Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
2.
Front Cell Infect Microbiol ; 13: 1113528, 2023.
Article in English | MEDLINE | ID: mdl-37065199

ABSTRACT

The Gram-negative bacterium Neisseria meningitidis, which causes meningitis in humans, has been demonstrated to manipulate or alter host signalling pathways during infection of the central nervous system (CNS). However, these complex signalling networks are not completely understood. We investigate the phosphoproteome of an in vitro model of the blood-cerebrospinal fluid barrier (BCSFB) based on human epithelial choroid plexus (CP) papilloma (HIBCPP) cells during infection with the N. meningitidis serogroup B strain MC58 in presence and absence of the bacterial capsule. Interestingly, our data demonstrates a stronger impact on the phosphoproteome of the cells by the capsule-deficient mutant of MC58. Using enrichment analyses, potential pathways, molecular processes, biological processes, cellular components and kinases were determined to be regulated as a consequence of N. meningitidis infection of the BCSFB. Our data highlight a variety of protein regulations that are altered during infection of CP epithelial cells with N. meningitidis, with the regulation of several pathways and molecular events only being detected after infection with the capsule-deficient mutant. Mass spectrometry proteomics data are available via ProteomeXchange with identifier PXD038560.


Subject(s)
Neisseria meningitidis , Humans , Neisseria meningitidis/physiology , Choroid Plexus/microbiology , Epithelial Cells/microbiology , Blood-Brain Barrier/microbiology , Cell Line, Tumor
3.
Cell Tissue Res ; 392(2): 393-412, 2023 May.
Article in English | MEDLINE | ID: mdl-36781482

ABSTRACT

Endothelial cells play a major part in the regulation of vascular permeability and angiogenesis. According to their duty to fit the needs of the underlying tissue, endothelial cells developed different subtypes with specific endothelial microdomains as caveolae, fenestrae and transendothelial channels which regulate nutrient exchange, leukocyte migration, and permeability. These microdomains can exhibit diaphragms that are formed by the endothelial cell-specific protein plasmalemma vesicle-associated protein (PLVAP), the only known protein component of these diaphragms. Several studies displayed an involvement of PLVAP in diseases as cancer, traumatic spinal cord injury, acute ischemic brain disease, transplant glomerulopathy, Norrie disease and diabetic retinopathy. Besides an upregulation of PLVAP expression within these diseases, pro-angiogenic or pro-inflammatory responses were observed. On the other hand, loss of PLVAP in knockout mice leads to premature mortality due to disrupted homeostasis. Generally, PLVAP is considered as a major factor influencing the permeability of endothelial cells and, finally, to be involved in the regulation of vascular permeability. Following these observations, PLVAP is debated as a novel therapeutic target with respect to the different vascular beds and tissues. In this review, we highlight the structure and functions of PLVAP in different endothelial types in health and disease.


Subject(s)
Diabetic Retinopathy , Endothelial Cells , Animals , Brain/metabolism , Capillary Permeability/physiology , Carrier Proteins/metabolism , Endothelial Cells/metabolism , Membrane Proteins/metabolism , Humans
4.
STAR Protoc ; 3(4): 101816, 2022 12 16.
Article in English | MEDLINE | ID: mdl-36386888

ABSTRACT

Choroid plexus, located in brain ventricles, is the site of blood-cerebrospinal fluid barrier that contains endothelial cells and an epithelial monolayer separated by stroma. We established a two-cell-type model of the human choroid plexus consisting of immortalized endothelial cells (iHCPEnC) and epithelial papilloma (HIBCPP) cells grown on opposite sides of filter supports. In this protocol, we describe the preparation of this model, the measurement of transepithelial electrical resistance (TEER), and immunofluorescence imaging-based analysis to determine the barrier function. For complete details on the use and execution of this protocol, please refer to Muranyi et al. (2022).


