• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 4
  • Tagged with
  • 7
  • 5
  • 3
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • 1
  • 1
  • 1
  • 1
  • 1
  • 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

STAT2 in the regulation of tumor growth and antitumor effects of type I interferons in a mouse model of melanoma

Yue, Chanyu January 2013 (has links)
Signal Transducer and Activator of Transcription (STAT) 2 is one of seven members of the STAT family of transcription factors with dual roles in signal transduction and gene activation. STAT2 is a central transcription factor that regulates the antiviral, apoptotic and cell growth inhibitory effects of type I interferons (IFN-α/β), a small family of secreted glycoproteins induced by the host after sensing the presence of tumor cells and pathogens. The creation of Stat2-/- mice established the pivotal role of STAT2 in type I IFN signaling and in antiviral immunity. In vitro studies conducted in STAT2 deficient tumor cell lines suggested a role in suppressing tumor cell growth in response to IFN treatment. Based on these properties STAT2 is presumed to have tumor suppressor functions but data to support this notion in animal models of cancer are limited. To address the role of STAT2 in cancer, I used the murine B16-F1 tumor transplantation model of human melanoma. The B16-F1 melanoma cell line was established from a spontaneous tumor that arose in mice. I discovered that tumor cells transplanted subcutaneously in Stat2-/- mice grew more aggressively than in the counterpart wild type mice. Closer examination of B16-F1 tumors harvested from wild type and Stat2-/- mice revealed an unexpected dramatic similar reduction of STAT2 and STAT1 proteins. Yet soluble factors secreted by B16-F1 tumors established in Stat2-/- mice alone were sufficient to enhance proliferation of B16-F1 tumor cells. I further showed that tumor-bearing wild type mice treated with IFN-β developed smaller tumors compared to Stat2-/- mice, whose tumors continued to grow and hence were unresponsive to IFN intervention. Lastly, to elucidate a mechanism that leads to enhanced tumor growth in Stat2-/- mice, I questioned the involvement of the host immune response in restricting tumor growth. I found that tumor specific T cell priming by Stat2-/- dendritic cells (DCs) was defective since generated cytotoxic T cells (CD8+ T lymphocytes) produced low levels of IFN-γ and IL-2 and adoptive transfer of these B16-F1 tumor specific CD8+ T cells in B16-F1 bearing Stat2-/- mice did not cause tumor regression with IFN-β intervention. Collectively, my findings reveal that host STAT2 restricts the establishment of melanoma tumors. More importantly, type I IFN/STAT2 signaling on DCs plays a pivotal role in tumor antigen cross-presentation to CD8+ T cells and in the development of a protective antitumor response resulting in tumor rejection. To now address whether STAT2 expression in cancer cells could influence tumor establishment and the antitumor effects of type I IFNs, STAT2 expression was silenced in B16-F1 tumor cells. Contrary to my expectation, silencing STAT2 augmented the growth inhibitory effects of IFN-β both in vitro and in vivo. However, loss of STAT2 expression in the tumor did not cause B16-F1 tumor cells to grow more aggressively compared to control B16-F1 cells. Furthermore, compared to B16-F1 control cells, STAT2-silenced B16-F1 cells showed an initial delay but later persistent STAT activation and formation of the ISGF3 transcriptional complex (consisting of STAT1, STAT2 and IRF9). This observation paralleled with an initial delay and then later an increase in the expression of IFN regulated genes. In addition, reduced activation of STAT5 induced by IFN-β was observed in STAT2-silenced B16-F1 cells. This may partially explain the enhanced growth inhibitory effects of type I IFNs. Together these results shed light on the unexpected role of tumor STAT2 expression in diminishing the efficacy of type I IFN treatment of melanoma. / Biochemistry
2

