Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
1.
J Virol ; 96(2): e0132621, 2022 01 26.
Article in English | MEDLINE | ID: mdl-34669461

ABSTRACT

Parvovirus B19 (B19V) infection can cause hematological disorders and fetal hydrops during pregnancy. Currently, no antivirals or vaccines are available for the treatment or prevention of B19V infection. To identify novel small-molecule antivirals against B19V replication, we developed a high-throughput screening (HTS) assay, which is based on an in vitro nicking assay using recombinant N-terminal amino acids 1 to 176 of the viral large nonstructural protein (NS1N) and a fluorescently labeled DNA probe (OriQ) that spans the nicking site of the viral DNA replication origin. We collectively screened 17,040 compounds and identified 2,178 (12.78%) hits that possess >10% inhibition of the NS1 nicking activity, among which 84 hits were confirmed to inhibit nicking in a dose-dependent manner. Using ex vivo-expanded primary human erythroid progenitor cells (EPCs) infected by B19V, we validated 24 compounds that demonstrated >50% in vivo inhibition of B19V infection at 10 µM, which can be categorized into 7 structure scaffolds. Based on the therapeutic index (half-maximal cytotoxic concentration [CC50]/half-maximal effective concentration [EC50] ratio) in EPCs, the top 4 compounds were chosen to examine their inhibitions of B19V infection in EPCs at two times of the 90% maximal effective concentration (EC90). A purine derivative (P7) demonstrated an antiviral effect (EC50 = 1.46 µM) without prominent cytotoxicity (CC50 = 71.8 µM) in EPCs and exhibited 92% inhibition of B19V infection in EPCs at 3.32 µM, which can be used as the lead compound in future studies for the treatment of B19V infection-caused hematological disorders. IMPORTANCE B19V encodes a large nonstructural protein, NS1. Its N-terminal domain (NS1N) consisting of amino acids 1 to 176 binds to viral DNA and serves as an endonuclease to nick the viral DNA replication origins, which is a pivotal step in rolling-hairpin-dependent B19V DNA replication. For high-throughput screening (HTS) of anti-B19V antivirals, we miniaturized a fluorescence-based in vitro nicking assay, which employs a fluorophore-labeled probe spanning the terminal resolution site (trs) and the NS1N protein, into a 384-well-plate format. The HTS assay showed high reliability and capability in screening 17,040 compounds. Based on the therapeutic index (half-maximal cytotoxic concentration [CC50]/half-maximal effective concentration [EC50] ratio) in EPCs, a purine derivative demonstrated an antiviral effect of 92% inhibition of B19V infection in EPCs at 3.32 µM (two times the EC90). Our study demonstrated a robust HTS assay for screening antivirals against B19V infection.


Subject(s)
Antiviral Agents/pharmacology , Erythroid Precursor Cells/virology , High-Throughput Screening Assays/methods , Parvovirus B19, Human/drug effects , Antiviral Agents/chemistry , Cell Survival/drug effects , DNA Replication/drug effects , DNA, Viral/metabolism , Erythroid Precursor Cells/drug effects , Fluorescent Dyes , Humans , Parvovirus B19, Human/physiology , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Replication Origin , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism , Virus Replication/drug effects
2.
Stem Cell Reports ; 16(5): 1165-1181, 2021 05 11.
Article in English | MEDLINE | ID: mdl-33979601

ABSTRACT

SARS-CoV-2 infection is associated with lower blood oxygen levels, even in patients without hypoxia requiring hospitalization. This discordance illustrates the need for a more unifying explanation as to whether SARS-CoV-2 directly or indirectly affects erythropoiesis. Here, we show significantly enriched CD71+ erythroid precursors/progenitors in the blood circulation of COVID-19 patients. We found that these cells have distinctive immunosuppressive properties. In agreement, we observed a strong negative correlation between the frequency of these cells with T and B cell proportions in COVID-19 patients. The expansion of these CD71+ erythroid precursors/progenitors was negatively correlated with the hemoglobin levels. A subpopulation of abundant erythroid cells, CD45+ CD71+ cells, co-express ACE2, TMPRSS2, CD147, and CD26, and these can be infected with SARS-CoV-2. In turn, pre-treatment of erythroid cells with dexamethasone significantly diminished ACE2/TMPRSS2 expression and subsequently reduced their infectivity with SARS-CoV-2. This provides a novel insight into the impact of SARS-CoV-2 on erythropoiesis and hypoxia seen in COVID-19 patients.


