Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 77
Filter
2.
J Clin Virol ; 63: 1-5, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25600595

ABSTRACT

BACKGROUND: Human herpesvirus 6 (HHV-6) A and B are lymphotropic viruses with life-long persistence, primarily associated with non-cardiac diseases, and discussed as a possible etiologic factor of myocarditis and cardiomyopathy. OBJECTIVE: To analyze the long-term spontaneous course of cardiac patients suffering from suspected inflammatory cardiomyopathy (CMi) with persisting HHV-6 A and B infections by follow-up biopsies. STUDY DESIGN: We prospectively evaluated patients (n=73) with biopsy-proven viral HHV-6 A and B infection in endomyocardial biopsies (EMBs), followed up by reanalysis of EMBs and left ventricular ejection fraction (LV-EF) measurements after a median period of 8.8 months (range 4-73 months). Beyond, we studied HHV-6 prevalence in isolated peripheral blood cells (PBCs) and HHV-6 species in EMBs. HHV-6 species-specific cellular infection sites within the myocardium were identified by immunohistochemistry (IHC). RESULTS: We identified 73 patients with cardiac HHV-6 A and B persistence or newly detected in follow-up EMB (95.0% B). Proof of HHV-6 in PBCs was primarily associated with A. Persistence of cardiac HHV-6 B genome was significantly associated with cardiac dysfunction at follow-up (LV-EF deteriorated from 58.2±16.0 to 51.8±17.2%, p<0.001), and LV improvement was observed when HHV-6 B persistence resolved (LV-EF increased from 54.9±15.4 to 60.7±13.1%, p<0.001). CONCLUSIONS: Persistence of cardiac HHV-6 B genomes was significantly associated with cardiac dysfunction, and hemodynamic parameters improved in association with HHV-6 B clearance.


Subject(s)
Biopsy , Cardiomyopathies/pathology , Cardiomyopathies/virology , Heart/virology , Herpesvirus 6, Human/isolation & purification , Roseolovirus Infections/complications , Ventricular Dysfunction, Left/physiopathology , Adult , Aged , Blood/virology , Female , Herpesvirus 6, Human/classification , Humans , Immunohistochemistry , Male , Middle Aged , Myocardium/pathology , Prospective Studies , Roseolovirus Infections/virology
4.
Basic Res Cardiol ; 109(3): 408, 2014 May.
Article in English | MEDLINE | ID: mdl-24691762

ABSTRACT

Adiponectin (APN) is an immunomodulatory adipocytokine that improves outcome in patients with virus-negative inflammatory cardiomyopathy and mice with autoimmune myocarditis. Here, we investigated whether APN modulates cardiac inflammation and injury in coxsackievirus B3 (CVB3) myocarditis. Myocarditis was induced by CVB3 infection of APN-KO and WT mice. APN reconstitution was performed by adenoviral gene transfer. Expression analyses were performed by qRT-PCR and immunoblot. Cardiac histology was analyzed by H&E-stain and immunohistochemistry. APN-KO mice exhibited diminished subacute myocarditis with reduced viral load, attenuated inflammatory infiltrates determined by NKp46, F4/80 and CD3/CD4/CD8 expression and reduced IFNß, IFNγ, TNFα, IL-1ß and IL-12 levels. Moreover, myocardial injury assessed by necrotic lesions and troponin I release was attenuated resulting in preserved left ventricular function. Those changes were reversed by APN reconstitution. APN had no influence on adhesion, uptake or replication of CVB3 in cardiac myocytes. In acute CVB3 myocarditis, cardiac viral load did not differ between APN-KO and WT mice. However, APN-KO mice displayed an enhanced acute immune response, i.e. increased expression of myocardial CD14, IFNß, IFNγ, IL-12, and TNFα resulting in increased cardiac infiltration with pro-inflammatory M1 macrophages and activated NK cells. Up-regulation of cardiac CD14 expression, type I and II IFNs and inflammatory cell accumulation in APN-KO mice was inhibited by APN reconstitution. Our observations indicate that APN promotes CVB3 myocarditis by suppression of toll-like receptor-dependent innate immune responses, polarization of anti-inflammatory M2 macrophages and reduction of number and activation of NK cells resulting in attenuated acute anti-viral immune responses.


