDV) is currently the only known member of the genus Amdovirus in the family Parvoviridae. It is the etiological agent of Aleutian disease of mink. We have previously shown that a small protein with a molecular mass of approximately 26 kDa was present during AMDV infection and following transfection of capsid expression constructs (J. Qiu, F. Cheng, L. R. Burger, and D. Pintel, J. Virol. 80:654-662, 2006). In this study, we report that the capsid proteins were specifically cleaved at aspartic acid residue 420 (D420) during virus infection, resulting in the previously observed cleavage product. Mutation of a single amino acid residue at D420 abolished the specific cleavage. Expression of the capsid proteins alone in Crandell feline kidney (CrFK) cells reproduced the cleavage of the capsid proteins in virus infection. More importantly, capsid protein expression alone induced active caspases, of which caspase-10 was the most active. Active caspases, in turn, cleaved capsid proteins in vivo. Our results also showed that active caspase-7 specifically cleaved capsid proteins at D420 in vitro. These results suggest that viral capsid proteins alone induce caspase activation, resulting in cleavage of capsid proteins. We also provide evidence that AMDV mutants resistant to caspase-mediated capsid cleavage increased virus production approximately 3- to 5-fold in CrFK cells compared to that produced from the parent virus AMDV-G at 37 degrees C but not at 31.8 degrees C. Collectively, our results indicate that caspase activity plays multiple roles in AMDV infection and that cleavage of the capsid proteins might have a role in regulating persistent infection of AMDV.


Subject(s)
Aleutian Mink Disease Virus/metabolism , Capsid Proteins/metabolism , Caspases/metabolism , Mink/virology , Aleutian Mink Disease/virology , Amino Acid Sequence , Animals , Capsid Proteins/genetics , Caspases/genetics , Cats , Cell Line , Enzyme Activation , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Molecular Sequence Data , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Temperature
12.
J Virol ; 84(23): 12385-96, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20861249

ABSTRACT

Parvovirus B19 (B19V) infection is highly restricted to human erythroid progenitor cells. Although previous studies have led to the theory that the basis of this tropism is receptor expression, this has been questioned by more recent observation. In the study reported here, we have investigated the basis of this tropism, and a potential role of erythropoietin (Epo) signaling, in erythroid progenitor cells (EPCs) expanded ex vivo from CD34(+) hematopoietic cells in the absence of Epo (CD36(+)/Epo(-) EPCs). We show, first, that CD36(+)/Epo(-) EPCs do not support B19V replication, in spite of B19V entry, but Epo exposure either prior to infection or after virus entry enabled active B19V replication. Second, when Janus kinase 2 (Jak2) phosphorylation was inhibited using the inhibitor AG490, phosphorylation of the Epo receptor (EpoR) was also inhibited, and B19V replication in ex vivo-expanded erythroid progenitor cells exposed to Epo (CD36(+)/Epo(+) EPCs) was abolished. Third, expression of constitutively active EpoR in CD36(+)/Epo(-) EPCs led to efficient B19V replication. Finally, B19V replication in CD36(+)/Epo(+) EPCs required Epo, and the replication response was dose dependent. Our findings demonstrate that EpoR signaling is absolutely required for B19V replication in ex vivo-expanded erythroid progenitor cells after initial virus entry and at least partly accounts for the remarkable tropism of B19V infection for human erythroid progenitors.


Subject(s)
Erythroid Precursor Cells/virology , Parvoviridae Infections/physiopathology , Parvovirus B19, Human/physiology , Receptors, Erythropoietin/metabolism , Signal Transduction/physiology , Viral Tropism/physiology , Virus Replication/physiology , Blotting, Southern , Blotting, Western , CD36 Antigens/metabolism , Flow Cytometry , Fluorescent Antibody Technique , Genetic Vectors/genetics , Humans , Janus Kinase 2/metabolism , Lentivirus , Phosphorylation/drug effects , RNA, Small Interfering , Reverse Transcriptase Polymerase Chain Reaction , Tyrphostins/pharmacology
13.
J Virol ; 83(8): 3956-67, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19211770

