Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 106
Filter
Add more filters

Publication year range
1.
Cell Mol Life Sci ; 77(24): 5223-5242, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32065241

ABSTRACT

Endocytosis of the amyloid precursor protein (APP) is critical for generation of ß-amyloid, aggregating in Alzheimer's disease. APP endocytosis depending on the intracellular NPTY motif is well investigated, whereas involvement of the YTSI (also termed BaSS) motif remains controversial. Here, we show that APP lacking the YTSI motif (ΔYTSI) displays reduced localization to early endosomes and decreased internalization rates, similar to APP ΔNPTY. Additionally, we show that the YTSI-binding protein, PAT1a interacts with the Rab5 activator RME-6, as shown by several independent assays. Interestingly, knockdown of RME-6 decreased APP endocytosis, whereas overexpression increased the same. Similarly, APP ΔNPTY endocytosis was affected by PAT1a and RME-6 overexpression, whereas APP ΔYTSI internalization remained unchanged. Moreover, we could show that RME-6 mediated increase of APP endocytosis can be diminished upon knocking down PAT1a. Together, our data identify RME-6 as a novel player in APP endocytosis, involving the YTSI-binding protein PAT1a.


Subject(s)
Alzheimer Disease/genetics , Amino Acid Motifs/genetics , Amyloid beta-Protein Precursor/genetics , rab5 GTP-Binding Proteins/genetics , Alzheimer Disease/pathology , Amyloid beta-Peptides/genetics , Animals , Carrier Proteins/genetics , Endocytosis/genetics , Endosomes/genetics , Humans , Mice , Protein Transport/genetics , Transport Vesicles/genetics
2.
Genes Dev ; 25(8): 875-88, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21498574

ABSTRACT

Spatial and timely coordination of cytokinesis is crucial for the maintenance of organelle inheritance and genome integrity. The mitotic exit network (MEN) pathway controls both the timely initiation of mitotic exit and cytokinesis in budding yeast. Here we identified the conserved F-BAR protein Hof1 as a substrate of the MEN kinase complex Dbf2-Mob1 during cytokinesis. We show that polo-like kinase Cdc5 first phosphorylates Hof1 to allow subsequent phosphorylation by Dbf2-Mob1. This releases Hof1 from the septin ring and facilitates Hof1 binding to the medial actomyosin ring (AMR), where Hof1 promotes AMR contraction and membrane ingression. Domain structure analysis established that the central, unstructured, region of Hof1, named the ring localization sequence (RLS), is sufficient to mediate Hof1's binding to the medial ring in a cell cycle-dependent manner. Genetic and functional data support a model in which Dbf2-Mob1 regulates Hof1 by inducing domain rearrangements, leading to the exposure of the Hof1 RLS domain during telophase.


Subject(s)
Cytokinesis/physiology , Microtubule-Associated Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Blotting, Western , Cell Cycle/genetics , Cell Cycle/physiology , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cytokinesis/genetics , Immunoprecipitation , Microtubule-Associated Proteins/genetics , Mitosis/genetics , Mitosis/physiology , Phosphoproteins/genetics , Phosphoproteins/metabolism , Phosphorylation , Protein Kinases/genetics , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Saccharomyces cerevisiae Proteins/genetics
3.
PLoS Genet ; 9(4): e1003373, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23593011

ABSTRACT

Non-coding RNAs are much more common than previously thought. However, for the vast majority of non-coding RNAs, the cellular function remains enigmatic. The two long non-coding RNA (lncRNA) genes DLEU1 and DLEU2 map to a critical region at chromosomal band 13q14.3 that is recurrently deleted in solid tumors and hematopoietic malignancies like chronic lymphocytic leukemia (CLL). While no point mutations have been found in the protein coding candidate genes at 13q14.3, they are deregulated in malignant cells, suggesting an epigenetic tumor suppressor mechanism. We therefore characterized the epigenetic makeup of 13q14.3 in CLL cells and found histone modifications by chromatin-immunoprecipitation (ChIP) that are associated with activated transcription and significant DNA-demethylation at the transcriptional start sites of DLEU1 and DLEU2 using 5 different semi-quantitative and quantitative methods (aPRIMES, BioCOBRA, MCIp, MassARRAY, and bisulfite sequencing). These epigenetic aberrations were correlated with transcriptional deregulation of the neighboring candidate tumor suppressor genes, suggesting a coregulation in cis of this gene cluster. We found that the 13q14.3 genes in addition to their previously known functions regulate NF-kB activity, which we could show after overexpression, siRNA-mediated knockdown, and dominant-negative mutant genes by using Western blots with previously undescribed antibodies, by a customized ELISA as well as by reporter assays. In addition, we performed an unbiased screen of 810 human miRNAs and identified the miR-15/16 family of genes at 13q14.3 as the strongest inducers of NF-kB activity. In summary, the tumor suppressor mechanism at 13q14.3 is a cluster of genes controlled by two lncRNA genes that are regulated by DNA-methylation and histone modifications and whose members all regulate NF-kB. Therefore, the tumor suppressor mechanism in 13q14.3 underlines the role both of epigenetic aberrations and of lncRNA genes in human tumorigenesis and is an example of colocalization of a functionally related gene cluster.


