Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
J Infect Dis ; 212(7): 1070-81, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-25805750

ABSTRACT

Human parvovirus B19 (B19V) is a common pathogen in microvascular disease and cardiomyopathy, owing to infection of endothelial cells. B19V replication, however, is almost restricted to erythroid progenitor cells (ErPCs). Endothelial regeneration attributable to bone marrow-derived circulating angiogenic cells (CACs) is a prerequisite for organ function. Because of many similarities of ErPCs and CACs, we hypothesized that B19V is a perpetrator of impaired endogenous endothelial regeneration. B19V DNA and messenger RNA from endomyocardial biopsy specimens, bone marrow specimens, and circulating progenitor cells were quantified by polymerase chain reaction analysis. The highest B19V DNA concentrations were found in CD34(+)KDR(+) cells from 17 patients with chronic B19V-associated cardiomyopathy. B19V replication intermediates could be detected in nearly half of the patients. Furthermore, chronic B19V infection was associated with impaired endothelial regenerative capacity. B19V infection of CACs in vitro resulted in expression of transcripts encoding B19V proteins. The capsid protein VP1 was identified as a novel inducer of apoptosis, as were nonstructural proteins. Inhibition studies identified so-called death receptor signaling with activation of caspase-8 and caspase-10 to be responsible for apoptosis induction. B19V causally impaired endothelial regeneration with spreading of B19V in CACs in an animal model in vivo. We thus conclude that B19V infection and damage to CACs result in dysfunctional endogenous vascular repair, supporting the emergence of primary bone marrow disease with secondary end-organ damage.


Subject(s)
Apoptosis , Cardiomyopathies/complications , Erythema Infectiosum/virology , Erythroid Precursor Cells/virology , Parvovirus B19, Human/physiology , Adult , Aged , Animals , Capsid Proteins/genetics , Capsid Proteins/metabolism , Case-Control Studies , Caspase 10/genetics , Caspase 10/metabolism , Cell Line , Endothelial Cells/physiology , Endothelial Cells/virology , Erythroid Precursor Cells/physiology , Female , Humans , Male , Mice , Middle Aged , Parvovirus B19, Human/genetics , Regeneration , Signal Transduction , Virus Replication
2.
J Virol ; 88(14): 8102-15, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24807719

ABSTRACT

Despite its strong host tropism for erythroid progenitor cells, human parvovirus B19 (B19V) can also infect a variety of additional cell types. Acute and chronic inflammatory cardiomyopathies have been associated with a high prevalence of B19V DNA in endothelial cells of the myocardium. To elucidate the mechanisms of B19V uptake into endothelium, we first analyzed the surface expression of the well-characterized primary B19V receptor P antigen and the putative coreceptors α5ß1 integrins and Ku80 antigen on primary and permanent endothelial cells. The receptor expression pattern and also the primary attachment levels were similar to those in the UT7/Epo-S1 cell line regarded as functional for B19V entry, but internalization of the virus was strongly reduced. As an alternative B19V uptake mechanism in endothelial cells, we demonstrated antibody-dependent enhancement (ADE), with up to a 4,000-fold increase in B19V uptake in the presence of B19V-specific human antibodies. ADE was mediated almost exclusively at the level of virus internalization, with efficient B19V translocation to the nucleus. In contrast to monocytes, where ADE of B19V has been described previously, enhancement does not rely on interaction of the virus-antibody complexes with Fc receptors (FcRs), but rather, involves an alternative mechanism mediated by the heat-sensitive complement factor C1q and its receptor, CD93. Our results suggest that ADE represents the predominant mechanism of endothelial B19V infection, and it is tempting to speculate that it may play a role in the pathogenicity of cardiac B19V infection. Importance: Both efficient entry and productive infection of human parvovirus B19 (B19V) seem to be limited to erythroid progenitor cells. However, in vivo, the viral DNA can also be detected in additional cell types, such as endothelial cells of the myocardium, where its presence has been associated with acute and chronic inflammatory cardiomyopathies. In this study, we demonstrated that uptake of B19V into endothelial cells most probably does not rely on the classical receptor-mediated route via the primary B19V receptor P antigen and coreceptors, such as α5ß1 integrins, but rather on antibody-dependent mechanisms. Since the strong antibody-dependent enhancement (ADE) of B19V entry requires the CD93 surface protein, it very likely involves bridging of the B19V-antibody complexes to this receptor by the complement factor C1q, leading to enhanced endocytosis of the virus.


