Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
Add more filters










Publication year range
1.
Contrast Media Mol Imaging ; 10(2): 153-62, 2015.
Article in English | MEDLINE | ID: mdl-25078884

ABSTRACT

Accurate determination of tissue concentration of ultrasmall superparamagnetic iron oxide nanoparticles (USPIO) using T2 * MR relaxometry is still challenging. We present a reliable quantification method for local USPIO amount with the estimation of the liver specific relaxivity r2 * using monodisperse (59) Fe-core-labeled USPIO ((59) FeUSPIO). Dynamic and relaxometric in vivo characteristics of unlabeled monodisperse USPIO were determined in MRI at 3 T. The in vivo MR studies were performed for liver tissue with (59) FeUSPIO using iron dosages of 9 (n = 3), 18 (n = 2) and 27 (n = 3) µmol Fe kg(-1) body weight. The R2 * of the liver before and after USPIO injection (∆R2 *) was measured and correlated with (59) Fe activity measurements of excised organs by a whole body radioactivity counter (HAMCO) to define the dependency of ∆R2 * and (59) FeUSPIO liver concentration and calculate the r2 * of (59) FeUSPIO for the liver. Ultrastructural analysis of liver uptake was performed by histology and transmission electron microscopy. ∆R2 * of the liver revealed a dosage-dependent accumulation of (59) FeUSPIO with a percentage uptake of 70-88% of the injection dose. Hepatic ∆R2 * showed a dose-dependent linear correlation to (59) FeUSPIO activity measurements (r = 0.92) and an r2 * in the liver of 481 ± 74.9 mm(-1) s(-1) in comparison to an in vitro r2 * of 60.5 ± 3.3 mm(-1) s(-1) . Our results indicate that core-labeled (59) FeUSPIO can be used to quantify the local amount of USPIO and to estimate the liver-specific relaxivity r2 *.


Subject(s)
Contrast Media , Ferric Compounds , Isotope Labeling/methods , Liver , Magnetic Resonance Imaging/methods , Nanoparticles/chemistry , Animals , Contrast Media/chemistry , Contrast Media/pharmacology , Dose-Response Relationship, Drug , Ferric Compounds/chemistry , Ferric Compounds/pharmacology , Iron Isotopes/chemistry , Iron Isotopes/pharmacology , Liver/diagnostic imaging , Liver/metabolism , Mice , Mice, SCID , Radiography
2.
Acta Crystallogr D Biol Crystallogr ; 69(Pt 2): 308-12, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23385466

ABSTRACT

Successful cryogenic X-ray structure determination from a single high-pressure-frozen bovine enterovirus 2 crystal is reported. The presented high-pressure-freezing procedure is based on a commercially available device and allows the cryocooling of macromolecular crystals directly in their mother liquor without the time- and crystal-consuming search for optimal cryoconditions. The method is generally applicable and will allow cryogenic data collection from all types of macromolecular crystals.


Subject(s)
Enterovirus, Bovine/chemistry , Freezing , Pressure , Animals , Cattle , Cryoelectron Microscopy/methods , Cryoprotective Agents/pharmacology , Crystallization , Crystallography, X-Ray/methods , Diffusion , Enterovirus, Bovine/radiation effects , Freezing/adverse effects
3.
J Hepatol ; 58(1): 112-8, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22989568

ABSTRACT

BACKGROUND & AIMS: The liver can mitigate the inflammatory activity of infiltrating T cells by mechanisms that are not entirely clear. Here we investigated the role of liver sinusoidal endothelial cells (LSECs) in regulating the activity of inflammatory CD4 T cells. METHODS: Interactions between T helper (Th) 1 or Th17 cells and LSEC were studied by intravital microscopy and by in vitro stimulation assays. RESULTS: Circulating CD4 T cells established lasting and repeated interactions with liver endothelium in vivo. Stimulation of Th1 and Th17 cells by LSEC greatly inhibited their capacity to secrete interferon-γ or interleukin-17 in vitro; in contrast, stimulation by dendritic cells (DCs) resulted in considerable secretion of both cytokines. Cytokine release by Th1 or Th17 cells seemed to be actively suppressed by LSEC, as indicated by the inhibition of cytokine secretion even in the presence of Th1- and Th17-promoting DC. This inhibition of CD4 T cell effector function seemed to depend on the dominance of inhibitory over activating co-stimulatory signals on LSEC, since (1) cytokine secretion could be restored by increased CD28 co-activation; (2) LSEC from interleukin-10(-/-) mice, which manifest increased activating signals, such as MHC II, and decreased inhibitory signals, such as PD-L1, failed to suppress cytokine secretion; and (3) cytokine secretion by Th1 or Th17 cells that lacked PD-1, the ligand for inhibitory PD-L1, could not be suppressed by LSEC. CONCLUSIONS: LSEC inhibit inflammatory cytokine secretion of Th1 and Th17 effector CD4 T cells in dependence of interleukin-10 and PD-1.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cell Communication/immunology , Endothelial Cells/immunology , Liver/immunology , Animals , CD4-Positive T-Lymphocytes/cytology , Cell Differentiation/immunology , Cells, Cultured , Dendritic Cells/cytology , Dendritic Cells/immunology , Endothelial Cells/cytology , Female , Immune Tolerance/immunology , Interleukin-10/genetics , Interleukin-10/immunology , Liver/cytology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/immunology , Signal Transduction/immunology , Th1 Cells/cytology , Th1 Cells/immunology , Th17 Cells/cytology , Th17 Cells/immunology
4.
ACS Nano ; 6(8): 7318-25, 2012 Aug 28.
Article in English | MEDLINE | ID: mdl-22793497