Subject(s)
Choroid Plexus , Endothelial Cells , Humans , Epithelial Cells , Blood-Brain Barrier , Cell Count
5.
iScience ; 25(6): 104383, 2022 Jun 17.
Article in English | MEDLINE | ID: mdl-35633941

ABSTRACT

The choroid plexus (CP) is a highly vascularized structure containing endothelial and epithelial cells located in the ventricular system of the central nervous system (CNS). The role of the fenestrated CP endothelium is under-researched and requires the generation of an immortalized CP endothelial cell line with preserved features. Transduction of primary human CP endothelial cells (HCPEnC) with the human telomerase reverse transcriptase (hTERT) resulted in immortalized HCPEnC (iHCPEnC), which grew as monolayer with contact inhibition, formed capillary-like tubes in Matrigel, and showed no colony growth in soft agar. iHCPEnC expressed pan-endothelial markers and presented characteristic plasmalemma vesicle-associated protein-containing structures. Cultivation of iHCPEnC and human epithelial CP papilloma (HIBCPP) cells on opposite sides of cell culture filter inserts generated an in vitro model with a consistently enhanced barrier function specifically by iHCPEnC. Overall, iHCPEnC present a tool that will contribute to the understanding of CP organ functions, especially endothelial-epithelial interplay.

6.
Methods Mol Biol ; 1663: 139-152, 2017.
Article in English | MEDLINE | ID: mdl-28924665

ABSTRACT

Single-molecule localization microscopy (SMLM) enables imaging of biological structures in the nanometre range. Long measurement times are the consequence of this kind of microscopy due to the need of acquiring thousands of images. We built a setup that automatically detects target structures using confocal microscopy and images them with SMLM. Utilizing the Konstanz Information Miner (KNIME), we were able to connect a confocal microscope with an SMLM unit for targeted screening. In this process, we developed KNIME plugins to communicate with the microscope components and combined them to a workflow. Thus, measuring biological nanometre-sized structures in a sufficient number to get statistical significance becomes feasible. For proof of principle HIV-1 assembly complexes in HeLa cells derived from transfection of replication deficient viral construct were imaged by a fully automated screen.


Subject(s)
Computational Biology/methods , HIV-1/physiology , Single Molecule Imaging/methods , HeLa Cells , Humans , Internet , Microscopy, Confocal/methods , Microscopy, Fluorescence/methods , Software , Transfection , Virus Assembly
7.
Elife ; 3: e04114, 2014 Dec 17.
Article in English | MEDLINE | ID: mdl-25517934

ABSTRACT

The steps from HIV-1 cytoplasmic entry until integration of the reverse transcribed genome are currently enigmatic. They occur in ill-defined reverse-transcription- and pre-integration-complexes (RTC, PIC) with various host and viral proteins implicated. In this study, we report quantitative detection of functional RTC/PIC by labeling nascent DNA combined with detection of viral integrase. We show that the viral CA (capsid) protein remains associated with cytoplasmic RTC/PIC but is lost on nuclear PIC in a HeLa-derived cell line. In contrast, nuclear PIC were almost always CA-positive in primary human macrophages, indicating nuclear import of capsids or capsid-like structures. We further show that the CA-targeted inhibitor PF74 exhibits a bimodal mechanism, blocking RTC/PIC association with the host factor CPSF6 and nuclear entry at low, and abrogating reverse transcription at high concentrations. The newly developed system is ideally suited for studying retroviral post-entry events and the roles of host factors including DNA sensors and signaling molecules.


Subject(s)
Active Transport, Cell Nucleus/drug effects , Capsid Proteins/metabolism , DNA, Viral/biosynthesis , HIV-1/genetics , Virus Replication/genetics , Capsid Proteins/antagonists & inhibitors , Capsid Proteins/genetics , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Nucleus/virology , Cytosol/metabolism , Cytosol/virology , Gene Expression Regulation , HEK293 Cells , HIV Integrase/genetics , HIV Integrase/metabolism , HIV-1/metabolism , HeLa Cells , Host-Pathogen Interactions , Humans , Indoles/pharmacology , Macrophages/metabolism , Macrophages/virology , Microscopy, Confocal/methods , Phenylalanine/analogs & derivatives , Phenylalanine/pharmacology , Primary Cell Culture , Reverse Transcription/drug effects , Signal Transduction , T-Lymphocytes/metabolism , T-Lymphocytes/virology , Virus Integration/drug effects , Virus Integration/genetics , Virus Internalization/drug effects , Virus Replication/drug effects , beta Karyopherins/genetics , beta Karyopherins/metabolism , mRNA Cleavage and Polyadenylation Factors/genetics , mRNA Cleavage and Polyadenylation Factors/metabolism
8.
PLoS Pathog ; 9(2): e1003198, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23468635