Subject(s)
Adaptive Immunity/immunology , COVID-19/pathology , Erythroid Precursor Cells/virology , Erythropoiesis/physiology , Hemoglobins/analysis , Oxygen/blood , Adolescent , Adult , Aged , Aged, 80 and over , Angiotensin-Converting Enzyme 2/metabolism , Animals , B-Lymphocytes/cytology , B-Lymphocytes/immunology , COVID-19/immunology , Dexamethasone/pharmacology , Erythroid Precursor Cells/immunology , Female , Humans , Lymphocyte Count , Male , Mice , Mice, Inbred BALB C , Middle Aged , SARS-CoV-2/immunology , Serine Endopeptidases/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Young Adult
3.
Stem Cell Reports ; 16(3): 428-436, 2021 03 09.
Article in English | MEDLINE | ID: mdl-33581053

ABSTRACT

We document here that intensive care COVID-19 patients suffer a profound decline in hemoglobin levels but show an increase of circulating nucleated red cells, suggesting that SARS-CoV-2 infection either directly or indirectly induces stress erythropoiesis. We show that ACE2 expression peaks during erythropoiesis and renders erythroid progenitors vulnerable to infection by SARS-CoV-2. Early erythroid progenitors, defined as CD34-CD117+CD71+CD235a-, show the highest levels of ACE2 and constitute the primary target cell to be infected during erythropoiesis. SARS-CoV-2 causes the expansion of colony formation by erythroid progenitors and can be detected in these cells after 2 weeks of the initial infection. Our findings constitute the first report of SARS-CoV-2 infectivity in erythroid progenitor cells and can contribute to understanding both the clinical symptoms of severe COVID-19 patients and how the virus can spread through the circulation to produce local inflammation in tissues, including the bone marrow.


Subject(s)
COVID-19/virology , Erythroid Precursor Cells/virology , Erythropoiesis/physiology , SARS-CoV-2/pathogenicity , Angiotensin-Converting Enzyme 2/metabolism , Animals , COVID-19/metabolism , Cell Line , Chlorocebus aethiops , Erythroid Precursor Cells/metabolism , Humans , Inflammation/metabolism , Inflammation/virology , Vero Cells
4.
Viruses ; 12(12)2020 12 18.
Article in English | MEDLINE | ID: mdl-33353185

ABSTRACT

Human parvovirus B19 (B19V) causes various human diseases, ranging from childhood benign infection to arthropathies, severe anemia and fetal hydrops, depending on the health state and hematological status of the patient. To counteract B19V blood-borne contamination, evaluation of B19 DNA in plasma pools and viral inactivation/removal steps are performed, but nucleic acid testing does not correctly reflect B19V infectivity. There is currently no appropriate cellular model for detection of infectious units of B19V. We describe here an improved cell-based method for detecting B19V infectious units by evaluating its host transcription. We evaluated the ability of various cell lines to support B19V infection. Of all tested, UT7/Epo cell line, UT7/Epo-STI, showed the greatest sensitivity to B19 infection combined with ease of performance. We generated stable clones by limiting dilution on the UT7/Epo-STI cell line with graduated permissiveness for B19V and demonstrated a direct correlation between infectivity and S/G2/M cell cycle stage. Two of the clones tested, B12 and E2, reached sensitivity levels higher than those of UT7/Epo-S1 and CD36+ erythroid progenitor cells. These findings highlight the importance of cell cycle status for sensitivity to B19V, and we propose a promising new straightforward cell-based method for quantifying B19V infectious units.


Subject(s)
Cell Cycle , Parvoviridae Infections/diagnosis , Parvoviridae Infections/virology , Parvovirus B19, Human/physiology , Biomarkers , Cell Line , Erythroid Cells/metabolism , Erythroid Cells/virology , Erythroid Precursor Cells/metabolism , Erythroid Precursor Cells/virology , Gene Expression , Gene Expression Regulation, Viral , Humans , Molecular Diagnostic Techniques , Parvoviridae Infections/metabolism , Sensitivity and Specificity , Viral Tropism
5.
Viruses ; 12(9)2020 08 25.
Article in English | MEDLINE | ID: mdl-32854437

ABSTRACT

Parvovirus B19 (B19V), an ssDNA virus in the family Parvoviridae, is a human pathogenic virus, responsible for a wide range of clinical manifestations, still in need of effective and specific antivirals. DNA structures, including G-quadruplex (G4), have been recognised as relevant functional features in viral genomes, and small-molecule ligands binding to these structures are promising antiviral compounds. Bioinformatic tools predict the presence of potential G4 forming sequences (PQSs) in the genome of B19V, raising interest as targets for antiviral strategies. Predictions locate PQSs in the genomic terminal regions, in proximity to replicative origins. The actual propensity of these PQSs to form G4 structures was investigated by circular dichroism spectroscopic analysis on synthetic oligonucleotides of corresponding sequences. No signature of G4 structures was detected, and the interaction with the G4 ligand BRACO-19 (N,N'-(9-{[4-(dimethylamino)phenyl]amino}acridine-3,6-diyl)bis(3-pyrrolidin-1-ylpropanamide) did not appear consistent with the stabilisation of G4 structures. Any potential role of PQSs in the viral lifecycle was then assessed in an in vitro infection model system, by evaluating any variation in replication or expression of B19V in the presence of the G4 ligands BRACO-19 and pyridostatin. Neither showed a significant inhibitory activity on B19V replication or expression. Experimental challenge did not support bioinformatic predictions. The terminal regions of B19V are characterised by relevant sequence and symmetry constraints, which are functional to viral replication. Our experiments suggest that these impose a stringent requirement prevailing over the propensity of forming actual G4 structures.