Subject(s)
DNA Methylation/genetics , Leukemia , RNA, Long Noncoding , Tumor Suppressor Proteins , Adult , Aged , Aged, 80 and over , Cell Transformation, Neoplastic , Chromatin/genetics , Chromosomes, Human, Pair 13/genetics , Down-Regulation , Epigenesis, Genetic/genetics , Female , HEK293 Cells , Humans , Leukemia/blood , Leukemia/genetics , Leukemia/physiopathology , Male , Middle Aged , Mutation , NF-kappa B/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Transcription Initiation Site , Transferases , Tumor Suppressor Proteins/blood , Tumor Suppressor Proteins/genetics , Up-Regulation
4.
J Cell Sci ; 125(Pt 5): 1353-62, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22349705

ABSTRACT

Centrioles are key structural elements of centrosomes and primary cilia. In mammals, only a few proteins including PLK4, CPAP (CENPJ), SAS6, CEP192, CEP152 and CEP135 have thus far been identified to be required for centriole duplication. STIL (SCL/TAL1 interrupting locus, also known as SIL) is a centrosomal protein that is essential for mouse and zebrafish embryonic development and mutated in primary microcephaly. Here, we show that STIL localizes to the pericentriolar material surrounding parental centrioles. Its overexpression results in excess centriole formation. siRNA-mediated depletion of STIL leads to loss of centrioles and abrogates PLK4-induced centriole overduplication. Additionally, we show that STIL is necessary for SAS6 recruitment to centrioles, suggesting that it is essential for daughter centriole formation, interacts with the centromere protein CPAP and rapidly shuttles between the cytoplasm and centrioles. Consistent with the requirement of centrioles for cilia formation, Stil(-/-) mouse embryonic fibroblasts lack primary cilia--a phenotype that can be reverted by restoration of STIL expression. These findings demonstrate that STIL is an essential component of the centriole replication machinery in mammalian cells.


Subject(s)
Centrioles/metabolism , Cilia/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Cell Cycle , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Division/physiology , Cell Line , Centrioles/genetics , Centrosome/physiology , Cytoplasm/physiology , HEK293 Cells , Humans , Microtubule-Associated Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , RNA Interference , RNA, Small Interfering
5.
Cell Microbiol ; 15(2): 227-36, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23051660

ABSTRACT

Foamy viruses (FVs), a unique type of retroviruses, are characterized by several unusual features in their replication strategy. FVs, common to all non-human primates and several other species, display an extremely broad tropism in vitro. Basically, all mammalian cells and species examined, but also cells of amphibian or bird origin, are permissive to FV glycoprotein (Env)-mediated capsid release into the cytoplasm. The nature of the broadly expressed, and potentially evolutionary conserved, FV entry receptor molecule(s) is poorly characterized. Although recent data indicate that proteoglycans serve as an important factor for FV Env-mediated target cell attachment, additional uncharacterized molecules appear to be essential for the pH-dependent fusion of viral and cellular lipid membranes after endocytic uptake of virions. Furthermore, FVs show a very special assembly strategy. Unlike other retroviruses, the FV capsid precursor protein (Gag) undergoes only very limited proteolytic processing during assembly. This results in an immature morphology of capsids found in released FV virions. In addition, the FV Gag protein appears to lack a functional membrane-targeting signal. As a consequence, FVs utilize a specific interaction between capsid and cognate viral glycoprotein for initiation of thebudding process. Genetic fusion of heterologous targeting domains for plasma but not endosomal membranes to FV Gag enables glycoprotein-independent particle egress. However, this is at the expense of normal capsid morphogenesis and infectivity. The low-level Gag precursor processing and the requirement for a reversible, artificial Gag membrane association for effective pseudotyping of FV capsids by heterologous glycoproteins strongly suggest that FVs require a transient interaction of capsids with cellular membranes for viral replication. Under natural condition, this appears to be achieved by the lack of a membrane-targeting function of the FV Gag protein and the accomplishment of capsid membrane attachment through an unusual specific interaction with the cognate glycoprotein.