ABSTRACT

The inner structural Gag proteins and the envelope (Env) glycoproteins of human immunodeficiency virus (HIV-1) traffic independently to the plasma membrane, where they assemble the nascent virion. HIV-1 carries a relatively low number of glycoproteins in its membrane, and the mechanism of Env recruitment and virus incorporation is incompletely understood. We employed dual-color super-resolution microscopy visualizing Gag assembly sites and HIV-1 Env proteins in virus-producing and in Env expressing cells. Distinctive HIV-1 Gag assembly sites were readily detected and were associated with Env clusters that always extended beyond the actual Gag assembly site and often showed enrichment at the periphery and surrounding the assembly site. Formation of these Env clusters depended on the presence of other HIV-1 proteins and on the long cytoplasmic tail (CT) of Env. CT deletion, a matrix mutation affecting Env incorporation or Env expression in the absence of other HIV-1 proteins led to much smaller Env clusters, which were not enriched at viral assembly sites. These results show that Env is recruited to HIV-1 assembly sites in a CT-dependent manner, while Env(ΔCT) appears to be randomly incorporated. The observed Env accumulation surrounding Gag assemblies, with a lower density on the actual bud, could facilitate viral spread in vivo. Keeping Env molecules on the nascent virus low may be important for escape from the humoral immune response, while cell-cell contacts mediated by surrounding Env molecules could promote HIV-1 transmission through the virological synapse.


Subject(s)
Cell Membrane/virology , HIV Envelope Protein gp120/metabolism , Microscopy, Fluorescence/methods , Virus Assembly/physiology , gag Gene Products, Human Immunodeficiency Virus/metabolism , Cell Membrane/metabolism , Cluster Analysis , HeLa Cells , Humans , Protein Conformation , Protein Structure, Tertiary , Virus Attachment , Virus Internalization , Virus Replication
9.
Histochem Cell Biol ; 139(1): 173-9, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22910843

ABSTRACT

We apply single-molecule super-resolution microscopy and coordinate-based cluster analysis to extract information on the distribution and on the morphology and size of clusters of the human immunodeficiency virus (HIV-1) Gag polyprotein in fixed cells. Three different patterns of Gag distribution could be distinguished. A major type of assembly observed was in accordance with previous electron microscopy analyses revealing ~140 nm-sized HIV-1 buds at the plasma membrane of virus-producing cells. The distribution of Gag molecules in the 2D projection at these sites was consistent with a semi-spherical 3D assembly. We compared different methods of cluster analysis and demonstrated that we can reliably distinguish different distribution patterns of the Gag polyprotein. These methods were applied to extract information on the properties of the different Gag clusters.


Subject(s)
Cell Membrane/metabolism , HIV-1/metabolism , Microscopy, Fluorescence/methods , Virion/metabolism , Virus Assembly , gag Gene Products, Human Immunodeficiency Virus/metabolism , Algorithms , Cell Line , Cell Membrane/virology , Cluster Analysis , HIV-1/genetics , Humans , Image Processing, Computer-Assisted , Transfection , gag Gene Products, Human Immunodeficiency Virus/genetics
10.
PLoS One ; 6(7): e22007, 2011.
Article in English | MEDLINE | ID: mdl-21799764

ABSTRACT

Fluorescently labeled human immunodeficiency virus (HIV) derivatives, combined with the use of advanced fluorescence microscopy techniques, allow the direct visualization of dynamic events and individual steps in the viral life cycle. HIV proteins tagged with fluorescent proteins (FPs) have been successfully used for live-cell imaging analyses of HIV-cell interactions. However, FPs display limitations with respect to their physicochemical properties, and their maturation kinetics. Furthermore, several independent FP-tagged constructs have to be cloned and characterized in order to obtain spectral variations suitable for multi-color imaging setups. In contrast, the so-called SNAP-tag represents a genetically encoded non-fluorescent tag which mediates specific covalent coupling to fluorescent substrate molecules in a self-labeling reaction. Fusion of the SNAP-tag to the protein of interest allows specific labeling of the fusion protein with a variety of synthetic dyes, thereby offering enhanced flexibility for fluorescence imaging approaches.Here we describe the construction and characterization of the HIV derivative HIV(SNAP), which carries the SNAP-tag as an additional domain within the viral structural polyprotein Gag. Introduction of the tag close to the C-terminus of the matrix domain of Gag did not interfere with particle assembly, release or proteolytic virus maturation. The modified virions were infectious and could be propagated in tissue culture, albeit with reduced replication capacity. Insertion of the SNAP domain within Gag allowed specific staining of the viral polyprotein in the context of virus producing cells using a SNAP reactive dye as well as the visualization of individual virions and viral budding sites by stochastic optical reconstruction microscopy. Thus, HIV(SNAP) represents a versatile tool which expands the possibilities for the analysis of HIV-cell interactions using live cell imaging and sub-diffraction fluorescence microscopy.


