Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Gesundheitswesen ; 79(5): 388-393, 2017 May.
Article in German | MEDLINE | ID: mdl-26110244

ABSTRACT

Background: In recent years quality assurance has become an essential part of today's health-care system in the wake of the modern patient-oriented quality management. With the statutory introduction of newborn hearing screening (NHS) in 2009, a quality assurance of these early detection methods has become necessary. The aim of the study was to determine patient satisfaction in relation to the NHS in Saxony-Anhalt. Patients/Methods: During the period from November 2013 to April 2014, 394 parents were retrospectively interviewed about their experiences and expectations in relation to the NHS, using a standardised questionnaire. In total, 21 child care centres and 6 paediatric primary care centres from all over Saxony-Anhalt were involved. Results: It turns out that the majority of parents are satisfied with the NHS and 97.7% are in favour of the offer of an NHS. Of the surveyed parents, 69.3% felt the information as sufficient. However, only 66.2% of parents took a closer look at the leaflet issued by the G-BA. In addition, 17.7% of respondents are dissatisfied with the professional competence of the examining staff. Conclusion: The study shows that the general attitude among parents towards newborn hearing screening was very positive. They felt reassured by it although there are some aspects still open to criticism.


Subject(s)
Hearing Tests/psychology , Hearing Tests/statistics & numerical data , Mass Screening/statistics & numerical data , Neonatal Screening/statistics & numerical data , Parents/psychology , Patient Compliance/statistics & numerical data , Patient Satisfaction/statistics & numerical data , Adult , Attitude to Health , Clinical Competence/statistics & numerical data , Germany/epidemiology , Health Care Surveys , Humans , Infant, Newborn , Mass Screening/psychology , National Health Programs/statistics & numerical data , Neonatal Screening/psychology , Refusal to Participate , Young Adult
2.
Gene Ther ; 22(6): 458-66, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25786873

ABSTRACT

Immunosuppressed (IS) patients, such as recipients of hematopoietic stem cell transplantation, occasionally develop severe and fatal adenovirus (Ad) infections. Here, we analyzed the potential of a virus receptor trap based on a soluble coxsackievirus and Ad receptor (sCAR) for inhibition of Ad infection. In vitro, a dimeric fusion protein, sCAR-Fc, consisting of the extracellular domain of CAR and the Fc portion of human IgG1 and a monomeric sCAR lacking the Fc domain, were expressed in cell culture. More sCAR was secreted into the cell culture supernatant than sCAR-Fc, but it had lower Ad neutralization activity than sCAR-Fc. Further investigations showed that sCAR-Fc reduced the Ad infection by a 100-fold and Ad-induced cytotoxicity by ~20-fold. Not only was Ad infection inhibited by sCAR-Fc applied prior to infection, it also inhibited infection when used to treat ongoing Ad infection. In vivo, sCAR-Fc was delivered to IS mice by an AAV9 vector, resulting in persistent and high (>40 µg ml(-1)) sCAR-Fc serum levels. The sCAR-Fc serum concentration was sufficient to significantly inhibit hepatic and cardiac wild-type Ad5 infection. Treatment with sCAR-Fc did not induce side effects. Thus, sCAR-Fc virus receptor trap may be a promising novel therapeutic for treatment of Ad infections.


Subject(s)
Adenoviridae Infections/therapy , Adenoviridae/metabolism , Enterovirus/metabolism , Genetic Therapy , Genetic Vectors , Receptors, Virus/metabolism , Adenoviridae/genetics , Animals , Carrier Proteins/genetics , Cell Line , Dependovirus/metabolism , Enterovirus/genetics , Humans , Liver/metabolism , Liver/pathology , Mice , Myocardium/metabolism , Myocardium/pathology , Receptors, Virus/genetics , Recombinant Proteins/genetics , Recombinant Proteins/therapeutic use
3.
J Mol Cell Cardiol ; 52(5): 1056-65, 2012 May.
Article in English | MEDLINE | ID: mdl-22326437

ABSTRACT

Tissue Factor (TF) is expressed in various cell types of the heart, such as cardiomyocytes. In addition to its role in the initiation of blood coagulation, the TF:FVIIa complex protects cells from apoptosis. There are two isoforms of Tissue Factor (TF): "full length" (fl)TF--an integral membrane protein, and alternatively spliced (as)TF--a protein that lacks a transmembrane domain and can thus be secreted in a soluble form. Whether asTF or flTF affects apoptosis of cardiomyocytes is unknown. In this study, we examined whether asTF or flTF protects murine cardiomyocytes from TNF-α-induced apoptosis. We used murine cardiomyocytic HL-1 cells and primary murine embryonic cardiomyocytes that overexpressed either murine asTF or murine flTF, and stimulated them with TNF-α to initiate cell death. Apoptosis was assessed by annexin-V assay, propidium iodide assay, as well as activation of caspase-3 and -9. In addition, signaling via integrins, Akt, NFκB and Erk1/2, and gene-expression of Bcl-2 family members were analyzed. We here report that overexpression of asTF reduced phosphatidylserine exposure upon TNF-α-stimulation. asTF overexpression led to an increased expression and phosphorylation of Akt, as well as up-regulation of the anti-apoptotic protein Bcl-x(L). The anti-apoptotic effects of asTF overexpression were mediated via α(V)ß(3)/Akt/NFκB signaling and were dependent on Bcl-x(L) expression in HL-1 cells. The anti-apoptotic activity of asTF was also observed using primary cardiomyocytes. Analogous yet less pronounced anti-apoptotic sequelae were observed due to overexpression of flTF. Importantly, cardiomyocytes deficient in TF exhibited increased apoptosis compared to wild type cells. We propose that asTF and flTF protect cardiomyocytes against TNF-α-induced apoptosis via activation of specific signaling pathways, and up-regulation of anti-apoptotic members of the Bcl-2 protein family.