ABSTRACT

Minute virus of canines (MVC) is a member of the genus Bocavirus in the family Parvoviridae. We have molecularly cloned and sequenced the 5'- and 3'-terminal palindromes of MVC. The MVC genome, 5,404 nucleotides (nt) in length, shared an identity of 52.6% and 52.1% with that of human bocavirus and bovine parvovirus, respectively. It had distinct palindromic hairpins of 183 nt and 198 nt at the left-end and right-end termini of the genome, respectively. The left-end terminus was also found in two alternative orientations (flip or flop). Both termini shared extensive similarities with those of bovine parvovirus. Four full-length molecular clones constructed with different orientations of the left-end terminus proved to be infectious in Walter Reed canine cell/3873D (WRD) canine cells. Both MVC infection and transfection of the infectious clone in WRD cells revealed an identical RNA transcription profile that was similar to that of bovine parvovirus. Mutagenesis of the infectious clone demonstrated that the middle open reading frame encodes the NP1 protein. This protein, unique to the genus Bocavirus, was essential for MVC DNA replication. Moreover, the phospholipase A2 motif in the VP1 unique region was also critical for MVC infection. Thus, our studies revealed important information about the genus Bocavirus that may eventually help us to clone the human bocavirus and study its pathogenesis.


Subject(s)
Bocavirus/genetics , DNA, Viral/genetics , Virus Replication , Animals , Bocavirus/physiology , Cell Line , Cloning, Molecular , DNA Mutational Analysis , DNA, Viral/chemistry , Dogs , Inverted Repeat Sequences , Molecular Sequence Data , Open Reading Frames , RNA Precursors/biosynthesis , Sequence Analysis, DNA , Sequence Homology, Nucleic Acid , Transcription, Genetic , Viral Proteins/genetics
14.
J Med Chem ; 50(22): 5372-81, 2007 Nov 01.
Article in English | MEDLINE | ID: mdl-17924615

ABSTRACT

Our earlier studies have shown that over 20 nonpolar 17beta-estradiol metabolite peaks were detected following incubations of radioactive 17beta-estradiol with human liver microsomes or recombinant human cytochrome P450 isoforms in the presence of NADPH as a cofactor. The structures of two representative nonpolar metabolites were identified earlier as dimers of 17beta-estradiol linked through a diaryl ether bond between the C-3 phenolic oxygen of one molecule and the C-2 or C-4 aromatic carbon of another. Six additional putative dimers between estrone and 17beta-estradiol with structures similar to the two identified ones were synthesized in this study. Using these newly synthesized estrogen dimers as reference standards, we demonstrated that incubations of human liver microsomes or various human cytochrome P450 isoforms with estrone or 17beta-estradiol alone or two estrogens in combination in the presence of NADPH as a cofactor resulted in the formation of all eight estrogen dimers in varying quantities.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Estradiol/analogs & derivatives , Estradiol/metabolism , Estrogens/metabolism , Estrone/analogs & derivatives , Estrone/metabolism , Catalysis , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme System/chemistry , Dimerization , Estradiol/chemistry , Estrogens/chemistry , Estrone/chemistry , Humans , In Vitro Techniques , Isoenzymes/chemistry , Isoenzymes/metabolism , Microsomes, Liver/metabolism , Recombinant Proteins/chemistry
15.
Diabetes ; 62(7): 2613-7, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23434931

ABSTRACT

Proliferative diabetic retinopathy (PDR) is the most severe vision-threatening complication of diabetes. For investigation of genetic association between TCF7L2 and PDR in Caucasian type 2 diabetes mellitus (T2DM) and its functional consequences, 383 T2DM patients with PDR (T2DM-PDR) and 756 T2DM patients without diabetic retinopathy (T2DM-no DR) were genotyped with rs7903146 in TCF7L2. We found that risk allele (T) frequency of rs7903146 was significantly higher in T2DM-PDR patients (allelic P = 2.52E-04). In lymphoblastoid cells induced to undergo endoplasmic reticulum (ER) stress by treatment of tunicamycin, higher fold change of TCF7L2 and VEGFA mRNA levels were observed in rs7903146-TT cells than in rs7903146-CC cells (P = 0.02 for TCF7L2; P = 0.004 for VEGFA), suggesting that ER stress plays a role in PDR pathogenesis. Silencing TCF7L2 resulted in decreased mRNA levels of both TCF7L2 and VEGFA (P < 0.001). Retinas of oxygen-induced retinopathy mice (a model for PDR) had higher TCF7L2 and VEGFA mRNA levels than those of controls (P = 2.9E-04 for TCF7L2; P = 1.9E-07 for VEGFA). Together, data from our study show that TCF7L2-rs7903146 is associated with PDR in Caucasian T2DM and suggest that TCF7L2 promotes pathological retinal neovascularization via ER stress-dependent upregulation of VEGFA.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Diabetic Retinopathy/genetics , Genetic Predisposition to Disease , Polymorphism, Single Nucleotide , Transcription Factor 7-Like 2 Protein/genetics , Alleles , Animals , Endoplasmic Reticulum Stress/genetics , Gene Frequency , Genetic Association Studies , Genotype , Humans , Mice , Retinal Neovascularization/genetics , Vascular Endothelial Growth Factor A/genetics
16.
Future Virol ; 5(6): 731-743, 2010 Nov.
Article in English | MEDLINE | ID: mdl-21331319