Subject(s)
Capsid/chemistry , Gene Products, gag/genetics , Spumavirus/chemistry , Virion/chemistry , Virus Assembly/physiology , Animals , Capsid/metabolism , Capsid/ultrastructure , Cell Membrane/chemistry , Cell Membrane/virology , Endocytosis , Gene Products, gag/metabolism , Glycoproteins/genetics , Glycoproteins/metabolism , HEK293 Cells , Humans , Hydrogen-Ion Concentration , Membrane Fusion , Spumavirus/metabolism , Spumavirus/ultrastructure , Virion/metabolism , Virion/ultrastructure , Virus Internalization , Virus Replication
6.
Nature ; 456(7222): 611-6, 2008 Dec 04.
Article in English | MEDLINE | ID: mdl-19052621

ABSTRACT

Cilia and flagella are highly conserved organelles that have diverse roles in cell motility and sensing extracellular signals. Motility defects in cilia and flagella often result in primary ciliary dyskinesia. However, the mechanisms underlying cilia formation and function, and in particular the cytoplasmic assembly of dyneins that power ciliary motility, are only poorly understood. Here we report a new gene, kintoun (ktu), involved in this cytoplasmic process. This gene was first identified in a medaka mutant, and found to be mutated in primary ciliary dyskinesia patients from two affected families as well as in the pf13 mutant of Chlamydomonas. In the absence of Ktu/PF13, both outer and inner dynein arms are missing or defective in the axoneme, leading to a loss of motility. Biochemical and immunohistochemical studies show that Ktu/PF13 is one of the long-sought proteins involved in pre-assembly of dynein arm complexes in the cytoplasm before intraflagellar transport loads them for the ciliary compartment.


Subject(s)
Axoneme/metabolism , Cilia/metabolism , Dyneins/metabolism , Fish Proteins/metabolism , Oryzias , Proteins/metabolism , Animals , Axoneme/chemistry , Axoneme/genetics , Axoneme/pathology , Chlamydomonas/genetics , Chlamydomonas/metabolism , Cilia/chemistry , Cilia/genetics , Cilia/pathology , Cloning, Molecular , Epithelial Cells/cytology , Fish Proteins/genetics , Genes, Recessive/genetics , HSP70 Heat-Shock Proteins/metabolism , Humans , Kartagener Syndrome/genetics , Kartagener Syndrome/pathology , Male , Mice , Molecular Sequence Data , Mutation/genetics , Oryzias/embryology , Oryzias/genetics , Oryzias/metabolism , Protein Binding , Proteins/genetics , Sequence Homology, Amino Acid , Sperm Motility , Testis/cytology
7.
EMBO J ; 28(4): 315-25, 2009 Feb 18.
Article in English | MEDLINE | ID: mdl-19131969

ABSTRACT

The recently identified type VI secretion systems (T6SS) have a crucial function in the virulence of various proteobacteria, including the human pathogen Vibrio cholerae. T6SS are encoded by a conserved gene cluster comprising approximately 15 open reading frames, mediating the appearance of Hcp and VgrG proteins in cell culture supernatants. Here, we analysed the function of the V. cholerae T6SS member ClpV, a specialized AAA+ protein. ClpV is crucial for a functional T6SS and interacts through its N-terminal domain with the VipA/VipB complex that is composed of two conserved and essential members of T6SS. Transferring ClpV substrate specificity to a distinct AAA+ protein involved in proteolysis caused degradation of VipA but not Hcp or VgrG2, suggesting that VipA rather than Hcp/VgrG2 functions as a primary ClpV substrate. Strikingly, VipA/VipB form tubular, cogwheel-like structures that are converted by a threading activity of ClpV into small complexes. ClpV-mediated remodelling of VipA/VipB tubules represents a crucial step in T6S, illuminating an unexpected role of an ATPase component in protein secretion.


Subject(s)
Adenosine Triphosphatases/physiology , Carrier Proteins/physiology , Vibrio cholerae/metabolism , Bacterial Proteins/metabolism , Biological Transport , Gene Expression Regulation, Bacterial , Microscopy, Electron , Molecular Chaperones/metabolism , Multigene Family , Open Reading Frames , Plasmids/metabolism , Protein Binding , Protein Structure, Tertiary , Virulence , Virulence Factors/metabolism
8.
Cancer Invest ; 31(9): 563-70, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24164297