Subject(s)
Apoptosis , Myocytes, Cardiac/physiology , Thromboplastin/physiology , Tumor Necrosis Factor-alpha/physiology , Animals , Cell Line , Gene Expression , MAP Kinase Signaling System , Mice , Myocytes, Cardiac/metabolism , NF-kappa B/metabolism , Phosphorylation , Primary Cell Culture , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Isoforms/physiology , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Thromboplastin/genetics , Thromboplastin/metabolism , Up-Regulation , bcl-X Protein/genetics , bcl-X Protein/metabolism
4.
J Virol ; 85(8): 3881-92, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21248045

ABSTRACT

The interaction of the rubella virus (RV) capsid (C) protein and the mitochondrial p32 protein is believed to participate in virus replication. In this study, the physiological significance of the association of RV with mitochondria was investigated by silencing p32 through RNA interference. It was demonstrated that downregulation of p32 interferes with microtubule-directed redistribution of mitochondria in RV-infected cells. However, the association of the viral C protein with mitochondria was not affected. When cell lines either pretreated with respiratory chain inhibitors or cultivated under (mild) hypoxic conditions were infected with RV, viral replication was reduced in a time-dependent fashion. Additionally, RV infection induces increased activity of mitochondrial electron transport chain complex III, which was associated with an increase in the mitochondrial membrane potential. These effects are outstanding among the examples of mitochondrial alterations caused by viruses. In contrast to the preferential localization of p32 to the mitochondrial matrix in most cell lines, RV-permissive cell lines were characterized by an almost exclusive membrane association of p32. Conceivably, this contributes to p32 function(s) during RV replication. The data presented suggest that p32 fulfills an essential function for RV replication in directing trafficking of mitochondria near sites of viral replication to meet the energy demands of the virus.


Subject(s)
Host-Pathogen Interactions , Microtubules/metabolism , Mitochondria/metabolism , Mitochondria/virology , Mitochondrial Proteins/metabolism , Rubella virus/pathogenicity , Viral Core Proteins/metabolism , Animals , Carrier Proteins , Cell Line , Electron Transport , Gene Silencing , Humans , Membrane Potential, Mitochondrial , Mitochondrial Proteins/antagonists & inhibitors , RNA Interference
5.
Gene Ther ; 18(2): 199-209, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21048795

ABSTRACT

Adeno-associated virus (AAV) vectors with capsids of AAV serotype 9 enable an efficient transduction of the heart upon intravenous injection of adult mice but also transduce the liver. The aim of this study was to improve specificity of AAV9 vector-mediated cardiac gene transfer by microRNA (miR)-dependent control of transgene expression. We constructed plasmids and AAV vectors containing target sites (TSs) of liver-specific miR122, miR192 and miR148a in the 3' untranslated region (3'UTR) of a luciferase expression cassette. Luciferase expression was efficiently suppressed in liver cell lines expressing high levels of the corresponding miRs, whereas luciferase expression was unaffected in cardiac myocytes. Intravenous injections of AAV9 vectors bearing three repeats of miR122 TS in the 3'UTR of an enhanced green fluorescent expression (EGFP) expression cassette resulted in the absence of EGFP expression in the liver of adult mice, whereas the control vectors without miR TS displayed significant hepatic EGFP expression. EGFP expression levels in the heart, however, were comparable between miR122-regulated and control vectors. The liver-specific de-targeting in vivo using miR122 was even more efficient than transcriptional targeting with a cardiac cytomegalovirus (CMV)-enhanced myosin light chain (MLC) promoter. These data indicate that miR-regulated targeting is a powerful new tool to further improve cardiospecificity of AAV9 vectors.


Subject(s)
Dependovirus/genetics , Gene Expression Regulation , Gene Transfer Techniques , Genetic Vectors , Heart , MicroRNAs/pharmacology , Animals , Injections, Intravenous , Liver , Mice , Organ Specificity , Transgenes , Untranslated Regions
6.
Curr Pharm Des ; 16(20): 2252-68, 2010.
Article in English | MEDLINE | ID: mdl-20459390

