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

Cellular, molecular, and evolutionary mechanisms of Wolbachia stem cell niche tropism in Drosophila

Olsen, Michelle Toomey 12 March 2016 (has links)
The intracellular bacteria Wolbachia infect up to 40% of all insect species, including the vectors of prevalent infectious diseases such as Dengue and malaria. Even though Wolbachia infections are the largest pandemic on this planet, the cellular and molecular mechanisms for bacterial spreading in nature are still unknown. Wolbachia are mainly vertically transmitted through the egg cytoplasm, however there is also evidence of extensive horizontal transmission. We have found that Wolbachia target the stem cell niches in the Drosophila ovary to enhance germline colonization and subsequent vertical transmission. This tropism is pervasive across the Drosophila genus, with the pattern of targeting being evolutionarily conserved. Phylogenetic analyses, confirmed by hybrid introgression and transinfection experiments, demonstrate that bacterial factors are the major determinants of differential patterns of niche tropism. Furthermore, bacterial load is increased in germline cells passing through infected niches, supporting previous findings suggesting a contribution of Wolbachia from stem cell niches towards vertical transmission. If niche tropism is important for Wolbachia transmission through the germline, evolutionary theory predicts that there should be no selective pressure to maintain niche tropism in males. Indeed, we have found that tropism to the stem cell niche in the testis, known as the hub, is not evolutionarily conserved. Towards identifying the cellular and molecular mechanisms of stem cell niche tropism, we investigated hub targeting of closely related Wolbachia strains (wMel-like strains: wMel, wMel2, and wMel3; wMelCS-like strains: wMelCS, wMelCS2, and wMelPop). wMel-like and wMelCS-like Wolbachia strains differ in their frequencies and densities of hub infection. The targeting differences of these strains of Wolbachia indicate that this phenotype is rapidly evolving, as they shared a common ancestor only 8,000 years ago. With the plethora of tools available in D. melanogaster, a candidate gene approach was used to target host proteins enriched in the stem cell niche in the testis for RNAi mediated gene knockdown in the hub. We have identified Drosophila stem cell related signaling pathways that promote Wolbachia accumulation. Unraveling the cellular and molecular bases of tissue tropism is fundamental to understanding Wolbachia-host interactions. / 2017-01-01T00:00:00Z
2

Random Mutagenesis for the Discovery of Obligate Intracellular Bacterial <i>In vivo</i> Virulence Genes

Bekebrede, Hannah S. January 2019 (has links)
No description available.
3

Investigation of Anaplasma phagocytophilum and Anaplasma marginale adhesin-host cell interactions

Hebert, Kathryn S. 01 January 2016 (has links)
Anaplasma phagocytophilum and A. marginale are the etiologic agents of bovine anaplasmosis and human granulocytic anaplasmosis, respectively. As obligate intracellular pathogens, binding and entry of host cells is a prerequisite for survival. The molecular events associated with these processes are poorly understood. Identifying the adhesins mediating binding, delineating their key functional domains, and determining the molecular determinants to which they bind not only benefits better understanding of Anaplasma spp. pathobiology, but could also benefit the development of novel approaches for protecting against infection. We previously demonstrated that A. phagocytophilum outer membrane protein A (ApOmpA) is critical for bacterial binding and entry host through recognition of α2,3-sialic acid and α1,3-fucose of its receptors, including 6-sulfo-sLex. In this study, we determined that two amino acids, G61 and K64, within its binding domain (ApOmpA59-74), are essential for ApOmpA function. We also confirmed the ability of ApOmpA to act as an adhesin and invasin as it conferred adhesiveness and invasiveness to inert beads. We next extended our studies to A. marginale as it also expresses OmpA (AmOmpA) and its role in infection has not been studied. Molecular models of ApOmpA and AmOmpA were nearly identical, especially in the ApOmpA binding domain and its counterpart in AmOmpA. Antisera raised against AmOmpA or its putative binding domain inhibit A. marginale infection. AmOmpA G55 and K58 are contributory and K59 is essential for AmOmpA to bind to host cells. AmOmpA binding is dependent on α2,3-sialic acid and α1,3-fucose. Coating inert beads with AmOmpA conferred the ability to bind to and be taken up by host cells, confirming that it acts as an adhesin and invasin. 6-sulfo-sLex is dispensable for AmOmpA binding and A. marginale infection. ApOmpA works cooperatively with Asp14 (14-kDa A. phagocytophilum surface protein) to promote optimal infection of host cells. We found that Asp14 is conserved across A. phagocytophilum strains and in A. marginale and confirmed the ability of Asp14 to act as an adhesin and invasin as it conferred adhesiveness and invasiveness to inert beads. Collectively, this work advances our understanding of A. phagocytophilum and A. marginale adhesion and invasion of host cells.
4

Orientia tsutsugamushi secretes two ankyrin repeat-containing effectors via a type 1 secretion system to inhibit host NF-κB function

Evans, Sean M. 01 January 2017 (has links)
Scrub typhus is a potentially fatal infection that threatens one billion persons in the Asia-Pacific region and is caused by the obligate intracellular bacterium, Orientia tsutsugamushi. How this organism facilitates its intracellular survival and pathogenesis is poorly understood. Intracellular bacterial pathogens utilize the Type 1 (T1SS) or Type 4 secretion system (T4SS) to translocate ankyrin repeat-containing proteins (Anks) into the host cell to modulate host cell processes. The O. tsutsugamushi genome encodes one of the largest known bacterial Ank libraries as well as Type 1 and Type 4 secretion systems (T1SS and T4SS), which are expressed during infection. In silico analyses of the Anks’ C-termini revealed that they possess characteristics of T1SS secretion signals. Escherichia coli expressing a functional T1SS was able to secrete chimeric hemolysin proteins bearing the C-termini of 19 of 20 O. tsutsugamushi Anks. In addition to infecting endothelial cells, O. tsutsugamushi infects professional phagocytes. To better understand why these innate immune cells are unable to eliminate O. tsutsugamushi, we addressed the activity of host NF-κB proinflammatory transcription factor. Screening of O. tsutsugamushi infected cells at an MOI of 1 revealed inhibition of NF-κB nuclear accumulation as early as 8 hours in HeLa and bone-marrow derived macrophage cells. When stimulating infected cells with TNF-α, IκBα degradation still occurs, however NF-κB dependent gene transcription remains downregulated. Immunofluorescence microscopic analysis of TNF-α treated cells ectopically expressing all O. tsutsugamushi Anks revealed that two nuclear trafficking Anks, Ank1 and Ank6, result in a significant decrease in NF-κB nuclear accumulation. Additionally, these Anks also significantly inhibited NF-κB dependent gene transcription. Co-immunoprecipitation experiments revealed that both Anks interact with importin-β1, exportin-1, and the p65 NF-κB subunit. Treating cells with importazole significantly reduces the nuclear accumulation of Ank1 and Ank6. Finally, treating infected cells or cells ectopically expressing Ank1 or Ank6 with leptomycin B resulted in restoration of NF-κB nuclear accumulation. With these data, we propose that O. tsutsugamushi secretes Ank1 and Ank6 to initially interact with importin-β1, which permits their nuclear entry where they then interact with NF-κB and subsequently exportin-1 to prevent NF-κB nuclear accumulation.

Page generated in 0.1474 seconds