ABSTRACT

A simple, fast, efficient, and widely applicable method to radiolabel the cores of monodisperse superparamagnetic iron oxide nanoparticles (SPIOs) with (59)Fe was developed. These cores can be used as precursors for a variety of functionalized nanodevices. A quality control using filtration techniques, size-exclusion chromatography, chemical degradation methods, transmission electron microscopy, and magnetic resonance imaging showed that the nanoparticles were stably labeled with (59)Fe. Furthermore, the particle structure and the magnetic properties of the SPIOs were unchanged. In a second approach, monodisperse SPIOs stabilized with (14)C-oleic acid were synthesized, and the stability of this shell labeling was studied. In proof of principle experiments, the (59)Fe-SPIOs coated with different shells to make them water-soluble were used to evaluate and compare in vivo pharmacokinetic parameters such as blood half-life. It could also be shown that our radiolabeled SPIOs embedded in recombinant lipoproteins can be used to quantify physiological processes in closer detail than hitherto possible. In vitro and in vivo experiments showed that the (59)Fe label is stable enough to be applied in vivo, whereas the (14)C label is rapidly removed from the iron core and is not adequate for in vivo studies. To obtain meaningful results in in vivo experiments, only (59)Fe-labeled SPIOs should be used.


Subject(s)
Dextrans/chemistry , Iron Radioisotopes , Magnetic Resonance Imaging/methods , Magnetite Nanoparticles/chemistry , Whole Body Imaging/methods , Animals , Contrast Media , Iron Radioisotopes/chemistry , Mice , Mice, Inbred BALB C , Organ Specificity , Radiopharmaceuticals , Tissue Distribution
5.
PLoS One ; 7(5): e36258, 2012.
Article in English | MEDLINE | ID: mdl-22590529

ABSTRACT

The human epithelial cell adhesion molecule (EpCAM) is highly expressed in a variety of clinical tumour entities. Although an antibody against EpCAM has successfully been used as an adjuvant therapy in colon cancer, this therapy has never gained wide-spread use. We have therefore investigated the possibilities and limitations for EpCAM as possible molecular imaging target using a panel of preclinical cancer models. Twelve human cancer cell lines representing six tumour entities were tested for their EpCAM expression by qPCR, flow cytometry analysis and immunocytochemistry. In addition, EpCAM expression was analyzed in vivo in xenograft models for tumours derived from these cells. Except for melanoma, all cell lines expressed EpCAM mRNA and protein when grown in vitro. Although they exhibited different mRNA levels, all cell lines showed similar EpCAM protein levels upon detection with monoclonal antibodies. When grown in vivo, the EpCAM expression was unaffected compared to in vitro except for the pancreatic carcinoma cell line 5072 which lost its EpCAM expression in vivo. Intravenously applied radio-labelled anti EpCAM MOC31 antibody was enriched in HT29 primary tumour xenografts indicating that EpCAM binding sites are accessible in vivo. However, bound antibody could only be immunohistochemically detected in the vicinity of perfused blood vessels. Investigation of the fine structure of the HT29 tumour blood vessels showed that they were immature and prone for higher fluid flux into the interstitial space. Consistent with this hypothesis, a higher interstitial fluid pressure of about 12 mbar was measured in the HT29 primary tumour via "wick-in-needle" technique which could explain the limited diffusion of the antibody into the tumour observed by immunohistochemistry.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antibodies, Neoplasm/pharmacology , Antigens, Neoplasm/metabolism , Cell Adhesion Molecules/metabolism , Neoplasm Proteins/metabolism , Neoplasms/diagnostic imaging , Neoplasms/metabolism , Animals , Antibodies, Monoclonal/immunology , Antibodies, Neoplasm/immunology , Antigens, Neoplasm/immunology , Binding Sites , Caco-2 Cells , Cell Adhesion Molecules/immunology , Epithelial Cell Adhesion Molecule , Humans , Magnetic Resonance Imaging , Male , Mice , Mice, SCID , Neoplasm Invasiveness , Neoplasm Proteins/immunology , Neoplasm Transplantation , Pressure , Radiography , Transplantation, Heterologous
6.
Acta Crystallogr Sect F Struct Biol Cryst Commun ; 68(Pt 4): 495-500, 2012 Apr 01.
Article in English | MEDLINE | ID: mdl-22505429