ABSTRACT

Understanding of the roles of RNAs within the cell has changed and expanded dramatically during the past few years. Based on fundamentally new insights it is now increasingly possible to employ RNAs as highly valuable tools in molecular biology and medicine. At present, the most important therapeutic strategies are based on non-coding regulatory RNAs inducing RNA interference (RNAi) to silence single genes, and on modulation of cellular microRNAs (miRNAs) to alter complex gene expression patterns in diseased organs. Only recently it became possible to target therapeutic RNAi to specific organs via organotropic viral vector systems and we discuss the most recent strategies in this field, e.g. heart failure treatment by cardiac-targeted RNAi. Due to the peculiar biochemical properties of small RNA molecules, true therapeutic translation of results in vitro is more demanding than with small molecule drugs or proteins. Specifically, there is a critical requirement for extensive studies in animal models of human disease after pre-testing of the RNAi tools in vitro. This requirement likewise applies for miRNA modulations which have complex consequences in the recipient dependent on biochemical stability and distribution of the therapeutic RNA. Problems not yet fully solved are the prediction of targets and specificity of the RNA tools. However, major progress has been made to achieve their tissue-specific and regulatable expression, and breakthroughs in vector technologies from the gene therapy field have fundamentally improved safety and efficacy of RNA-based therapeutic approaches, too. In summary, insight into the molecular mechanisms of action of regulatory RNAs in combination with new delivery tools for RNA therapeutics will significantly expand our cardiovascular therapeutic repertoire beyond classical pharmacology.


Subject(s)
Cardiovascular Diseases/therapy , Genetic Therapy/methods , MicroRNAs/therapeutic use , Pharmacogenetics/methods , RNA, Small Interfering/therapeutic use , Animals , Cardiovascular Diseases/genetics , Drug Design , Gene Expression Regulation , Humans
7.
Gene Ther ; 14(18): 1319-29, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17611587

ABSTRACT

Recently it was shown that several new pseudotyped adeno-associated virus (AAV) vectors support cardioselective expression of transgenes. The molecular mechanisms underlying this propensity for cardiac cell transduction are not well understood. We comparatively analyzed AAV vector attachment, internalization, intracellular trafficking, and nuclear uncoating of recombinant self-complementary (sc) AAV2.2 versus pseudotyped scAAV2.6 vectors expressing green fluorescence protein (GFP) in cells of cardiac origin. In cardiac-derived HL-1 cells and primary neonatal rat cardiomyocytes (PNCMs), expression of GFP increased rapidly after incubation with scAAV2.6-GFP, but remained low after scAAV2.2-GFP. Internalization of scAAV2.6-GFP was more efficient than that of scAAV2.2-GFP. Nuclear translocation was similarly efficient for both, but differential nuclear uncoating rates emerged as a key additional determinant of transduction: 30% of all scAAV2.6-GFP genomes translocated to the nucleus became uncoated within 48 h, but only 16% of scAAV2.2-GFP genomes. In contrast to this situation in cells of cardiac origin, scAAV2.2-GFP displayed more efficient internalization and similar (tumor cell line HeLa) or higher (human microvascular endothelial cell (HMEC)) uncoating rates than scAAV.2.6-GFP in non-cardiac cell types. In summary, both internalization and nuclear uncoating are key determinants of cardiac transduction by scAAV2.6 vectors. Any in vitro screening for the AAV pseudotype most suitable for cardiac gene therapy - which is desirable since it may allow significant reductions in vector load in upcoming clinical trials--needs to quantitate both key steps in transduction.


Subject(s)
Dependovirus/genetics , Genetic Therapy/methods , Genetic Vectors/pharmacokinetics , Myocytes, Cardiac/virology , Transduction, Genetic/methods , Animals , Biological Transport , Cardiovascular Diseases/therapy , Cell Line , Cell Line, Tumor , Cell Nucleus/virology , Endothelial Cells/metabolism , Endothelial Cells/virology , Female , Gene Expression , Genetic Vectors/genetics , Green Fluorescent Proteins/genetics , HeLa Cells , Humans , Rats , Transgenes , Virus Attachment , Virus Integration , Virus Internalization
8.
Gene Ther ; 14(12): 960-71, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17377597

ABSTRACT

As coxsackievirus B3 (CoxB3) and adenoviruses may cause acute myocarditis and inflammatory cardiomyopathy, isolation of the common coxsackievirus-adenovirus-receptor (CAR) has provided an interesting new target for molecular antiviral therapy. Whereas many viruses show high mutation rates enabling them to develop escape mutants, mutations of their cellular virus receptors are far less likely. We report on antiviral efficacies of CAR gene silencing by short hairpin (sh)RNAs in the cardiac-derived HL-1 cell line and in primary neonatal rat cardiomyocytes (PNCMs). Treatment with shRNA vectors mediating RNA interference against the CAR resulted in almost complete silencing of receptor expression both in HL-1 cells and PNCMs. Whereas CAR was silenced in HL-1 cells as early as 24 h after vector treatment, its downregulation in PNCMs did not become significant before day 6. CAR knockout resulted in inhibition of CoxB3 infections by up to 97% in HL-1 cells and up to 90% in PNCMs. Adenovirus was inhibited by only 75% in HL-1 cells, but up to 92% in PNCMs. We conclude that CAR knockout by shRNA vectors is efficient against CoxB3 and adenovirus in primary cardiac cells, but the efficacy of this approach in vivo may be influenced by cell type-specific silencing kinetics in different tissues.