Subject(s)
DNA, Viral/chemistry , G-Quadruplexes , Parvovirus B19, Human/genetics , Acridines/pharmacology , Aminoquinolines/pharmacology , Antiviral Agents/pharmacology , Cells, Cultured , Circular Dichroism , Computational Biology , DNA, Viral/metabolism , Erythroid Precursor Cells/virology , Genome, Viral , Humans , Parvovirus B19, Human/drug effects , Parvovirus B19, Human/physiology , Picolinic Acids/pharmacology , Virus Replication/drug effects
6.
Viral Immunol ; 33(8): 539-549, 2020 10.
Article in English | MEDLINE | ID: mdl-32412895

ABSTRACT

Human parvovirus B19 (B19), a human pathogen of the erythroparvovirus genus, is responsible for a variety of diseases. B19 cause less symptoms in healthy individuals, also cause acute and chronic anemia in immunodeficiency patients. Transient aplastic crisis and pure red cell aplasia are two kinds of anemic hemogram, respectively, in acute and chronic B19 infection phase, especially occurring in patients with a shortened red cell survival or with immunodeficiency. In addition, B19-infected pregnant women may cause hydrops fetalis or fetal loss. B19 possesses high affinity to bone marrow and fetal liver due to its extremely restricted cytotoxicity to erythroid progenitor cells (EPCs) mediated by viral proteins. The nonstructural protein NS1 is considered to be the major pathogenic factor, which has been shown to inhibit the differentiation and maturation of EPCs through inducing viral DNA damage responses and cell cycle arrest. The time phase property of NS1 activity during DNA replication and conformity to transient change of hemogram are suggestive of its role in regulating differentiation of hematopoietic cells, which is not completely understood. In this review, we summarized the bridge between B19 NS1 and Notch signaling pathway or transcriptional factors GATA, which play an important role in erythroid cell proliferation and differentiation, to provide a new insight of the potential mechanism of B19-induced differential inhibition of EPCs.


Subject(s)
Cell Differentiation , Erythroid Precursor Cells/physiology , Erythroid Precursor Cells/virology , Parvovirus B19, Human/pathogenicity , Viral Nonstructural Proteins/metabolism , Animals , DNA Replication , Female , Humans , Mice , Parvoviridae Infections/virology , Pregnancy , Signal Transduction , Viral Nonstructural Proteins/genetics , Virus Replication
7.
mBio ; 11(2)2020 03 10.
Article in English | MEDLINE | ID: mdl-32156816

ABSTRACT

During infection of human parvovirus B19 (B19V), one viral precursor mRNA (pre-mRNA) is transcribed by a single promoter and is alternatively spliced and alternatively polyadenylated. Here, we identified a novel cis-acting sequence (5'-GUA AAG CUA CGG GAC GGU-3'), intronic splicing enhancer 3 (ISE3), which lies 72 nucleotides upstream of the second splice acceptor (A2-2) site of the second intron that defines the exon of the mRNA encoding the 11-kDa viral nonstructural protein. RNA binding motif protein 45 (RBM45) specifically binds to ISE3 with high affinity (equilibrium dissociation constant [KD ] = 33 nM) mediated by its RNA recognition domain and 2-homo-oligomer assembly domain (RRM2-HOA). Knockdown of RBM45 expression or ectopic overexpression of RRM2-HOA in human erythroid progenitor cells (EPCs) expanded ex vivo significantly decreased the level of viral mRNA spliced at the A2-2 acceptor but not that of the mRNA spliced at A2-1 that encodes VP2. Moreover, silent mutations of ISE3 in an infectious DNA of B19V significantly reduced 11-kDa expression. Notably, RBM45 also specifically interacts in vitro with ISE2, which shares the octanucleotide (GGGACGGU) with ISE3. Taken together, our results suggest that RBM45, through binding to both ISE2 and ISE3, is an essential host factor for maturation of 11-kDa-encoding mRNA.IMPORTANCE Human parvovirus B19 (B19V) is a human pathogen that causes severe hematological disorders in immunocompromised individuals. B19V infection has a remarkable tropism with respect to human erythroid progenitor cells (EPCs) in human bone marrow and fetal liver. During B19V infection, only one viral precursor mRNA (pre-mRNA) is transcribed by a single promoter of the viral genome and is alternatively spliced and alternatively polyadenylated, a process which plays a key role in expression of viral proteins. Our studies revealed that a cellular RNA binding protein, RBM45, binds to two intron splicing enhancers and is essential for the maturation of the small nonstructural protein 11-kDa-encoding mRNA. The 11-kDa protein plays an important role not only in B19V infection-induced apoptosis but also in viral DNA replication. Thus, the identification of the RBM45 protein and its cognate binding site in B19V pre-mRNA provides a novel target for antiviral development to combat B19V infection-caused severe hematological disorders.