ABSTRACT

The cytopathic effects induced during parvovirus infection have been widely documented. Parvovirus infection-induced cell death is often directly associated with disease outcomes (e.g., anemia resulting from loss of erythroid progenitors during parvovirus B19 infection). Apoptosis is the major form of cell death induced by parvovirus infection. However, nonapoptotic cell death, namely necrosis, has also been reported during infection of the minute virus of mice, parvovirus H-1 and bovine parvovirus. Recent studies have revealed multiple mechanisms underlying the cell death during parvovirus infection. These mechanisms vary in different parvoviruses, although the large nonstructural protein (NS)1 and the small NS proteins (e.g., the 11 kDa of parvovirus B19), as well as replication of the viral genome, are responsible for causing infection-induced cell death. Cell cycle arrest is also common, and contributes to the cytopathic effects induced during parvovirus infection. While viral NS proteins have been indicated to induce cell cycle arrest, increasing evidence suggests that a cellular DNA damage response triggered by an invading single-stranded parvoviral genome is the major inducer of cell cycle arrest in parvovirus-infected cells. Apparently, in response to infection, cell death and cell cycle arrest of parvovirus-infected cells are beneficial to the viral cell lifecycle (e.g., viral DNA replication and virus egress). In this article, we will discuss recent advances in the understanding of the mechanisms underlying parvovirus infection-induced cell death and cell cycle arrest.

17.
PLoS One ; 5(12): e15113, 2010 Dec 03.
Article in English | MEDLINE | ID: mdl-21151930

ABSTRACT

The transcription profile of chipmunk parvovirus (ChpPV), a tentative member of the genus Erythrovirus in the subfamily Parvovirinae of the family Parvoviridae, was characterized by transfecting a nearly full-length genome. We found that it is unique from the profiles of human parvovirus B19 and simian parvovirus, the members in the genus Erythrovirus so far characterized, in that the small RNA transcripts were not processed for encoding small non-structural proteins. However, like the large non-structural protein NS1 of the human parvovirus B19, the ChpPV NS1 is a potent inducer of apoptosis. Further phylogenetic analysis of ChpPV with other parvoviruses in the subfamily Parvovirinae indicates that ChpPV is distinct from the members in genus Erythrovirus. Thus, we conclude that ChpPV may represent a new genus in the family Parvoviridae.


Subject(s)
Erythrovirus/genetics , Parvoviridae Infections/genetics , Parvoviridae/genetics , Animals , Apoptosis , COS Cells , Chlorocebus aethiops , Erythrovirus/classification , Humans , Parvoviridae/classification , Phylogeny , Plasmids/metabolism , Sciuridae , Transcription, Genetic
18.
Virology ; 403(2): 145-54, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20457462

ABSTRACT

We have generated a quantitative transcription profile of human bocavirus type 1 (HBoV1) by transfecting a nearly full-length clone in human lung epithelial A549 cells as well as in a replication competent system in 293 cells. The overall transcription profile of HBoV1 is similar to that of two other members of genus Bocavirus, minute virus of canines and bovine parvovirus 1. In particular, a spliced NS1-transcript that was not recognized previously expressed the large non-structural protein NS1 at approximately 100kDa; and the NP1-encoding transcripts were expressed abundantly. In addition, the protein expression profile of human bocavirus type 2 (HBoV2) was examined in parallel by transfection of a nearly full-length clone in A549 cells, which is similar to that of HBoV1. Moreover, our results showed that, unlike human parvovirus B19 infection, expression of the HBoV1 proteins only does not induce cell cycle arrest and apoptosis of A549 cells.