Subject(s)
Adenoviridae Infections/therapy , Coxsackievirus Infections/therapy , Genetic Therapy/methods , Myocarditis/therapy , RNA Interference , Receptors, Virus/genetics , Adenoviridae , Animals , Cell Line , Cells, Cultured , Coxsackie and Adenovirus Receptor-Like Membrane Protein , Enterovirus B, Human , Gene Silencing , Genetic Engineering , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Myocarditis/virology , Myocytes, Cardiac/virology , RNA, Small Interfering/administration & dosage , Rats , Virus Replication/genetics
9.
Gene Ther ; 14(3): 211-8, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17024101

ABSTRACT

Impaired function of the phospholamban (PLB)-regulated sarcoplasmic reticulum Ca(2+) pump (SERCA2a) contributes to cardiac dysfunction in heart failure (HF). PLB downregulation may increase SERCA2a activity and improve cardiac function. Small interfering (si)RNAs mediate efficient gene silencing by RNA interference (RNAi). However, their use for in vivo gene therapy is limited by siRNA instability in plasma and tissues, and by low siRNA transfer rates into target cells. To address these problems, we developed an adenoviral vector (AdV) transcribing short hairpin (sh)RNAs against rat PLB and evaluated its potential to silence the PLB gene and to modulate SERCA2a-mediated Ca(2+) sequestration in primary neonatal rat cardiomyocytes (PNCMs). Over a period of 13 days, vector transduction resulted in stable > 99.9% ablation of PLB-mRNA at a multiplicity of infection of 100. PLB protein gradually decreased until day 7 (7+/-2% left), whereas SERCA, Na(+)/Ca(2+) exchanger (NCX1), calsequestrin and troponin I protein remained unchanged. PLB silencing was associated with a marked increase in ATP-dependent oxalate-supported Ca(2+) uptake at 0.34 microM of free Ca(2+), and rapid loss of responsiveness to protein kinase A-dependent stimulation of Ca(2+) uptake was maintained until day 7. In summary, these results indicate that AdV-derived PLB-shRNA mediates highly efficient, specific and stable PLB gene silencing and modulation of active Ca(2+) sequestration in PNCMs. The availability of the new vector now enables employment of RNAi for the treatment of HF in vivo.


Subject(s)
Calcium-Binding Proteins/genetics , Calcium/metabolism , Genetic Therapy/methods , Heart Failure/therapy , Myocytes, Cardiac/metabolism , RNA Interference , Animals , Blotting, Western/methods , COS Cells , Cells, Cultured , Chlorocebus aethiops , Genetic Engineering , Green Fluorescent Proteins/genetics , Heart Failure/metabolism , Homeostasis , RNA, Small Interfering/administration & dosage , Rats , Sarcoplasmic Reticulum/metabolism , Transfection/methods
10.
J Mol Med (Berl) ; 85(3): 257-71, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17106732

ABSTRACT

The clinical phenotype of human dilated cardiomyopathy (DCM) encompasses a broad spectrum of etiologically distinct disorders. As targeting of etiology-related pathogenic pathways may be more efficient than current standard heart failure treatment, we obtained the genomic expression profile of a DCM subtype characterized by cardiac inflammation to identify possible new therapeutic targets in humans. In this inflammatory cardiomyopathy (DCMi), a distinctive cardiac expression pattern not described in any previous study of cardiac disorders was observed. Two significantly altered gene networks of particular interest and possible interdependence centered around the cysteine-rich angiogenic inducer 61 (CYR61) and adiponectin (APN) gene. CYR61 overexpression, as in human DCMi hearts in situ, was similarly induced by inflammatory cytokines in vascular endothelial cells in vitro. APN was strongly downregulated in DCMi hearts and completely abolished cytokine-dependent CYR61 induction in vitro. Dysbalance between the CYR61 and APN networks may play a pathogenic role in DCMi and contain novel therapeutic targets. Multiple immune cell-associated genes were also deregulated (e.g., chemokine ligand 14, interleukin-17D, nuclear factors of activated T cells). In contrast to previous investigations in patients with advanced or end-stage DCM where etiology-related pathomechanisms are overwhelmed by unspecific processes, the deregulations detected in this study occurred at a far less severe and most probably fully reversible disease stage.


Subject(s)
Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/therapy , Gene Expression Profiling , Genome, Human/genetics , Adiponectin/genetics , Adiponectin/metabolism , Adult , Aged , Cysteine-Rich Protein 61 , Cytokines/pharmacology , Gene Expression Regulation/drug effects , Gene Regulatory Networks , Humans , Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Middle Aged , Models, Biological , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism
11.
J Mol Med (Berl) ; 84(10): 842-51, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16924471

ABSTRACT

Coxsackie adenovirus receptor (CAR) is involved in immunological processes, and its soluble isoforms have antiviral effects on coxsackievirus B3 (CVB3) infection in vitro. We explored in this study the impact of CAR4/7, a soluble CAR isoform, on CVB3-induced myocarditis in BALB/c mice. BALB/c mice were treated daily with recombinant CAR4/7, beta-galactosidase (beta-Gal; as control protein) or buffer for 9 days. Half of each group was infected with CVB3 on day 3, and all mice were killed on day 9. Myocardial CVB3 titer, histology, and serology were analyzed. Treatment with CAR4/7 led to a significant reduction of myocardial CVB3 titer, whereas the application of beta-Gal had no detectable effect on the myocardial virus load. CAR4/7 application, however, resulted in increased myocardial inflammation and tissue damage in CVB3-infected hearts, whereas beta-Gal caused a degree of cardiac inflammation and injury similar to that in buffer-treated CVB3-infected control animals. CAR4/7 and beta-Gal treatment induced the production of antibodies against the respective antigens. CAR4/7-, but not beta-Gal-specific, virus-negative sera reacted against myocardial tissue and cellular membranous CAR, and significantly inhibited CVB3 infection in vitro. Thus, CAR4/7 suppressed CVB3 infection in vivo, supporting the concept of receptor analog in antiviral therapy. However, CAR4/7 treatment also leads to an aggravation of myocardial inflammation and injury most likely secondary to an autoimmune process.


