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

Functions of connexin 46 in lens and solid tumors during hypoxia

Molina, Samuel A. January 1900 (has links)
Doctor of Philosophy / Graduate Biochemistry Group / Dolores J. Takemoto / Eukaryotic cells possess a unique way to communicate with each other by passing metabolites and small molecules through protein pores that connect adjacent cells. Although there are many types and families of protein pores, connexins comprise a unique family. Six connexin monomers assemble into a hemichannel, which is transported to the cell membrane. An opposing cell membrane containing compatible connexin hemichannels is located and connected, forming an intercellular dodecameric protein complex. This results in a protein channel that connects two separate cytoplasmic compartments to each other. This type of channel is known as a gap junction. Connexin expression and function is commonly tissue specific. Of the 21 known human connexins, less than half are currently well characterized. Three connexins are expressed in the lens, connexin 43 (Cx43), 46 (Cx46), and 50 (Cx50). Of these three, Cx46 and Cx50 both have major functions in the mature lens. Cx46 functions as a major gap junction channel, which maintains mature lens homeostasis, while Cx50 possesses growth control properties in the lens. Cx46 expression is modulated in breast and bone tumors, and during ischemia. It is hypothesized that Cx46 provides resistance to hypoxia mediated cell death by prolonging survival. In this study, Cx46 expression was detected in human Y79 retinoblastoma cells. Decreasing the expression of Cx46 in nude mice carrying Y79 xenografts slowed early stage tumor growth. Y79 cells in culture survive for over 72 hours in 1% oxygen in vitro. C46 was upregulated in cultured lens cells when grown under hypoxia. Human lens epithelial cells, rabbit N/N1003A lens cells, and Y79 cells proliferated in 1% oxygen until Cx46 expression was depleted by use of siRNA. Protection from hypoxia-induced cell death was provided by transfection with the C-terminus of Cx46. We further determined that the promoter activity of Cx46 was increased in 1% oxygen. These results indicate that Cx46 would increase in response to hypoxia and suggest a role for Cx46 in protection from hypoxia. The studies demonstrate a novel function for Cx46 in cell survival during hypoxia.
2

Study of the involvement of autophagy in the acquisition of tumor resistance to Natural Killer-mediated lysis / Etude de l'implication de l'autophagie dans l'acquisition de résistance tumorale à la lyse par les lymphocytes "Natural Killer"

Baginska, Joanna 28 November 2013 (has links)
Les lymphocytes « Natural Killer » (NK) sont des effecteurs de l’immunité innée, capables de lyser les cellules cancéreuses grâce au relargage de la protéase cytotoxique Granzyme B (GzmB). Récemment, de nouvelles stratégies anti-cancéreuses, basées sur l’utilisation des cellules NK, ont émergé et se sont révélées très prometteuses. Il est maintenant clairement établi que le microenvironnement tumoral hypoxique influence la réponse immunitaire et constitue, de ce fait, un obstacle majeur pour établir des protocoles d’immunothérapies efficaces. Des études récentes ont montré que l’autophagie est un régulateur important de l’immunité innée dans le microenvironnement tumoral, mais les mécanismes de régulation impliqués restent encore peu connus. Nous avons montré in vitro que l'hypoxie diminue la sensibilité des cellules de carcinome mammaire à la lyse dépendante des cellules NK par un mécanisme impliquant l'activation de l'autophagie. De manière intéressante, cette diminution de lyse est reversée par l’inhibition de l’autophagie. Nous avons démontré que la résistance des cellules tumorales hypoxiques à la lyse par les cellules NK n'est liée ni à un défaut de reconnaissance des cellules cibles, ni à une altération de l’activité cytotoxique des effecteurs. Nous avons mis en évidence que l'activation de l’autophagie conduit à la dégradation de GzmB dans les lysosomes des cellules hypoxiques. Ainsi, ces cellules deviennent résistantes à l’apoptose, qui est normalement induite par GzmB, transféré par les cellules NK. L’invalidation génétique et pharmacologique de l'autophagie permet de restaurer le niveau intracellulaire de GzmB et réduit la résistance des cellules cibles hypoxiques in vitro. Nos résultats mettent en évidence que l'autophagie est un régulateur primordial de la réponse immunitaire anti-tumorale dépendante des cellules NK. Nous avons validé ce concept in vivo en montrant que l’inhibition de l'autophagie favorise de manière significative la prise en charge de la tumeur par les cellules NK dans des modèles murins de mélanome et de carcinome mammaire. Cette étude contribue à l’avancé des connaissances sur la manière dont l'autophagie, induite par l'hypoxie, affecte la lyse dépendante des cellules NK et ouvre la voie à la formulation de nouvelles stratégies thérapeutiques anti-tumorales combinant l’utilisation des cellules NK à des inhibiteurs d'autophagie. / Natural killer (NK) cells are effectors of the antitumor immunity, able to kill cancer cells through the release of the cytotoxic protease granzyme B. NK-based therapies have recently emerged as promising anticancer strategies. However, it is well established that hypoxic microenvironment interferes with the function of antitumor immune cells and constitutes a major obstacle for cancer immunotherapies. Recent studies demonstrated that autophagy is an important regulator of innate immune response in this microenvironment, but the mechanism by which autophagy regulates NK cell-mediated antitumor immune responses remains elusive. Here, we demonstrate that hypoxia impairs breast cancer cell susceptibility to NK-mediated lysis in vitro via the activation of autophagy. This impairment was not related to a defect in target cell recognition by NK cells but to the degradation of NK-derived granzyme B in autophagosomes of hypoxic cells. Inhibition of autophagy by targeting beclin1 (BECN1) restored granzyme B levels in hypoxic cells in vitro and induced tumor regression in vivo by facilitating NK-mediated tumor cell killing. Together, our data highlight autophagy as a mechanism underlying the resistance of hypoxic tumor cells to NK-mediated lysis and provides a cutting-edge advance in our understanding of the underlying mechanism. This study might pave the way for the formulation of more effective NK cell-based antitumor therapies.

Page generated in 0.0554 seconds