• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 5
  • 1
  • 1
  • Tagged with
  • 13
  • 7
  • 7
  • 6
  • 5
  • 4
  • 4
  • 3
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • About
  • The Global ETD Search service is a free service for researchers to find electronic theses and dissertations. This service is provided by the Networked Digital Library of Theses and Dissertations.
    Our metadata is collected from universities around the world. If you manage a university/consortium/country archive and want to be added, details can be found on the NDLTD website.
1

KSHV vGPCR: A VIRAL ONCOPROTEIN THAT TRIGGERS AUTOPHAGY AND CELLULAR SENESCENCE

Cyr, David P 16 June 2011 (has links)
Autophagy (literally to ‘self-eat’) is an intracellular, catabolic mechanism to degrade and recycle cytoplasmic contents in response to metabolic, oxidative, and genotoxic stresses. Autophagy plays an important role in cellular homeostasis, and dysfunctional autophagic activity has been implicated in an array of human diseases. Importantly, autophagy has recently been identified to function in host defence against intracellular pathogens, including viruses. For this reason, many viruses have evolved strategies to subvert or exploit autophagy and block its antiviral effects. Kaposi’s sarcoma-associated herpesvirus (KSHV) is the etiological agent of Kaposi’s sarcoma (KS), an AIDS-related cancer of the endothelium. KSHV gene products have evolved to support viral replication and evade immune surveillance. Some of these same gene products impact KS tumourigenesis, but the precise mechanisms have yet to be elucidated. Furthermore, the impact of autophagy on KSHV replication and KS tumourigenesis remains unexplored. The KSHV viral G-protein-coupled receptor (vGPCR) is a constitutively active signalling molecule that stimulates a number of host regulatory pathways that would be expected to impact autophagy, including PI3K/Akt/mTOR and JNK. Moreover, vGPCR is expressed during lytic replication when the antiviral autophagic response may threaten virion production. Here, vGPCR activity has been definitively shown to trigger autophagy in endothelial cells using immunoblot analysis, fluorescent reporter proteins, and transmission electron microscopy. Furthermore, preliminary data suggest that this stimulatory effect is evoked through JNK activation. Taken together, these findings indicate that vGPCR likely elicits autophagic responses during KSHV lytic replication. Recently, autophagy has been recognized as a molecular barrier to tumourigenesis, influencing cell survival, cell death, or a form of cell cycle arrest called oncogene-induced senescence (OIS). Remarkably, like many host oncogenes, ectopic expression of vGPCR triggers OIS in endothelial cells. This response is dependent on vGPCR signalling activity, as an inactive form of vGPCR (R143A) fails to trigger OIS. Furthermore, vGPCR OIS is atypical in that it does not involve DNA damage responses (DDRs). Together, these autophagic and senescence responses to ectopic vGPCR expression illustrate the potency of its oncogenic potential. The significance of vGPCR-induced autophagy and senescence during KSHV replication and KS development is presently unclear. I speculate that autophagy represents a hurdle that the virus must overcome in vivo. In my working model, potent vGPCR oncogenic signalling activity sets off the alarm, eliciting autophagic responses. It seems likely that additional lytic viral gene products may serve to undermine these autophagic responses and permit viral replication and dissemination in vivo.
2

Study of a tumor virus unveils a novel function for the miRNA biogenesis machinery