Subject(s)
Coxsackievirus Infections/drug therapy , Enterovirus B, Human/drug effects , Receptors, Virus/therapeutic use , Animals , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Cell Survival/drug effects , Coxsackie and Adenovirus Receptor-Like Membrane Protein , Coxsackievirus Infections/pathology , Coxsackievirus Infections/virology , Creatine Kinase/blood , Enterovirus B, Human/growth & development , Enzyme-Linked Immunosorbent Assay , HeLa Cells , Humans , Immune Sera/pharmacology , Immunohistochemistry , Injections, Intraperitoneal , Male , Mice , Mice, Inbred BALB C , Myocarditis/chemically induced , Myocarditis/pathology , Myocarditis/virology , Random Allocation , Receptors, Virus/genetics , Receptors, Virus/immunology , Recombinant Proteins/administration & dosage , Recombinant Proteins/adverse effects , Recombinant Proteins/therapeutic use , Serum Amyloid A Protein/analysis , Solubility
12.
Gene Ther ; 13(2): 173-86, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16136163

ABSTRACT

Pharmacological control is a desirable safety feature of oncolytic adenoviruses (oAdV). It has recently been shown that oAdV replication may be controlled by drug-dependent transcriptional regulation of E1A expression. Here, we present a novel concept that relies on tamoxifen-dependent regulation of E1A activity through functional linkage to the mutated hormone-binding domain of the murine estrogen receptor (Mer). Four different E1A-Mer chimeras (ME, EM, E(DeltaNLS)M, MEM) were constructed and inserted into the adenoviral genome under control of a lung-specific surfactant protein B promoter. The highest degree of regulation in vitro was seen for the corresponding oAdVs Ad.E(DeltaNLS)M and Ad.MEM, which exhibited an up to 100-fold higher oAdV replication in the presence as compared with the absence of 4-OH-tamoxifen. Moreover, destruction of nontarget cells was six- and 13-fold reduced for Ad.E(DeltaNLS)M and Ad.MEM, respectively, as compared with Ad.E. Further investigations supported tamoxifen-dependent regulation of Ad.E(DeltaNLS)M and Ad.MEM in vivo. Induction of Ad.E(DeltaNLS)M inhibited growth of H441 lung tumors as efficient as a control oAdV expressing E1A. E(DeltaNLS)M and the MEM chimeras can be easily inserted into a single vector genome, which extends their application to existing oAdVs and strongly facilitates in vivo application.


Subject(s)
Adenocarcinoma/therapy , Adenovirus E1A Proteins/genetics , Genetic Therapy/methods , Lung Neoplasms/therapy , Receptors, Estrogen/genetics , Tamoxifen/therapeutic use , Adenocarcinoma/virology , Animals , Cytopathogenic Effect, Viral , Female , Gene Expression Regulation , Genetic Engineering , HeLa Cells , Humans , Lung Neoplasms/virology , Mice , Mice, Nude , Recombinant Proteins/therapeutic use , Tamoxifen/analogs & derivatives , Transcription, Genetic , Tumor Cells, Cultured , Virus Replication/drug effects
13.
Article in English | MEDLINE | ID: mdl-16329668

ABSTRACT

Dilated cardiomyopathy (DCM) is a prevalent heart muscle disease characterized by impaired contractility and dilation of the ventricles. Recent clinical research suggests that cardiotropic viruses are important environmental pathogenic factors in human DCM, which may therefore be considered as a chronic viral cardiomyopathy. All virus-positive DCM patients thus come into the focus of virological research and should be considered for antiviral strategies. Interferon-beta therapy has been shown to mediate virus elimination in patients with adenovirus or coxsackievirus persistence. We discuss here several possible new molecular targets for patients infected with cardiotropic viruses in (1) the cellular virus uptake system, (2) virus-induced cellular signaling pathways, and (3) interactions between virus-encoded proteins with important cellular target proteins. The potential of these approaches in the setting of a chronic viral infection is significantly different from that in an acute viral infection. Specific problems encountered in a chronic situation and possible solutions are discussed.


