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.
PLoS Pathog ; 6(1): e1000742, 2010 Jan 29.
Article in English | MEDLINE | ID: mdl-20126446

ABSTRACT

Upregulation of xCT, the inducible subunit of a membrane-bound amino acid transporter, replenishes intracellular glutathione stores to maintain cell viability in an environment of oxidative stress. xCT also serves as a fusion-entry receptor for the Kaposi's sarcoma-associated herpesvirus (KSHV), the causative agent of Kaposi's sarcoma (KS). Ongoing KSHV replication and infection of new cell targets is important for KS progression, but whether xCT regulation within the tumor microenvironment plays a role in KS pathogenesis has not been determined. Using gene transfer and whole virus infection experiments, we found that KSHV-encoded microRNAs (KSHV miRNAs) upregulate xCT expression by macrophages and endothelial cells, largely through miR-K12-11 suppression of BACH-1-a negative regulator of transcription recognizing antioxidant response elements within gene promoters. Correlative functional studies reveal that upregulation of xCT by KSHV miRNAs increases cell permissiveness for KSHV infection and protects infected cells from death induced by reactive nitrogen species (RNS). Interestingly, KSHV miRNAs simultaneously upregulate macrophage secretion of RNS, and biochemical inhibition of RNS secretion by macrophages significantly reduces their permissiveness for KSHV infection. The clinical relevance of these findings is supported by our demonstration of increased xCT expression within more advanced human KS tumors containing a larger number of KSHV-infected cells. Collectively, these data support a role for KSHV itself in promoting de novo KSHV infection and the survival of KSHV-infected, RNS-secreting cells in the tumor microenvironment through the induction of xCT.


Subject(s)
Amino Acid Transport System y+/biosynthesis , Gene Expression Regulation/physiology , Herpesvirus 8, Human/pathogenicity , MicroRNAs/metabolism , Oxidative Stress/physiology , Sarcoma, Kaposi/virology , Animals , Basic-Leucine Zipper Transcription Factors/metabolism , Cell Line , Endothelial Cells/metabolism , Endothelial Cells/virology , Fluorescent Antibody Technique , Herpesvirus 8, Human/genetics , Herpesvirus 8, Human/metabolism , Humans , Immunoblotting , Immunohistochemistry , Macrophages/metabolism , Macrophages/virology , Mice , MicroRNAs/genetics , Neoplasm Staging , Polymerase Chain Reaction , Reactive Nitrogen Species/metabolism , Sarcoma, Kaposi/metabolism , Sarcoma, Kaposi/pathology , Transfection , Up-Regulation
2.
J Leukoc Biol ; 87(1): 25-34, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20052801

ABSTRACT

Macrophages are an important source of inflammatory cytokines generated during the innate immune response,but in the microenvironment of certain tumors,macrophages promote tumor progression through their preferential secretion of cytokines that support tumor cell growth and suppress antitumoral immune responses. KSHV is the causative agent of KS and lymphomas preferentially arising in immuno compromised patients, and specific cytokines, including IL-6 and IL-10, have been implicated in KSHV-associated cancer pathogenesis. However, the contribution of KSHV-infected macrophages to the cytokine milieu within KSHV-related tumors is unclear. We found that individual KSHV-encoded miRNA induce IL-6 and IL-10 secretion independently and additively by murine macrophages and human myelomonocytic cells. Bioinformatics analysis identified KSHV miRNA binding sites formiR-K12-3 and miR-K12-7 within the 3'UTR of the basic region/leucine zipper motif transcription factor C/EBPbeta, a known regulator of IL-6 and IL-10 transcriptional activation.Subsequent immunoblot analyses revealed that miR-K12-3 and miR-K12-7 preferentially reduce expression of C/EBPbeta p20 (LIP), an isoform of C/EBPbeta known to function as a negative transcription regulator. In addition,RNA interference specifically targeting LIP induced basal secretion of IL-6 and IL-10 by macrophages.Taken together, these data support a role for KSHV miRNA in the programming of macrophage cytokine responses in favor of KSHV-related tumor progression.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/antagonists & inhibitors , Gene Expression Regulation, Viral , Herpesvirus 8, Human/physiology , Interleukin-10/metabolism , Interleukin-6/metabolism , Macrophages/virology , MicroRNAs/physiology , Monocytes/virology , RNA, Viral/physiology , 3' Untranslated Regions , Animals , Cell Line/metabolism , Cell Line/virology , Cell Line, Tumor/metabolism , Cell Line, Tumor/virology , Herpesvirus 8, Human/genetics , Host-Pathogen Interactions , Humans , Interleukin-10/biosynthesis , Interleukin-10/genetics , Interleukin-6/biosynthesis , Interleukin-6/genetics , Macrophages/metabolism , Mice , Monocytes/metabolism , Protein Isoforms/antagonists & inhibitors , RNA, Viral/genetics
3.
Genes Dev ; 24(2): 195-205, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-20080955