ABSTRACT

High-pressure freezing (HPF) is a method which allows sample vitrification without cryoprotectants. In the present work, protein crystals were cooled to cryogenic temperatures at a pressure of 210 MPa. In contrast to other HPF methods published to date in the field of cryocrystallography, this protocol involves rapid sample cooling using a standard HPF device. The fast cooling rates allow HPF of protein crystals directly in their mother liquor without the need for cryoprotectants or external reagents. HPF was first attempted with hen egg-white lysozyme and cubic insulin crystals, yielding good to excellent diffraction quality. Non-cryoprotected crystals of the membrane protein photosystem II have been successfully cryocooled for the first time. This indicates that the presented HPF method is well suited to the vitrification of challenging systems with large unit cells and weak crystal contacts.


Subject(s)
Crystallography, X-Ray/methods , Insulin/analysis , Muramidase/analysis , Animals , Chickens , Crystallography, X-Ray/instrumentation , Freezing , Pressure , Time Factors
7.
J Infect Dis ; 205(11): 1654-64, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22457281

ABSTRACT

Many enveloped viruses, including herpes viruses, hepatitis B virus (HBV), and hepatitis C virus (HCV), and human immunodeficiency virus (HIV), are among the most important human pathogens and are often responsible for coinfections involving ≥2 types of viruses. However, therapies that are effective against multiple virus classes are rare. Here we present a new class of synthetic anti-lipopolysaccharide peptides (SALPs) that bind to heparan sulfate moieties on the cell surface and inhibit infection with a variety of enveloped viruses. We demonstrate that SALPs inhibit entry of human immunodeficiency virus type 1 (HIV-1), herpes simplex virus (HSV) 1 and 2, HBV, and HCV to their respective host cells. Despite their high antiviral efficiency, SALPs were well tolerated, and neither toxicity nor measurable inhibitor-induced adverse effects were observed. Since these broad-spectrum antiviral peptides target a host cell rather than a viral component, they may also be useful for suppression of viruses that are resistant to antiviral drugs.


Subject(s)
Antiviral Agents/pharmacology , Peptides/pharmacology , Virus Attachment/drug effects , Virus Internalization/drug effects , Viruses/drug effects , Antiviral Agents/toxicity , Cell Line , Cell Survival , Heparitin Sulfate/metabolism , Humans , Lipopolysaccharides/metabolism , Peptides/toxicity , Protein Binding
8.
Cell Microbiol ; 14(5): 774-89, 2012 May.
Article in English | MEDLINE | ID: mdl-22289149

ABSTRACT

The mechanism of budding of influenza A virus revealed important deviation from the consensus mechanism of budding of retroviruses and of a growing number of negative-strand RNA viruses. This study is focused on the role of the influenza A virus matrix protein M1 in virus release. We found that a mutation of the proline residue at position 16 of the matrix protein induces inhibition of virus detachment from cells. Depletion of the M1-binding protein RACK1 also impairs virus release and RACK1 binding requires the proline residue at position 16 of M1. The impaired M1-RACK1 interaction does not affect the plasma membrane binding of M1; in contrast, RACK1 is recruited to detergent-resistant membranes in a M1-proline-16-dependent manner. The proline-16 mutation in M1 and depletion of RACK1 impairs the pinching-off of the budding virus particles. These findings reveal the active role of the viral matrix protein in the release of influenza A virus particles that involves a cross-talk with a RACK1-mediated pathway.


Subject(s)
GTP-Binding Proteins/metabolism , Host-Pathogen Interactions , Influenza A Virus, H1N1 Subtype/physiology , Neoplasm Proteins/metabolism , Receptors, Cell Surface/metabolism , Viral Matrix Proteins/metabolism , Virus Release , Cell Line , Humans , Influenza A Virus, H1N1 Subtype/genetics , Mutant Proteins/genetics , Mutant Proteins/metabolism , Mutation, Missense , Proline/genetics , Proline/metabolism , Protein Interaction Mapping , Receptors for Activated C Kinase , Viral Matrix Proteins/genetics
9.
PLoS Pathog ; 8(2): e1002517, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22319449