Subject(s)
Antiviral Agents/therapeutic use , Cardiomyopathy, Dilated , Virus Diseases/therapy , Animals , Cardiomyopathy, Dilated/pathology , Cardiomyopathy, Dilated/therapy , Cardiomyopathy, Dilated/virology , Chronic Disease , Endocytosis , Heart/virology , Humans , Immunity, Innate/physiology , RNA Interference , Signal Transduction/physiology , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Diseases/pathology , Viruses/genetics , Viruses/metabolism
14.
J Mol Med (Berl) ; 83(8): 579-86, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15931504

ABSTRACT

Dilated cardiomyopathy (DCM) is a heart muscle disease characterized by impaired contractility and dilation of the ventricles. In a subset of DCM patients, classical inheritance patterns occur (familial DCM), which have led to the identification of specific genomic loci and gene defects causing monogenic DCM subtypes. In the majority of DCM patients, however, there is no evidence for a monogenic etiology of the disorder (sporadic DCM), and in the absence of other recognizable etiological factors, these cases were classified as "idiopathic". Recent research suggests that cardiotropic viruses are important environmental factors in the pathogenesis of "idiopathic" cases and that DCM commonly results from interactions between genetic and environmental factors, whereas "pure" genetic forms are rather rare. Regarding genetics, the clinical cardiomyopathic phenotype associated with single gene defects may be highly variable for unknown reasons. Furthermore, a novel class of genetic defects was identified recently which provide a molecular basis for abnormal reactions of cardiomyocytes to environmental stress. These defects are paradigms of specific molecular links between genome and environment during the pathogenesis of DCM. Regarding environmental factors, a recent molecular virological study based on myocardial biopsies in a large series of sporadic DCM patients has detected cardiac viral infections in the majority of patients, with a broad spectrum of virus species being involved. Apparently, DCM does not only occur as a late sequela of acute viral myocarditis, but also in patients without clinical history of cardiac viral disease. Cardiotropic viruses thus emerge as prevalent environmental factors which may cause or influence the course of DCM in a large fraction of cases. Synopsis of current data suggests that a comprehensive picture of DCM pathogenesis can only be drawn if both genetic and environmental pathogenetic factors are considered. The course of cardiac viral infections depends strongly on genetic host factors and may range from rapid and complete virus elimination or silencing without clinical symptoms, to rapidly progressive or fatal disease. Viruses interact not only with genetically heterogenous host systems of virus uptake, migration, and antiviral immunity, but, due to their prevalence in DCM hearts, are also likely to encounter multiple structural proteins of cardiac cells known to be defective in familial DCM. The combined knowledge on DCM-associated gene defects and viruses therefore suggests in-depth studies on genome-environment interactions in DCM pathogenesis which may underlie the high clinical variability observed both in monogenic and virus-associated DCM and have implications for the clinical management of DCM patients.


Subject(s)
Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/virology , Genome, Viral , Disease Progression , Humans
15.
Z Kardiol ; 93(3): 171-93, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15024585

ABSTRACT

During the past few years major conceptual and technical advances have been made towards the therapeutic modulation of cardiac gene expression for the treatment of cardiac diseases. Among these are 1) the identification of new molecular therapy targets in cardiac disorders, often derived from genetic animal models. 2) A better understanding of the molecular and cellular determinants of cardiac gene transfer in vivo, in animal models and in first clinical trials. 3) The development of novel regulatable and long-term stable vector systems. This review is focused on nucleic acid-based modulation of cardiac calcium homeostasis as a paradigm for the new gene therapeutic approaches, since recent landmark papers have suggested this to be a molecular target of key importance in heart failure. In particular, the development of severe heart failure in the genetic MLP(-/-) animal model could be completely abolished by the targeted ablation of phospholamban (PL), a key regulator of cardiac calcium homeostasis. This impressive effect of permanent germline PL ablation provides-in conjunction with former important work on disturbed calcium handling in the failing human heart-a rationale for the gene therapeutic approach of ad hoc suppression of PL by antisense strategies (antisense RNAs, ribozymes, RNA interference) or PL variants. Based on the broad spectrum of methods employed to characterize this general strategy, PL-targeted approaches may be considered as a paradigm of future genetic treatments of cardiac disorders, although the differences between animal models and humans must be kept in mind. High safety of any such therapy will be a prerequisite for any possible clinical application and therefore novel methods to improve control are being devised: 1) The regulation of gene therapy vectors by biochemical abnormalities associated with the target disease itself (" Induction-by-Disease" gene therapy). 2) External control of vector activity by the employment of drug-sensitive promotors. In addition, the important goal of cardiac long-term stability of the therapeutic vectors has recently been achieved in animal models using vectors derived from adeno-associated viruses (AAVs).


Subject(s)
Gene Expression/physiology , Gene Transfer Techniques , Genetic Therapy/methods , Heart Diseases/genetics , Heart Diseases/therapy , Nucleic Acids/genetics , Animals , Calcium/metabolism , Calcium-Binding Proteins/genetics , Disease Models, Animal , Genetic Vectors/genetics , Heart Failure/genetics , Heart Failure/therapy , Humans , Mutagenesis, Site-Directed/genetics , Oligonucleotides, Antisense , RNA Interference/physiology
16.
Gene Ther ; 10(19): 1680-90, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12923567

ABSTRACT

The use of restricted replication-competent adenoviruses (RRCAs) inducing tumor cell-specific lysis is a promising approach in cancer gene therapy. However, the use of RRCAs in humans carries considerable risk, since after injection into the patient, further regulation or inhibition of virus replication from the outside is impossible. Therefore, we have developed a novel system allowing external pharmacological control of RRCA replication. We show here that a tumor-selective E1B-deleted RRCA can be tightly regulated by use of doxycycline (dox)-controlled adenoviral E1A gene expression, which in turn determines vector replication. RRCA replication is switched on by addition and switched off by withdrawal of dox. The system results in efficient tumor cell killing after induction by dox, whereas cells are unaffected by the uninduced system. It was also employed for efficient external control of transgene expression from cotransfected replication-deficient adenovectors. Furthermore, the use of a liver cell-specific human alpha1-antitrypsin (hAAT)-promoter driving a tetracycline-controlled transcriptional silencer allowed specific protection of cells with hAAT-promoter activity in the absence of dox in vitro and in vivo, delineating a new principle of 'tissue protective' gene therapy. The concept of external control of RRCAs may help to improve the safety of cancer gene therapy.


Subject(s)
Adenoviridae/genetics , Doxycycline/pharmacology , Genetic Therapy/methods , Neoplasms/therapy , Protein Synthesis Inhibitors/pharmacology , Virus Replication/genetics , Adenovirus E1A Proteins/metabolism , Adenovirus E1B Proteins/genetics , Animals , Gene Deletion , Humans , Mice , Mice, Nude , Tumor Cells, Cultured , Virus Replication/drug effects
17.
Z Kardiol ; 91(12): 978-91, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12490988

ABSTRACT

Coxsackieviruses and adenoviruses are common agents of viral heart disease. In the majority of exposed individuals they do not cause myocardial disease, however, since they are not primarily cardiotropic. Until recently the molecular basis of their anomalous tropism in patients who develop viral heart disease was unknown. An important step towards clarification of the molecular basis of cardiotropic viral infections was achieved in 1997, when a common receptor for the two structurally unrelated viruses was cloned. This coxsackievirus-adenovirus receptor (CAR) is a key determinant for the cellular uptake of both viruses and for the molecular pathogenesis of coxsackievirus and adenovirus diseases. We have mapped the CAR expression in human hearts and observed highly variable expression patterns. Healthy donor hearts had low CAR expression levels, whereas explanted hearts of patients with dilated cardiomyopathy (DCM) displayed high CAR expression in the myocardium. Remarkably, however, heart failure per se was not associated with CAR induction, since in heart failure of non-DCM origin no induction was found. Additional studies on the molecular mechanisms of CAR induction in cardiomyocytes indicated the existence of a cell-cell contact-dependent molecular mechanism regulating CAR expression, whereas cellular virus uptake and low level replication had no effect. Recombinant expression of human CAR in cardiomyocytes strongly increased their virus uptake rate suggesting that CAR induction enhances cardiac vulnerability to viral disease, whereas healthy myocardium is rather resistant to CAR-dependent viruses. Receptor induction may significantly aggravate the clinical course of viral heart disease, so that the blockade of receptor expression or receptor-virus interactions opens new therapeutic perspectives. Elucidation of the molecular mechanism of CAR induction in DCM, but not in heart failure per se, may reveal a particular pathogenetic process in this disease. A broader analysis of the cardiovascular expression patterns of receptors for other potentially cardiotropic viruses (CMV, EBV, HIV, HHV-6, Parvo-B19, etc.) should lead to a better understanding of individual risk factors for viral heart diseases and of their highly variable clinical courses, and offer new therapeutic options.


Subject(s)
Cardiomyopathy, Dilated/virology , Heart Diseases/etiology , Heart/virology , Receptors, Virus , Virus Diseases/complications , Adenoviridae , Adenovirus Infections, Human/complications , Animals , Cardiomyopathy, Dilated/metabolism , Coxsackie and Adenovirus Receptor-Like Membrane Protein , Coxsackievirus Infections/complications , Disease Susceptibility , Enterovirus , Gene Expression Regulation, Viral , Genetic Therapy , Heart Diseases/metabolism , Humans , Immunohistochemistry , Myocarditis/etiology , Myocytes, Cardiac/metabolism , Rats , Receptors, Virus/genetics , Receptors, Virus/metabolism , Risk Factors
18.
Circulation ; 104(3): 275-80, 2001 Jul 17.
Article in English | MEDLINE | ID: mdl-11457744

ABSTRACT

BACKGROUND: The coxsackievirus and adenovirus receptor (CAR) was identified as a common cellular receptor for both viruses, but its biological and pathogenic relevance is uncertain. Knowledge of CAR localization in the human cardiovascular system is limited but important with respect to CAR-dependent viral infections and gene transfer using CAR-dependent viral vectors. METHODS AND RESULTS: Explanted failing hearts from 13 patients (8 with dilated cardiomyopathy [DCM] and 5 with other heart diseases [non-DCM]) and normal donor hearts (n=7) were investigated for the expression levels and subcellular localization of CAR and the adenovirus coreceptors alpha(v)beta(3) and alpha(v)beta(5) integrins. CAR immunoreactivity was very low in normal and non-DCM hearts, whereas strong CAR signals occurred at the intercalated discs and sarcolemma in 5 of the 8 DCM hearts (62.5%); these strong signals colocalized with both integrins. In all hearts, CAR was detectable in subendothelial layers of the vessel wall, but not on the luminal endothelial surface, and on interstitial cells. Human CAR (hCAR) expressed in rat cardiomyocytes was targeted to cell-cell contacts, which resembled CAR localization in DCM hearts and resulted in 15-fold increased adenovirus uptake. CONCLUSIONS: Low hCAR abundance may render normal human myocardium resistant to CAR-dependent viruses, whereas re-expression of hCAR, such as that observed in DCM, may be a key determinant of cardiac susceptibility to viral infections. Asymmetric expression of hCAR in the vessel wall may be an important determinant of adenovirus tropism in humans. hCAR subcellular localization in human myocardium and hCAR targeting to cell-cell contacts in cardiomyocyte cultures suggest that hCAR may play a role in cell-cell contact formation.


Subject(s)
Cardiomyopathy, Dilated/metabolism , Integrins/metabolism , Myocardium/metabolism , Receptors, Virus/metabolism , Receptors, Vitronectin/metabolism , Adenoviridae/metabolism , Adenoviridae Infections/metabolism , Adult , Aged , Animals , Animals, Newborn , Cardiomyopathy, Dilated/pathology , Cells, Cultured , Coxsackie and Adenovirus Receptor-Like Membrane Protein , Female , Gene Expression , Humans , Immunohistochemistry , Male , Middle Aged , Myocardium/cytology , Myocardium/pathology , Rats , Receptors, Virus/genetics , Sarcolemma/metabolism , Transfection , Up-Regulation
19.
J Lipid Res ; 41(12): 2009-16, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11108734

ABSTRACT

The scavenger receptor class B type I (SR-BI) mediates the selective uptake of cholesterol and cholesteryl ester (CE) from high density lipoprotein (HDL) into cells. The high expression in liver and steroidogenic tissues is compatible with a role of SR-BI in reverse cholesterol transport and steroid hormone synthesis. Ways of regulation thus far described include induction by trophic hormones via cAMP-activated protein kinase A (PKA) and the effects of cellular and plasma cholesterol. Here we show that vitamin E (vitE) has a major effect on the expression of SR-BI in rat liver and in a human hepatoma-derived cell line, HepG2. Feeding rats a vitE-depleted diet resulted in an 11-fold increase in the SR-BI protein level in liver tissue. This effect was readily reversed by feeding a vitE-enriched chow. In HepG2 cells, the expression of the human SR-BI homolog was reduced when the vitE content was increased by incubating the cells with vitE-loaded HDL or with phosphatidylcholine/vitE vesicles. The downregulation of human SR-BI (hSR-BI) was accompanied by a reduced level of protein kinase C (PKC) in the particulate cell fraction, and PKC inhibition decreased the expression of hSR-BI and the uptake of vitE and cholesterol from HDL. Our results are consistent with the view that the cellular level of vitE exerts a tight control over the expression of SR-BI. Furthermore, the inhibitory effect of vitE on PKC seems to be involved in the signaling pathway.


Subject(s)
CD36 Antigens/metabolism , Liver/drug effects , Membrane Proteins , Receptors, Immunologic , Receptors, Lipoprotein , Vitamin E/pharmacology , Animals , Down-Regulation , Humans , Lipoproteins, HDL/metabolism , Liver/metabolism , Male , Protein Kinase C/metabolism , Rats , Rats, Wistar , Receptors, Scavenger , Scavenger Receptors, Class B , Signal Transduction , Tumor Cells, Cultured , Vitamin E Deficiency/metabolism
20.
Article in English | MEDLINE | ID: mdl-10900827

ABSTRACT

Bovine leukaemia virus (BLV) is an oncogenic retrovirus that causes B-cell lymphocytosis and in the terminal stage of the disease lymphosarcoma. The comparison of the previously published BLV provirus sequence from Belgium, Australia and Japan showed that the protease gene (prt) of the Australian and the Japanese isolate contain a nucleotide deletion when compared to the Belgian isolate. Because all these proviruses were isolated from tumour tissue, the prt gene of functionally active and infectious proviruses from peripheral blood leucocytes (PBLs) of BLV-infected cattle and from BLV-infected fetal lamb kidney cells were sequenced. The only variations between these sequences and the Belgian isolate consist of nucleotide substitutions. The delection of one nucleotide of the prt gene of the Japanese and the Australian BLV tumour isolate caused a changed reading frame and a premature translational stop. It was shown that the Japanese provirus is non-infectious in transfected cell culture and in injected sheep. To analyse the impact of the prt mutation on viral protein expression and infectivity, the prt region of the Japanese provirus was exchanged with the prt region from the Belgian provirus. The resulting pBLVprtbelg was infectious in transfected cells and enabled the expression of gag and gag-precursor proteins. One sheep was injected with this mutated provirus and became positive in BLV-PCR, but no seroconversion was developed. The prt mutation of the Japanese tumour isolates was shown to be responsible for the loss of infectivity and changed viral expression. These results and the occurrence of this mutation in only two isolates from lymphosarcoma indicate a possible relation between the prt mutation and the induction of cell transformation.


Subject(s)
Enzootic Bovine Leukosis/virology , Gene Deletion , Gene Expression Regulation, Viral , Leukemia Virus, Bovine/genetics , Viral Proteins/metabolism , Amino Acid Sequence , Animals , Base Sequence , Cattle , DNA Primers , DNA, Viral/isolation & purification , Endopeptidases/genetics , Molecular Sequence Data , Polymerase Chain Reaction/veterinary , Protein Biosynthesis , Reading Frames/genetics , Sequence Alignment/veterinary , Sheep , Viral Proteins/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...