Subject(s)
Gene Expression Regulation, Viral , Introns , Nerve Tissue Proteins/metabolism , Parvovirus B19, Human/genetics , RNA Splicing , RNA-Binding Proteins/metabolism , Viral Nonstructural Proteins/genetics , Cells, Cultured , Erythroid Precursor Cells/virology , Genome, Viral , Hematopoietic Stem Cells , Humans , Nerve Tissue Proteins/genetics , Parvovirus B19, Human/metabolism , Protein Binding , RNA Precursors/genetics , RNA Precursors/metabolism , RNA-Binding Proteins/genetics , Viral Nonstructural Proteins/metabolism
8.
Viruses ; 11(9)2019 09 05.
Article in English | MEDLINE | ID: mdl-31491892

ABSTRACT

Piscine orthoreovirus (PRV-1) can cause heart and skeletal muscle inflammation (HSMI) in farmed Atlantic salmon (Salmo salar). The virus targets erythrocytes in the acute peak phase, followed by cardiomyocytes, before the infection subsides into persistence. The persistent phase is characterized by high level of viral RNA, but low level of viral protein. The origin and nature of persistent PRV-1 are not clear. Here, we analyzed for viral persistence and activity in various tissues and cell types in experimentally infected Atlantic salmon. Plasma contained PRV-1 genomic dsRNA throughout an 18-week long infection trial, indicating that viral particles are continuously produced and released. The highest level of PRV-1 RNA in the persistent phase was found in kidney. The level of PRV-1 ssRNA transcripts in kidney was significantly higher than that of blood cells in the persistent phase. In-situ hybridization assays confirmed that PRV-1 RNA was present in erythroid progenitor cells, erythrocytes, macrophages, melano-macrophages and in some additional un-characterized cells in kidney. These results show that PRV-1 establishes a productive, persistent infection in Atlantic salmon and that erythrocyte progenitor cells are PRV target cells.


Subject(s)
Erythroid Precursor Cells/virology , Fish Diseases/virology , Orthoreovirus/physiology , Reoviridae Infections/veterinary , Animals , Orthoreovirus/genetics , Orthoreovirus/growth & development , RNA, Viral/genetics , RNA, Viral/metabolism , Reoviridae Infections/virology , Salmo salar/virology , Viral Proteins/genetics , Viral Proteins/metabolism
9.
Antiviral Res ; 162: 22-29, 2019 02.
Article in English | MEDLINE | ID: mdl-30529090

ABSTRACT

Parvovirus B19 (B19V), a single-stranded DNA virus in the family Parvoviridae, is a human pathogenic virus responsible for a wide range of clinical manifestations. Currently there is no approved antiviral therapy for parvovirus infection. The acyclic nucleoside phosphonate cidofovir (CDV) has been demonstrated to inhibit replication of B19V in vitro. The aim of the present study was to evaluate whether brincidofovir (BCV), a novel lipid conjugate of CDV, could also inhibit B19V replication. Experiments were carried out in erythroid progenitor cells (EPCs) and UT7/EpoS1 cells, infected with B19V and cultured in the presence of different concentrations of BCV and CDV for comparison. The dynamics of viral replication was evaluated by a qPCR-based assay and the extent of inhibition of viral replication exerted by the compounds determined, along with the effect of the compounds on cell viability and cell proliferation rates. Results confirmed that BCV showed significantly higher antiviral activity against B19V compared to CDV in both cell-based systems. For BCV, the calculated EC50 values were in the range 6.6-14.3 µM in EPCs and 0.22-0.63 µM in UT7/EpoS1 cells. In comparison, the EC50 values for CDV were >300 µM in EPCs and 16.1 µM in UT7/EpoS1 cells. Concurrently, the effects on cell viability were observed at a much higher concentration of BCV, with calculated CC50 values in the range 93.4-102.9 µM in EPCs and 59.9-66.8 µM in UT7/Epos1. The antiviral activity was observed specifically with the metabolically active stereoisomer of BCV suggesting that CDV-diphosphate, the metabolite of both BCV and CDV, was the active antiviral. Our results support a selective role for BCV in the inhibition of B19 viral replication.