ABSTRACT

The early host response to viral infections involves transient activation of pattern recognition receptors leading to an induction of inflammatory cytokines such as interleukin-1ß (IL-1ß) and tumor necrosis factor α (TNFα). Subsequent activation of cytokine receptors in an autocrine and paracrine manner results in an inflammatory cascade. The precise mechanisms by which viruses avert an inflammatory cascade are incompletely understood. Nuclear factor (NF)-κB is a central regulator of the inflammatory signaling cascade that is controlled by inhibitor of NF-κB (IκB) proteins and the IκB kinase (IKK) complex. In this study we show that murine cytomegalovirus inhibits the inflammatory cascade by blocking Toll-like receptor (TLR) and IL-1 receptor-dependent NF-κB activation. Inhibition occurs through an interaction of the viral M45 protein with the NF-κB essential modulator (NEMO), the regulatory subunit of the IKK complex. M45 induces proteasome-independent degradation of NEMO by targeting NEMO to autophagosomes for subsequent degradation in lysosomes. We propose that the selective and irreversible degradation of a central regulatory protein by autophagy represents a new viral strategy to dampen the inflammatory response.


Subject(s)
I-kappa B Kinase/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Muromegalovirus/metabolism , NF-kappa B/antagonists & inhibitors , Phagosomes/immunology , Animals , Autophagy , Inflammation , Interleukin-1beta/metabolism , Mice , NF-kappa B/metabolism , NIH 3T3 Cells , Receptors, Interleukin-1/antagonists & inhibitors , Receptors, Interleukin-1/immunology , Receptors, Interleukin-1/metabolism , Signal Transduction/immunology , Toll-Like Receptors/antagonists & inhibitors , Toll-Like Receptors/immunology , Toll-Like Receptors/metabolism , Tumor Necrosis Factor-alpha/metabolism
10.
AIDS Res Ther ; 9(1): 2, 2012 Jan 19.
Article in English | MEDLINE | ID: mdl-22260499

ABSTRACT

Recently, it has been shown that human ejaculate enhances human immunodeficiency virus 1 (HIV-1) infectivity. Enhancement of infectivity is conceived to be mediated by amyloid filaments from peptides that are proteolytically released from prostatic acid phosphatase (PAP), termed Semen-derived Enhancer of Virus Infection (SEVI). The aim of this study was to test the range of HIV-1 infectivity enhancing properties of a large number of individual semen samples (n = 47) in a TZM-bl reporter cell HIV infection system. We find that semen overall increased infectivity to 156% of the control experiment without semen, albeit with great inter- and intraindividual variability (range -53%-363%). Using transmission electron microscopy, we provide evidence for SEVI fibrils in fresh human semen for the first time. Moreover, we confirm that the infectivity enhancing property can be inhibited by the major green tea ingredient epigallocatechin-3-gallate (EGCG) at non-toxic concentrations. The median inhibition of infection by treatment with 0.4 mM EGCG was 70.6% (p < 0.0001) in our cohort. Yet, there were substantial variations of inhibition and in a minority of samples, infectivity enhancement was not inhibited by EGCG treatment at all. Thus, topical application of EGCG may be a feasible additional measure to prevent the sexual transmission of HIV. However, the reasons for the variability in the efficacy of the abrogation of semen-mediated enhancement of HIV-1 infectivity and EGCG efficacy have to be elucidated before therapeutic trials can be conducted.

11.
J Virol ; 86(5): 2826-36, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22205742

ABSTRACT

In macrophages, HIV-1 accumulates in intracellular vesicles designated virus-containing compartments (VCCs). These might play an important role in the constitution of macrophages as viral reservoirs and allow HIV-1 to evade the immune system by sequestration in an internal niche, which is difficult to access from the exterior. However, until now, evidence of whether internal virus accumulations are protected from the host's humoral immune response is still lacking. In order to be able to study the formation and antibody accessibility of VCCs, we generated HIV-1 with green fluorescent protein (GFP)-tagged Gag replicating in primary macrophages. Live-cell observations revealed faint initial cytosolic Gag expression and subsequent large intracellular Gag accumulations which stayed stable over days. Taking advantage of the opportunity to study the accessibility of intracellular VCCs via the cell surface, we demonstrate that macrophage internal HIV-1-containing compartments cannot be targeted by neutralizing antibodies. Furthermore, HIV-1 was efficiently transferred from antibody-treated macrophages to T cells. Three-dimensional reconstruction of electron microscopic slices revealed that Gag accumulations correspond to viral particles within enclosed compartments and convoluted membranes. Thus, although some VCCs were connected to the plasma membrane, the complex membrane architecture of the HIV-1-containing compartment might shield viral particles from neutralizing antibodies. In sum, our study provides evidence that HIV-1 is sequestered into a macrophage internal membranous web, posing an obstacle for the elimination of this viral reservoir.


