@phdthesis{Konrad2023, author = {Konrad, Johannes}, title = {Biomechanische Eigenschaften eines biomaterialbasierten Kreuzbandkonstruktes in-vivo und in-vitro}, doi = {10.25972/OPUS-23555}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-235555}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2023}, abstract = {Kreuzbandrupturen stellen nach wie vor eine Herausforderung in der klinischen Praxis hinsichtlich kurz- und langfristiger unerw{\"u}nschter Nebenwirkungen dar (z.B. Reruptur und Arthrosebildung). In der vorliegenden Arbeit wird der entwickelte Ansatz eines Kollagen-I-basierten k{\"u}nstlichen Kreuzbandkonstruktes hinsichtlich der Reißfestigkeit, Lagerung, Verst{\"a}rkungsm{\"o}glichkeit mittels Fiber-tape und langfristigen Arthroseentstehung untersucht mittels in-vitro und in-vivo Untersuchungen unter zur Hilfe nahme des Minipig Tiermodels. Die Ergebnisse zeigen keinen Einfluss der Lagerungstemperatur sowie des Lagerungszeitraums auf die Reißfestigkeit des Konstruktes, sowie eine m{\"o}gliche initiale Verst{\"a}rkung mittels Fibertape im Minipig. Dar{\"u}ber hinaus wurde mikroskopisch wie makroskopische Arthroseentstehung nachgewiesen. Das Ausmaß der Arthroseentstehung ist diesbez{\"u}glich mit einer Abweichung der Konstruktimplantation von der urspr{\"u}nglichen Kreuzbandinsertion mittels MRT best{\"a}tigt worden.}, subject = {Ligamentum cruciatum anterius}, language = {de} } @phdthesis{Kupczyk2023, author = {Kupczyk, Eva Katharina}, title = {Charakterisierung von Zellen aus dem vorderen Kreuzband nach Vorderer- Kreuzband-Ruptur im Hinblick auf das Rupturalter}, doi = {10.25972/OPUS-28056}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-280568}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2023}, abstract = {Die Vordere Kreuzband (VKB)-Ruptur ist eine h{\"a}ufige Verletzung, welche eine hohe individuelle und sozio{\"o}konomische Belastung verursacht. Eine etablierte Therapie ist die VKB-Plastik, problematisch sind jedoch die hohen Rerupturraten nach operativer Versorgung. In der Annahme, dass Mesenchymale Stammzellen (MSC) eine bedeutende Rolle f{\"u}r die Heilung spielen, sollte in der vorliegenden Arbeit untersucht werden, ob ein Zusammenhang zwischen Zahl und Qualit{\"a}t der aus dem VKB isolierten MSC sowie der Latenz zwischen Ruptur und Rekonstruktion besteht und so ein optimaler Therapiezeitraum eingegrenzt werden kann. Zun{\"a}chst erfolgte die Zellisolierung aus intraoperativ gewonnenen VKB-Biopsien. Je nach Latenz zwischen Ruptur und Operation wurden drei Gruppen (akute ≙ ≤ 30 d, subakute ≙ 31-90 d, verz{\"o}gerte Rekonstruktion ≙ > 90 d) gebildet. Zum Nachweis von MSC wurden die Zellen hinsichtlich ihrer Plastikadh{\"a}renz, eines multipotenten Differenzierungspotentials sowie eines spezifischen Oberfl{\"a}chenantigenmusters (CD73+, CD90+, CD105+, CD34-) untersucht. Zudem wurde ihr Einflusses auf die biomechanischen und histologischen Eigenschaften eines analysiert. Der Nachweis von MSC war in allen Gruppen m{\"o}glich. Das Proliferationspotential war in Gruppe II am gr{\"o}ßten, ebenso der Anteil der MSC an allen Zellen. Er war 5,4\% (4,6\% - 6,3\%, 95\% CI; p < 0,001) h{\"o}her als in Gruppe I und 18,9\% (18,2\% - 19,6\%, 95\% CI; p < 0,001) h{\"o}her als in Gruppe III. In den mit Zellen kultivierten Bandkonstrukten konnte im Gegensatz zu zellfreien Konstrukten humanes Kollagen I nachgewiesen werden. Die Stabilit{\"a}t nahm bei Kultivierung mit Zellen ab. Die Ergebnisse legen nahe, dass das Regenerationspotential bei subakuter VKB-Rekonstruktion (31-90 d) am h{\"o}chsten ist. Potenziell urs{\"a}chlich sind die Regeneration hemmende Entz{\"u}ndungsprozesse zu Beginn sowie degenerative Prozesse im l{\"a}ngerfristigen Verlauf. Zudem konnte gezeigt werden, dass die isolierten Zellen die Eigenschaften eines Bandkonstruktes durch Bildung von Kollagen I und Reduktion der Stabilit{\"a}t im kurzfristigen Verlauf ver{\"a}ndern und dementsprechend den Therapieerfolg beeinflussen k{\"o}nnten. Zur Verifizierung der Ergebnisse bedarf es weiterer Untersuchungen.}, subject = {Ligamentum cruciatum anterius}, language = {de} } @phdthesis{SchmidtgebSchmid2023, author = {Schmidt [geb. Schmid], Freia Florina}, title = {Ein dreidimensionales kutanes Melanommodell f{\"u}r den Einsatz in der pr{\"a}klinischen Testung}, doi = {10.25972/OPUS-32925}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-329255}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2023}, abstract = {Das maligne Melanom nimmt als Tumorerkrankung mit hoher Metastasierungsrate und steigenden Inzidenzraten bei h{\"o}chster Mortalit{\"a}t aller Hauttumoren eine zunehmende Bedeutung in der modernen Onkologie ein. Fr{\"u}hzeitige Diagnosem{\"o}glichkeiten und moderne Behandlungen konnten das {\"U}berleben der Patienten bereits