ABSTRACT

We used a [(32)P] p53 sequence-specific oligodeoxynucleotide and Electrophoretic-Mobility-Shift-Assays to monitor p53 DNA sequence-specific binding with p53-R267W, a nonbinding point mutant; and p53-Δ30, a deletion-mutant which lacks the carboxy-terminus that recognizes DNA-strand-breaks. Recombinant p53 and poly(ADP-ribose)polymerase-1 (PARP-1) were incubated with labeled ßNAD(+) with/without DNA. The poly(ADP-ribosyl)ation of each protein increased with incubation-time and ßNAD(+) and p53 concentration(s). Since p53-Δ30 was efficiently labeled, poly(ADP-ribosyl)ation target site(s) of wt-p53 must reside outside its carboxy-terminal-domain. The poly(ADP-ribosyl)ation of p53-Δ30 did not diminish its DNA binding; Instead, it enhanced DNA-sequence-specific-binding. Therefore, we conclude that DNA-sequence-specific-binding and DNA-nick-sensing of mutant-p53 are differentially regulated by poly(ADP-ribosyl)ation.


Subject(s)
DNA Breaks, Single-Stranded , DNA/metabolism , Poly Adenosine Diphosphate Ribose/metabolism , Transcriptional Activation , Tumor Suppressor Protein p53/metabolism , Binding Sites , Electrophoretic Mobility Shift Assay , Humans , NAD/metabolism , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/metabolism , Protein Processing, Post-Translational , Recombinant Proteins/metabolism , Sequence Deletion , Time Factors , Tumor Suppressor Protein p53/genetics , Up-Regulation
9.
J Pathol ; 227(3): 325-35, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22262369

ABSTRACT

Involvement of dysregulated autophagy in cancer growth and progression has been shown in different tumour entities, including pancreatic ductal adenocarcinoma (PDA). PDA is an extremely aggressive tumour characterized by a small population of highly therapy-resistant cancer stem cells (CSCs) capable of self-renewal and migration. We examined whether autophagy might be involved in the survival of CSCs despite nutrition and oxygen deprivation typical for the hypoxic tumour microenvironment of PDA. Immunohistochemistry revealed that markers for hypoxia, CSCs and autophagy are co-expressed in patient-derived tissue of PDA. Hypoxia starvation (H/S) enhanced clonogenic survival and migration of established pancreatic cancer cells with stem-like properties (CSC(high)), while pancreatic tumour cells with fewer stem cell markers (CSC(low)) did not survive these conditions. Electron microscopy revealed more advanced autophagic vesicles in CSC(high) cells, which exhibited higher expression of autophagy-related genes under normoxic conditions and relative to CSC(low) cells, as found by RT-PCR and western blot analysis. LC3 was already fully converted to the active LC3-II form in both cell lines, as evaluated by western blot and detection of accumulated GFP-LC3 protein by fluorescence microscopy. H/S increased formation of autophagic and acid vesicles, as well as expression of autophagy-related genes, to a higher extent in CSC(high) cells. Modulation of autophagy by inhibitors and activators resensitized CSC(high) to apoptosis and diminished clonogenicity, spheroid formation, expression of CSC-related genes, migratory activity and tumourigenicity in mice. Our data suggest that enhanced autophagy levels may enable survival of CSC(high) cells under H/S. Interference with autophagy-activating or -inhibiting drugs disturbs the fine-tuned physiological balance of enhanced autophagy in CSC and switches survival signalling to suicide.


Subject(s)
Autophagy , Carcinoma, Pancreatic Ductal/pathology , Neoplastic Stem Cells/pathology , Pancreatic Neoplasms/pathology , Tumor Microenvironment , Animals , Antineoplastic Agents/pharmacology , Autophagy/drug effects , Autophagy/genetics , Biomarkers, Tumor/metabolism , Blotting, Western , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/ultrastructure , Cell Hypoxia , Cell Line, Tumor , Cell Movement , Cell Survival , Female , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Mice , Mice, Nude , Microscopy, Electron , Microscopy, Fluorescence , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/ultrastructure , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/ultrastructure , Polymerase Chain Reaction , Time Factors , Tumor Burden
10.
J Biol Chem ; 286(34): 30010-21, 2011 Aug 26.
Article in English | MEDLINE | ID: mdl-21733841

ABSTRACT

Ring-forming AAA(+) ATPases act in a plethora of cellular processes by remodeling macromolecules. The specificity of individual AAA(+) proteins is achieved by direct or adaptor-mediated association with substrates via distinct recognition domains. We investigated the molecular basis of substrate interaction for Vibrio cholerae ClpV, which disassembles tubular VipA/VipB complexes, an essential step of type VI protein secretion and bacterial virulence. We identified the ClpV recognition site within VipB, showed that productive ClpV-VipB interaction requires the oligomeric state of both proteins, solved the crystal structure of a ClpV N-domain-VipB peptide complex, and verified the interaction surface by mutant analysis. Our results show that the substrate is bound to a hydrophobic groove, which is formed by the addition of a single α-helix to the core N-domain. This helix is absent from homologous N-domains, explaining the unique substrate specificity of ClpV. A limited interaction surface between both proteins accounts for the dramatic increase in binding affinity upon ATP-driven ClpV hexamerization and VipA/VipB tubule assembly by coupling multiple weak interactions. This principle ensures ClpV selectivity toward the VipA/VipB macromolecular complex.