Subject(s)
Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , HIV Infections/immunology , HIV-1/immunology , Macrophages/immunology , Cell Line , Gene Expression Regulation, Viral , HIV Infections/virology , HIV-1/physiology , Humans , Immune Evasion , Macrophages/virology , Virus Replication
12.
PLoS One ; 6(12): e28030, 2011.
Article in English | MEDLINE | ID: mdl-22162753

ABSTRACT

Members of the carcinoembryonic antigen cell adhesion molecules (CEACAMs) family are the prototype of tumour markers. Classically they are used as serum markers, however, CEACAMs could serve as targets for molecular imaging as well.In order to test the anti CEACAM monoclonal antibody T84.1 for imaging purposes, CEACAM expression was analysed using this antibody. Twelve human cancer cell lines from different entities were screened for their CEACAM expression using qPCR, Western Blot and FACS analysis. In addition, CEACAM expression was analyzed in primary tumour xenografts of these cells. Nine of 12 tumour cell lines expressed CEACAM mRNA and protein when grown in vitro. Pancreatic and colon cancer cell lines showed the highest expression levels with good correlation of mRNA and protein level. However, when grown in vivo, the CEACAM expression was generally downregulated except for the melanoma cell lines. As the CEACAM expression showed pronounced expression in FemX-1 primary tumours, this model system was used for further experiments. As the accessibility of the antibody after i.v. application is critical for its use in molecular imaging, the binding of the T84.1 monoclonal antibody was assessed after i.v. injection into SCID mice harbouring a FemX-1 primary tumour. When applied i.v., the CEACAM specific T84.1 antibody bound to tumour cells in the vicinity of blood vessels. This binding pattern was particularly pronounced in the periphery of the tumour xenograft, however, some antibody binding was also observed in the central areas of the tumour around blood vessels. Still, a general penetration of the tumour by i.v. application of the anti CEACAM antibody could not be achieved despite homogenous CEACAM expression of all melanoma cells when analysed in tissue sections. This lack of penetration is probably due to the increased interstitial fluid pressure in tumours caused by the absence of functional lymphatic vessels.


Subject(s)
Carcinoembryonic Antigen/metabolism , Cell Adhesion Molecules/metabolism , Albumins/metabolism , Animals , Antigens, CD/biosynthesis , Biomarkers/metabolism , Caco-2 Cells , Carcinoembryonic Antigen/chemistry , Cell Adhesion Molecules/biosynthesis , Cell Adhesion Molecules/chemistry , Cell Line, Tumor , Evans Blue/pharmacology , Female , Humans , Immunohistochemistry/methods , Lymphatic Vessels/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, SCID , Molecular Imaging/methods , Neoplasm Metastasis , Neoplasm Transplantation , RNA, Messenger/metabolism
13.
PLoS One ; 6(12): e29112, 2011.
Article in English | MEDLINE | ID: mdl-22216177

ABSTRACT

Merkel Cell Polyomavirus (MCPyV) genomes are clonally integrated in tumor tissues of approximately 85% of all Merkel cell carcinoma (MCC) cases, a highly aggressive tumor of the skin which predominantly afflicts elderly and immunosuppressed patients. All integrated viral genomes recovered from MCC tissue or MCC cell lines harbor signature mutations in the early gene transcript encoding for the large T-Antigen (LT-Ag). These mutations selectively abrogate the ability of LT-Ag to support viral replication while still maintaining its Rb-binding activity, suggesting a continuous requirement for LT-Ag mediated cell cycle deregulation during MCC pathogenesis. To gain a better understanding of MCPyV biology, in vitro MCPyV replication systems are required. We have generated a synthetic MCPyV genomic clone (MCVSyn) based on the consensus sequence of MCC-derived sequences deposited in the NCBI database. Here, we demonstrate that transfection of recircularized MCVSyn DNA into some human cell lines recapitulates efficient replication of the viral genome, early and late gene expression together with virus particle formation. However, serial transmission of infectious virus was not observed. This in vitro culturing system allows the study of viral replication and will facilitate the molecular dissection of important aspects of the MCPyV lifecycle.


