Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
J Virol ; 88(22): 13161-72, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25187543

ABSTRACT

UNLABELLED: Kaposi's sarcoma herpesvirus (KSHV) (or human herpesvirus 8) is the cause of Kaposi's sarcoma, primary effusion lymphoma (PEL), and the plasma cell variant of multicentric Castleman's disease (MCD). The transmembrane K15 protein, encoded by KSHV, has been shown to activate NF-κB and the mitogen-activated protein kinases (MAPKs) c-jun-N-terminal kinase (JNK) and extracellular signal-regulated kinase (Erk) as well as phospholipase C gamma (PLCγ) and to contribute to KSHV-induced angiogenesis. Here we investigate how the K15 protein activates the NF-κB pathway. We show that activation of NF-κB involves the recruitment of NF-κB-inducing kinase (NIK) and IKK α/ß to result in the phosphorylation of p65/RelA on Ser536. A K15 mutant devoid in NIK/IKK recruitment fails to activate NF-κB but remains proficient in the stimulation of both NFAT- and AP1-dependent promoters, showing that the structural integrity of the mutant K15 protein has not been altered dramatically. Direct recruitment of NIK represents a novel way for a viral protein to activate and manipulate the NF-κB pathway. IMPORTANCE: KSHV K15 is a viral protein involved in the activation of proinflammatory and angiogenic pathways. Previous studies reported that K15 can activate the NF-κB pathway. Here we show the molecular mechanism underlying the activation of this signaling pathway by K15, which involves direct recruitment of the NF-κB-inducing kinase NIK to K15 as well as NIK-mediated NF-κB p65 phosphorylation on Ser536. K15 is the first viral protein shown to activate NF-κB through direct recruitment of NIK. These results indicate a new mechanism whereby a viral protein can manipulate the NF-κB pathway.


Subject(s)
Herpesvirus 8, Human/immunology , I-kappa B Kinase/metabolism , NF-kappa B/metabolism , Protein Serine-Threonine Kinases/metabolism , Transcription Factor RelA/metabolism , Viral Proteins/metabolism , Cell Line , Humans , Phosphorylation , Protein Processing, Post-Translational , NF-kappaB-Inducing Kinase
2.
PLoS Pathog ; 8(9): e1002927, 2012 Sep.
Article in English | MEDLINE | ID: mdl-23028325

ABSTRACT

Kaposi's Sarcoma (KS), caused by Kaposi's Sarcoma Herpesvirus (KSHV), is a highly vascularised angiogenic tumor of endothelial cells, characterized by latently KSHV-infected spindle cells and a pronounced inflammatory infiltrate. Several KSHV proteins, including LANA-1 (ORF73), vCyclin (ORF72), vGPCR (ORF74), vIL6 (ORF-K2), vCCL-1 (ORF-K6), vCCL-2 (ORF-K4) and K1 have been shown to exert effects that can lead to the proliferation and atypical differentiation of endothelial cells and/or the secretion of cytokines with angiogenic and inflammatory properties (VEGF, bFGF, IL6, IL8, GROα, and TNFß). To investigate a role of the KSHV K15 protein in KSHV-mediated angiogenesis, we carried out a genome wide gene expression analysis on primary endothelial cells infected with KSHV wildtype (KSHVwt) and a KSHV K15 deletion mutant (KSHVΔK15). We found RCAN1/DSCR1 (Regulator of Calcineurin 1/Down Syndrome critical region 1), a cellular gene involved in angiogenesis, to be differentially expressed in KSHVwt- vs KSHVΔK15-infected cells. During physiological angiogenesis, expression of RCAN1 in endothelial cells is regulated by VEGF (vascular endothelial growth factor) through a pathway involving the activation of PLCγ1, Calcineurin and NFAT1. We found that K15 directly recruits PLCγ1, and thereby activates Calcineurin/NFAT1-dependent RCAN1 expression which results in the formation of angiogenic tubes. Primary endothelial cells infected with KSHVwt form angiogenic tubes upon activation of the lytic replication cycle. This effect is abrogated when K15 is deleted (KSHVΔK15) or silenced by an siRNA targeting the K15 expression. Our study establishes K15 as one of the KSHV proteins that contribute to KSHV-induced angiogenesis.


Subject(s)
Herpesvirus 8, Human/metabolism , Human Umbilical Vein Endothelial Cells/virology , Intracellular Signaling Peptides and Proteins/metabolism , Muscle Proteins/metabolism , Neovascularization, Pathologic/virology , Phospholipase C gamma/metabolism , Viral Proteins/metabolism , Angiogenesis Inducing Agents , Animals , Calcineurin/metabolism , Cell Line , Chlorocebus aethiops , DNA-Binding Proteins , HEK293 Cells , Herpesvirus 8, Human/genetics , Herpesvirus 8, Human/growth & development , Humans , Intracellular Signaling Peptides and Proteins/genetics , Molecular Sequence Data , Muscle Proteins/genetics , NFATC Transcription Factors/metabolism , RNA Interference , RNA, Small Interfering , Sarcoma, Kaposi/virology , Sequence Deletion , Vero Cells , Viral Proteins/genetics
3.
PLoS One ; 7(9): e44358, 2012.
Article in English | MEDLINE | ID: mdl-22970203