Subject(s)
Adenosine Triphosphatases/chemistry , Bacterial Secretion Systems/physiology , Molecular Chaperones/chemistry , Protein Multimerization/physiology , Vibrio cholerae/enzymology , Binding Sites , Crystallography, X-Ray , Protein Structure, Secondary , Protein Structure, Tertiary
11.
J Biol Chem ; 286(16): 14237-45, 2011 Apr 22.
Article in English | MEDLINE | ID: mdl-21357692

ABSTRACT

Nephronophthisis is the most common genetic cause of end-stage renal failure during childhood and adolescence. Genetic studies have identified disease-causing mutations in at least 11 different genes (NPHP1-11), but the function of the corresponding nephrocystin proteins remains poorly understood. The two evolutionarily conserved proteins nephrocystin-1 (NPHP1) and nephrocystin-4 (NPHP4) interact and localize to cilia in kidney, retina, and brain characterizing nephronophthisis and associated pathologies as result of a ciliopathy. Here we show that NPHP4, but not truncating patient mutations, negatively regulates tyrosine phosphorylation of NPHP1. NPHP4 counteracts Pyk2-mediated phosphorylation of three defined tyrosine residues of NPHP1 thereby controlling binding of NPHP1 to the trans-Golgi sorting protein PACS-1. Knockdown of NPHP4 resulted in an accumulation of NPHP1 in trans-Golgi vesicles of ciliated retinal epithelial cells. These data strongly suggest that NPHP4 acts upstream of NPHP1 in a common pathway and support the concept of a role for nephrocystin proteins in intracellular vesicular transport.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cilia/metabolism , Focal Adhesion Kinase 2/metabolism , Gene Expression Regulation , Membrane Proteins/metabolism , Proteins/physiology , Tyrosine/chemistry , Cell Line , Cytoskeletal Proteins , Golgi Apparatus/metabolism , Humans , Kidney Diseases, Cystic/metabolism , Models, Biological , Mutation , Phosphorylation , Protein Binding , Tissue Distribution
12.
Am J Hum Genet ; 85(6): 883-9, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19944400

ABSTRACT

Genetic defects affecting motility of cilia and flagella cause chronic destructive airway disease, randomization of left-right body asymmetry, and, frequently, male infertility in primary ciliary dyskinesia (PCD). The most frequent defects involve outer and inner dynein arms (ODAs and IDAs) that are large multiprotein complexes responsible for cilia-beat generation and regulation, respectively. Here, we demonstrate that large genomic deletions, as well as point mutations involving LRRC50, are responsible for a distinct PCD variant that is characterized by a combined defect involving assembly of the ODAs and IDAs. Functional analyses showed that LRRC50 deficiency disrupts assembly of distally and proximally DNAH5- and DNAI2-containing ODA complexes, as well as DNALI1-containing IDA complexes, resulting in immotile cilia. On the basis of these findings, we assume that LRRC50 plays a role in assembly of distinct dynein-arm complexes.


Subject(s)
Dyneins/genetics , Gene Deletion , Kartagener Syndrome/genetics , Microtubule-Associated Proteins/physiology , Point Mutation , Proteins/genetics , Adolescent , Adult , Alleles , Animals , Chromosomes/ultrastructure , DNA Mutational Analysis , Female , Flagella , Genomics , Humans , Male , Mice , Microtubule-Associated Proteins/genetics , Models, Genetic , Mutation , Proteins/metabolism
13.
J Cell Sci ; 123(Pt 11): 1851-61, 2010 Jun 01.
Article in English | MEDLINE | ID: mdl-20442249

ABSTRACT

The mitotic-exit network (MEN) is a signaling pathway that is essential for the coordination of mitotic exit and cytokinesis. Whereas the role of the MEN in mitotic exit is well established, the molecular mechanisms by which MEN components regulate cytokinesis remain poorly understood. Here, we show that the MEN controls components involved in septum formation, including Inn1, Cyk3 and Chs2. MEN-deficient mutants, forced to exit mitosis as a result of Cdk1 inactivation, show defects in targeting Cyk3 and Inn1 to the bud-neck region. In addition, we found that the chitin synthase Chs2 did not efficiently localize at the bud neck in the absence of MEN activity. Ultrastructural analysis of the bud neck revealed that low MEN activity led to unilateral, uncoordinated extension of the primary and secondary septa. This defect was partially suppressed by increased levels of Cyk3. We therefore propose that the MEN directly controls cytokinesis via targeting of Inn1, Cyk3 and Chs2 to the bud neck.