Lin, Yao-Tang 04 March 2014 (has links)
Kaposi’s Sarcoma-associated Herpes Virus (KSHV) is a human herpesvirus associated with cancers. To date, KSHV miRNAs have been mostly identified via analysis of cells that are undergoing latent infection. This work presented here is a novel approach to profile small RNAs from populations of cells undergoing predominantly lytic infection. Using two different next generation sequencing platforms, I cloned and sequenced both pre-microRNAs and derivative microRNAs (miRNAs). This analysis shows that the vast majority of viral and host 5p miRNAs are co-terminal with the 5 prime end of the cloned pre-miRNAs, consistent with both being defined by microprocessor cleavage. I report the complete repertoire (25 total) of 5p and 3p derivative miRNAs from all 12 previously described KSHV pre-miRNAs. Two KSHV pre-miRNAs, pre-miRs-K8 and K12, encode abundant derivative miRNAs from the previously unreported strands of the pre-miRNA. I identify several novel small RNAs of low abundance, including viral microRNA-offset-RNAs (moRNAs), and antisense viral miRNAs (miRNA-AS) that are encoded antisense to previously reported KSHV pre-miRNAs. This work also shows that much of the KSHV genome is transcribed in both the top and bottom strand orientations during lytic replication. Despite the enormous potential to form double-stranded RNA in KSHV-infected cells, I observe no evidence for the existence of abundant viral-derived small interfering RNAs (siRNAs). From the small RNA deep-sequencing, I also detected a low abundant small RNA fragment (23 nt) that maps to a putative hairpin structure (named hairpin K) within the KSHV PAN transcript. I demonstrate that hairpin K is a cis-negative regulatory element in PAN. It is well-appreciated that viruses utilize host effectors for macromolecular synthesis and as regulators of viral gene expression. Viruses can encode their own regulators, but often utilize host-encoded factors to optimize replication. This work shows that Drosha, an endoribonuclease best known for its role in the biogenesis of miRNAs, can also function to directly regulate viral gene expression. Kaposin B (KapB) is a KSHV-encoded protein associated with cytokine production and cytotoxicity. I demonstrate that in addition to previously known transcriptional mechanisms, differences in Drosha levels contribute to low levels of KapB expression in latency and robust increases in expression during lytic replication. Thus, KSHV modulates Drosha activity differentially depending on the mode of replication. This regulation is dependent on Drosha-mediated cleavage, and KapB transcripts lacking the Drosha cleavage sites express higher levels of KapB resulting in increased cell death. This work increases the known functions of Drosha and implies that tying viral gene expression to Drosha activity is advantageous for viruses. / text
3

Studies on a new human herpesvirus, Kaposi's sarcoma-associated herpesvirus

Elzinger, Bianca Ariane January 2000 (has links)
Kaposi's sarcoma (KS)-associated herpesvirus (KSHV), also called human herpesvirus 8 (HHV8), has been been identified in all epidemiological forms of KS as well as in tissue obtained from primary effusion lymphoma (PEL) and Multicentric Castleman's disease (MCD). The KSHV genome contains several putative oncogenes, suggesting that viral infection may induce cellular transformation and tumorgenesis. Herpesviruses encode a number of different surface glycoproteins, which are involved in virus-host interactions. Studies have shown that the viral glycoproteins H and L form a complex that plays an essential role in viral attachment and cell to cell fusion. Both glycoproteins have been identified in KSHV and were expressed in mammalian cells. Expression studies revealed that KSHV gH and gL exhibit similar features to those seen in other herpesviruses. However, KSI-IV gL appears to traffic independently and may function in cell to cell fusion processes even when expressed alone. KSI-IV de novo infections are rare and the lack of a reliable cell culture system has delayed pathogenesis studies. As part of this thesis the hepatoma cell line HepG2 has been shown to allow limited KSHV infection, as judged by nested PCR. Studies have shown that infection leads to increased apoptosis, although viral replication could not be detected. Furthermore, Epstein Barr Virus (EBV) appeared to modulate the ability of KSHV to infect HepG2 cells. Finally, a microtitre plate assay has been established for the quantification of the KSHV genome. A comparison of plasma and serum samples obtained at the same time point showed that plasma is more reliable in testing for KSHV, the DNA copy number in serum samples being reduced up to 10 fold. In conclusion, this new assay is a potentially useful tool for both diagnostic proposes and research studies.
4