erheblich verbessern. Jedoch besteht nach wie vor Bedarf an geeigneten Modellen, um die Melanomprogression vollst{\"a}ndig zu verstehen und neue wirksame Therapien zu entwickeln. Hierf{\"u}r werden h{\"a}ufig Tiermodelle verwendet, diese spiegeln jedoch nicht die menschliche Mikroumgebung wider. Zweidimensionalen Zellkulturen fehlen dagegen entscheidende Elemente der Tumormikroumgebung. Daher wurde in dieser Arbeit ein dreidimensionales epidermales Tumormodell des malignen Melanoms, welches aus prim{\"a}ren humanen Keratinozyten und verschiedenen Melanomzelllinien besteht, entwickelt. Die eingesetzten Melanomzelllinien variieren in ihren Treibermutationen, wodurch das Modell in der Lage ist, Wirkstoffe zu untersuchen, die spezifisch auf diese Mutationen wirken. Mit Techniken des Tissue Engineerings konnte ein dreidimensionales Hautmodell aufgebaut werden, das alle charakteristischen Schichten der Epidermis aufweist und im Bereich des stratum basale Melanomcluster ausbildet. Diese reichen je nach Gr{\"o}ße und Ausdehnung bis in suprabasale Epidermisschichten hinein. Die Tumor-Histopathologie, der Tumorstoffwechsel sowie tumorassoziierte Proteinsekretionen ließen sich im in vitro Modell nachweisen. Dar{\"u}ber hinaus konnte ein Protokoll entwickelt werden, mit dem einzelne Zellen aus den Modellen reisoliert werden k{\"o}nnen. Dies erm{\"o}glichte es, den Proliferationszustand innerhalb des jeweiligen Modells zu charakterisieren und die Wirkung von Antitumortherapien gezielt zu bewerten. Die Anwendbarkeit als Testsystem im Bereich der Tumortherapeutika wurde mit dem in der Klinik h{\"a}ufig verwendeten v-raf-Maus-Sarkom-Virus-Onkogen-Homolog B (BRAF)-Inhibitor Vemurafenib demonstriert. Der selektive BRAF-Inhibitor reduzierte erfolgreich das Tumorwachstum in den Modellen mit BRAF-mutierten Melanomzellen, was durch eine Verringerung der metabolischen Aktivit{\"a}t, der proliferierenden Zellen und des Glukoseverbrauchs gezeigt wurde. F{\"u}r die Implementierung des Modells in die pr{\"a}klinische Therapieentwicklung wurde B-B-Dimethylacrylshikonin, ein vielversprechender Wirkstoffkandidat, welcher einen Zellzyklusarrest mit anschließender Apoptose bewirkt, im Modell getestet. Bei einer Anwendung der Modelle im Bereich der Testung topischer Behandlungen ist eine Barrierefunktion der Modelle notwendig, die der in vivo Situation nahe kommt. Die Barriereeigenschaften der Haut{\"a}quivalente wurden durch die Melanomzellen nachweislich nicht beeinflusst, sind aber im Vergleich zur in vivo Situation noch unzureichend. Eine signifikante Steigerung der Hautbarriere konnte durch die Bereitstellung von Lipiden und die Anregung hauteigener Regenerationsprozesse erreicht werden. {\"U}ber den Nachweis des transepidermalen Wasserverlusts konnte eine Messmethode zur nicht-invasiven Bestimmung der Hautbarriere etabliert und {\"u}ber den Vergleich zur Impedanzspektroskopie validiert werden. Hierbei gelang es, erstmals die Korrelation der Hautmodelle zur in vivo Situation {\"u}ber ein solches Verfahren zu zeigen. Das entwickelte epidermale Modell konnte durch die Integration eines dermalen Anteils und einer Endothelzellschicht noch weiter an die komplexe Struktur und Physiologie der Haut angepasst werden um Untersuchungen, die mit der Metastierung und Invasion zusammenh{\"a}ngen, zu erm{\"o}glichen. Die artifizielle Dermis basiert auf einem Kollagen-Hydrogel mit prim{\"a}ren Fibroblasten. Eine dezellularisierte Schweinedarmmatrix ließ sich zur Erweiterung des Modells um eine Endothelzellschicht nutzen. Dabei wanderten die prim{\"a}ren Fibroblasten apikal in die nat{\"u}rliche Schweindarmmatrix ein, w{\"a}hrend die Endothelzellen basolateral eine geschlossene Schicht bildeten. Die in dieser Arbeit entwickelten Gewebemodelle sind in der Lage, die Vorhersagekraft der in vitro Modelle und die in vitro - in vivo Korrelation zu verbessern. Durch die Kombination des Melanommodells mit einer darauf abgestimmten Analytik wurde ein neuartiges Werkzeug f{\"u}r die pr{\"a}klinische Forschung zur Testung von pharmazeutischen Wirkstoffen geschaffen.