Subject(s)
CDC2 Protein Kinase/metabolism , Cell Cycle Proteins/metabolism , Chitin Synthase/metabolism , Microtubule-Associated Proteins/deficiency , Saccharomyces cerevisiae Proteins/metabolism , Saccharomycetales/physiology , CDC2 Protein Kinase/genetics , Cloning, Molecular , Cytokinesis/genetics , Microscopy, Fluorescence , Mitosis/genetics , Mutagenesis, Site-Directed , Myosin Heavy Chains/deficiency , Sequence Deletion/genetics , Signal Transduction/genetics
14.
J Virol ; 85(4): 1452-63, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21106749

ABSTRACT

Prototype foamy virus (PFV) Gag lacks the characteristic orthoretroviral Cys-His motifs that are essential for various steps of the orthoretroviral replication cycle, such as RNA packaging, reverse transcription, infectivity, integration, and viral assembly. Instead, it contains three glycine-arginine-rich boxes (GR boxes) in its C terminus that putatively represent a functional equivalent. We used a four-plasmid replication-deficient PFV vector system, with uncoupled RNA genome packaging and structural protein translation, to analyze the effects of deletion and various substitution mutations within each GR box on particle release, particle-associated protein composition, RNA packaging, DNA content, infectivity, particle morphology, and intracellular localization. The degree of viral particle release by all mutants was similar to that of the wild type. Only minimal effects on Pol encapsidation, exogenous reverse transcriptase (RT) activity, and genomic viral RNA packaging were observed. In contrast, particle-associated DNA content and infectivity were drastically reduced for all deletion mutants and were undetectable for all alanine substitution mutants. Furthermore, GR box I mutants had significant changes in particle morphology, and GR box II mutants lacked the typical nuclear localization pattern of PFV Gag. Finally, it could be shown that GR boxes I and III, but not GR box II, can functionally complement each other. It therefore appears that, similar to the orthoretroviral Cys-His motifs, the PFV Gag GR boxes are important for RNA encapsidation, genome reverse transcription, and virion infectivity as well as for particle morphogenesis.


Subject(s)
Gene Products, gag/chemistry , Reverse Transcription , Spumavirus/growth & development , Virion/growth & development , Virus Assembly , Amino Acid Sequence , Arginine/chemistry , Cell Line , Gene Products, gag/genetics , Gene Products, gag/metabolism , Glycine/chemistry , HeLa Cells , Humans , Molecular Sequence Data , RNA, Viral/genetics , RNA, Viral/metabolism , Spumavirus/genetics , Spumavirus/metabolism , Spumavirus/pathogenicity , Virion/genetics , Virion/metabolism , Virus Replication
15.
J Cell Biol ; 177(4): 573-8, 2007 May 21.
Article in English | MEDLINE | ID: mdl-17517959

ABSTRACT

Considerable efforts are being undertaken to elucidate the processes of ribosome biogenesis. Although various preribosomal RNP complexes have been isolated and molecularly characterized, the order of ribosomal protein (r-protein) addition to the emerging ribosome subunits is largely unknown. Furthermore, the correlation between the ribosome assembly pathway and the structural organization of the dedicated ribosome factory, the nucleolus, is not well established. We have analyzed the nucleolar localization of several early binding r-proteins in human cells, applying various methods, including live-cell imaging and electron microscopy. We have located all examined r-proteins (S4, S6, S7, S9, S14, and L4) in the granular component (GC), which is the nucleolar region where later pre-ribosomal RNA (rRNA) processing steps take place. These results imply that early binding r-proteins do not assemble with nascent pre-rRNA transcripts in the dense fibrillar component (DFC), as is generally believed, and provide a link between r-protein assembly and the emergence of distinct granules at the DFC-GC interface.