Subject(s)
Antiviral Agents/pharmacology , Cytosine/analogs & derivatives , Organophosphonates/pharmacology , Parvovirus B19, Human/drug effects , Cells, Cultured , Cytosine/pharmacology , Erythroid Precursor Cells/virology , Humans , Parvovirus B19, Human/physiology , Virus Replication/drug effects
10.
Expert Rev Mol Diagn ; 18(9): 821-832, 2018 09.
Article in English | MEDLINE | ID: mdl-30028234

ABSTRACT

INTRODUCTION: Parvovirus B19 (B19V), a single-stranded DNA virus in the family Parvoviridae, is a human pathogenic virus, characterized by a selective but not exclusive tropism for erythroid progenitor cells. Widely diffuse, it is responsible for an ample range of clinical manifestations, whose characteristics and outcomes depend on the interplay between the viral properties and the physiological and immune status of the infected individuals. The complexity of virus-host relationship and the diversity of the clinical course of infection pose a diagnostic challenge that may require non-trivial solutions. Areas covered: The review includes an updated description of the course of B19V infection in its complexity and diversity of pathogenetic mechanisms, discusses the consequent requirements for different and appropriated diagnostic approaches, presents the main diagnostic techniques, more recent technical advancements, and their application to the diverse clinical situations. Expert commentary: The complex scenario of the infectious process and the diversity in possible pathogenetic mechanisms make necessary a multi-parametric approach for an accurate and informative laboratory diagnosis of B19V infection, combining as much as possible the molecular detection of viral components, mainly viral DNA, to commonly followed immunological detection of virus-specific antibodies and a critical assessment of laboratory findings.


Subject(s)
Parvoviridae Infections/diagnosis , Parvoviridae Infections/virology , Parvovirus B19, Human/pathogenicity , DNA, Viral/genetics , Erythroid Precursor Cells/virology , Host-Pathogen Interactions/genetics , Humans , Parvovirus B19, Human/genetics
11.
Article in English | MEDLINE | ID: mdl-29922597

ABSTRACT

Parvovirus B19 (B19V) is pathogenic to humans and causes bone marrow failure diseases and various other inflammatory disorders. B19V infection exhibits high tropism for human erythroid progenitor cells (EPCs) in the bone marrow and fetal liver. The exclusive restriction of B19V replication to erythroid lineage cells is partly due to the expression of receptor and co-receptor(s) on the cell surface of human EPCs and partly depends on the intracellular factors essential for virus replication. We first summarize the latest developments in the viral entry process and the host cellular factors or pathways critical for B19V replication. We discuss the role of hypoxia, erythropoietin signaling and STAT5 activation in the virus replication. The B19V infection-induced DNA damage response (DDR) and cell cycle arrest at late S-phase are two key events that promote B19V replication. Lately, the virus infection causes G2 arrest, followed by the extensive cell death of EPCs that leads to anemia. We provide the current understanding of how B19V exploits the cellular resources and manipulate pathways for efficient virus replication. B19V encodes a single precursor mRNA (pre-mRNA), which undergoes alternate splicing and alternative polyadenylation to generate at least 12 different species of mRNA transcripts. The post-transcriptional processing of B19V pre-mRNA is tightly regulated through cis-acting elements and trans-acting factors flanking the splice donor or acceptor sites. Overall, in this review, we focus on the recent advances in the molecular virology and pathogenesis of B19V infection.


Subject(s)
Parvoviridae Infections/virology , Parvovirus B19, Human/physiology , Parvovirus B19, Human/pathogenicity , Viral Proteins/metabolism , Virus Replication/physiology , Cell Cycle Checkpoints , DNA Damage , Erythroid Precursor Cells/virology , Gene Expression Regulation, Viral , Humans , Parvovirus B19, Human/genetics , STAT5 Transcription Factor/metabolism , Viral Tropism , Virulence
12.
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 δ 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
13.
Nat Commun ; 8(1): 423, 2017 09 04.
Article in English | MEDLINE | ID: mdl-28871080

ABSTRACT

A short half-life in the circulation limits the application of therapeutics such as single-domain antibodies (VHHs). We utilize red blood cells to prolong the circulatory half-life of VHHs. Here we present VHHs against botulinum neurotoxin A (BoNT/A) on the surface of red blood cells by expressing chimeric proteins of VHHs with Glycophorin A or Kell. Mice whose red blood cells carry the chimeric proteins exhibit resistance to 10,000 times the lethal dose (LD50) of BoNT/A, and transfusion of these red blood cells into naive mice affords protection for up to 28 days. We further utilize an improved CD34+ culture system to engineer human red blood cells that express these chimeric proteins. Mice transfused with these red blood cells are resistant to highly lethal doses of BoNT/A. We demonstrate that engineered red blood cells expressing VHHs can provide prolonged prophylactic protection against bacterial toxins without inducing inhibitory immune responses and illustrates the potentially broad translatability of our strategy for therapeutic applications.The therapeutic use of single-chain antibodies (VHHs) is limited by their short half-life in the circulation. Here the authors engineer mouse and human red blood cells to express VHHs against botulinum neurotoxin A (BoNT/A) on their surface and show that an infusion of these cells into mice confers long lasting protection against a high dose of BoNT/A.