}, subject = {Tissue Engineering}, language = {de} } @phdthesis{Alzheimer2023, author = {Alzheimer, Mona}, title = {Development of tissue-engineered three-dimensional infection models to study pathogenesis of \(Campylobacter\) \(jejuni\)}, doi = {10.25972/OPUS-19344}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-193440}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2023}, abstract = {Infectious diseases caused by pathogenic microorganisms are one of the largest socioeconomic burdens today. Although infectious diseases have been studied for decades, in numerous cases, the precise mechanisms involved in the multifaceted interaction between pathogen and host continue to be elusive. Thus, it still remains a challenge for researchers worldwide to develop novel strategies to investigate the molecular context of infectious diseases in order to devise preventive or at least anti-infective measures. One of the major drawbacks in trying to obtain in-depth knowledge of how bacterial pathogens elicit disease is the lack of suitable infection models to authentically mimic the disease progression in humans. Numerous studies rely on animal models to emulate the complex temporal interactions between host and pathogen occurring in humans. While they have greatly contributed to shed light on these interactions, they require high maintenance costs, are afflicted with ethical drawbacks, and are not always predictive for the infection outcome in human patients. Alternatively, in-vitro two-dimensional (2D) cell culture systems have served for decades as representatives of human host environments to study infectious diseases. These cell line-based models have been essential in uncovering virulence-determining factors of diverse pathogens as well as host defense mechanisms upon infection. However, they lack the morphological and cellular complexity of intact human tissues, limiting the insights than can be gained from studying host-pathogen interactions in these systems. The focus of this thesis was to establish and innovate intestinal human cell culture models to obtain in-vitro reconstructed three-dimensional (3D) tissue that can faithfully mimic pathogenesis-determining processes of the zoonotic bacterium Campylobacter jejuni (C. jejuni). Generally employed for reconstructive medicine, the field of tissue engineering provides excellent tools to generate organ-specific cell culture models in vitro, realistically recapitulating the distinctive architecture of human tissues. The models employed in this thesis are based on decellularized extracellular matrix (ECM) scaffolds of porcine intestinal origin. Reseeded with intestinal human cells, application of dynamic culture conditions promoted the formation of a highly polarized mucosal epithelium maintained by functional tight and adherens junctions. While most other in-vitro infection systems are limited to a flat monolayer, the tissue models developed in this thesis can display the characteristic 3D villi and crypt structure of human small intestine. First, experimental conditions were established for infection of a previously developed, statically cultivated intestinal tissue model with C. jejuni. This included successful isolation of bacterial colony forming units (CFUs), measurement of epithelial barrier function, as well as immunohistochemical and histological staining techniques. In this way, it became possible to follow the number of viable bacteria during the infection process as well as their translocation over the polarized epithelium of the tissue model. Upon infection with C. jejuni, disruption of tight and adherens junctions could be observed via confocal microscopy and permeability measurements of the epithelial barrier. Moreover, C. jejuni wildtype-specific colonization and barrier disruption became apparent in addition to niche-dependent bacterial localization within the 3D microarchitecture of the tissue model. Pathogenesis-related phenotypes of C. jejuni mutant strains in the 3D host environment deviated from those obtained with conventional in-vitro 2D monolayers but mimicked observations made in vivo. Furthermore, a genome-wide screen of a C. jejuni mutant library revealed significant differences for bacterial factors required or dispensable for interactions with unpolarized host cells or the highly prismatic epithelium provided by the intestinal tissue model. Elucidating the role of several previously uncharacterized factors specifically important for efficient colonization of a 3D human environment, promises to be an intriguing task for future research. At the frontline of the defense against invading pathogens is the protective, viscoelastic mucus layer overlying mucosal surfaces along the human gastrointestinal tract (GIT). The development of a mucus-producing 3D tissue model in this thesis was a vital step towards gaining a deeper understanding of the interdependency between bacterial pathogens and host-site specific mucins. The presence of a mucus layer conferred C. jejuni wildtype-specific protection against epithelial barrier disruption by the pathogen and prevented a high bacterial burden during the course of infection. Moreover, results obtained in this thesis provide evidence in vitro that the characteristic corkscrew morphology of C. jejuni indeed grants a distinct advantage in colonizing mucous surfaces. Overall, the results obtained within this thesis highlight the strength of the tissue models to combine crucial features of native human