ABSTRACT

The Rho GTPases Rac1 and Cdc42 regulate a variety of cellular functions by signaling to different signal pathways. It is believed that the presence of a specific effector at the location of GTPase activation determines the route of downstream signaling. We previously reported about EGF-induced Ser-71 phosphorylation of Rac1/Cdc42. By using the phosphomimetic S71E-mutants of Rac1 and Cdc42 we investigated the impact of Ser-71 phosphorylation on binding to selected effector proteins. Binding of the constitutively active (Q61L) variants of Rac1 and Cdc42 to their specific interaction partners Sra-1 and N-WASP, respectively, as well as to their common effector protein PAK was abrogated when Ser-71 was exchanged to glutamate as phosphomimetic substitution. Interaction with their common effector proteins IQGAP1/2/3 or MRCK alpha was, however, hardly affected. This ambivalent behaviour was obvious in functional assays. In contrast to Rac1 Q61L, phosphomimetic Rac1 Q61L/S71E was not able to induce increased membrane ruffling. Instead, Rac1 Q61L/S71E allowed filopodia formation, which is in accordance with abrogation of the dominant Sra-1/Wave signalling pathway. In addition, in contrast to Rac1 transfected cells Rac1 S71E failed to activate PAK1/2. On the other hand, Rac1 Q61L/S71E was as effective in activation of NF-kappaB as Rac1 Q61L, illustrating positive signal transduction of phosphorylated Rac1. Together, these data suggest that phosphorylation of Rac1 and Cdc42 at serine-71 represents a reversible mechanism to shift specificity of GTPase/effector coupling, and to preferentially address selected downstream pathways.


Subject(s)
Neuropeptides/metabolism , Phosphoserine/metabolism , Signal Transduction , rac GTP-Binding Proteins/metabolism , Animals , Enzyme Activation , HEK293 Cells , Humans , Mice , Mutant Proteins/metabolism , NF-kappa B/metabolism , Neuropeptides/deficiency , Phenotype , Phosphorylation , Protein Binding , Pseudopodia/metabolism , Structure-Activity Relationship , cdc42 GTP-Binding Protein/metabolism , p21-Activated Kinases/metabolism , rac GTP-Binding Proteins/deficiency , rac1 GTP-Binding Protein
4.
J Virol ; 84(16): 8231-40, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20534855

ABSTRACT

The Kaposi's sarcoma-associated herpesvirus (KSHV) contains several open reading frames (ORFs) that encode proteins capable of initiating and modulating cellular signaling pathways. Among them is ORF K15, encoding a 12-transmembrane-spanning protein with a cytoplasmic C-terminal domain. Through conserved binding motifs, such as Src homology 2 (SH2) and SH3 binding sites, K15 interacts with cellular proteins, activates the NF-kappaB, MEK/Erk, and Jun N-terminal protein kinase (JNK) pathways, and induces the expression of several inflammatory and angiogenic genes. In this study, we investigated the role of an SH3 domain binding site centered on a PPLP motif in K15. We screened libraries of cellular SH3 domains to identify signaling molecules interacting with the KSHV PPLP motif. We found its affinities for two Src kinase family members, Lyn and Hck, to exceed those of other viral proteins. While the SH2 binding motif YEEV is essential for the inflammatory response induced by KSHV K15, recruitment of Lyn and Hck to the K15 PPLP motif seems to be dispensable for this inflammatory response. However, the PPLP motif is essential for the decrease in B-cell receptor-mediated signaling induced by K15, as measured by calcium mobilization assays.


Subject(s)
B-Lymphocytes/immunology , Herpesvirus 8, Human/immunology , Herpesvirus 8, Human/physiology , Host-Pathogen Interactions , Lymphocyte Activation , Signal Transduction , Viral Proteins/immunology , HeLa Cells , Humans , Protein Binding , Protein Interaction Mapping , Proto-Oncogene Proteins c-hck/metabolism , Viral Proteins/metabolism , src-Family Kinases/metabolism
5.
J Gen Virol ; 90(Pt 5): 1190-1201, 2009 May.
Article in English | MEDLINE | ID: mdl-19264656

ABSTRACT

Rhesus monkey rhadinovirus (RRV) is a gamma-2 herpesvirus related to the human Kaposi's sarcoma-associated herpesvirus (KSHV or human herpesvirus 8). This study identified an alternatively spliced gene at the right side of the RRV genome (strain 17577) between open reading frame 75 and the terminal repeat region. Of its eight exons, the first seven encoded up to 12 transmembrane domains, whilst the eighth exon encoded a predicted C-terminal cytoplasmic domain. Structurally and positionally, this RRV gene therefore resembles the K15 gene of KSHV; it was provisionally named RK15 to avoid confusion with other RRV17577 genes. In ectopic expression studies, the 55 kDa RK15 protein isoform activated the JNK and NF-kappaB pathways, like the 45 kDa KSHV K15-encoded protein isoform. In contrast to K15, which activates angiogenic and inflammatory cytokines such as interleukin (IL)-8, IL-6 and CCL20, the range of cellular transcripts activated by the RRV K15 homologue was much more restricted, but included IL-6, IL-8 and FGF21. These data suggest functional differences between terminal membrane proteins at the right end of the genomes of Old World primate gamma-2 herpesviruses.


Subject(s)
Herpesvirus 8, Human/genetics , Rhadinovirus/genetics , Alternative Splicing , Amino Acid Sequence , Animals , Base Sequence , Cell Line , Cloning, Molecular , Gene Expression Regulation, Viral/physiology , Genes, Viral , Humans , MAP Kinase Kinase 4/metabolism , Macaca mulatta , Mitogen-Activated Protein Kinase Kinases/metabolism , Molecular Sequence Data , NF-kappa B/metabolism , Rhadinovirus/classification , Rhadinovirus/metabolism , Sequence Homology , Transcription, Genetic , Viral Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...