Subject(s)
Botulinum Toxins, Type A/toxicity , Erythrocytes/metabolism , Genetic Engineering , Single-Domain Antibodies/genetics , Animals , Antibodies, Neutralizing/administration & dosage , Antibodies, Neutralizing/genetics , Antibodies, Neutralizing/metabolism , Botulinum Toxins, Type A/metabolism , Botulism/etiology , Botulism/therapy , Erythrocyte Transfusion , Erythrocytes/virology , Erythroid Precursor Cells/metabolism , Erythroid Precursor Cells/transplantation , Erythroid Precursor Cells/virology , Genetic Vectors/genetics , Genetic Vectors/metabolism , Glycophorins/genetics , Glycophorins/metabolism , Humans , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Metalloendopeptidases/genetics , Metalloendopeptidases/metabolism , Mice , Mice, Inbred C57BL , Retroviridae/genetics , Retroviridae/metabolism , Single-Domain Antibodies/administration & dosage , Single-Domain Antibodies/metabolism
14.
Virology ; 511: 40-48, 2017 11.
Article in English | MEDLINE | ID: mdl-28806616

ABSTRACT

The pathogenic autonomous human parvovirus B19 (B19V) productively infects erythroid progenitor cells (EPCs). Functional similarities between B19V nonstructural protein (NS1), a DNA binding endonuclease, and the Rep proteins of Adeno-Associated Virus (AAV) led us to hypothesize that NS1 may facilitate targeted nicking of the human genome and B19 vDNA integration. We adapted an integration capture sequencing protocol (IC-Seq) to screen B19V infected human CD36+ EPCs for viral integrants, and discovered 40,000 unique B19V integration events distributed throughout the human genome. Computational analysis of integration patterns revealed strong correlations with gene intronic regions, H3K9me3 sites, and the identification of 41 base pair consensus sequence with an octanucleotide core motif. The octanucleotide core has homology to a single region of B19V, adjacent to the P6 promoter TATA box. We present the first direct evidence that B19V infection of erythroid progenitor cells disrupts the human genome and facilitates viral DNA integration.


Subject(s)
DNA-Binding Proteins/metabolism , DNA/metabolism , Endonucleases/metabolism , Erythroid Precursor Cells/virology , Parvovirus B19, Human/physiology , Viral Nonstructural Proteins/metabolism , Virus Integration , CD36 Antigens/analysis , Cells, Cultured , Erythroid Precursor Cells/chemistry , Humans
15.
PLoS Pathog ; 13(5): e1006370, 2017 May.
Article in English | MEDLINE | ID: mdl-28459842

ABSTRACT

Productive infection of human parvovirus B19 (B19V) exhibits high tropism for burst forming unit erythroid (BFU-E) and colony forming unit erythroid (CFU-E) progenitor cells in human bone marrow and fetal liver. This exclusive restriction of the virus replication to human erythroid progenitor cells is partly due to the intracellular factors that are essential for viral DNA replication, including erythropoietin signaling. Efficient B19V replication also requires hypoxic conditions, which upregulate the signal transducer and activator of transcription 5 (STAT5) pathway, and phosphorylated STAT5 is essential for virus replication. In this study, our results revealed direct involvement of STAT5 in B19V DNA replication. Consensus STAT5-binding elements were identified adjacent to the NS1-binding element within the minimal origins of viral DNA replication in the B19V genome. Phosphorylated STAT5 specifically interacted with viral DNA replication origins both in vivo and in vitro, and was actively recruited within the viral DNA replication centers. Notably, STAT5 interacted with minichromosome maintenance (MCM) complex, suggesting that STAT5 directly facilitates viral DNA replication by recruiting the helicase complex of the cellular DNA replication machinery to viral DNA replication centers. The FDA-approved drug pimozide dephosphorylates STAT5, and it inhibited B19V replication in ex vivo expanded human erythroid progenitors. Our results demonstrated that pimozide could be a promising antiviral drug for treatment of B19V-related diseases.


Subject(s)
DNA Replication , Minichromosome Maintenance Proteins/metabolism , Parvovirus B19, Human/genetics , STAT5 Transcription Factor/metabolism , Virus Replication , Erythroid Precursor Cells/virology , Erythropoietin/genetics , Erythropoietin/metabolism , Humans , Minichromosome Maintenance Proteins/genetics , Parvovirus B19, Human/physiology , Phosphorylation , STAT5 Transcription Factor/genetics , Signal Transduction
16.
Biochem Pharmacol ; 136: 32-39, 2017 07 15.
Article in English | MEDLINE | ID: mdl-28377277