Subject(s)
HIV-1/physiology , Host-Pathogen Interactions , Molecular Probes/metabolism , gag Gene Products, Human Immunodeficiency Virus/metabolism , Amino Acid Sequence , Cell Line , Humans , Intracellular Space/metabolism , Molecular Probes/chemistry , Molecular Sequence Data , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Virus Internalization , Virus Replication , gag Gene Products, Human Immunodeficiency Virus/chemistry
11.
J Virol ; 82(21): 10625-33, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18701596

ABSTRACT

The human pathogenic poxvirus molluscum contagiosum virus (MCV) is the causative agent of benign neoplasm, with worldwide incidence, characterized by intraepidermal hyperplasia and hypertrophy of cells. Here, we present evidence that the MC007L protein of MCV targets retinoblastoma protein (pRb) via a conserved LxCxE motif, which is present in many viral oncoproteins. The deregulation of the pRb pathway plays a central role in tumor pathogenesis. The oncoproteins of small DNA viruses contain amino acid sequences that bind to and inactivate pRb. Isolated expression of these oncoproteins induces apoptosis, cell proliferation, and cellular transformation. The MC007L gene displays no homology to other genes within the poxvirus family. The protein anchors into the outer mitochondrial membrane via an N-terminal mitochondrial targeting sequence. Through the LxCxE motifs, MC007L induces a cytosolic sequestration of pRb at mitochondrial membranes, leading to the inactivation of the protein by mislocalization. MC007L precipitates the endogenous pRb/E2F-1 complex. Moreover, MC007L is able to cooperate to transform primary rat kidney cells. The interaction between MC007L and pRb provides a novel mechanism by which a virus can perturb the cell cycle.


Subject(s)
Molluscum contagiosum virus/physiology , Oncogene Proteins/metabolism , Retinoblastoma Protein/antagonists & inhibitors , Retinoblastoma Protein/metabolism , Viral Proteins/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , Cell Line , Conserved Sequence , Humans , Mitochondrial Membranes/chemistry , Molecular Sequence Data , Protein Binding , Protein Interaction Domains and Motifs , Rats
12.
Clin Transplant ; 20 Suppl 17: 25-9, 2006.
Article in English | MEDLINE | ID: mdl-17100698

ABSTRACT

Mycophenolic acid (MPA) is a highly selective, non-competitive and reversible inhibitor of the inosine monophosphate dehydrogenase (IMPDH), the rate-limiting enzyme in the de novo biosynthesis of guanosine nucleotides. Mycophenolate mofetil (MMF, the ester prodrug of MPA) strongly inhibits both T- and B-lymphocyte proliferation and has now been widely used in the prevention of acute and chronic allograft rejection. Recent evidence, however, suggests that MMF is also capable of inhibiting the proliferation of non-immune cells. In various cell lines, e.g. smooth muscle cells, renal tubular cells, mesangial cells, and fibroblasts, MPA reduced or even abrogated proliferation in response to proliferative stimuli. In animal studies, MMF ameliorated renal lesions in immune-mediated disease, e.g. in the Anti-Thy 1.1 model and experimental lupus nephritis, but was also effective in non-immune-mediated renal damage, e.g. in the rat remnant kidney model or in a model of chronic cyclosporine nephrotoxicity in the rat. In humans, MMF reduced proteinuria in steroid-resistant nephrotic syndrome and had beneficial effects in the prevention and treatment of chronic allograft nephropathy and calcineurin inhibitor toxicity through the reduction of immune- and non-immune-mediated renal damage. MMF is well tolerated and has proven to be a relatively safe drug. Taken together, there is a growing body of evidence pointing to therapeutic applications of MMF other than immunosuppression, in particular the prevention of fibrosis.