Regulation of type I interferons in murine dendritic cells

Xu, Jun January 2014 (has links)
Conventional Dendritic cells (cDCs), a specialized group of immunological sentinels with tree-like or dendritic shapes, are critical for recognition of danger signals, presentation of antigens and control of a spectrum of innate and adaptive immune responses. Type I interferons (IFNs), as important danger signals, activate cDCs through the canonical type I IFN receptor signaling. Type I IFNs are the first line of host defense against viral infection by up-regulating IFN-stimulated genes (ISGs). However, there are circumstances in which the silencing of excessive type I IFNs could be beneficial to the host, such as IFN-dependent autoimmune diseases, gene therapy that uses viral vectors and transplantation. The role of type I IFNs in DC development, activation and antigen presentation function remains to be completely investigated. In this dissertation, we studied the regulation of Type I IFNs in murine DCs, both cDCs and plasmacytoid DCs (pDCs), and specifically we investigated the role of two molecules, Signal Transducer and Activator of Transcription 2 (STAT2) and Three prime Repair EXonuclease 1 (Trex1), in DC biology. Our research furthers our understanding of DC development, activation and function, and provides important data for the therapeutic application of modified DCs to induce immunological tolerance in gene therapy, IFN-dependent autoimmune diseases and transplantation. STAT2 is a nuclear transcription factor downstream of type I IFN receptor-mediated signaling, the role of which has been mostly explored in antiviral responses mediated by type I IFNs. However, the involvement of STAT2 in cDC activation and function such as cross-presentation remains hitherto unclear. We report that STAT2 is essential for murine cDC activation upon TLR3, -4, -7 and -9 stimulation. In the absence of STAT2, cDCs displayed impaired up-regulation of type I IFN response (costimulatory molecules and type I IFN-stimulated genes), and reduced inflammatory cytokine production when stimulated with TLR ligands. STAT2 was required in all of the DC responses to exogenous IFNα, suggesting that the canonical STAT1-STAT2 heterodimers are the major signaling transducers downstream of type I IFNs in DCs. Of interest, LPS-induced TNFα and IL6 production were reduced in STAT2-/- DCs but not in IFNAR1-/- DCs, suggesting a novel STAT2-dependent pathway mediated by LPS, bypassing type I IFN-receptor signaling. STAT2-deficient cDCs showed impaired cross-presentation leading to decreased CD8+ T cell response both in vitro and CTL killing in vivo, indicating that STAT2 is essential for TLR-induced cross-presentation. These results demonstrate that STAT2 is critical in the regulation of TLR-induced DC activation and cross-presentation, suggesting an essential role for STAT2 in anti-viral and anti-tumor immune responses. We also propose a novel regulatory function of STAT2 in the LPS response independent of type I IFN receptor signaling. Trex1 mutations are associated with a spectrum of type I IFN-dependent autoimmune diseases such as Aicardi-Goutières syndrome and systemic lupus erythematosus. Trex1 plays an essential role in preventing accumulation of excessive cytoplasmic DNA, avoiding cell-intrinsic innate DNA sensor activation and suppressing activation of both type I IFN-stimulated and IFN-independent antiviral genes. Trex1 also helps HIV escape cytoplasmic detection by DNA sensors. However, regulation of Trex1 in DC biology is lacking. We report that murine cDCs have high constitutive expression of Trex1 in vitro and in vivo in the spleen. In resting bone marrow-derived cDCs, type I IFNs up-regulate Trex1 expression via the canonical IFN receptor signaling pathway (STAT1-, STAT2-dependent). DC activation induced by TLR3, -4, -7 and -9 ligands also augments Trex1 expression through autocrine IFNß production and triggering of the IFN signaling pathway, while TLR4 ligand LPS also stimulates an early expression of Trex1 through an IFN-independent NFΚB-dependent signaling pathway. Furthermore, retroviral infection also induces Trex1 up-regulation in cDCs, as we found that a gene therapy HIV-1-based lentiviral vector induces significant Trex1 expression, suggesting that Trex1 may affect local and systemic administration of gene therapy vehicles. Our data indicate that Trex1 is induced in cDCs during activation upon IFN- and TLR- stimulation through the canonical IFN signaling pathway, and suggest that Trex1 may play a role in cDC activation during infection and autoimmunity. Finally, these results suggest that HIV-like viruses may up-regulate Trex1 to increase their ability to escape immunosurveillance. In order to dissect the role of Trex1 in DC functions, we compared DCs from Trex1-/- and wild-type mice. We report that Trex1 deficiency reduces absolute number of pDCs in BM but not in spleen of male over female mice. Furthermore, Trex1 deficiency preferentially represses Flt3L-induced DC development both in vitro and in vivo but not GM-CSF-dependent DC development, suggesting that Trex1 plays an indispensable role in Flt3L-induced DC development and GM-CSF may compensate the effect of Trex1 deficiency. This defect is only limited to male Trex1-/- DCs, and mimics the effect of mTOR inhibition. Furthermore, although Flt3L-induced Trex1-/- DCs expressed a type I IFN signature, they also exhibited decreased pDC development markers, indicating Trex1 regulates pDC development at the transcriptional level. Thus, we propose a novel and essential role of Trex1 in Flt3L-induced DC development, and the effect of Trex1 regulation is gender-dependent. Together, these findings enhance our understanding of regulatory roles of Type I IFNs in DC development, activation and function, supporting the beneficial role of STAT2/type I IFN axis in TLR-induced DC activation and cross-presentation. Our study in Trex1 reveals Trex1 induction by viral infection, type I IFNs and TLRs in DCs, and a new role of Trex1 in early development of Flt3L-induced DCs in a gender-dependent manner, whereby a balance between type I IFNs and Trex1 is important for DC activation and hemostasis. / Microbiology and Immunology
3