Subject(s)
Gene Expression Profiling , Human bocavirus/physiology , Bocavirus/physiology , Cell Line , Humans , RNA Splicing , RNA, Messenger/metabolism , RNA, Viral/metabolism
19.
PLoS One ; 4(11): e7703, 2009 Nov 05.
Article in English | MEDLINE | ID: mdl-19890385

ABSTRACT

BACKGROUND: Dinitrochlorobenzene-induced contact hypersensitivity is widely considered as a cell-mediated rather than antibody-mediated immune response. At present, very little is known about the role of antigen-specific antibodies and B cells in the development of dinitrochlorobenzene-induced hypersensitivity reactions, and this is the subject of the present investigation. METHODOLOGY/PRINCIPAL FINDINGS: Data obtained from multiple lines of experiments unequivocally showed that the formation of dinitrochlorobenzene-specific Abs played an important role in the development of dinitrochlorobenzene-induced contact hypersensitivity. The appearance of dinitrochlorobenzene-induced skin dermatitis matched in timing the appearance of the circulating dinitrochlorobenzene-specific antibodies. Adoptive transfer of sera containing dinitrochlorobenzene-specific antibodies from dinitrochlorobenzene-treated mice elicited a much stronger hypersensitivity reaction than the adoptive transfer of lymphocytes from the same donors. Moreover, dinitrochlorobenzene-induced contact hypersensitivity was strongly suppressed in B cell-deficient mice with no DNCB-specific antibodies. It was also observed that treatment of animals with dinitrochlorobenzene polarized Th cells into Th2 differentiation by increasing the production of Th2 cytokines while decreasing the production of Th1 cytokines. CONCLUSIONS/SIGNIFICANCE: In striking contrast to the long-held belief that dinitrochlorobenzene-induced contact hypersensitivity is a cell-mediated immune response, the results of our present study demonstrated that the production of dinitrochlorobenzene-specific antibodies by activated B cells played an indispensible role in the pathogenesis of dinitrochlorobenzene-induced CHS. These findings may provide new possibilities in the treatment of human contact hypersensitivity conditions.


Subject(s)
Antibodies/metabolism , Dermatitis, Allergic Contact/pathology , Dinitrochlorobenzene/toxicity , Animals , B-Lymphocytes/drug effects , Cell Differentiation , Cytokines/metabolism , Female , Immunity, Cellular/immunology , Irritants/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Skin/drug effects , Th1 Cells/drug effects , Th2 Cells/drug effects
20.
Virology ; 394(2): 276-85, 2009 Nov 25.
Article in English | MEDLINE | ID: mdl-19758675

ABSTRACT

We have characterized the transcription profiles of parvovirus B19 (B19V) genotype-2 A6 and genotype-3 V9 variants. The A6 RNA profile differs from that of the prototype B19V in both B19V non-permissive and permissive cells, whereas the overall profile of the V9 RNA in these cells is similar to that of the prototype. A unique feature we have identified is that the genotype-2 A6 variant used only one splice acceptor to remove the first intron. We also demonstrated that the inverted terminal repeats (ITRs) of the prototype B19V support replication of the V9 genome, which produces infectious virus, but not that of the A6 genome, in B19V-permissive cells. Similar to the proapoptotic nature of the prototype B19V large non-structural protein (NS1), the A6 and V9 NS1 proteins also are potent inducers of apoptosis in B19V-permissive cells.


Subject(s)
Parvovirus B19, Human/genetics , Apoptosis , Base Sequence , Cell Line , Chromosome Mapping , DNA Primers/genetics , Gene Expression Profiling , Genome, Viral , Genotype , Humans , Parvovirus B19, Human/isolation & purification , Parvovirus B19, Human/pathogenicity , Parvovirus B19, Human/physiology , RNA, Messenger/genetics , RNA, Viral/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Terminal Repeat Sequences , Transfection , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/physiology
SELECTION OF CITATIONS
SEARCH DETAIL