Subject(s)
Enzyme Inhibitors/pharmacology , IMP Dehydrogenase/antagonists & inhibitors , Kidney Transplantation , Kidney/pathology , Mycophenolic Acid/pharmacology , Animals , Fibrosis/prevention & control , Humans
13.
Virus Genes ; 33(2): 229-34, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16972039

ABSTRACT

Hantavirus serotype Puumala (PUUV) is the etiologic agent of Nephropathia epidemica (NE), a mild variant of Hemorrhagic Fever with Renal Syndrome (HFRS) in Europe, with lethality rates up to 1% among human patients. The serotype PUUV is composed of numerous geographically restricted strains forming a great number of phylogeographic lineages, sublineages, variants, and clusters. We describe a new, geographically and phylogenetically well-defined pathogenic PUUV sublineage in Southwest Germany (strain Heidelberg/hu) originated from an HFRS patient. The genetic analysis of PUUV strain Heidelberg/hu may contribute in future to better diagnostics, the development of vaccines, and the understanding of the factors that determine hantavirus pathogenesis.


Subject(s)
Hemorrhagic Fever with Renal Syndrome/virology , Puumala virus/classification , Puumala virus/genetics , Adult , Base Sequence , Genotype , Germany , Hemorrhagic Fever with Renal Syndrome/pathology , Humans , Kidney/pathology , Male , Molecular Sequence Data , Sequence Alignment
15.
Exp Cell Res ; 306(1): 192-202, 2005 May 15.
Article in English | MEDLINE | ID: mdl-15878344

ABSTRACT

Diaphanous-related formins (DRFs) are actin nucleators that mediate rearrangements of the actin cytoskeleton downstream of specific Rho GTPases. The DRF Formin Homology 2 Domain containing 1 (FHOD1) interacts with the Rac1 GTPase and induces the formation of and associates with bundled actin stress fibers. Here we report that active FHOD1 also coordinates microtubules with these actin stress fibers. Expression of a constitutive active FHOD1 variant in HeLa cells not only resulted in pronounced formation of FHOD1-actin fibers but also caused marked cell elongation and parallel alignment of microtubules without affecting cytokinesis of these cells. The analysis of deletions in the FH1 and FH2 functional regions revealed that the integrity of both domains was strictly required for FHOD1's effects on the cytoskeleton. Dominant-negative approaches demonstrated that filament coordination and cell elongation depended on the activity of the Rho-ROCK cascade, but did not involve Rac or Cdc42 activity. Experimental depolymerization of actin filaments or microtubules revealed that the formation of FHOD1-actin fibers was a prerequisite for the polarization of microtubules. However, only simultaneous disruption of both filament systems reversed the cell elongation induced by activated FHOD1. Thus, sustained cell elongation was a consequence of FHOD1-mediated actin-microtubule coordination. These results suggest filament coordination as a conserved function of mammalian DRFs.


Subject(s)
Actin Cytoskeleton/metabolism , Fetal Proteins/physiology , Microtubules/metabolism , Nuclear Proteins/physiology , Actin Cytoskeleton/drug effects , Animals , Cell Shape/drug effects , Cytochalasin D/pharmacology , Cytokinesis/drug effects , Fetal Proteins/genetics , Formins , HeLa Cells , Humans , Mice , Microtubules/drug effects , Mutation , NIH 3T3 Cells , Nocodazole/pharmacology , Nuclear Proteins/genetics , Signal Transduction/physiology , Stress Fibers/drug effects , Stress Fibers/metabolism , Transfection , rho GTP-Binding Proteins/genetics , rho GTP-Binding Proteins/physiology
16.
Virus Genes ; 30(2): 157-80, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15744574