Subject(s)
Adiponectin/metabolism , Coxsackievirus Infections/metabolism , Enterovirus B, Human/immunology , Myocarditis/metabolism , Myocardium/metabolism , Adiponectin/deficiency , Adiponectin/genetics , Animals , Animals, Newborn , Cells, Cultured , Coxsackievirus Infections/genetics , Coxsackievirus Infections/immunology , Coxsackievirus Infections/pathology , Coxsackievirus Infections/physiopathology , Coxsackievirus Infections/virology , Disease Models, Animal , Enterovirus B, Human/genetics , Enterovirus B, Human/pathogenicity , Immunity, Innate , Inflammation Mediators/metabolism , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Killer Cells, Natural/virology , Macrophages/immunology , Macrophages/metabolism , Macrophages/virology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocarditis/genetics , Myocarditis/immunology , Myocarditis/pathology , Myocarditis/physiopathology , Myocarditis/virology , Myocardium/immunology , Myocardium/pathology , Necrosis , Rats , Rats, Wistar , Toll-Like Receptors/metabolism , Ventricular Function, Left , Viral Load
5.
Eur Heart J ; 35(32): 2186-95, 2014 Aug 21.
Article in English | MEDLINE | ID: mdl-24667923

ABSTRACT

AIMS: Improvement of clinical diagnostics of idiopathic giant cell myocarditis (IGCM) and cardiac sarcoidosis (CS), two frequently fatal human myocardial diseases. Currently, IGCM and CS are diagnosed based on differential patterns of inflammatory cell infiltration and non-caseating granulomas in histological sections of endomyocardial biopsies (EMBs), after heart explantation or postmortem. We report on a method for improved differential diagnosis by myocardial gene expression profiling in EMBs. METHODS AND RESULTS: We examined gene expression profiles in EMBs from 10 patients with histopathologically proven IGCM, 10 with CS, 18 with active myocarditis (MCA), and 80 inflammation-free control subjects by quantitative RT-QPCR. We identified distinct differential profiles that allowed a clear discrimination of tissues harbouring giant cells (IGCS, CS) from those with MCA or inflammation-free controls. The expression levels of genes coding for cytokines or chemokines (CCL20, IFNB1, IL6, IL17D; P < 0.05), cellular receptors (ADIPOR2, CCR5, CCR6, TLR4, TLR8; P < 0.05), and proteins involved in the mitochondrial energy metabolism (CPT1, CYB, DHODH; P < 0.05) were deregulated in 2- to 300-fold, respectively. Bioinformatic analyses and correlation of the gene expression data with immunohistochemical findings provided novel information regarding the differential cellular and molecular pathomechanisms in IGCM, CS, and MCA. CONCLUSION: Myocardial gene expression profiling is a reliable method to predict the presence of multinuclear giant cells in the myocardium, even without a direct histological proof, in single small EMB sections, and thus to reduce the risk of sampling errors. This profiling also facilitates the discrimination between IGCM and CS, as two different clinical entities that require immediate and tailored differential therapy.


Subject(s)
Cardiomyopathies/diagnosis , Gene Expression Profiling/methods , Sarcoidosis/diagnosis , DNA, Complementary/metabolism , Female , Humans , Immunohistochemistry , Male , Middle Aged , Myocarditis/diagnosis , Retrospective Studies
6.
Basic Res Cardiol ; 108(5): 372, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23934091

ABSTRACT

Recent studies have detected erythrovirus genomes in the hearts of cardiomyopathy and cardiac transplant patients. Assessment of the functional status of viruses may provide clinically important information beyond detection of the viral genomes. Here, we report transcriptional activation of cardiotropic erythrovirus to be associated with strongly altered myocardial gene expression in a distinct subgroup of cardiomyopathy patients. Endomyocardial biopsies (EMBs) from 415 consecutive cardiac erythrovirus (B19V)-positive patients with clinically suspected cardiomyopathy were screened for virus-encoded VP1/VP2 mRNA indicating transcriptional activation of the virus, and correlated with cardiac host gene expression patterns in transcriptionally active versus latent infections, and in virus-free control hearts. Transcriptional activity was detected in baseline biopsies of only 66/415 patients (15.9 %) harbouring erythrovirus. At the molecular level, significant differences between cardiac B19V-positive patients with transcriptionally active versus latent virus were revealed by expression profiling of EMBs. Importantly, latent B19V infection was indistinguishable from controls. Genes involved encode proteins of antiviral immune response, B19V receptor complex, and mitochondrial energy metabolism. Thus, functional mapping of erythrovirus allows definition of a subgroup of B19V-infected cardiomyopathy patients characterized by virus-encoded VP1/VP2 transcripts and anomalous host myocardial transcriptomes. Cardiac B19V reactivation from latency, as reported here for the first time, is a key factor required for erythrovirus to induce altered cardiac gene expression in a subgroup of cardiomyopathy patients. Virus genome detection is insufficient to assess pathogenic potential, but additional transcriptional mapping should be incorporated into future pathogenetic and therapeutic studies both in cardiology and transplantation medicine.