Subject(s)
Antibodies, Viral/metabolism , Antibody-Dependent Enhancement , Endothelial Cells/virology , Membrane Glycoproteins/metabolism , Parvovirus B19, Human/physiology , Receptors, Complement/metabolism , Receptors, Virus/metabolism , Virus Internalization , Cell Line , Healthy Volunteers , Humans , Parvovirus B19, Human/immunology
3.
J Virol ; 86(9): 5099-109, 2012 May.
Article in English | MEDLINE | ID: mdl-22357277

ABSTRACT

Despite its very narrow tropism for erythroid progenitor cells, human parvovirus B19 (B19V) has recently been shown to replicate and form infectious progeny virus in 293 cells in the presence of early adenoviral functions provided either by infection with adenovirus type 5 or by addition of the pHelper plasmid encoding the E2a, E4orf6, and VA RNA functions. In the present study we dissected the individual influence of these functions on B19V genome replication and expression of structural proteins VP1 and VP2. We show that, in the presence of the constitutively expressed E1A and E1B, E4orf6 alone is able to promote B19V DNA replication, resulting in a concomitant increase in VP expression levels. The stimulatory effects of E4orf6 require the integrity of the BC box motifs, which target cellular proteins such as p53 and the Mre11 DNA repair complex for proteosomal degradation through formation of an E3 ubiquitin ligase complex with E1B. VA RNA also strongly induces VP expression but, in contrast to E4orf6, in a replication-independent manner. This stimulation could be attributed exclusively to the VA I RNA transcript and does not involve major activating effects at the level of the B19V p6 promoter, but the nucleotide residues required for the well-defined pathway of VA I RNA mediated stimulation of translation through functional inactivation of protein kinase R. These data show that the cellular pathways regulating B19V replication may be very similar to those governing the productive cycle of the helper-dependent parvoviruses, the adeno-associated viruses.


Subject(s)
Adenoviridae/metabolism , Adenovirus E4 Proteins/metabolism , Capsid Proteins/genetics , DNA Replication , Gene Expression Regulation, Viral , Parvovirus B19, Human/genetics , RNA, Viral/metabolism , Adenoviridae/genetics , Cell Line , Cullin Proteins/metabolism , DNA, Viral/biosynthesis , Humans , Multiprotein Complexes/metabolism , Parvovirus B19, Human/metabolism , Parvovirus B19, Human/physiology , Protein Binding , Ubiquitin-Protein Ligases/metabolism , eIF-2 Kinase/metabolism
4.
Antivir Ther ; 21(7): 559-566, 2016.
Article in English | MEDLINE | ID: mdl-27032991

ABSTRACT

Adenovirus (Ad) infections are usually mild and self-limiting, but severe systemic infections and fatal diseases can occur, especially in immunosuppressed patients. Anti-adenoviral pharmacotherapy has been proven to inhibit Ad infection, but its efficiency is limited. This review addresses biological antiviral agents as a new class of therapeutics for treatment of Ad infections. One group of agents is composed of short double-stranded RNA molecules that have been developed to inhibit Ad receptor and Ad protein expression. The second group of agents includes soluble virus receptor traps which inhibit Ad uptake into cells. Anti-Ad-adoptive T-cell therapy constitutes a third approach. We also outline how the combination of biological antiviral agents and combinations of these agents with the classical antiviral drugs can increase therapeutic efficiency in anti-adenoviral treatments.