ABSTRACT

Parvovirus B19 (B19V) infection is restricted to erythroid progenitor cells (EPCs) of the human bone marrow, leading to transient arrest of erythropoiesis and severe complications mainly in subjects with underlying hematological disorders or with immune system deficits. Currently, there are no specific antiviral drugs for B19V treatment, but identification of compounds inhibiting B19V replication can be pursued by a drug repositioning strategy. In this frame, the present study investigates the activity of hydroxyurea (HU), the only disease-modifying therapy approved for sickle cell disease (SCD), towards B19V replication in the two relevant cellular systems, the UT7/EpoS1 cell line and EPCs. Results demonstrate that HU inhibits B19V replication with EC50 values of 96.2µM and 147.1µM in UT7/EpoS1 and EPCs, respectively, providing experimental evidence of the antiviral activity of HU towards B19V replication, and confirming the efficacy of a drug discovery process by drug repositioning strategy. The antiviral activity occurs in vitro at concentrations lower than those affecting cellular DNA replication and viability, and at levels measured in plasma samples of SCD patients undergoing HU therapy. HU might determine a dual beneficial effect on SCD patients, not only for the treatment of the disease but also towards a virus responsible for severe complications.


Subject(s)
Erythroid Precursor Cells/drug effects , Erythroid Precursor Cells/physiology , Hydroxyurea/pharmacology , Parvovirus B19, Human/physiology , Virus Replication/drug effects , Virus Replication/physiology , Antiviral Agents/pharmacology , Cell Proliferation/drug effects , Cell Proliferation/physiology , Cells, Cultured , Dose-Response Relationship, Drug , Erythroid Precursor Cells/virology , Humans , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/physiology
17.
PLoS Pathog ; 13(3): e1006266, 2017 03.
Article in English | MEDLINE | ID: mdl-28264028

ABSTRACT

Human parvovirus B19 (B19V) infection of primary human erythroid progenitor cells (EPCs) arrests infected cells at both late S-phase and G2-phase, which contain 4N DNA. B19V infection induces a DNA damage response (DDR) that facilitates viral DNA replication but is dispensable for cell cycle arrest at G2-phase; however, a putative C-terminal transactivation domain (TAD2) within NS1 is responsible for G2-phase arrest. To fully understand the mechanism underlying B19V NS1-induced G2-phase arrest, we established two doxycycline-inducible B19V-permissive UT7/Epo-S1 cell lines that express NS1 or NS1mTAD2, and examined the function of the TAD2 domain during G2-phase arrest. The results confirm that the NS1 TAD2 domain plays a pivotal role in NS1-induced G2-phase arrest. Mechanistically, NS1 transactivated cellular gene expression through the TAD2 domain, which was itself responsible for ATR (ataxia-telangiectasia mutated and Rad3-related) activation. Activated ATR phosphorylated CDC25C at serine 216, which in turn inactivated the cyclin B/CDK1 complex without affecting nuclear import of the complex. Importantly, we found that the ATR-CHK1-CDC25C-CDK1 pathway was activated during B19V infection of EPCs, and that ATR activation played an important role in B19V infection-induced G2-phase arrest.


Subject(s)
G2 Phase Cell Cycle Checkpoints/physiology , Parvoviridae Infections/metabolism , Signal Transduction/physiology , Viral Nonstructural Proteins/metabolism , Ataxia Telangiectasia Mutated Proteins/metabolism , Blotting, Western , CDC2 Protein Kinase , Cell Line , Cyclin-Dependent Kinases/metabolism , Erythroid Precursor Cells/virology , Flow Cytometry , Humans , Immunoprecipitation , Oligonucleotide Array Sequence Analysis , Parvovirus B19, Human , cdc25 Phosphatases/metabolism
18.
Virus Res ; 220: 47-51, 2016 07 15.
Article in English | MEDLINE | ID: mdl-27071853

ABSTRACT

Human parvovirus B19 (B19V) commonly induces self-limiting infections but can also cause severe clinical manifestations in patients with underlying haematological disorders or with immune system deficits. Currently, therapeutic options for B19V entirely rely on symptomatic and supportive treatments since a specific antiviral therapy is not yet available. Recently a first step in the research for active compounds inhibiting B19V replication has allowed identifying the acyclic nucleoside phosphonate cidofovir (CDV). Herein, the effect of CDV against B19V replication was characterized in human erythroid progenitor cells (EPCs) cultured and infected following different experimental approaches to replicate in vitro the infection of an expanding erythroid cell population in the bone marrow. B19V replication was selectively inhibited both in infected EPCs extendedly exposed to CDV 500µM (viral inhibition 82%) and in serially infected EPCs cultures with passage of the virus progeny, constantly under drug exposure (viral inhibition 99%). In addition, a potent inhibitory effect against B19V (viral inhibition 92%) was assessed in a short-term infection of EPCs treated with CDV 500µM 1day before viral infection. In the evaluated experimental conditions, the enhanced effect of CDV against B19V might be ascribed both to the increased intracellular drug concentration achieved by extended exposure, and to a progressive reduction in efficiency of the replicative process within treated EPCs population.


