Show simple item record

Files in this item

Thumbnail

Item metadata

dc.contributor.advisorAndrews, Melissa
dc.contributor.advisorPowis, Simon John
dc.contributor.authorForbes, Lindsey H.
dc.coverage.spatialx, 251 p.en_US
dc.date.accessioned2018-11-28T13:03:28Z
dc.date.available2018-11-28T13:03:28Z
dc.date.issued2018-01-16
dc.identifier.urihttps://hdl.handle.net/10023/16567
dc.description.abstractRepair of the adult mammalian spinal cord is prohibited by several extrinsic and intrinsic factors. As the CNS matures, growth-promoting proteins such as integrins are developmentally downregulated resulting in a reduced capacity for axonal outgrowth. Integrins are heterodimeric receptors involved in cell-cell and cell-matrix interactions. Specifically, within mature corticospinal tract (CST) axons, integrins are not transported into the axonal compartment. One integrin heterodimer, α9β1, is of particular interest for its ability to promote neurite outgrowth when bound to a component of the injury-induced milieu, tenascin-C. This project aimed to increase integrin expression within the CNS using induced pluripotent stem cell-derived human neural progenitor cells (iPSC-hNPCs). Using immunocytochemistry and western blotting, endogenous integrin expression within iPSC-hNPCs was determined. In addition, overexpression of α9 integrin was achieved using transfection and lentiviral transduction. The capacity of wild type (WT) and α9-hNPCs to extend neurites on tenascin-C was assessed using neurite outgrowth assays. Results revealed increasing α9 integrin expression in hNPCs significantly promoted neurite outgrowth when cultured on tenascin-C. Interestingly, increasing the concentration of human tenascin-C, resulted in increasingly longer neurites from WT hNPCs suggesting hNPCs could actively upregulate integrin expression. Subsequently, WT and α9-hNPCs were transplanted into layer V of the neonatal rat sensorimotor cortex, which projects to the CST. WT and α9-hNPCs survived up to 8 weeks post-transplantation and produced projections along white matter tracts, including areas of the CST. Additionally, hNPCs retained α9-eYFP protein expression in vivo over time and was localised within axonal projections. These results highlight the capabilities of iPSC-hNPCs to promote integrin expression within the rodent CNS presenting one potential avenue to target neuronal replacement following spinal injury. Future research should focus on assessing the regenerative capacity of WT and α9-hNPCs within an injury model concentrating on the ability of these cells to adapt within an injured environment.en_US
dc.language.isoenen_US
dc.publisherUniversity of St Andrews
dc.rightsAttribution-NonCommercial-NoDerivatives 4.0 International*
dc.rights.urihttp://creativecommons.org/licenses/by-nc-nd/4.0/*
dc.subjectNeuroregenerationen_US
dc.subjectStem cellsen_US
dc.subjectIntegrinen_US
dc.subjectBrain and spinal cord repairen_US
dc.subjectTransplantationen_US
dc.subjectCNSen_US
dc.subject.lccQP370.F7
dc.subject.lcshCentral nervous system--Regenerationen
dc.subject.lcshIntegrinsen
dc.titleUsing human iPSC-derived neural progenitor cells to increase integrin expression in the CNSen_US
dc.typeThesisen_US
dc.contributor.sponsorUniversity of St Andrews. 600th Anniversary Scholarshipen_US
dc.type.qualificationlevelDoctoralen_US
dc.type.qualificationnamePhD Doctor of Philosophyen_US
dc.publisher.institutionThe University of St Andrewsen_US
dc.publisher.departmentSchool of Medicineen_US
dc.identifier.doihttps://doi.org/10.17630/10023-16567


The following licence files are associated with this item:

    This item appears in the following Collection(s)

    Show simple item record

    Attribution-NonCommercial-NoDerivatives 4.0 International
    Except where otherwise noted within the work, this item's licence for re-use is described as Attribution-NonCommercial-NoDerivatives 4.0 International