Subject(s)
Adenoviridae Infections/therapy , Antiviral Agents/therapeutic use , Biological Products/therapeutic use , Animals , Coxsackie and Adenovirus Receptor-Like Membrane Protein/physiology , Humans , Immunotherapy, Adoptive , RNA, Double-Stranded/therapeutic use
5.
Antiviral Res ; 120: 72-8, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26026665

ABSTRACT

Adenoviruses (Ad) generally induce mild self-limiting respiratory or intestinal infections but can also cause serious disease with fatal outcomes in immunosuppressed patients. Antiviral drug therapy is an important treatment for adenoviral infections but its efficiency is limited. Recently, we have shown that gene silencing by RNA interference (RNAi) is a promising new approach to inhibit adenoviral infection. In the present in vitro study, we examined whether the efficiency of an RNAi-based anti-adenoviral therapy can be further increased by combination with a virus receptor trap sCAR-Fc and with the antiviral drug cidofovir. Initially, three siRNAs, siE1A_4, siIVa2_2 and Pol-si2, targeting the adenoviral E1A, IVa2 and DNA polymerase mRNAs, respectively, were used for gene silencing. Replication of the Ad was inhibited in a dose dependent manner by each siRNA, but the efficiency of inhibition differed (Pol-si2>siIVa2_2>siE1A_4). Double or triple combinations of the siRNAs compared with single siRNAs did not result in a measurably higher suppression of Ad replication. Combination of the siRNAs (alone or mixes of two or three siRNAs) with sCAR-Fc markedly increased the suppression of adenoviral replication compared to the same siRNA treatment without sCAR-Fc. Moreover, the triple combination of a mix of all three siRNAs, sCAR-Fc and cidofovir was about 23-fold more efficient than the combination of siRNAs mix/sCAR-Fc and about 95-fold more efficient than the siRNA mix alone. These data demonstrate that co-treatment of cells with sCAR-Fc and cidofovir is suitable to increase the efficiency of anti-adenoviral siRNAs.


Subject(s)
Adenoviridae/drug effects , Antiviral Agents/metabolism , Cytosine/analogs & derivatives , Organophosphonates/metabolism , RNA, Small Interfering/metabolism , Receptors, Virus/metabolism , Virus Internalization/drug effects , Virus Replication/drug effects , Adenoviridae/physiology , Cidofovir , Cytosine/metabolism , DNA, Viral/analysis , Drug Synergism , HeLa Cells , Humans
6.
Virus Res ; 176(1-2): 155-60, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23770153

ABSTRACT

Human parvovirus B19 (B19V) has been considered to cause acute and chronic myocarditis, which is accompanied by endothelial dysfunction. Currently, no causative treatment option for B19V-infections is available. Since RNA interference (RNAi) has proven to be a highly potent antiviral approach, the aim of the current study was to develop an RNAi-based strategy to inhibit B19V replication. Three B19V-VP2-specific short hairpin RNAs (shRNAs) were designed and tested for their silencing activity in reporter assays and the expression cassette of the best one was introduced into an adenoviral shuttle vector (Ad5). B19V-permissive UT7/Epo-S1 cells were infected with B19V and the RNAi triggers were delivered by the adenoviral vector (Ad5shVP2) 24h thereafter. The shRNA targeting the B19V-VP2 gene significantly suppressed VP2 mRNA levels as determined by quantitative RT-PCR. Additionally, also the expression levels of the non-targeted non-structural B19V-NS1 mRNA were strongly reduced. Our results demonstrate that vector-mediated delivery of shRNA expression cassettes targeting the structural B19-VP2 gene is a suitable approach to inhibit B19V replication.


Subject(s)
Adenoviruses, Human/genetics , Genetic Vectors , Parvovirus B19, Human/physiology , RNA Interference , Virus Replication , Antiviral Agents/metabolism , Cell Line , Gene Expression Profiling , Humans , Parvovirus B19, Human/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Real-Time Polymerase Chain Reaction
7.
Virology ; 411(1): 50-64, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21236463

ABSTRACT

Human parvovirus B19 (B19V) DNA is highly prevalent in endothelial cells lining up intramyocardial arterioles and postcapillary venules of patients with chronic myocarditis and cardiomyopathies. We addressed the question of a possible stimulation of B19V gene expression in endothelial cells by infection with adenoviruses. Adenovirus infection led to a strong augmentation of B19V structural and nonstructural proteins in individual endothelial cells infected with B19V or transfected with an infectious B19V genome. Transactivation was mostly mediated at the level of transcription and not due to adenovirus-mediated induction of second-strand synthesis from the single-stranded parvoviral genome. The main adenoviral functions required were E1A and E4orf6, which displayed synergistic effects. Furthermore, a limited B19V genome replication could be demonstrated in endothelial cells and adenovirus infection induced the appearance of putative dimeric replication intermediates. Thus the almost complete block in B19V gene expression seen in endothelial cells can be abrogated by infection with other viruses.