Subject(s)
DNA Replication , Gene Expression , Genome, Viral , Merkel cell polyomavirus/genetics , Virion , Virus Replication , Cell Line, Tumor , Humans , Merkel cell polyomavirus/physiology , Mutation , Transcription, Genetic
14.
Mol Imaging ; 9(4): 211-22, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20643024

ABSTRACT

The objective of this study was to quantify enzymatic activity on the surface of T cells by magnetic resonance imaging (MRI) using R2 and R2* relaxometry. Lymphoma cells expressing adenosine diphosphate (ADP)-ribosyltransferase 2 (ART2) were incubated with increasing doses of its substrate etheno-nicotinamide adenine dinucleotide (NAD), resulting in increasing amounts of surface protein ADP-ribosylation. Etheno-ADP-ribosylated proteins were detected with monoclonal antibody 1G4 and superparamagnetic iron oxide conjugated secondary antibodies (Ab-SPIO). Labeling efficiency was determined with R2 and R2* relaxometry on a clinical 3.0 T scanner. Parallel aliquots of cells were analyzed by flow cytometry. Cell-bound SPIO conjugates were detected by immunofluorescence and electron microscopy and quantified by atomic absorption spectroscopy. To mimic an inflammatory site in vivo, Ab-SPIO-labeled cells were injected subcutaneously in mice and analyzed by MRI. Immunofluorescence and electron microscopy confirmed cell-surface localization of Ab-SPIO. MRI of Ab-SPIO-labeled cells showed a corresponding signal reduction. Increases in R2 and R2* determined by magnetic resonance relaxometry correlated linearly with the expression level of ART2 and the concentration of the ART2 substrate etheno-NAD. R2 and R2* increases correlated linearly with the results from flow cytometry and atomic absorption spectroscopy analyses. Quantitative R2 and R2* mapping enable noninvasive determination of enzymatic activity on T cells and holds promise for characterization of inflammatory sites in vivo by MRI.


Subject(s)
ADP Ribose Transferases/metabolism , Cell Membrane/enzymology , Ferric Compounds/metabolism , Magnetic Resonance Imaging/methods , T-Lymphocytes/enzymology , Adenosine Diphosphate Ribose/metabolism , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/metabolism , Cell Line, Tumor , Ferric Compounds/chemistry , Flow Cytometry , Linear Models , Mice , Mice, Inbred BALB C , Microscopy, Electron, Transmission , Microscopy, Fluorescence , NAD/metabolism , NAD/pharmacology , Reproducibility of Results , Whole Body Imaging
15.
PLoS One ; 5(2): e9174, 2010 Feb 11.
Article in English | MEDLINE | ID: mdl-20161802

ABSTRACT

Recombinant viruses labelled with fluorescent proteins are useful tools in molecular virology with multiple applications (e.g., studies on intracellular trafficking, protein localization, or gene activity). We generated by homologous recombination three recombinant cytomegaloviruses carrying the enhanced yellow fluorescent protein (EYFP) fused with the viral proteins IE-2, ppUL32 (pp150), and ppUL83 (pp65). In growth kinetics, the three viruses behaved all like wild type, even at low multiplicity of infection (MOI). The expression of all three fusion proteins was detected, and their respective localizations were the same as for the unmodified proteins in wild-type virus-infected cells. We established the in vivo measurement of fluorescence intensity and used the recombinant viruses to measure inhibition of viral replication by neutralizing antibodies or antiviral substances. The use of these viruses in a pilot screen based on fluorescence intensity and high-content analysis identified cellular kinase inhibitors that block viral replication. In summary, these viruses with individually EYFP-tagged proteins will be useful to study antiviral substances and the dynamics of viral infection in cell culture.


Subject(s)
Cytomegalovirus/metabolism , Luminescent Proteins/metabolism , Recombinant Fusion Proteins/metabolism , Viral Proteins/metabolism , Active Transport, Cell Nucleus/drug effects , Antiviral Agents/pharmacology , Cell Nucleus/metabolism , Cells, Cultured , Cytomegalovirus/drug effects , Cytomegalovirus/genetics , Drug Evaluation, Preclinical , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/virology , Ganciclovir/pharmacology , Humans , Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , Luminescent Proteins/genetics , Male , Microscopy, Fluorescence , Phosphoproteins/genetics , Phosphoproteins/metabolism , Protein Transport/drug effects , Recombinant Fusion Proteins/genetics , Spectrometry, Fluorescence , Trans-Activators/genetics , Trans-Activators/metabolism , Viral Matrix Proteins/genetics , Viral Matrix Proteins/metabolism , Viral Proteins/genetics , Virus Replication/drug effects
16.
J Immunol ; 183(9): 5847-60, 2009 Nov 01.
Article in English | MEDLINE | ID: mdl-19812190