intestine into accessible in-vitro infection models. Translation of these systems into infection research demonstrated their ability to expose in-vivo like infection outcomes. While displaying complex organotypic architecture and highly prismatic cellular morphology, these tissue models still represent an imperfect reflection of human tissue. Future advancements towards inclusion of human primary and immune cells will strive for even more comprehensive model systems exhibiting intricate multicellular networks of in-vivo tissue. Nevertheless, the work presented in this thesis emphasizes the necessity to investigate host-pathogen interactions in infection models authentically mimicking the natural host environment, as they remain among the most vital parts in understanding and counteracting infectious diseases.}, subject = {Campylobacter jejuni}, language = {en} } @phdthesis{SchliermanngebStratmann2023, author = {Schliermann [geb. Stratmann], Anna Theresa}, title = {The Role of FGF Receptor 2 in GDF5 mediated Signal Transduction}, doi = {10.25972/OPUS-19288}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-192889}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2023}, abstract = {Bone morphogenetic proteins (BMPs) are involved in various aspects of cell-cell communication in complex life forms. They act as morphogens, help differentiate different cell types from different progenitor cells in development, and are involved in many instances of intercellular communication, from forming a body axis to healing bone fractures, from sugar metabolism to angiogenesis. If the same protein or protein family carries out many functions, there is a demand to regulate and fine-tune their biological activities, and BMPs are highly regulated to generate cell- and context-dependent outcomes. Not all such instances can be explained yet. Growth/differentiation factor (GDF)5 (or BMP14) synergizes with BMP2 on chondrogenic ATDC5 cells, but antagonizes BMP2 on myoblastic C2C12 cells. Known regulators of BMP2/GDF5 signal transduction failed to explain this context-dependent difference, so a microarray was performed to identify new, cell-specific regulatory components. One identified candidate, the fibroblast growth factor receptor (FGFR)2, was analyzed as a potential new co-receptor to BMP ligands such as GDF5: It was shown that FGFR2 directly binds BMP2, GDF5, and other BMP ligands in vitro, and FGFR2 was able to positively influence BMP2/GDF5-mediated signaling outcome in cell-based assays. This effect was independent of FGFR2s kinase activity, and independent of the downstream mediators SMAD1/5/8, p42/p44, Akt, and p38. The elevated colocalization of BMP receptor type IA and FGFR2 in the presence of BMP2 or GDF5 suggests a signaling complex containing both receptors, akin to other known co-receptors of BMP ligands such as repulsive guidance molecules. This unexpected direct interaction between FGF receptor and BMP ligands potentially opens a new category of BMP signal transduction regulation, as FGFR2 is the second receptor tyrosine kinase to be identified as BMP co-receptor, and more may follow. The integration of cell surface interactions between members of the FGF and BMP family especially may widen the knowledge of such cellular communication mechanisms which involve both growth factor families, including morphogen gradients and osteogenesis, and may in consequence help to improve treatment options in osteochodnral diseases.}, subject = {Molekularbiologie}, language = {en} } @phdthesis{Reuter2023, author = {Reuter, Christian Steffen}, title = {Development of a tissue-engineered primary human skin infection model to study the pathogenesis of tsetse fly-transmitted African trypanosomes in mammalian skin}, doi = {10.25972/OPUS-25114}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-251147}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2023}, abstract = {Many arthropods such as mosquitoes, ticks, bugs, and flies are vectors for the transmission of pathogenic parasites, bacteria, and viruses. Among these, the unicellular parasite Trypanosoma brucei (T. brucei) causes human and animal African trypanosomiases and is transmitted to the vertebrate host by the tsetse fly. In the fly, the parasite goes through a complex developmental cycle in the alimentary tract and salivary glands ending with the cellular differentiation into the metacyclic life cycle stage. An infection in the mammalian host begins when the fly takes a bloodmeal, thereby depositing the metacyclic form into the dermal skin layer. Within the dermis, the cell cycle-arrested metacyclic forms are activated, re-enter the cell cycle, and differentiate into proliferative trypanosomes, prior to dissemination throughout the host. Although T. brucei has been studied