ABSTRACT

In the last decades a significant number of so far unknown or underestimated pathogens have emerged as fundamental health hazards of the human population despite intensive research and exceptional efforts of modern medicine to embank and eradicate infectious diseases. Almost all incidents caused by such emerging pathogens could be ascribed to agents that are zoonotic or expanded their host range and crossed species barriers. Many different factors influence the status of a pathogen to remain unnoticed or evolves into a worldwide threat. The ability of an infectious agent to adapt to changing environmental conditions and variations in human behavior, population development, nutrition, education, social, and health status are relevant factors affecting the correlation between pathogen and host. Hantaviruses belong to the emerging pathogens having gained more and more attention in the last decades. These viruses are members of the family Bunyaviridae and are grouped into a separate genus known as Hantavirus. The serotypes Hantaan (HTN), Seoul (SEO), Puumala (PUU), and Dobrava (DOB) virus predominantly cause hemorrhagic fever with renal syndrome (HFRS), a disease characterized by renal failure, hemorrhages, and shock. In the recent past, many hantavirus isolates have been identified and classified in hitherto unaffected geographic regions in the New World (North, Middle, and South America) with characteristic features affecting the lungs of infected individuals and causing an acute pulmonary syndrome. Hantavirus outbreaks in the United States of America at the beginning of the 10th decade of the last century fundamentally changed our knowledge about the appearance of the hantavirus specific clinical picture, mortality, origin, and transmission route in human beings. The hantavirus pulmonary syndrome (HPS) was first recognized in 1993 in the Four Corners Region of the United States and had a lethality of more than 50%. Although the causative virus was first termed in connection with the geographic name of its outbreak region the analysis of the individual viruses indicate that the causing virus of HPS was a genetically distinct hantavirus and consequently termed as Sin Nombre virus. Hantaviruses are distributed worldwide and are assumed to share a long time period of co-evolution with specific rodent species as their natural reservoir. The degree of relatedness between virus serotypes normally coincides with the relatedness between their respective hosts. There are no known diseases that are associated with hantavirus infections in rodents underlining the amicable relationship between virus and host developed by mutual interaction in hundreds of thousands of years. Although rodents are the major reservoir, antibodies against hantaviruses are also present in domestic and wild animals like cats, dogs, pigs, cattle, and deer. Domestic animals and rodents live jointly in a similar habitat. Therefore the transmission of hantaviruses from rodents to domestic animals seems to be possible, if the target organs, tissues, and cell parenchyma of the co-habitat domestic animals possess adequate virus receptors and are suitable for hantavirus entry and replication. The most likely incidental infection of species other than rodents as for example humans turns hantaviruses from harmless to life-threatening pathogenic agents focusing the attention on this virus group, their ecology and evolution in order to prevent the human population from a serious health risk. Much more studies on the influence of non-natural hosts on the ecology of hantaviruses are needed to understand the directions that the hantavirus evolution could pursue. At least, domestic animals that share their environmental habitat with rodents and humans particularly in areas known as high endemic hantavirus regions have to be copiously screened. Each transfer of hantaviruses from their original natural hosts to other often incidental hosts is accompanied by a change of ecology, a change of environment, a modulation of numerous factors probably influencing the pathogenicity and virulence of the virus. The new environment exerts a modified evolutionary pressure on the virus forcing it to adapt and probably to adopt a form that is much more dangerous for other host species compared to the original one.


Subject(s)
Hantavirus Infections/transmission , Animals , Disease Outbreaks , Disease Reservoirs , Ecosystem , Orthohantavirus/classification , Orthohantavirus/isolation & purification , Orthohantavirus/pathogenicity , Hantavirus Infections/epidemiology , Hantavirus Infections/etiology , Hantavirus Infections/prevention & control , Hantavirus Pulmonary Syndrome/transmission , Hantavirus Pulmonary Syndrome/virology , Humans , Zoonoses/transmission , Zoonoses/virology
17.
BMC Mol Biol ; 5: 16, 2004 Sep 06.
Article in English | MEDLINE | ID: mdl-15350211

ABSTRACT

BACKGROUND: Foamy virus Bel1/Tas trans-activators act as key regulators of gene expression and directly bind to Bel1 response elements (BRE) in both the internal and the 5'LTR promoters leading to strong transcriptional trans-activation. Cellular coactivators interacting with Bel1/Tas are unknown to date. RESULTS: Transient expression assays, co-immunoprecipitation experiments, pull-down assays, and Western blot analysis were used to demonstrate that the coactivator p300 and histone acetyltransferase PCAF specifically interact with the retroviral trans-activator Bel1/Tas in vivo. Here we show that the Bel1/Tas-mediated trans-activation was enhanced by the coactivator p300, histone acetyltransferases PCAF and SRC-1 based on the crucial internal promoter BRE. The Bel1/Tas-interacting region was mapped to the C/H1 domain of p300 by co-immunoprecipitation and pull-down assays. In contrast, coactivator SRC-1 previously reported to bind to the C-terminal domain of p300 did not directly interact with the Bel1 protein but nevertheless enhanced Bel1/Tas-mediated trans-activation. Cotransfection of Bel1/Tas and p300C with an expression plasmid containing the C/H1domain partially inhibited the p300C-driven trans-activation. CONCLUSIONS: Our data identify p300 and PCAF as functional partner molecules that directly interact with Bel1/Tas. Since the acetylation activities of the three coactivators reside in or bind to the C-terminal regions of p300, a C/H1 expression plasmid was used as inhibitor. This is the first report of a C/H1 domain-interacting retroviral trans-activator capable of partially blocking the strong Bel1/Tas-mediated activation of the C-terminal region of coactivator p300. The potential mechanisms and functional roles of the three histone and factor acetyltransferases p300, PCAF, and SRC-1 in Bel1/Tas-mediated trans-activation are discussed.


Subject(s)
Acetyltransferases/metabolism , Cell Cycle Proteins/metabolism , DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Retroviridae Proteins/metabolism , Trans-Activators/metabolism , Transcription Factors/metabolism , Transcriptional Activation/genetics , Cell Line , Cell Line, Tumor , DNA-Binding Proteins/physiology , HeLa Cells/chemistry , HeLa Cells/metabolism , Histone Acetyltransferases , Humans , Kidney/chemistry , Kidney/cytology , Kidney/embryology , Kidney/metabolism , Mutation/genetics , Mutation/physiology , Peptides/metabolism , Protein Interaction Mapping/methods , Protein Structure, Tertiary , Response Elements/genetics , Retroviridae Proteins/physiology , Spumavirus/genetics , Trans-Activators/physiology , p300-CBP Transcription Factors
18.
J Virol ; 78(8): 4085-97, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15047825

ABSTRACT

The Nef protein of human immunodeficiency virus type 1 is an important factor in AIDS pathogenesis. In addition to downregulating CD4 and major histocompatibility complex class I molecules from the cell surface, as well as increasing virion infectivity, Nef triggers activation of the T-cell receptor (TCR) cascade to facilitate virus spread. Signaling pathways that are induced by Nef have been identified; however, it is unclear how and in which subcellular compartment Nef triggers signaling. Nef recruits a multiprotein complex to activate the cellular Pak kinase that mediates downstream effector functions. Since a subpopulation of Nef is present in detergent-insoluble microdomains (lipid rafts) from where physiological TCR signaling is initiated, we tested whether lipid rafts are instrumental for Nef-mediated Pak activation. In flotation analysis, Nef-associated Pak activity exclusively fractionated with lipid rafts. Activation of Pak in the presence of Nef coincided with lipid raft recruitment of the kinase, which was otherwise excluded from detergent-insoluble microdomains. Experimental solubilization of lipid rafts interfered with the association of Pak activity with Nef. To analyze the importance of the raft localization for Nef function more rigorously, we generated a palmitoylated Nef (PalmNef). PalmNef was highly enriched in lipid rafts and associated with significantly higher levels of Pak activity than Nef. Notably, activation of Pak by its physiological activators, Cdc42 and Rac, also occurred in lipid rafts and required raft integrity. Together, these data suggest that Nef induces signal transduction via the recruitment of a signaling machinery including Pak into lipid rafts, thereby mimicking a physiological cellular mechanism to initiate the TCR cascade.