ABSTRACT

Autophagy is a central lysosomal degradation process that is essential for the maintenance of cellular homeostasis. Autophagy has furthermore emerged as integral part of the host immune response. Autophagic processes promote the separation and degradation of intracellular microorganisms which contributes to the development of innate and adaptive immunity. Some pathogenic microbes have therefore evolved mechanisms to evade or impede autophagy. We analyzed the effects of the enteropathogenic bacterium Yersinia enterocolitica on autophagy in macrophages. Yersiniae use a number of defined adhesins and secreted proteins to manipulate host immune responses. Our results showed that Y. enterocolitica defective in type III protein secretion efficiently activated autophagy in macrophages. Autophagy was mediated by the Yersinia adhesins invasin and YadA and particularly depended on the engagement of beta(1) integrin receptors. Several autophagy-related events followed beta(1) integrin-mediated engulfment of the bacteria including the formation of autophagosomes, processing of the marker protein LC3, redistribution of GFP-LC3 to bacteria-containing vacuoles, and the segregation of intracellular bacteria by autophagosomal compartments. These results provide direct evidence for the linkage of beta(1) integrin-mediated phagocytosis and autophagy induction. Multiple microbes signal through integrin receptors, and our results suggest a general principle by which the sensing of an extracellular microbe triggers autophagy. Owing to the importance of autophagy as host defense response, wild-type Y. enterocolitica suppressed autophagy by mobilizing type III protein secretion. The subversion of autophagy may be part of the Y. enterocolitica virulence strategy that supports bacterial survival when beta(1) integrin-dependent internalization and autophagy activation by macrophages are deleterious for the pathogen.


Subject(s)
Autophagy/immunology , Bacterial Proteins/classification , Bacterial Proteins/metabolism , Integrin beta1/physiology , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/microbiology , Yersinia enterocolitica/immunology , Adhesins, Bacterial/physiology , Animals , Autophagy/genetics , Bacterial Proteins/genetics , Biomarkers/metabolism , Cell Line , Female , Green Fluorescent Proteins/metabolism , Humans , Integrin beta1/biosynthesis , Macrophages, Peritoneal/metabolism , Mice , Mice, Inbred C3H , Mice, Knockout , Microtubule-Associated Proteins/physiology , Plasmids/immunology , Protein Transport/genetics , Protein Transport/immunology , Virulence/genetics , Virulence/immunology , Yersinia enterocolitica/genetics , Yersinia enterocolitica/pathogenicity
17.
Circ Res ; 105(3): 239-48, 2009 Jul 31.
Article in English | MEDLINE | ID: mdl-19590044

ABSTRACT

RATIONALE: Mutations in the MYBPC3 gene encoding cardiac myosin-binding protein (cMyBP)-C are frequent causes of hypertrophic cardiomyopathy, but the mechanisms leading from mutations to disease remain elusive. OBJECTIVE: The goal of the present study was therefore to gain insights into the mechanisms controlling the expression of MYBPC3 mutations. METHODS AND RESULTS: We developed a cMyBP-C knock-in mouse carrying a point mutation. The level of total cMyBP-C mRNAs was 50% and 80% lower in heterozygotes and homozygotes, respectively. Surprisingly, the single G>A transition on the last nucleotide of exon 6 resulted in 3 different mutant mRNAs: missense (exchange of G for A), nonsense (exon skipping, frameshift, and premature stop codon) and deletion/insertion (as nonsense but with additional partial retention of downstream intron, restoring of the reading frame, and almost full-length protein). Inhibition of nonsense-mediated mRNA decay in cultured cardiac myocytes or in vivo with emetine or cycloheximide increased the level of nonsense mRNAs severalfold but not of the other mRNAs. By using sequential protein fractionation and a new antibody directed against novel amino acids produced by the frameshift, we showed that inhibition of the proteasome with epoxomicin via osmotic minipumps increased the level of (near) full-length mutants but not of truncated proteins. Homozygotes exhibited myocyte and left ventricular hypertrophy, reduced fractional shortening, and interstitial fibrosis; heterozygotes had no major phenotype. CONCLUSIONS: These data reveal (1) an unanticipated complexity of the expression of a single point mutation in the whole animal and (2) the involvement of both nonsense-mediated mRNA decay and the ubiquitin-proteasome system in lowering the level of mutant proteins.


Subject(s)
Carrier Proteins/genetics , Carrier Proteins/metabolism , Codon, Nonsense/genetics , Hypertrophy, Left Ventricular/genetics , Hypertrophy, Left Ventricular/metabolism , Proteasome Endopeptidase Complex/metabolism , RNA Stability/genetics , Ubiquitin/metabolism , Animals , Cells, Cultured , Cycloheximide/pharmacology , Disease Models, Animal , Emetine/pharmacology , Exons/genetics , Gene Knock-In Techniques , Homozygote , Mice , Mice, Mutant Strains , Mice, Transgenic , Muscle Cells/drug effects , Muscle Cells/metabolism , Muscle Cells/pathology , Point Mutation/genetics , Protein Synthesis Inhibitors/pharmacology
18.
PLoS One ; 4(6): e6084, 2009 Jun 29.
Article in English | MEDLINE | ID: mdl-19562035