for decades, very little is known about the early events in the skin prior to systemic dissemination. The precise timing and the mechanisms controlling differentiation of the parasite in the skin continue to be elusive, as does the characterization of the proliferative skin-residing trypanosomes. Understanding the first steps of an infection is crucial for developing novel strategies to prevent disease establishment and its progression. A major shortcoming in the study of human African trypanosomiasis is the lack of suitable infection models that authentically mimic disease progression. In addition, the production of infectious metacyclic parasites requires tsetse flies, which are challenging to keep. Thus, although animal models - typically murine - have produced many insights into the pathogenicity of trypanosomes in the mammalian host, they were usually infected by needle injection into the peritoneal cavity or tail vein, bypassing the skin as the first entry point. Furthermore, animal models are not always predictive for the infection outcome in human patients. In addition, the relatively small number of metacyclic parasites deposited by the tsetse flies makes them difficult to trace, isolate, and study in animal hosts. The focus of this thesis was to develop and validate a reconstructed human skin equivalent as an infection model to study the development of naturally-transmitted metacyclic parasites of T. brucei in mammalian skin. The first part of this work describes the development and characterization of a primary human skin equivalent with improved mechanical properties. To achieve this, a computer-assisted compression system was designed and established. This system allowed the improvement of the mechanical stability of twelve collagen-based dermal equivalents in parallel through plastic compression, as evaluated by rheology. The improved dermal equivalents provided the basis for the generation of the skin equivalents and reduced their contraction and weight loss during tissue formation, achieving a high degree of standardization and reproducibility. The skin equivalents were characterized using immunohistochemical and histological techniques and recapitulated key anatomical, cellular, and functional aspects of native human skin. Furthermore, their cellular heterogeneity was examined using single-cell RNA sequencing - an approach which led to the identification of a remarkable repertoire of extracellular matrix-associated genes expressed by different cell subpopulations in the artificial skin. In addition, experimental conditions were established to allow tsetse flies to naturally infect the skin equivalents with trypanosomes. In the second part of the project, the development of the trypanosomes in the artificial skin was investigated in detail. This included the establishment of methods to successfully isolate skin-dwelling trypanosomes to determine their protein synthesis rate, cell cycle and metabolic status, morphology, and transcriptome. Microscopy techniques to study trypanosome motility and migration in the skin were also optimized. Upon deposition in the artificial skin by feeding tsetse, the metacyclic parasites were rapidly activated and established a proliferative population within one day. This process was accompanied by: (I) reactivation of protein synthesis; (II) re-entry into the cell cycle; (III) change in morphology; (IV) increased motility. Furthermore, these observations were linked to potentially underlying developmental mechanisms by applying single-cell parasite RNA sequencing at five different timepoints post-infection. After the initial proliferative phase, the tsetse-transmitted trypanosomes appeared to enter a reversible quiescence program in the skin. These quiescent skin-residing trypanosomes were characterized by very slow replication, a strongly reduced metabolism, and a transcriptome markedly different from that of the deposited metacyclic forms and the early proliferative trypanosomes. By mimicking the migration from the skin to the bloodstream, the quiescent phenotype could be reversed and the parasites returned to an active proliferating state. Given that previous work has identified the skin as an anatomical reservoir for T. brucei during disease, it is reasonable to assume that the quiescence program is an authentic facet of the parasite's behavior in an infected host. In summary, this work demonstrates that primary human skin equivalents offer a new and promising way to study vector-borne parasites under close-to-natural conditions as an alternative to animal experimentation. By choosing the natural transmission route - the bite of an infected tsetse fly - the early events of trypanosome infection have been detailed with unprecedented resolution. In addition, the evidence here for a quiescent, skin-residing trypanosome population may explain the persistence of T. brucei in the skin of aparasitemic and asymptomatic individuals. This could play an important role in maintaining an infection over long time periods.