Subject(s)
Antiviral Agents/pharmacology , Cytosine/analogs & derivatives , Organophosphonates/pharmacology , Parvovirus B19, Human/drug effects , Virus Replication/drug effects , Cidofovir , Cytosine/pharmacology , DNA, Viral/antagonists & inhibitors , DNA, Viral/biosynthesis , Erythroid Precursor Cells/drug effects , Erythroid Precursor Cells/virology , Humans , Parvovirus B19, Human/growth & development , Parvovirus B19, Human/metabolism , Primary Cell Culture
19.
PLoS One ; 11(2): e0148547, 2016.
Article in English | MEDLINE | ID: mdl-26845771

ABSTRACT

The pathogenic Parvovirus B19 (B19V) is characterized by a strict adaptation to erythroid progenitor cells (EPCs), a heterogeneous population of differentiating cells with diverse phenotypic and functional properties. In our work, we studied the dynamics of B19V infection in EPCs in dependence on the cell differentiation stage, in terms of distribution of infected cells, synthesis of viral nucleic acids and production of infectious virus. EPCs at early differentiation stage led to an abortive infection, without viral genome replication and a very low transcriptional activity. EPCs at later stages were permissive, with highest levels of viral replicative activity at day 9 (+3.0 Log from 2 to 48 hpi) and lower levels at day 18 (+1.5 Log from 2 to 48 hpi). B19V DNA increment was in accordance with the percentage of cells positive to flow-FISH assay (41.4% at day 9, 1.1% at day 18). Quantitation of total RNA indicated a close association of genome replication and transcription with viral RNA accumulation within infected cells related to viral DNA increase during the course of infection. Analysis of the different classes of mRNAs revealed two distinct pattern of genome expression profile with a fine regulation in the frequency utilization of RNA processing signals: an early phase, when cleavage at the proximal site leading to a higher relative production of mRNA for NS protein, and a late phase, when cleavage at the distal site was more frequent leading to higher relative abundance of mRNA for VP and 11 kDA proteins. Infectious virus was released from cells at day 6-15, but not at day 18. Our results, providing a detailed description of B19V replication and expression profile in differentiating EPCs, highlight the very tight adaptation of B19V to a specific cellular target defined both by its erythroid lineage and its differentiation stage.


Subject(s)
Erythroid Precursor Cells/virology , Gene Expression Regulation, Viral , Parvovirus B19, Human/physiology , Virus Replication , Biomarkers , Cell Differentiation , Erythroid Precursor Cells/cytology , Erythroid Precursor Cells/metabolism , Humans , Models, Biological , Phenotype , Transcription, Genetic , Virus Release
20.
Mol Biol Rep ; 43(3): 175-81, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26878856

ABSTRACT

Human Parvovirus B19 (PVB19) is one of the most important pathogens that targets erythroid lineage. Many factors were mentioned for restriction to erythroid progenitor cells (EPCs). Previous studies showed that in non-permissive cells VP1 and VP2 (structural proteins) mRNAs were detected but could not translate to proteins. A bioinformatics study showed that this inhibition might be due to specific microRNAs (miRNAs) present in non-permissive cells but not in permissive EPCs. To confirm the hypothesis, we evaluated the effect of miRNAs on VP expression. CD34(+) HSCs were separated from cord blood. Then, CD34(+) cells were treated with differentiation medium to obtain CD36(+) EPCs. To evaluate the effect of miRNAs on VP expression in MCF7 and HEK-293 cell lines (non-permissive cells) and CD36(+) EPCs, dual luciferase assay was performed in presence of shRNAs against Dicer and Drosha to disrupt miRNA biogenesis. QRT-PCR was performed to check down-regulation of Dicer and Drosha after transfection. All measurements were done in triplicate. Data means were compared using one-way ANOVAs. MicroRNA prediction was done by the online microRNA prediction tools. No significant difference was shown in luciferase activity of CD36(+) EPCs after co-transfection with shRNAs, while it was significant in non-permissive cells. Our study revealed that miRNAs may be involved in inhibition of VP expression in non-permissive cells, although further studies are required to demonstrate which miRNAs exactly are involved in regulation of PVB19 replication.


Subject(s)
Capsid Proteins/genetics , Erythroid Precursor Cells/metabolism , MicroRNAs , Parvoviridae Infections/metabolism , Parvovirus B19, Human/metabolism , Tropism , Computational Biology , Erythroid Precursor Cells/virology , Gene Expression Regulation , HEK293 Cells , Humans , MCF-7 Cells , Parvoviridae Infections/genetics , Parvovirus B19, Human/physiology , RNA, Messenger
SELECTION OF CITATIONS
SEARCH DETAIL
...