ABSTRACT

Kaposi sarcoma herpesvirus (KSHV) induces transcriptional reprogramming of endothelial cells. In particular, KSHV-infected lymphatic endothelial cells (LECs) show an up-regulation of genes associated with blood vessel endothelial cells (BECs). Consequently, KSHV-infected tumor cells in Kaposi sarcoma are poorly differentiated endothelial cells, expressing markers of both LECs and BECs. MicroRNAs (miRNAs) are short noncoding RNA molecules that act post-transcriptionally to negatively regulate gene expression. Here we validate expression of the KSHV-encoded miRNAs in Kaposi sarcoma lesions and demonstrate that these miRNAs contribute to viral-induced reprogramming by silencing the cellular transcription factor MAF (musculoaponeurotic fibrosarcoma oncogene homolog). MAF is expressed in LECs but not in BECs. We identify a novel role for MAF as a transcriptional repressor, preventing expression of BEC-specific genes, thereby maintaining the differentiation status of LECs. These findings demonstrate that viral miRNAs could influence the differentiation status of infected cells, and thereby contribute to KSHV-induced oncogenesis.


Subject(s)
Cellular Reprogramming , Endothelial Cells/cytology , Endothelial Cells/pathology , Herpesvirus 8, Human/metabolism , MicroRNAs/metabolism , Oncogene Protein v-maf/metabolism , Sarcoma, Kaposi/physiopathology , Cell Differentiation , Cell Line , Cell Line, Tumor , Down-Regulation , Endothelial Cells/metabolism , Endothelial Cells/virology , Gene Expression Regulation, Viral , Gene Silencing , HeLa Cells , Herpesviridae Infections/physiopathology , Herpesvirus 8, Human/genetics , Humans
4.
Trends Microbiol ; 17(12): 544-53, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19828316

ABSTRACT

MicroRNAs (miRNAs) are short RNAs of about 22 nucleotides in length that post-transcriptionally regulate gene expression by binding to 3' untranslated regions of mRNAs, thereby inducing translational silencing. Recently, more than 140 miRNAs have been identified in the genomes of herpesviruses. Deciphering their role in viral biology requires the identification of target genes, a challenging task because miRNAs require only limited complementarity. The subject of this review will be the herpesvirus miRNAs and their respective target genes that have been determined experimentally to date. These miRNAs regulate fundamental cellular processes including immunity, angiogenesis, apoptosis, and key steps in the herpesvirus life cycle, latency and the switch from latent to lytic replication.


Subject(s)
Gene Expression Regulation, Viral , Herpesviridae/genetics , Herpesviridae/metabolism , MicroRNAs/metabolism , RNA, Viral/metabolism , MicroRNAs/genetics , RNA, Viral/genetics
5.
J Virol ; 81(23): 12836-45, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17881434

ABSTRACT

MicroRNAs (miRNAs) are small noncoding RNAs that posttranscriptionally regulate gene expression by binding to 3'-untranslated regions (3'UTRs) of target mRNAs. Kaposi's sarcoma-associated herpesvirus (KSHV), a virus linked to malignancies including primary effusion lymphoma (PEL), encodes 12 miRNA genes, but only a few regulatory targets are known. We found that KSHV-miR-K12-11 shares 100% seed sequence homology with hsa-miR-155, an miRNA frequently found to be up-regulated in lymphomas and critically important for B-cell development. Based on this seed sequence homology, we hypothesized that both miRNAs regulate a common set of target genes and, as a result, could have similar biological activities. Examination of five PEL lines showed that PELs do not express miR-155 but do express high levels of miR-K12-11. Bioinformatic tools predicted the transcriptional repressor BACH-1 to be targeted by both miRNAs, and ectopic expression of either miR-155 or miR-K12-11 inhibited a BACH-1 3'UTR-containing reporter. Furthermore, BACH-1 protein levels are low in cells expressing either miRNA. Gene expression profiling of miRNA-expressing stable cell lines revealed 66 genes that were commonly down-regulated. For select genes, miRNA targeting was confirmed by reporter assays. Thus, based on our in silico predictions, reporter assays, and expression profiling data, miR-K12-11 and miR-155 regulate a common set of cellular targets. Given the role of miR-155 during B-cell maturation, we speculate that miR-K12-11 may contribute to the distinct developmental phenotype of PEL cells, which are blocked in a late stage of B-cell development. Together, these findings indicate that KSHV miR-K12-11 is an ortholog of miR-155.


Subject(s)
Herpesvirus 8, Human/genetics , MicroRNAs/genetics , RNA, Viral/genetics , Sequence Homology, Nucleic Acid , 3' Untranslated Regions/genetics , Basic-Leucine Zipper Transcription Factors/biosynthesis , Basic-Leucine Zipper Transcription Factors/genetics , Cell Line, Tumor/chemistry , Cell Line, Tumor/virology , Down-Regulation , Fanconi Anemia Complementation Group Proteins/biosynthesis , Fanconi Anemia Complementation Group Proteins/genetics , Gene Expression , Gene Expression Profiling , Genes, Reporter , Humans , Luciferases/biosynthesis , Luciferases/genetics , Oligonucleotide Array Sequence Analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...