}, subject = {Trypanosoma brucei}, language = {en} } @phdthesis{Kruse2024, author = {Kruse, Daniel}, title = {Quantitative Analyse histologischer Aufnahmen der Haut}, doi = {10.25972/OPUS-35294}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-352946}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2024}, abstract = {Diese Arbeit hatte zum Ziel quantitative Analysen histologischer Aufnahmen der Haut nach unterschiedlichen Gesichtspunkten zu etablieren. Im ersten Abschnitt wurde die bildgest{\"u}tzte Quantifizierung der epidermalen Histomorphologie untersucht. Nach Sichtung und Beurteilung von 2145 hochaufl{\"o}senden Fotografien HE-gef{\"a}rbter Epidermis- und Vollhautmodellen jeglichen Zustands, wurde der BSGC-Score als Facettenklassifikation mit seinen insgesamt 40 Beurteilungskriterien aufgestellt. Die unterschiedlichen epidermalen Strata wurden mit Wichtungsfaktoren belegt. Die Bewertungskategorien sind mit einem Ampelsystem unterlegt. Eine Befundungsformel wurde aufgestellt. Weitere Bestandteile des BSGC-Scores sind eine Anleitung mit Bildbeilage sowie Dokumentationselemente. Die Anwendung erfolgte erfolgreich im Rahmen der Qualit{\"a}tssicherung an Chargentests und zur Verlaufsbeurteilung eines In-vitro-Verbrennungsmodells aus humaner Epidermis durch Schneider et al. (2021) Der BSGC-Score dient als z{\"u}gig durchf{\"u}hrbares Evaluationstool zur Befundung von In-vitro-Epidermismodellen und nicht als diagnostisches Mittel. Der zweite Abschnitt besch{\"a}ftigt sich mit der Vaskularisierung als Parameter der kutanen Wundheilung. Es wurden aSMA-IF-gef{\"a}rbte Abbildungen porciner Verwundungsmodelle betrachtet und nach der Entfernung dr{\"u}siger Strukturen Gef{\"a}ßanschnitte zu Beginn manuell ausgez{\"a}hlt. Hieraus wurden die n{\"o}tigen Einstellungen f{\"u}r die Bildbearbeitungssoftware ImageJ ermittelt und die Abbildungen dieser anschließend zugef{\"u}hrt. Es erfolgte die automatisierte Quantifizierung elliptischer Formationen mit einer Gr{\"o}ße ≥ 30 Pixel. Im n{\"a}chsten Schritt wurden die Abbildungen in die Bereiche Wundrand, Wundgrund und Wundheilung unterteilt. In dem Bereich Wundheilung zeigte sich eine signifikant gr{\"o}ßere Revaskularisierung als in Wundgrund. Abschließend erfolgte der Vergleich sekund{\"a}rer Wundauflagen. Der Vergleich der Quotienten Wundheilung/Wundgrund nicht-okklusiver und okklusiver Wundauflagen zeigte keinen signifikanten Unterschied in der Neovaskularisierung. Die isolierte Betrachtung der Revaskularisierung als einzelner Prozess der Wundheilung kann nicht als generelles Kriterium f{\"u}r die Gesamtbeurteilung dienen. Hier findet die gew{\"a}hlte Methodik ihre Limitation. Zuk{\"u}nftige Anwendungsbereiche des BSGC-Scores sind die Ausweitung auf Vollhautmodelle und andere Verwundungsmodalit{\"a}ten. Eine automatisierte und durch eine KI-gest{\"u}tzte Befundung ist ebenfalls aufgrund des zugrundeliegenden umfangreichen Datensatzes denkbar. Auch kann eine automatisierte softwaregest{\"u}tzte Quantifizierung der Vaskularisierung als {\"u}berblickende und z{\"u}gige Beurteilung der Wundheilung sinnvoll erscheinen.}, subject = {Wundheilung}, language = {de} } @phdthesis{Kuehnemundt2024, author = {K{\"u}hnemundt, Johanna}, title = {Defined microphysiologic 3D tumour models with aspects from the tumour microenvironment for the evaluation of cellular immunotherapies}, doi = {10.25972/OPUS-27667}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-276674}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2024}, abstract = {Adoptive cellular immunotherapy with chimeric antigen receptor (CAR) T cells is highly effective in haematological malignancies. This success, however, has not been achieved in solid tumours so far. In contrast to hematologic malignancies, solid tumours include a hostile tumour microenvironment (TME), that poses additional challenges for curative effects and consistent therapeutic outcome. These challenges manifest in physical and immunological barriers that dampen efficacy of the CAR T cells. Preclinical testing of novel cellular immunotherapies is performed mainly in 2D cell culture and animal experiments. While 2D cell culture is an easy technique for efficacy analysis, animal studies reveal information about toxicity in vivo. However, 2D cell culture cannot fully reflect the complexity observed in vivo, because cells are cultured without anchorage to a matrix and only short-term periods are feasible. Animal studies provide a more complex tissue environment, but xenografts often lack human stroma and