ABSTRACT

Nestin is the characteristic intermediate filament (IF) protein of rapidly proliferating progenitor cells and regenerating tissue. Nestin copolymerizes with class III IF-proteins, mostly vimentin, into heteromeric filaments. Its expression is downregulated with differentiation. Here we show that a strong nestin expression in mouse embryo tissue coincides with a strong accumulation of the glucocorticoid receptor (GR), a key regulator of growth and differentiation in embryonic development. Microscopic studies on cultured cells show an association of GR with IFs composed of vimentin and nestin. Cells lacking nestin, but expressing vimentin, or cells expressing vimentin, but lacking nestin accumulate GR in the nucleus. Completing these networks with an exogenous nestin, respectively an exogenous vimentin restores cytoplasmic anchoring of GR to the IF system. Thus, heteromeric filaments provide the basis for anchoring of GR. The reaction pattern with phospho-GR specific antibodies and the presence of the chaperone HSC70 suggest that specifically the unliganded receptor is anchored to the IF system. Ligand addition releases GR from IFs and shifts the receptor into the nucleus. Suppression of nestin by specific shRNA abolishes anchoring of GR, induces its accumulation in the nucleus and provokes an irreversible G1/S cell cycle arrest. Suppression of GR prior to that of nestin prevents entry into the arrest. The data give evidence that nestin/vimentin specific anchoring modulates growth suppression by GR. We hypothesize that expression of nestin is a major determinant in suppression of anti-proliferative activity of GR in undifferentiated tissue and facilitates activation of this growth control in a precise tissue and differentiation dependent manner.


Subject(s)
Cytoplasm/metabolism , Gene Expression Regulation, Developmental , Intermediate Filament Proteins/biosynthesis , Nerve Tissue Proteins/biosynthesis , Receptors, Glucocorticoid/metabolism , Vimentin/biosynthesis , Animals , Cell Cycle , Cell Differentiation , Cell Nucleus/metabolism , Cytoskeleton/metabolism , Glioma/metabolism , Humans , Intermediate Filament Proteins/metabolism , Mice , Mice, Inbred BALB C , Nerve Tissue Proteins/metabolism , Nestin , RNA/metabolism , Rats , Vimentin/metabolism
19.
Proc Natl Acad Sci U S A ; 106(22): 9033-8, 2009 Jun 02.
Article in English | MEDLINE | ID: mdl-19451623

ABSTRACT

Peptide fragments, derived from prostatic acidic phosphatase, are secreted in large amounts into human semen and form amyloid fibrils. These fibrillar structures, termed semen-derived enhancer of virus infection (SEVI), capture HIV virions and direct them to target cells. Thus, SEVI appears to be an important infectivity factor of HIV during sexual transmission. Here, we are able to demonstrate that epigallocatechin-3-gallate (EGCG), the major active constituent of green tea, targets SEVI for degradation. Furthermore, it is shown that EGCG inhibits SEVI activity and abrogates semen-mediated enhancement of HIV-1 infection in the absence of cellular toxicity. Therefore, EGCG appears to be a promising supplement to antiretroviral microbicides to reduce sexual transmission of HIV-1.


Subject(s)
Anti-HIV Agents/pharmacology , Catechin/analogs & derivatives , HIV Infections/prevention & control , HIV-1 , Semen/drug effects , Amyloid/antagonists & inhibitors , Amyloid/metabolism , Camellia sinensis/chemistry , Catechin/pharmacology , Cells, Cultured , HIV Infections/transmission , Humans , Male , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/metabolism , Protein Tyrosine Phosphatases/antagonists & inhibitors , Protein Tyrosine Phosphatases/metabolism , Semen/virology
20.
J Am Chem Soc ; 130(23): 7315-20, 2008 Jun 11.
Article in English | MEDLINE | ID: mdl-18484723

ABSTRACT

Magnetic nanoparticles have been assembled into the bilayer membrane of block copolymer vesicles. The nanoparticles decorate the hydrophobic/hydrophilic interface, which leads to bridging of adjacent bilayers and the formation of oligo-lamellar vesicles. The nanoparticle uptake of the vesicles is sufficiently high to become magnetophoretic in external magnetic fields as shown by video microscopy.


Subject(s)
Hemiterpenes/chemistry , Latex/chemistry , Lipid Bilayers/chemistry , Magnetics , Nanoparticles/chemistry , Polyethylene Glycols/chemistry , Ferric Compounds/chemistry , Hydrophobic and Hydrophilic Interactions , Neutron Diffraction , Scattering, Small Angle
SELECTION OF CITATIONS
SEARCH DETAIL
...