• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 61
  • 60
  • 17
  • 6
  • 4
  • 3
  • 3
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • Tagged with
  • 179
  • 36
  • 25
  • 23
  • 20
  • 19
  • 18
  • 17
  • 17
  • 16
  • 16
  • 15
  • 15
  • 15
  • 14
  • 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.
171

The Sweet Side of the Extracellular Matrix -: Glycosaminoglycans in Matrix Remodeling, Endothelial Cell Activation and Functional Biomaterials

Rother, Sandra 19 October 2017 (has links)
Bone fractures and pathologic conditions like chronic wounds significantly reduce the quality of life for the patients, which is especially dramatic in an elderly population with considerable multi-morbidity and lead to substantial socio-economic costs. To improve the wound healing capacity of these patients, new strategies for the design of novel multi-functional biomaterials are required: they should be able to decrease extensive pathologic tissue degradation and specifically control angiogenesis in damaged vascularized tissues like bone and skin. Glycosaminoglycans (GAGs) like hyaluronan (HA) and chondroitin sulfate (CS) as important extracellular matrix (ECM) components are involved in several biological processes such as matrix remodeling and growth factor signaling, either by directly influencing the cellular response or by interacting with mediator proteins. This could be useful in functionalizing biomaterials, but native sulfated GAGs (sGAGs) show a high batch-to-batch variability and are limited in their availability. Chemically modified HA and CS derivatives with much more defined characteristics regarding their carbohydrate backbone, sulfate group distribution and sulfation degree are favorable to study the structure-function relationship of GAGs in their interaction with mediator proteins and/or cells and this might be used to precisely modulate activity profiles to stimulate wound healing. By combining collagen type I as the main structural protein of the bone and skin ECM with these GAG derivatives, 2.5-dimensional (2.5D) and 3D artificial ECM (aECM) coatings and hydrogels were developed. These biomaterials as well as the respective GAG derivatives alone were compared to native GAGs and used to analyze how the sulfation degree, pattern and carbohydrate backbone of GAGs influence: i) the activity of tissue inhibitor of metalloproteinase-3 (TIMP-3) and vascular endothelial growth factor-A (VEGF-A) as main regulators of ECM remodeling and angiogenesis, ii) the composition and characteristics of the developed 2.5D and 3D aECMs, iii) the enzymatic degradation of collagen-based aECMs and HA/collagen-based hydrogels, iv) the proliferation and functional morphology of endothelial cells. Surface plasmon resonance (SPR) and enzyme linked immunosorbent assay (ELISA) binding studies revealed that sulfated HA (sHA) derivatives interact with TIMP-3 and VEGF-A in a sulfation-dependent manner. sHA showed an enhanced interplay with these proteins compared to native GAGs like heparin (HEP) or CS, suggesting a further impact of the carbohydrate backbone and sulfation pattern. sGAGs alone were weak modulators of the matrix metalloproteinase-1 and -2 (MMP-1 and -2) activity and did not interfere with the inhibitory potential of TIMP-3 against these proteinases during enzyme kinetic analyses. However, the formation of TIMP 3/GAG complexes reduced the binding of TIMP-3 to cluster II and IV of its endocytic receptor low-density lipoprotein receptor-related protein-1 (LRP-1, mediates the up-take and degradation of TIMP-3 from the extracellular environment) in a sulfation- and GAG type-dependent manner. It is of note that the determined complex stabilities of TIMP-3 with cluster II and IV were almost identical indicating for the first time that both clusters contribute to the TIMP-3 binding. Competitive SPR experiments demonstrated that GAG polysaccharides interfere stronger with the TIMP 3/LRP-1 interplay than GAG oligosaccharides. The importance of the position of sulfation is highlighted by the finding that a sHA tetrasaccharide exclusively sulfated at the C6 position of the N-acetylglucosamine residues significantly blocked the receptor binding, while CS and HEP hexasaccharides had no detectable effects. Thus, sHA derivatives as part of biomaterials could be used to sequester and accumulate TIMP 3 in aECMs in a defined manner where sHA-bound TIMP-3 could decrease the matrix breakdown by potentially restoring the MMP/TIMP balance. GAG binding might extend the beneficial presence of TIMP-3 into wounds characterized by excessive pathologic tissue degradation (e.g. chronic wounds, osteoarthritis). Mediator protein interaction studies with sHA coated surfaces showed the simultaneous binding of TIMP-3 and VEGF-A, even though the sHA/VEGF-A interplay was preferred. Moreover, kinetic analysis revealed almost comparable affinities of both proteins for VEGF receptor-2 (VEGFR-2), explaining their competition that mainly regulates the activation of endothelial cells. Additional SPR measurements demonstrated that the binding of sGAGs to TIMP-3 or VEGF-A decreases the binding of the respective mediator protein to VEGFR-2. Likewise, a sulfation-dependent reduction of the binding signal was observed after pre-incubation of a mixture of TIMP-3 and VEGF-A with sGAG poly- and oligosaccharides. The biological consequences of GAGs interfering with VEGF-A/VEGFR-2 and TIMP-3/VEGFR 2 were assessed in vitro using porcine aortic endothelial cells stably transfected with VEGFR 2 (PAE/KDR cells). The presence of sHA both