tumour inoculation occurs mostly ectopically. This emphasises the need for standardisable and scalable tumour models with incorporated TME-aspects, which enable preclinical testing with enhanced predictive value for the clinical outcome of immunotherapies. Therefore, microphysiologic 3D tumour models based on the biological SISmuc (Small Intestinal mucosa and Submucosa) matrix with preserved basement membrane were engaged and improved in this work to serve as a modular and versatile tumour model for efficacy testing of CAR T cells. In order to reflect a variety of cancer entities, TME-aspects, long-term stability and to enhance the read-out options they were further adapted to achieve scalable and standardisable defined microphysiologic 3D tumour models. In this work, novel culture modalities (semi-static, sandwich-culture) were characterised and established that led to an increased and organised tissue generation and long-term stability. Application of the SISmuc matrix was extended to sarcoma and melanoma models and serial bioluminescence intensity (BLI)-based in vivo imaging analysis was established in the microphysiologic 3D tumour models, which represents a time-efficient read-out method for quality evaluation of the models and treatment efficacy analysis, that is independent of the cell phenotype. Isolation of cancer-associated-fibroblasts (CAFs) from lung (tumour) tissue was demonstrated and CAF-implementation further led to stromal-enriched microphysiologic 3D tumour models with in vivo-comparable tissue-like architecture. Presence of CAFs was confirmed by CAF-associated markers (FAP, α-SMA, MMP-2/-9) and cytokines correlated with CAF phenotype, angiogenesis, invasion and immunomodulation. Additionally, an endothelial cell barrier was implemented for static and dynamic culture in a novel bioreactor set-up, which is of particular interest for the analysis of immune cell diapedesis. Studies in microphysiologic 3D Ewing's sarcoma models indicated that sarcoma cells could be sensitised for GD2-targeting CAR T cells. After enhancing the scale of assessment of the microphysiologic 3D tumour models and improving them for CAR T cell testing, the tumour models were used to analyse their sensitivity towards differently designed receptor tyrosine kinase-like orphan receptor 1 (ROR1) CAR T cells and to study the effects of the incorporated TME-aspects on the CAR T cell treatment respectively. ROR1 has been described as a suitable target for several malignancies including triple negative breast cancer (TNBC), as well as lung cancer. Therefore, microphysiologic 3D TNBC and lung cancer models were established. Analysis of ROR1 CAR T cells that differed in costimulation, spacer length and targeting domain, revealed, that the microphysiologic 3D tumour models are highly sensitive and can distinguish optimal from sub-optimal CAR design. Here, higher affinity of the targeting domain induced stronger anti-tumour efficacy and anti-tumour function depended on spacer length, respectively. Long-term treatment for 14 days with ROR1 CAR T cells was demonstrated in dynamic microphysiologic 3D lung tumour models, which did not result in complete tumour cell removal, whereas direct injection of CAR T cells into TNBC and lung tumour models represented an alternative route of application in addition to administration via the medium flow, as it induced strong anti-tumour response. Influence of the incorporated TME-aspects on ROR1 CAR T cell therapy represented by CAF-incorporation and/or TGF-β supplementation was analysed. Presence of TGF-β revealed that the specific TGF-β receptor inhibitor SD-208 improves ROR1 CAR T cell function, because it effectively abrogated immunosuppressive effects of TGF-β in TNBC models. Implementation of CAFs should provide a physical and immunological barrier towards ROR1 CAR T cells, which, however, was not confirmed, as ROR1 CAR T cell function was retained in the presence of CAFs in stromal-enriched microphysiologic 3D lung tumour models. The absence of an effect of CAF enrichment on CAR T cell efficacy suggests a missing component for the development of an immunosuppressive TME, even though immunomodulatory cytokines were detected in co-culture models. Finally, improved gene-edited ROR1 CAR T cells lacking exhaustion-associated genes (PD-1, TGF-β-receptor or both) were challenged by the combination of CAF-enrichment and TGF-β in microphysiologic 3D TNBC models. Results indicated that the absence of PD-1 and TGF-β receptor leads to improved CAR T cells, that induce strong tumour cell lysis, and are protected against the hostile TME. Collectively, the microphysiologic 3D tumour models presented in this work reflect aspects of the hostile TME of solid tumours, engage BLI-based analysis and provide long-term tissue homeostasis. Therefore, they present a defined, scalable, reproducible, standardisable and exportable model for translational research with enhanced predictive value for efficacy testing and candidate selection of cellular immunotherapy, as exemplified by ROR1 CAR T cells.