Non-canonical roles for STAT1 and STAT2 in mitochondrial biogenesis and fasting homeostasis.

Sisler, Jennifer 01 January 2012 (has links)
The signal transducer and activator of transcription 1 (STAT1) and 2 (STAT2) are primarily activated by interferons and play a central role in orchestrating responses to a variety of pathogens by activating the transcription of nuclear encoded genes that mediate antiviral, antigrowth and immune surveillance responses. In addition to their nuclear effects, we report that STAT1 and STAT2 inhibit the expression of mitochondrial encoded mRNAs by activating type I interferons (IFNβ) under basal conditions. STAT1-/- livers also exhibit elevated levels of nuclear encoded components of the electron transport chain (ETC). Treatment of wild-type mice with IFNβ inhibits both mitochondrial and nuclear transcription of the ETC components. The inhibition of mitochondrial encoded transcription by IFNβ is both STAT1 independent and dependent because it is also seen in STAT1-/- mice. This inhibitory action of IFNβ on mitochondrial transcription is mediated by a small pool of STAT1 and STAT2 residing within the mitochondria. This study would suggest a novel mechanism for STAT1 and STAT2, wherein they negatively regulate mitochondrial transcription and STAT1 coordinately regulates transcription of both mitochondrial and nuclear ETC components upon IFNβ stimulation. PGC1α has been described as the master regulator of mitochondrial biogenesis, and upon starvation its levels are elevated within the liver to increase mitochondrial biogenesis. The levels of PGC1α are increased in the STAT1-/- mice basally. However they are not increased further under starvation. Additionally, we report a novel phenotype of the STAT1-/- mice that suggests that they have a dysregulation of energy expenditure during starvation. We see that activation of hormone sensitive lipase which is the rate limiting step of lipolysis is attenuated in the adipose tissue of STAT1-/- mice and that there is less lipid accumulation in the livers of STAT1-/- compared to wild-type mice. The mechanism of STAT1’s role in energy regulation is not fully understood; however, this report would suggest that STAT1 does play a role within fasting homeostasis that is independent of IFNβ.
4

MECHANISMS OF TYPE-I IFN INHIBITION: EQUINE HERPESVIRUS-1 ESCAPE FROM THE ANTIVIRAL EFFECT OF TYPE-1 INTERFERON RESPONSE IN HOST CELL

Oladunni, Fatai S. 01 January 2019 (has links)
Equine herpesvirus-1 (EHV-1) is one of the most important and prevalent viral pathogens of horses causing a major threat to the equine industry throughout most of the world. EHV-1 primarily causes respiratory disease but viral spread to distant organs enables the development of more severe sequelae; abortion and neurologic disease. In order to produce disease, EHV-1 has to overcome the innate barrier of the type-I interferon (IFN) system in host cells. However, the underlying mechanisms employed by EHV-1 to circumvent the type-I IFN response in host cells are not well understood. In this project study, using molecular techniques, we explored how EHV-1 is able to escape the type-I IFN response in host cells during infection. We also investigated whether EHV-4, a closely related but less pathogenic virus, has similar effects on type-I IFN as a clue to understanding how widespread IFN suppressive function is found among equine alphaherpesviruses. Our data showed that inhibition of the type-I IFN response in host cells is not a function of neuropathogenicity of EHV-1 strains. However, a reduced type-I IFN response correlated with pathogenicity as EHV-4, unlike EHV-1, was unable to down-regulate the type-I IFN response in equine endothelial cells (EECs). Investigation of the mechanisms employed by EHV-1 to suppress type-I IFN revealed that the virus sequentially prevented outside-in signaling events that lead to type-I IFN production. Specifically, EHV-1 blocked the expression of Toll-like receptors (TLR) 3 and TLR4 at 6 hours post-infection (hpi) and 12 hpi. EHV-1 also prevented the transcription of IRF7 and IRF9 at different time-points during infection. The virus also perturbed the JAK-STAT signaling pathway by negatively regulating the cellular levels of TYK2 and phosphorylation-mediated activation of STAT2 molecules. Immunofluorescence data revealed that during infection, EHV-1 was able to sequester STAT2 molecules from nuclear translocation. This may be a limiting step preventing the formation of interferon- stimulated gene factor 3 (ISGF3) whose nuclear translocation is required to transactivate interferon-stimulated genes (ISGs) including IRF7. Further investigation showed that unlike EHV-1, EHV-4 only interfered with phosphorylation-mediated activated STAT1 and STAT2 molecules at 3 and 6 hpi. EHV-4 was unable to block TLR3/4 and IRF7/9 mRNA expression at any time-point. Intriguingly, while viral late gene of EHV-1 mediates inhibition of STAT phosphorylation, our data showed that for EHV-4, a virus late gene did not mediate the inhibition of STAT phosphorylation. The findings from this study help illuminate how EHV-1 strategically interferes with limiting steps required for type-I IFN response in host cells to promote pathology. Our data also strengthen the hypothesis that the ability to shut off host factors required for type-I IFN production might be directly related to the degree of pathogenicity of the EHV subtypes.
5