Subject(s)
Cell Nucleolus/metabolism , Ribosomal Proteins/biosynthesis , Ribosomes/metabolism , Cell Line, Tumor , Cell Nucleolus/ultrastructure , Humans , Ribosomal Proteins/metabolism , Ribosomal Proteins/ultrastructure , Ribosomes/ultrastructure
16.
J Biol Chem ; 285(28): 21644-54, 2010 Jul 09.
Article in English | MEDLINE | ID: mdl-20452983

ABSTRACT

PEA-15/PED (phosphoprotein enriched in astrocytes 15 kDa/phosphoprotein enriched in diabetes) is a death effector domain-containing protein which is known to modulate apoptotic cell death. The mechanism by which PEA-15 inhibits caspase activation and increases ERK (extracellular-regulated kinase) activity is well characterized. Here, we demonstrate that PEA-15 is not only pivotal in the activation of the ERK pathway but also modulates JNK (c-Jun N-terminal kinase) signaling. Upon overexpression of PEA-15 in malignant glioma cells, JNK is potently activated. The PEA-15-induced JNK activation depends on the phosphorylation of PEA-15 at both phosphorylation sites (serine 104 and serine 116). The activation of JNK is substantially inhibited by siRNA-mediated down-regulation of endogenous PEA-15. Moreover, we demonstrate that glioma cells overexpressing PEA-15 show increased signs of autophagy in response to classical autophagic stimuli such as ionizing irradiation, serum deprivation, or rapamycin treatment. In contrast, the non-phosphorylatable mutants of PEA-15 are not capable of promoting autophagy. The inhibition of JNK abrogates the PEA-15-mediated increase in autophagy. In conclusion, our data show that PEA-15 promotes autophagy in glioma cells in a JNK-dependent manner. This might render glioma cells more resistant to adverse stimuli such as starvation or ionizing irradiation.


Subject(s)
Autophagy , Brain Neoplasms/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation, Neoplastic , Glioma/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Phosphoproteins/metabolism , Apoptosis , Apoptosis Regulatory Proteins , Cell Line, Tumor , Flow Cytometry , Humans , Models, Biological , Phosphorylation , RNA, Small Interfering/metabolism , Transfection
17.
Retrovirology ; 8: 66, 2011 Aug 15.
Article in English | MEDLINE | ID: mdl-21843316

ABSTRACT

BACKGROUND: Foamy viruses (FVs) unlike orthoretroviruses express Pol as a separate precursor protein and not as a Gag-Pol fusion protein. A unique packaging strategy, involving recognition of briding viral RNA by both Pol precursor and Gag as well as potential Gag-Pol protein interactions, ensures Pol particle encapsidation. RESULTS: Several Prototype FV (PFV) Gag-Pol fusion protein constructs were generated to examine whether PFV replication is compatible with an orthoretroviral-like Pol expression. During their analysis, non-particle-associated secreted Pol precursor protein was discovered in extracellular wild type PFV particle preparations of different origin, copurifying in simple virion enrichment protocols. Different analysis methods suggest that extracellular wild type PFV particles contain predominantly mature p85(PR-RT) and p40(IN) Pol subunits. Characterization of various PFV Gag-Pol fusion constructs revealed that PFV Pol expression in an orthoretroviral manner is compatible with PFV replication as long as a proteolytic processing between Gag and Pol proteins is possible. PFV Gag-Pol translation by a HIV-1 like ribosomal frameshift signal resulted in production of replication-competent virions, although cell- and particle-associated Pol levels were reduced in comparison to wild type. In-frame fusion of PFV Gag and Pol ORFs led to increased cellular Pol levels, but particle incorporation was only marginally elevated. Unlike that reported for similar orthoretroviral constructs, a full-length in-frame PFV Gag-Pol fusion construct showed wildtype-like particle release and infectivity characteristics. In contrast, in-frame PFV Gag-Pol fusion with C-terminal Gag ORF truncations or non-removable Gag peptide addition to Pol displayed wildtype particle release, but reduced particle infectivity. PFV Gag-Pol precursor fusion proteins with inactivated protease were highly deficient in regular particle release, although coexpression of p71(Gag) resulted in a significant copackaging of these proteins. CONCLUSIONS: Non-particle associated PFV Pol appears to be naturally released from infected cells by a yet unknown mechanism. The absence of particle-associated Pol precursor suggests its rapid processing upon particle incorporation. Analysis of different PFV Gag-Pol fusion constructs demonstrates that orthoretroviral-like Pol expression is compatible with FV replication in principal as long as fusion protein processing is possible. Furthermore, unlike orthoretroviruses, PFV particle release and infectivity tolerate larger differences in relative cellular Gag/Pol levels.