Subject(s)
Cardiomyopathies/genetics , Cardiomyopathies/virology , Parvoviridae Infections/virology , Transcriptome , Cardiomyopathies/complications , DNA, Viral/analysis , Female , Humans , Male , Middle Aged , Parvoviridae Infections/complications , Parvoviridae Infections/genetics , Parvovirus B19, Human/genetics , Reverse Transcriptase Polymerase Chain Reaction
7.
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
8.
Amino Acids ; 43(3): 1399-403, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22160263

ABSTRACT

Light microscopic evidence suggested a synaptic role for agmatinase, an enzyme capable of inactivating the putative neurotransmitter and endogenous anti-depressant agmatine. Using electron microscopy and an alternative pre-embedding approach referred to as virtual pre-embedding, agmatinase was localised pre- and postsynaptically, to dendritic spines, spine and non-spine terminals, and dendritic profiles. In dendritic spines, labelling displayed a tendency towards the postsynaptic density. These results further strengthen a synaptic role for agmatine and strongly suggest a regulatory role for synaptically expressed agmatinase.


Subject(s)
Dendritic Spines/enzymology , Parietal Lobe/enzymology , Plastic Embedding , Synapses/enzymology , Ureohydrolases/metabolism , Animals , Dendritic Spines/ultrastructure , Fluorescent Dyes/chemistry , Parietal Lobe/ultrastructure , Rats , Rhodamines/chemistry , Staining and Labeling , Synapses/ultrastructure
9.
Neuroscience ; 193: 205-16, 2011 Oct 13.
Article in English | MEDLINE | ID: mdl-21798320

ABSTRACT

The lateral habenular complex (LHb) is a key signal integrator between limbic forebrain regions and monoaminergic hindbrain nuclei. Major projections of LHb neurons target the dopaminergic ventral tegmental area (VTA) and the serotonergic dorsal (DR) and median raphe nuclei (MnR). Both monoaminergic neurotransmitter systems play a central role in reward processing and reward-related decision-making. Glutamatergic LHb efferents terminate on GABAergic neurons in the VTA, the rostromedial tegmental nucleus (RMTg), and the raphe nuclei, thereby suppressing monoamine release when required by the present behavioral context. Recent studies suggest that the LHb exerts a strong tonic inhibition on monoamine release when no reward is to be obtained. It is yet unknown whether this inhibition is the result of a continuous external activation by other brain areas, or if it is intrinsically generated by LHb projection neurons. To analyze whether the tonic inhibition may be the result of a hyperpolarization-activated cyclic nucleotid-gated cation channel (HCN)-mediated pacemaker activity of LHb projection neurons, we combined retrograde tracing in rats with in situ hybridization of HCN1 to HCN4 mRNAs. In fact, close to all LHb neurons targeting VTA or raphe nuclei are equipped with HCN subunit mRNAs. While HCN1 mRNA is scarce, most neurons display strong expression of HCN2 to HCN4 mRNAs, in line with the potential formation of heteromeric channels. These results are supported by quantitative PCR and immunocytochemical analyses. Thus, our data suggest that the tonic inhibition of monoamine release is intrinsically generated in LHb projection neurons and that their activity may only be modulated by synaptic inputs to the LHb.


Subject(s)
Biogenic Monoamines/metabolism , Cyclic Nucleotide-Gated Cation Channels/metabolism , Habenula/cytology , Neurons/physiology , Animals , Cell Count , Gene Expression Regulation/physiology , Male , Neurons/ultrastructure , RNA, Messenger , Raphe Nuclei/cytology , Raphe Nuclei/metabolism , Rats , Rats, Wistar , Reticular Formation/cytology , Reticular Formation/metabolism , Silver Staining , Statistics, Nonparametric , Ventral Tegmental Area/cytology , Ventral Tegmental Area/metabolism , Wheat Germ Agglutinin-Horseradish Peroxidase Conjugate/metabolism
10.
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
11.
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
12.
J Thromb Haemost ; 7(5): 871-8, 2009 May.
Article in English | MEDLINE | ID: mdl-19228282

ABSTRACT

SUMMARY BACKGROUND: Myocardial inflammation is associated with an increase in circulating microparticles (MPs) and procoagulability. OBJECTIVES: We determined whether acute inflammation was associated with altered full-length tissue factor (flTF) expression and increased procoagulability in cardiomyocytic cells. METHODS: This study examined the transcriptional regulation of flTF expression in murine cardiomyocytic (HL-1) cells. Also, the generation of MPs by HL-1 cells and their ability to diffuse through an artificial endothelium was evaluated. RESULTS: Constitutive and tumor necrosis factor-alpha (TNF-alpha)-induced flTF expression of HL-1 was reduced when c-Jun N-terminal kinase (JNK) was inhibited. Tissue factor (TF)-positive procoagulant MPs were released from HL-1 cells in response to TNF-alpha. JNK inhibition potentiated the release of MPs from HL-1 cells without affecting MP-associated TF activity. MP generation was dependent on RhoA activation and associated with a reorganization of the actin cytoskeleton. Increased diffusion of HL-1-derived MPs through an endothelial monolayer was found after TNF-alpha treatment. The increased diffusion was dependent not only on TNF-alpha but also on HL-1-released mediators. CONCLUSIONS: Full-length TF expression in HL-1 cells was regulated through JNK. The TNF-alpha-induced increase in procoagulability was mediated through RhoA-dependent release of flTF-bearing MPs. These MPs were able to diffuse through an endothelial barrier adjacent to HL-1 cells and increased the procoagulability of the extracellular endothelial space. Cardiomyocytes seem to be a likely source of flTF-bearing procoagulant MPs.


Subject(s)
Inflammation/metabolism , Myocardium/metabolism , Thromboplastin/metabolism , Actins/metabolism , Animals , Cell Line , Coculture Techniques , Mice , Polymerase Chain Reaction , RNA, Messenger/genetics , Thromboplastin/genetics , Transcription, Genetic , Tumor Necrosis Factor-alpha/pharmacology
13.
J Med Virol ; 80(7): 1243-51, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18461615

ABSTRACT

Parvovirus B19 (PVB19) is a member of the human erythrovirus family detected frequently in endomyocardial biopsies from patients with dilated cardiomyopathy. Human erythroviruses cluster into three genotypes 1-3 which share a high degree of homology between major structural proteins and may cause indistinguishable infections clinically and serologically. In human cardiac tissue erythrovirus genotypes other than PVB19 have not yet been reported. Three hundred seventeen consecutive patients with symptomatic dilated cardiomyopathy (median left ventricular ejection fraction: 28.6%, range 5-45%) who underwent endomyocardial biopsy for the elucidation of the etiology, were analyzed using a new consensus PCR assay designed for the detection of the three erythrovirus genotype sequences. Endomyocardial biopsies of 151 (47.6%) patients were erythrovirus-positive. Genotype 1 specific sequences were detected in 43/151 (28.5%) of positive biopsy samples, whereas genotype 2-specific sequences so far considered rare in human disease and not yet been described in human heart tissue was identified in 108/151 (71.5%) of virus-positive endomyocardial biopsies with a preference in patients above 50 years of age. In spite of younger age, systolic left ventricular dysfunction of genotype 1-positive patients was significantly reduced as compared to genotype 2-positive patients (24.4+/-10.4% vs. 31.0+/-9.5%, P=0.0001) at the initial presentation. The data show that two genetically distinct erythrovirus variants with a different age distribution are detectable in endomyocardial biopsies of patients with dilated cardiomyopathy. The erythrovirus genotype 2, not described previously in human heart tissue, is highly prevalent in the heart but the less prevalent genotype 1 is associated with more severe disturbed cardiac function.


Subject(s)
Cardiomyopathy, Dilated/virology , Erythrovirus/isolation & purification , Heart/virology , Parvoviridae Infections/virology , Adult , Aged , Amino Acid Sequence , Base Sequence , Capsid Proteins/chemistry , Erythrovirus/genetics , Female , Genotype , Humans , Male , Middle Aged , Molecular Sequence Data , Prevalence , Sequence Alignment , Viral Load
14.
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
15.
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
16.
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
17.
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
18.
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
19.
Article in English | MEDLINE | ID: mdl-16329670

ABSTRACT

Ongoing viral persistence in the myocardium is associated with an adverse prognosis of cardiomyopathy eventually resulting in a reduced capacity for work and thus it is associated with enormous social costs. Experimental and clinical data highlight that an imbalance of the cytokine network and a defect in the cytokine-induced immune response may constitute major causes leading to the development of virus persistence and progression of myocardial dysfunction. Reversibility of cardiac impairment during the early stages of the disease and the arising chance of specific treatment options demand early diagnosis and treatment of the disease. Our pilot data on anti-viral treatment using INF-beta showed beneficial clinical effects and suggest that some of the ventricular dysfunction and wall motion abnormalities resolved after elimination of the responsible agents. The data also suggest that elimination of cardiotropic viruses and associated clinical effects may occur even in DCM patients presenting with a long history.


Subject(s)
Antiviral Agents/therapeutic use , Cardiomyopathies , Virus Diseases/drug therapy , Animals , Cardiomyopathies/drug therapy , Cardiomyopathies/virology , Heart/virology , Humans , Immunity/physiology , Immunologic Factors/therapeutic use , Interferons/therapeutic use , Myocardium/cytology , Myocardium/immunology , Myocardium/pathology , Treatment Outcome , Ventricular Dysfunction, Left/therapy , Ventricular Dysfunction, Left/virology , Virus Diseases/immunology
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...