The Biology of Dendritic Cells in the Context of Autoimmunity

Qiu, Connie Claire January 2019 (has links)
Systemic lupus erythematosus (SLE) is a complex autoimmune disease that affects at least five million people worldwide. An increased expression of type I interferon (IFN) regulated genes is a hallmark of SLE, but the precise etiology of SLE initiation and flares is poorly understood. Because plasmacytoid dendritic cells (pDCs) are the primary type I IFN producers, their role in SLE has long been suspected, with murine pDC depletion models successfully delaying the progression of murine lupus-like disease. However, the mechanism behind how exactly how pDCs contribute to lupus autoimmunity is unknown, contributing to the current dearth lack of disease modifying treatments; current treatments only succeed in suppressing symptoms, and do not halt disease progression. In this study, we take a multifactorial approach to understanding the biology of pDCs in the context of lupus autoimmunity. Although the exact etiology of lupus is unknown, infections are an important environmental trigger for / Infectious Disease & Immunity
6

Mechanism of inteferon-beta-mediated inhibition of IL-8 gene expression

Laver, Travis. January 2008 (has links) (PDF)
Thesis (Ph. D.)--University of Alabama at Birmingham, 2008. / Title from first page of PDF file (viewed June 6, 2008). Includes bibliographical references.
7

Development of a novel cell-based screening platform to identify inhibitors of viral interferon antagonists from clinically important viruses

Vasou, Andri January 2016 (has links)
All viruses encode for at least one viral interferon (IFN) antagonist, which is used to subvert the cellular IFN response, a powerful antiviral innate immune response. Numerous in vitro and in vivo studies have demonstrated that IFN antagonism is crucial for virus survival, suggesting that viral IFN antagonists could represent promising therapeutic targets. This study focuses on Respiratory Syncytial Virus (RSV), an important human pathogen for which there is no vaccine or virus-specific antiviral drug. RSV encodes two IFN antagonists NS1 and NS2, which play a critical role in RSV replication and pathogenicity. We developed a high-throughput screening (HTS) assay to target NS2 via our A549.pr(ISRE)GFP-RSV/NS2 cell-line, which contains a GFP gene under the control of an IFN-stimulated response element (ISRE) to monitor IFN- signalling pathway. NS2 inhibits the IFN-signalling pathway and hence GFP expression in the A549.pr(ISRE)GFP-RSV/NS2 cell-line by mediating STAT2 degradation. Using a HTS approach, we screened 16,000 compounds to identify small molecules that inhibit NS2 function and therefore relinquish the NS2 imposed block to IFN-signalling, leading to restoration of GFP expression. A total of twenty-eight hits were identified; elimination of false positives left eight hits, four of which (AV-14, -16, -18, -19) are the most promising. These four hit compounds have EC₅₀ values in the single μM range and three of them (AV-14, -16, -18) represent a chemically related series with an indole structure. We demonstrated that the hit compounds specifically inhibit the STAT2 degradation function of NS2, not the function of NS1 or unrelated viral IFN antagonists. At the current time, compounds do not restrict RSV replication in vitro, hence hit optimization is required to improve their potency. Nonetheless, these compounds could be used as chemical tools to determine the unknown mechanism by which NS2 mediates STAT2 degradation and tackle fundamental questions about RSV biology.

Page generated in 0.0252 seconds