Subject(s)
Fusion Proteins, gag-pol/metabolism , Gene Expression Regulation, Viral , Retroviridae Infections/virology , Spumavirus/physiology , Virus Replication , Animals , Cell Line , Cricetinae , Fusion Proteins, gag-pol/genetics , Humans , Spumavirus/genetics
18.
Am J Hum Genet ; 82(4): 959-70, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18371931

ABSTRACT

Many genetic diseases have been linked to the dysfunction of primary cilia, which occur nearly ubiquitously in the body and act as solitary cellular mechanosensory organelles. The list of clinical manifestations and affected tissues in cilia-related disorders (ciliopathies) such as nephronophthisis is broad and has been attributed to the wide expression pattern of ciliary proteins. However, little is known about the molecular mechanisms leading to this dramatic diversity of phenotypes. We recently reported hypomorphic NPHP3 mutations in children and young adults with isolated nephronophthisis and associated hepatic fibrosis or tapetoretinal degeneration. Here, we chose a combinatorial approach in mice and humans to define the phenotypic spectrum of NPHP3/Nphp3 mutations and the role of the nephrocystin-3 protein. We demonstrate that the pcy mutation generates a hypomorphic Nphp3 allele that is responsible for the cystic kidney disease phenotype, whereas complete loss of Nphp3 function results in situs inversus, congenital heart defects, and embryonic lethality in mice. In humans, we show that NPHP3 mutations can cause a broad clinical spectrum of early embryonic patterning defects comprising situs inversus, polydactyly, central nervous system malformations, structural heart defects, preauricular fistulas, and a wide range of congenital anomalies of the kidney and urinary tract (CAKUT). On the functional level, we show that nephrocystin-3 directly interacts with inversin and can inhibit like inversin canonical Wnt signaling, whereas nephrocystin-3 deficiency leads in Xenopus laevis to typical planar cell polarity defects, suggesting a role in the control of canonical and noncanonical (planar cell polarity) Wnt signaling.


Subject(s)
Abnormalities, Multiple/genetics , Fetal Death/genetics , Kidney Diseases, Cystic/genetics , Kinesins/genetics , Situs Inversus/genetics , Adolescent , Animals , Child , Female , Humans , Infant, Newborn , Kidney/abnormalities , Kinesins/metabolism , Liver/abnormalities , Male , Mice , Mice, Mutant Strains , Mutation , Pancreas/abnormalities , Pedigree , Syndrome , Transcription Factors/metabolism , Wnt Proteins/metabolism , Xenopus laevis
19.
Nat Methods ; 5(5): 439-45, 2008 May.
Article in English | MEDLINE | ID: mdl-18391960

ABSTRACT

The complexity of the angiogenic cascade limits cellular approaches to studying angiogenic endothelial cells (ECs). In turn, in vivo assays do not allow the analysis of the distinct cellular behavior of ECs during angiogenesis. Here we show that ECs can be grafted as spheroids into a matrix to give rise to a complex three-dimensional network of human neovessels in mice. The grafted vasculature matures and is connected to the mouse circulation. The assay is highly versatile and facilitates numerous applications including studies of the effects of different cytokines on angiogenesis. Modifications make it possible to study human lymphangiogenic processes in vivo. EC spheroids can also be coimplanted with other cell types for tissue engineering purposes.


Subject(s)
Cell Culture Techniques/methods , Endothelial Cells/cytology , Neovascularization, Physiologic/physiology , Spheroids, Cellular/cytology , Animals , Cell Communication , Endothelial Cells/drug effects , Fibroblast Growth Factor 2/pharmacology , Humans , Mice , Tissue Engineering , Vascular Endothelial Growth Factor A/pharmacology
20.
J Virol ; 84(12): 5909-22, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20375165

ABSTRACT

The rat parvovirus H-1 (H-1PV) attracts high attention as an anticancer agent, because it is not pathogenic for humans and has oncotropic and oncosuppressive properties. The viral nonstructural NS1 protein is thought to mediate H-1PV cytotoxicity, but its exact contribution to this process remains undefined. In this study, we analyzed the effects of the H-1PV NS1 protein on human cell proliferation and cell viability. We show that NS1 expression is sufficient to induce the accumulation of cells in G(2) phase, apoptosis via caspase 9 and 3 activation, and cell lysis. Similarly, cells infected with wild-type H-1PV arrest in G(2) phase and undergo apoptosis. Furthermore, we also show that both expression of NS1 and H-1PV infection lead to higher levels of intracellular reactive oxygen species (ROS), associated with DNA double-strand breaks. Antioxidant treatment reduces ROS levels and strongly decreases NS1- and virus-induced DNA damage, cell cycle arrest, and apoptosis, indicating that NS1-induced ROS are important mediators of H-1PV cytotoxicity.


Subject(s)
Apoptosis , H-1 parvovirus/metabolism , Parvoviridae Infections/metabolism , Parvoviridae Infections/physiopathology , Reactive Oxygen Species/metabolism , Viral Nonstructural Proteins/metabolism , Cell Cycle , Cell Line , DNA Damage , H-1 parvovirus/genetics , Humans , Parvoviridae Infections/genetics , Parvoviridae Infections/virology , Viral Nonstructural Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL