Investigating the homing and healing properties of adipose-derived mesenchymal stromal cells using a model of wound repair

Please be advised that the site will be down for maintenance on Sunday, September 1, 2024, from 08:00 to 18:00, and again on Monday, September 2, 2024, from 08:00 to 09:00. We apologize for any inconvenience this may cause.

Show simple item record

dc.contributor.advisor Pepper, Michael Sean
dc.contributor.coadvisor Pittet-Cuénod, Brigitte
dc.contributor.coadvisor Modarressi, Ali
dc.contributor.coadvisor Martinou, Jean-Claude
dc.contributor.postgraduate Kallmeyer, Karlien
dc.date.accessioned 2019-12-05T13:15:08Z
dc.date.available 2019-12-05T13:15:08Z
dc.date.created 2020-04
dc.date.issued 2019
dc.description Thesis (PhD (Medical Immunology))--University of Pretoria, 2019. en_ZA
dc.description.abstract Adult mesenchymal stromal cells show promise therapeutically due to their multipotent differentiation capacity, immunomodulatory properties, paracrine signalling and ability to migrate to sites of injury. The potential use of adipose-derived stromal cells (ASCs) as a cellular therapy for treating delayed healing, non-healing and chronic wounds, their optimal route of administration, bio-distribution and fate needs to be investigated. Therefore, this study aimed first to establish the location and survival of systemically and locally administered ASCs during wound repair under physiological conditions (model 1) and second, the effect of locally administered ASCs during wound repair under pathological conditions of hyperglycaemia and ischemia (model 2). A dual tracking approach was used to follow firefly luciferase (Fluc) and green fluorescent protein (GFP) expressing ASCs by bioluminescence imaging (BLI) and histological analysis. The immuno-phenotype and differentiation capacity of rat ASCs transduced to express Fluc and GFP were assessed before the cells were used in the wound repair models. In model 1, full-thickness bilateral wounds were created on the dorsal aspect of the hind paws in healthy rats and two treatment modes were assessed: a single dose of 2 x 106 ASCs or NaCl administered systemically into the tail vein or 2 x 105 ASCs injected locally around each wound. Animals were followed by digital photography, BLI and sacrificed for histological analysis. In model 2, ischemia was induced unilaterally by resection of the femoral artery in hyperglycaemic rats before full-thickness bilateral wounds were created and 2 x 105 ASCs or NaCl administered locally around each non-ischemic and ischemic wound. Animals were followed by digital photography and sacrificed for histology and immunohistochemistry (IHC). Haematoxylin/eosin (H/E) staining as well as Masson’s trichrome staining and IHC for alpha-smooth muscle actin (αSMA), ionised calcium binding adaptor molecule 1 (Iba1) and GFP were performed on sample sections. Wound closure time and the contraction/epithelialisation ratio were assessed in both models. Transduced ASCs maintained their immuno-phenotype and differentiation capacity. Under physiological conditions, systemically administered ASCs were filtered out in the lungs, whereas locally administered ASCs survived at the injection site and migrated into the wound bed. Wound closure was accelerated by 5 and 7 days with systemic and local ASC treatment Page | iv respectively compared to control animals, and this was the result of increased epithelialisation. Under pathological conditions, locally administered ASCs significantly enhanced wound closure in non-ischemic and ischemic wounds by 9 days compared to control wounds. Semi-quantitative analysis revealed that ASC treatment led to enhanced cellularity in the wound. No changes in collagen deposition, vascularisation (as determined by αSMA staining) or macrophage infiltration were observed between ASC treated and control groups. However, αSMA staining was detected earlier and remained higher at wound closure in the former without enhancing wound closure by contraction. Despite the limited systemic homing capacity of ASCs, wound healing was improved. Locally injected ASCs migrated from the wound edge into the wound bed where they promoted wound repair. Under pathological conditions, ASCs enhanced wound closure. A significant increase in wound cellularity was observed, possibly through a mechanism of paracrine signalling that recruited more immune regulating and tissue repair cells into the granulation tissue. Administration of ASCs for delayed healing wounds show promise as a cellular treatment for enhancing wound repair. Key words: Adipose-derived mesenchymal stromal cells (ASCs), in vivo imaging, bioluminescence imaging (BLI), green fluorescent protein (GFP), firefly luciferase (Fluc), re-epithelialisation, contraction, wound healing, wound repair, homing en_ZA
dc.description.availability Unrestricted en_ZA
dc.description.degree PhD (Medical Immunology) en_ZA
dc.description.department Immunology en_ZA
dc.description.sponsorship NRF, Scarce Skills Doctoral Scholarship, Grant UID: 88799 Swiss National Science Foundation Project (#310030_170132) The South African Medical Research Council University Flagship program (SAMRC-RFA-UFSP-01-2013/STEM CELLS) Medical faculty of University of Geneva The SAMRC Extramural Unit for Stem Cell Research and Therapy, and the Institute for Cellular and Molecular Medicine (ICMM) of the University of Pretoria Ernst & Ethel Eriksen Trust en_ZA
dc.identifier.citation Kallmeyer, K 2019, Investigating the homing and healing properties of adipose-derived mesenchymal stromal cells using a model of wound repair. PhD thesis, University of Pretoria, Pretoria, viewed yymmdd http://hdl.handle.net/2263/72532 en_ZA
dc.identifier.other A2020
dc.identifier.uri http://hdl.handle.net/2263/72532
dc.language.iso en en_ZA
dc.publisher University of Pretoria
dc.rights © 2019 University of Pretoria. All rights reserved. The copyright in this work vests in the University of Pretoria. No part of this work may be reproduced or transmitted in any form or by any means, without the prior written permission of the University of Pretoria.
dc.subject UCTD en_ZA
dc.title Investigating the homing and healing properties of adipose-derived mesenchymal stromal cells using a model of wound repair en_ZA
dc.type Thesis en_ZA


Files in this item

This item appears in the following Collection(s)

Show simple item record