}, subject = {Immuntherapie}, language = {en} } @phdthesis{Jihyoung2024, author = {Jihyoung, Choi}, title = {Development of an Add-On Electrode for Non-Invasive Monitoring in Bioreactor Cultures and Medical Devices}, doi = {10.25972/OPUS-35823}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-358232}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2024}, abstract = {Electrochemical impedance spectroscopy (EIS) is a valuable technique analyzing electrochemical behavior of biological systems such as electrical characterization of cells and biomolecules, drug screening, and biomaterials in biomedical field. In EIS, an alternating current (AC) power signal is applied to the biological system, and the impedance of the system is measured over a range of frequencies. In vitro culture models of endothelial or epithelial barrier tissue can be achieved by culturing barrier tissue on scaffolds made with synthetic or biological materials that provide separate compartments (apical and basal sides), allowing for further studies on drug transport. EIS is a great candidate for non-invasive and real-time monitoring of the electrical properties that correlate with barrier integrity during the tissue modeling. Although commercially available transendothelial/transepithelial electrical resistance (TEER) measurement devices are widely used, their use is particularly common in static transwell culture. EIS is considered more suitable than TEER measurement devices in bioreactor cultures that involve dynamic fluid flow to obtain accurate and reliable measurements. Furthermore, while TEER measurement devices can only assess resistance at a single frequency, EIS measurements can capture both resistance and capacitance properties of cells, providing additional information about the cellular barrier's characteristics across various frequencies. Incorporating EIS into a bioreactor system requires the careful optimization of electrode integration within the bioreactor setup and measurement parameters to ensure accurate EIS measurements. Since bioreactors vary in size and design depending on the purpose of the study, most studies have reported using an electrode system specifically designed for a particular bioreactor. The aim of this work was to produce multi-applicable electrodes and established methods for automated non-invasive and real-time monitoring using the EIS technique in bioreactor cultures. Key to the electrode material, titanium nitride (TiN) coating was fabricated on different substrates (materials and shape) using physical vapor deposition (PVD) and housed in a polydimethylsiloxane (PDMS) structure to allow the electrodes to function as independent units. Various electrode designs were evaluated for double-layer capacitance and morphology using EIS and scanning electron microscopy (SEM), respectively. The TiN-coated tube electrode was identified as the optimal choice. Furthermore, EIS measurements were performed to examine the impact of influential parameters related to culture conditions on the TiN-coated electrode system. In order to demonstrate the versatility of the electrodes, these electrodes were then integrated into in different types of perfusion bioreactors for monitoring barrier cells. Blood-brain barrier (BBB) cells were cultured in the newly developed dynamic flow bioreactor, while human umblical vascular endothelial cells (HUVECs) and Caco-2 cells were cultured in the miniature hollow fiber bioreactor (HFBR). As a result, the TiN-coated tube electrode system enabled investigation of BBB barrier integrity in long-term bioreactor culture. While EIS measurement could not detect HUVECs electrical properties in miniature HFBR culture, there was the possibility of measuring the barrier integrity of Caco-2 cells, indicating potential usefulness for evaluating their barrier function. Following the bioreactor cultures, the application of the TiN-coated tube electrode was expanded to hemofiltration, based on the hypothesis that the EIS system may be used to monitor clotting or clogging phenomena in hemofiltration. The findings suggest that the EIS monitoring system can track changes in ion concentration of blood before and after hemofiltration in real-time, which may serve as an indicator of clogging of filter membranes. Overall, our research demonstrates the potential of TiN-coated tube electrodes for sensitive and versatile non-invasive monitoring in bioreactor cultures and medical devices.}, subject = {Monitoring}, language = {en} }