Assessment of Mechanical Cues to Enhance the Clinical Translation of Extracellular Vesicles

dc.contributor.advisorJay, Steven Men_US
dc.contributor.authorKronstadt, Stephanie Marieen_US
dc.contributor.departmentBioengineeringen_US
dc.contributor.publisherDigital Repository at the University of Marylanden_US
dc.contributor.publisherUniversity of Maryland (College Park, Md.)en_US
dc.date.accessioned2023-02-01T06:37:40Z
dc.date.available2023-02-01T06:37:40Z
dc.date.issued2022en_US
dc.description.abstractMesenchymal stem cells (MSCs) are a common source for cell-based therapies due to their innate regenerative properties. However, these cells often die shortly after injection and, if they do survive, run the risk of forming tumors. Cell-secreted nanoparticles known as extracellular vesicles (EVs) have been identified as having therapeutic effects similar to those of their parental cells without the safety risks. Specifically, MSC EVs have emerged as a promising therapeutic modality in a multitude of applications, including autoimmune and cardiovascular diseases, cancer, and wound healing. Despite this promise, low levels of naturally occurring EV cargo may necessitate repeated doses to achieve clinical benefit, countering the advantages of EVs over MSCs. The current techniques to combat low EV potency (e.g., loading external molecules or using chemicals) are not agreeable to large-scale manufacturing techniques and would substantially increase the regulatory burden associated with EV translation. Fortunately, mechanical cues within the microenvironment have potential to overcome these translational barriers as they can alter EV therapeutic effects but are also cost-effective and can be precisely manipulated in a reproducible manner. The goal of this project is to understand how these cues impact MSC EV secretion and physiological effects. We showed that flow-derived shear stress applied to MSCs seeded within a 3D-printed scaffold (i.e., the bioreactor) can significantly upregulate EV production (EVs/cell) while maintaining the in vitro pro-angiogenic effects of MSC EVs. Interestingly, we demonstrated that MSC EVs generated using the bioreactor system significantly improved wound healing in a diabetic mouse model, with increased CD31+ staining in wound bed tissue compared to animals treated with flask cell culture-generated MSC EVs. Furthermore, for the first time, we showed that mechanical confinement of MSCs within micropillars could augment MSC EV production and bioactivity. Lastly, we demonstrated that soft substrates composed of various polydimethylsiloxane (PDMS) formulations could increase MSC EV production and activity as well. Through the work performed here, we have laid the groundwork to elucidate the relationship between cell mechanobiology and EV activity that will ultimately enable an adaptable and scalable EV therapeutic platform.en_US
dc.identifierhttps://doi.org/10.13016/dwzj-qlcl
dc.identifier.urihttp://hdl.handle.net/1903/29581
dc.language.isoenen_US
dc.subject.pqcontrolledBiomedical engineeringen_US
dc.subject.pqcontrolledCellular biologyen_US
dc.subject.pquncontrolledbioreactoren_US
dc.subject.pquncontrolledexosomesen_US
dc.subject.pquncontrolledextracellular vesiclesen_US
dc.subject.pquncontrolledmechanical cueen_US
dc.subject.pquncontrolledmesenchymal stem cellen_US
dc.subject.pquncontrolledsubstrateen_US
dc.titleAssessment of Mechanical Cues to Enhance the Clinical Translation of Extracellular Vesiclesen_US
dc.typeDissertationen_US

Files

Original bundle
Now showing 1 - 1 of 1
No Thumbnail Available
Name:
Kronstadt_umd_0117E_22930.pdf
Size:
5.7 MB
Format:
Adobe Portable Document Format
Download
(RESTRICTED ACCESS)