Subject(s)
Gene Products, nef/physiology , HIV-1/physiology , HIV-1/pathogenicity , Membrane Microdomains/enzymology , Membrane Microdomains/virology , Protein Serine-Threonine Kinases/metabolism , Cell Line , Enzyme Activation , Gene Products, nef/genetics , HIV-1/genetics , Humans , Jurkat Cells , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , T-Lymphocytes/physiology , T-Lymphocytes/virology , cdc42 GTP-Binding Protein/metabolism , nef Gene Products, Human Immunodeficiency Virus , p21-Activated Kinases , rac GTP-Binding Proteins/metabolism
19.
J Biol Chem ; 278(40): 38902-12, 2003 Oct 03.
Article in English | MEDLINE | ID: mdl-12857739

ABSTRACT

Diaphanous related formins (DRFs) are part of the formin protein family that control morphogenesis, embryonic differentiation, cytokinesis, and cell polarity. DRFs organize the cytoskeleton in eukaryotic cells via the interaction with specific members of the Rho family of small GTPases including Rho, Rac, and Cdc42. This is best understood for Rho, which transmits signals to the actin cytoskeleton through the cooperation of its DRF effector mDia with ROCK (Rho-associated kinase). Here, we show that a constitutive active form of the Rac-interacting DRF FHOD1 (formin homology 2 domain containing 1) associates with F-actin in NIH3T3 cells, resulting in the formation of thick actin fibers. Cytoskeletal changes induced by FHOD1 correlated with the induction of serum response element transcription and were mediated by formin homology domains 1 and 2 of FHOD1. FHOD1-induced effects required the activity of the Rho-ROCK cascade that is targeted at a level downstream of Rho by the DRF. However, when the functional interaction of FHOD1 with individual GTPases was addressed, Rac but not Rho or Cdc42 bound to FHOD1 in cells and induced its recruitment to actin filaments and lamellipodia/membrane ruffles. Furthermore, activated FHOD1 interfered with lamellipodia formation. These results indicate that FHOD1 acts as an effector of Rac in actin rearrangements and transcriptional regulation and may provide a link for the Rac-dependent activation of the Rho cascade.


Subject(s)
Actins/metabolism , Fetal Proteins/metabolism , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , rac GTP-Binding Proteins/metabolism , 3T3 Cells , Actins/chemistry , Animals , Blotting, Western , Cytoskeleton/metabolism , Formins , GTP Phosphohydrolases/chemistry , Glutathione Transferase/metabolism , Green Fluorescent Proteins , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins , Luminescent Proteins/metabolism , Mice , Microscopy, Fluorescence , Plasmids/metabolism , Protein Binding , Protein Structure, Tertiary , Pseudopodia/chemistry , Time Factors , Transcription, Genetic , Transfection , rho-Associated Kinases
20.
J Biol Chem ; 278(17): 14961-70, 2003 Apr 25.
Article in English | MEDLINE | ID: mdl-12556465

ABSTRACT

Misfolding of the mammalian prion protein (PrP) is implicated in the pathogenesis of prion diseases. We analyzed wild type PrP in comparison with different PrP mutants and identified determinants of the in vivo folding pathway of PrP. The complete N terminus of PrP including the putative transmembrane domain and the first beta-strand could be deleted without interfering with PrP maturation. Helix 1, however, turned out to be a major determinant of PrP folding. Disruption of helix 1 prevented attachment of the glycosylphosphatidylinositol (GPI) anchor and the formation of complex N-linked glycans; instead, a high mannose PrP glycoform was secreted into the cell culture supernatant. In the absence of a C-terminal membrane anchor, however, helix 1 induced the formation of unglycosylated and partially protease-resistant PrP aggregates. Moreover, we could show that the C-terminal GPI anchor signal sequence, independent of its role in GPI anchor attachment, mediates core glycosylation of nascent PrP. Interestingly, conversion of high mannose glycans to complex type glycans only occurred when PrP was membrane-anchored. Our study indicates a bipartite function of helix 1 in the maturation and aggregation of PrP and emphasizes a critical role of a membrane anchor in the formation of complex glycosylated PrP.


Subject(s)
PrPC Proteins/chemistry , Protein Folding , Protein Processing, Post-Translational , Animals , Cell Membrane/chemistry , Glycosylation , Glycosylphosphatidylinositols , Mannose , Mice , Polysaccharides/biosynthesis , Polysaccharides/chemistry , PrPC Proteins/biosynthesis , PrPC Proteins/genetics , PrPC Proteins/metabolism , Protein Structure, Tertiary , Protein Transport , Transfection , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...