Subject(s)
Adenoviruses, Human/genetics , Endothelial Cells/virology , Gene Expression Regulation, Viral , Parvovirus B19, Human/physiology , Trans-Activators/metabolism , Transcriptional Activation , Viral Proteins/metabolism , Adenovirus E1A Proteins/metabolism , Adenovirus E4 Proteins/metabolism , Cells, Cultured , Humans , Parvovirus B19, Human/genetics , Transcription, Genetic
8.
Antiviral Res ; 88(1): 86-94, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20708037

ABSTRACT

Adenoviruses are pathological agents inducing mild respiratory and gastrointestinal infections. Under certain circumstances, for example in immunosuppressed patients, they induce severe infections of the liver, heart and lung, sometimes leading to death. Currently, adenoviral infections are treated by palliative care with no curative antiviral therapy yet available. Gene silencing by RNA interference (RNAi) has been shown to be a potent new therapeutic option for antiviral therapy. In the present study, we examined the potential of RNAi-mediated inhibition of adenovirus 5 infection by the use of small interfering (si)RNAs targeting both early (E1A) and late (hexon, IVa2) adenoviral genes. Several of the initially analyzed siRNAs directed against E1A, hexon and IVa2 showed a distinct antiviral activity. Among them, one siRNA for each gene was selected and used for the further comparative investigations of their efficiency to silence adenoviruses. Silencing of the late genes was more efficient in inhibiting adenoviral replication than comparable silencing of the E1A early gene. A combination strategy involving down-regulation of any two or all three of the targeted genes did not result in an enhanced inhibition of viral replication as compared to the single siRNA approaches targeting the late genes. However, protection against adenovirus-mediated cytotoxicity was substantially improved by combining siRNAs against either of the two late genes with the siRNA against the E1A early gene. Thus, an enhanced anti-adenoviral efficiency of RNAi-based inhibition strategies can be achieved by co-silencing of early and late adenoviral genes, with down regulation of the E1A as a crucial factor.


Subject(s)
Adenoviridae/genetics , Adenovirus E1A Proteins/genetics , RNA Interference , RNA, Small Interfering/genetics , Viral Proteins/genetics , Adenoviridae/physiology , Adenovirus Infections, Human/therapy , Adenovirus Infections, Human/virology , Adenoviruses, Human/genetics , Adenoviruses, Human/physiology , Antiviral Agents , Cells, Cultured , Down-Regulation , Fluorescent Antibody Technique, Indirect , Genes, Viral , HeLa Cells , Humans , Luciferases , Polymerase Chain Reaction , RNA, Double-Stranded/drug effects , RNA, Viral/genetics , Transcription, Genetic , Viral Plaque Assay , Virus Replication
9.
Vet Microbiol ; 137(3-4): 375-9, 2009 Jun 12.
Article in English | MEDLINE | ID: mdl-19282113

ABSTRACT

Sera and selected tissue homogenates collected from gnotobiotic swine never exposed to the environment or other swine tissues were tested for the presence of porcine torque teno virus (TTV) DNAs by nested and non-nested polymerase chain reactions (PCR) using primers specific for the untranslated region of porcine genogroups (g) 1 and 2. Twenty-three of 105 (21.9%) gnotobiotic piglets were g1- and/or g2-TTV DNA positive. Twenty-three of 27 (85.2%) sow sera, collected at the time of Caesarian derivation of the litters contained either or both TTV genogroup DNAs. These data demonstrate that porcine TTV may be transmitted to piglets by the in utero route and that the incidence of fetal infection is high.


Subject(s)
DNA Virus Infections/veterinary , Infectious Disease Transmission, Vertical/veterinary , Swine Diseases/transmission , Torque teno virus , Animals , DNA Virus Infections/transmission , DNA Virus Infections/virology , Female , Germ-Free Life , Pregnancy , Swine , Swine Diseases/virology
SELECTION OF CITATIONS
SEARCH DETAIL