Régulation de l'apoptose par les microARN du virus associé au sarcome de Kaposi / Regulation of apoptosis by Kaposi’s sarcoma associated herpesvirus microRNAs

Suffert, Guillaume 07 May 2013 (has links)
Le virus associé au sarcome de Kaposi (KSHV) code pour un cluster de 12 précurseurs de micro (mi)ARN abondamment exprimés pendant les phases lytiques et latentes de l’infection. Des études précédentes ont rapporté que KSHV est capable d’inhiber l’apoptose pendant l’infection latente ; nous avons donc testé si les miARN du virus étaient impliqués dans ce processus. Nous avons trouvé que des cellules HEK293 et DG-75 exprimant de manière stable les miARN de KSHV étaient protégées de l’apoptose. Les cibles cellulaires potentielles qui étaient significativement négativement régulées lors de l’expression des miARNs de KSHV ont été identifiées par analyse transcriptomique par microarray. Parmi celles-ci, nous avons validé par tests rapporteurs luciférase, PCR quantitative, et western blot, Caspase 3 (CASP3), un facteur jouant un rôle critique dans le contrôle de l’apoptose. Via le biais de mutagenèse dirigée, nous avons montré que trois miARN de KSHV, miR- 12-1, 3 et 4-3p, étaient responsables du ciblage de CASP3. L’inhibition spécifique de ces miARN dans des cellules infectées par KSHV a résulté en une augmentation des niveaux d’expression de CASP3 endogène, et en une apoptose plus accrue. Vus dans leur ensemble, nos résultats suggèrent que les miARN de KSHV participent directement à l’inhibition précédemment rapportée de l’apoptose par le virus, et donc qu’ils jouent probablement un rôle dans l’oncogenèse induite par KSHV. / Kaposi’s sarcoma herpesvirus (KSHV) encodes a cluster of twelve micro (mi)RNA precursors, which are abundantly expressed during both latent and lytic infection. Previous studies reported that KSHV is able to inhibit apoptosis; we thus tested the involvement of viral miRNAs in this process. We found that both HEK293 epithelial cells and DG-75 cells stably expressing KSHV miRNAs were protected from apoptosis. Potential cellular targets that were significantly down-regulated upon KSHV miRNAs expression were identified by microarray profiling. Among them, we validated by luciferase reporter assays, quantitative PCR and western blotting Caspase 3 (CASP3), a critical factor for the control of apoptosis. Using site-directed mutagenesis, we found that three KSHV miRNAs, miR-K12-1, 3 and 4-3p, were responsible for the targeting of CASP3. Specific inhibition of these miRNAs in KSHV infected cells resulted in increased expression levels of endogenous CASP3 and enhanced apoptosis. Altogether, our results suggest that KSHV miRNAs directly participate to the previously reported inhibition of apoptosis by the virus, and are thus likely to play a role in KSHV-induced oncogenesis.
5

Prevalent and differential herpesviral gene regulation mediated by 3'-untranslated regions

McClure, Lydia Virginia 16 September 2014 (has links)
Herpesviral infections are currently incurable and are associated with severe human diseases, such as cancer. Kaposi’s Sarcoma-associated Herpesvirus (KSHV), like all herpesviruses, undergoes a long-term, latent infection where few viral products are made as a mechanism to evade the host immune system. Recently, the KSHV latent genome was shown to have bivalent histone marks thought to keep the virus poised for replication. However, it is unclear how the virus prevents spurious leaky transcription from this primed state. The 3' untranslated region (3'-UTR) of transcripts is a common site of gene expression regulation, however less than half of the KSHV 3'-UTRs have been mapped and few studies have interrogated their role during infection. The work presented here is the first large-scale map and analysis of the KSHV 3'-UTRs. Four methods were used to identify the 3'-UTRs expressed by the ~85 KSHV genes, including prediction algorithms, 3'-RACE, DNA tiling array, and next generation deep sequencing analysis. The role of each KSHV 3'-UTR in gene expression was then examined using luciferase reporter assays and showed a surprising prevalence of negative regulation conveyed during latent infection. Sequential deletions across numerous 3'-UTRs indicated RNA structure is likely involved in this regulation. In addition, several KSHV 3'-UTRs conveyed an increase in translation during lytic infection through enhanced recognition by the cap-dependent translation initiation machinery activated via the MNK1 kinase. A second mechanism of KSHV gene regulation was identified through motifs encoded in the K7 3'-UTR. This work indicated that a previously characterized RNA element and a novel putative hairpin are both partially responsible for negative regulation conveyed by the K7 3'-UTR. We hypothesize that these structural motifs control expression of the K7 transcript by altering its sub-cellular location and/or via RNA stability. This work represents a broad 3'-UTR study that mapped the KSHV 3'-UTRs and is the first large-scale functional analysis of 3'-UTRs from a large genome virus. We have implicated post-transcriptional mechanisms, along with known transcriptional regulation, in viral evasion of the immune response during latency and the escape of viral-mediated host shutoff. These results identify new potential targets for therapeutic intervention of KSHV-associated disease. / text
6

Dynamic Interplay Between Kaposi's Sarcoma-Associated Herpesvirus Latent Proteins in the Control of Oncogene-Induced Senescence

Leidal, Andrew Michael 10 April 2012 (has links)
Acute oncogenic stress can activate autophagy and facilitate permanent arrest of the cell cycle through a failsafe mechanism known as oncogene-induced senescence (OIS). Kaposi's sarcoma-associated herpesvirus (KSHV) is the etiologic agent of Kaposi’s Sarcoma (KS) and has been reported to encode oncoproteins within its latency program that engage host autophagy and OIS pathways; however, the mechanisms by which KSHV oncoproteins promote KS tumorigenesis remain unclear. Here, I demonstrate that ectopic expression of the latent KSHV protein viral cyclin (v-cyclin) deregulates the cell cycle, induces DNA-damage responses (DDRs) and promotes OIS through an autophagy-dependent mechanism. During latency, v-cyclin is co-expressed from a single transcript with a viral homolog of FLICE-inhibitory protein (v-FLIP) that blocks autophagy by binding and inhibiting Atg3. Co-expression of v-FLIP has no effect on DDRs, but efficiently blocks v-cyclin-induced autophagy and senescence. Remarkably, suppression of v-FLIP function during KSHV latency, through specific inhibitory peptides, rescues host cell autophagy and induces senescence of infected cells. Together, these data reveal a coordinated viral gene-expression program that subverts autophagy, impairs senescence, and facilitates the proliferation of KSHV-infected cells.
7

The SHAPE of U: Mapping Out Protective Elements in mRNA Escapees

Miles, Jacob 18 December 2020 (has links)
A crucial step of the viral life cycle of Kaposi’s Sarcoma Herpesvirus (KSHV) lytic infection is the triggering of a massive RNA decay event termed “Host Shutoff”. Host Shutoff is driven by the viral endonuclease SOX which leads to the destruction of over 70% of the total transcriptome. This process cripples cellular gene expression and allows for viral reprograming of the cell for the purpose of viral replication. Co-evolution has led to the host developing a multitude of antiviral defenses aimed at preserving certain cellular RNAs linked to antiviral responses. One such defense are RNA secondary structures located within the 3’UTR of select host transcripts that protect them from SOX degradation. This structure, known as the SOX Resistant Element or SRE, has previously been isolated to a 200-nucleotide region found within the 3’UTR of the host transcript Interleukin-6. In this thesis, I sought to further define the structure of the IL-6 and other SREs using SHAPE-MaP to generate chemically-probed RNA structural models. Through this work, I demonstrated that the IL-6 SRE confers a form of active resistance to SOX cleavage, and based on structural analyses, likely acts as a scaffold for the recruitment of a protective ribonucleoprotein complex. This research highlights the importance of RNA secondary structures in influencing mRNA fate during viral infection and establishes the groundwork for understanding how these structural features can facilitate escape of cellular transcripts from viral endonucleases.
8

The Role of Viral Interleukin-6 in Tumor Development of Kaposi's Sarcoma-Associated Herpesvirus Lymphomas

Fullwood, Rebecca A. 01 December 2016 (has links)
Kaposi's sarcoma herpesvirus (KSHV) is a cancer-causing virus, primarily affecting AIDS patients. KSHV is found in 3-10% of the U.S. population and can cause a range of cancers in the highly immunosuppressed; these cancers include Kaposi's sarcoma, pleural effusion lymphoma (PEL) and multicentric Castleman's disease (MCD). The current techniques for treating these cancers are relatively ineffective, largely due to their inefficiency at targeting tumors formed by the infection. One protein produced by KSHV, the viral homolog of interleukin-6 (vIL-6), is thought to play a major role in tumor development post-infection. Here a novel animal model is implemented to study the ways vIL-6 affects tumor development through growth factors and other cytokines within infected highly immune-deficient Rag2-/-γc-/- mice. Mice were subcutaneously injected with one of three types of cells: B cells infected with a wild-type (WT) KSHV, B cells infected with mutant KSHV without the gene for viral interleukin 6, and a negative control of uninfected B cells. After allowing time for tumors to develop the mice were sacrificed and the tumors assessed. Analysis of the physical properties of the tumors, as well as markers expressed by the tumors, were used to help determine whether vIL-6 could be an appropriate target when treating these cancers. In this study vIL-6 was seen to influence certain B cell markers (CD30), as well as onset of tumors (with no significant increase in overall tumor mass, but with marginally statistically significant increase in tumor number). This indicates that although vIL-6 could play a small role as a target for cancer, further investigation into the relationship of CD30 in these types of cancers needs to be explored. It was also found that the KSHV viral-infection decreases the development of tumors compared with uninfected immortalized B cells (BJAB). Not only would results from this experiment help develop new treatments, and change the lives of those suffering with cancers induced by KSHV, but they would provide a foundation for future studies with these types of cancers.
9

Využití metody RNA interference (RNAi) ke studiu onkogenních vlastností viru Kaposiho sarkomu (KSHV). / Employing an RNA interference method (RNAi) to sudy oncogenic properties of Kaposi's sarcoma-associated herpesvirus (KSHV)

Riegerová, Petra January 2017 (has links)
Kaposi's sarcoma-associated herpesvirus (KSHV) is a DNA tumor virus that has been associated with all epidemiological forms of Kaposi's sarcoma, primary effusion lymphoma (PEL) and multicentric Castleman disease (MCD). Like other herpesviruses, KSHV undergoes two phases of life cycle (latent and lytic replication). During latency, the viral genome persists as a circular episome in the nucleus of the host cell and only a few viral genes are expressed, namely LANA (latency- associated nuclear antigen), Kaposin, vFLIP (viral FLICE inhibitory protein), vCyclin, and vIRF3/LANA2 (viral interferon regulatory factor 3). These viral genes are responsible for regulation of host cell proliferation, prevention of apoptosis, facilitation of immune evasion, and maintenance of the extrachromosomal viral genome during cell divisions. vIRF3 is a multifunctional nuclear protein that is constitutively expressed in KSHV positive PEL cells and Castleman's disease tumors, which expression causes dramatic changes of critical host pathways that are involved in the regulation of apoptosis, cell cycle, antiviral immunity, and tumorigenesis. In our study, we have demonstrated and elucidated predicted mechanism, by which vIRF3 enhances transcription activity of c-Myc. Moreover, we have clarified the previously unappreciated...
10

Cell and Receptor Tropism of γ2-Herpesviruses

Großkopf, Anna Katharina 23 March 2020 (has links)
No description available.

Page generated in 0.0143 seconds