decreased VEGF-A activity and the activity of TIMP-3 to inhibit the VEGF-A-induced VEGFR-2 phosphorylation. The same decreased activities could be observed for the migration of endothelial cells. However, if sHA, TIMP-3 and VEGF-A were present simultaneously, sHA partially restored the TIMP-3-mediated blocking of VEGF-A activity. These findings provide novel insights into the regulatory potential of sHA during endothelial cell activation as an important aspect of angiogenesis, which could be translated into the design of biomaterials to treat abnormal angiogenesis. These sHA-containing materials might control the angiogenic response by modulating the activity of TIMP 3 and VEGF-A. The in vitro fibrillogenesis of collagen type I in the presence of sHA derivatives led to 2.5D collagen-based aECM coatings with stable collagen contents and GAG contents that resemble the organic part of the bone ECM. A burst release of GAGs was observed during the first hour of incubation in buffer with the GAG content remaining almost constant afterwards, implying that the number of GAG-binding sites of collagen restricts the amounts of associated GAGs. Moreover, two differently sulfated HA derivatives could for the first time be incorporated into one multi-GAG aECM as verified via agarose gel electrophoresis and fluorescence measurements. This illustrates the multiple options to modify the aECM composition and thereby potentially their functionality. Atomic force microscopy showed that the presence of sHA derivatives during fibrillogenesis significantly reduced the resulting fibril diameter in a concentration- and sulfation-dependent manner, indicating an interference of the GAGs with the self-assembly of collagen monomers. In line with enzyme kinetic results, none of the GAGs as part of aECMs altered the enzymatic collagen degradation via a bacterial collagenase. Thus aECMs were proven to be biodegradable independent from their composition, which is favorable concerning a potential biomedical usage of the aECMs e.g. as implant coatings. HA/collagen-based hydrogels containing fibrillar collagen embedded into a network of crosslinked HA and sGAGs were developed as 3D aECMs. Scanning electron microscopy demonstrated a porous structure of the gels after lyophilization, which could favor the cultivation of cells. The presence of collagen markedly enhanced the stability of the gels against the enzymatic degradation via hyaluronidase, something beneficial to clinical use as this is often limited by the generally fast breakdown of HA. Binding and release experiments with lysozyme, as positively charged model protein for e.g. pro-inflammatory cytokines, and VEGF A revealed that the sulfation of GAGs increased the protein binding capacity for pure GAG coatings and retarded the protein release from hydrogels compared to hydrogels without sGAGs. Moreover, the additional acrylation of sHA was shown to strongly reduce the interaction with both proteins when the primary hydroxyl groups were targets of acrylation. This stresses the influence of the substitution pattern on the protein binding properties of the GAG derivatives. However, hydrogel characteristics like the elastic modulus remained unaffected. The different interaction profiles of lysozyme and VEGF-A with GAGs demonstrated a protein-specific preference of different monosaccharide compositions, suggesting that the mediator protein binding could be simultaneously adjusted for several proteins by combining different GAG derivatives. This might allow the scavenging of pro-inflammatory cytokines and at the same time a binding and release of wound healing stimulating growth factors. Since there is a growing demand for biomaterials to regenerate injured vascularized tissues like bone and skin, endothelial cells were used to examine the direct effects of solute GAGs and hydrogels containing these GAGs in vitro. In both cases, sHA strongly enhanced the proliferation of PAE/KDR cells. A VEGFR-2-mediated effect of GAGs on endothelial cells as underlying mechanism is unlikely since GAGs alone did not bind to VEGFR-2 and had no influence on VEGFR-2 phosphorylation. Other factors like GAG-induced alterations of cell-matrix interactions and cell signaling could be responsible. In accordance with SPR results, a decreased endothelial cell proliferation stimulating activity of VEGF-A was observed in the presence of solute GAGs or after binding to hydrogels compared to the respective treatment without VEGF-A. However, tube formation could be observed in the presence of solute VEGF A and GAGs and within hydrogels with sGAGs that released sufficient VEGF-A amounts over time. Overall the presence of GAGs and VEGF-A strongly promoted the endothelial cell proliferation compared to the treatment with GAGs or VEGF-A alone. Thus, HA/collagen-based hydrogels functionalized with sHA derivatives offer a promising option for the design of “intelligent” biomaterials that direct and regulate the cellular behavior instead of simply acting as inert filling material. They could be used for the controlled delivery and/or scavenging of multiple mediator proteins, thus enhancing the local availability or reducing the activity of these GAG-interacting mediator proteins, or by directly influencing the cellular response. This might be applied to a range of pathological conditions by tuning the biomaterial compositions to patient-specific needs. However, extensive in vivo validation is required to show whether these in vitro findings could be used to control the biological activity of for instance TIMP-3 and VEGF-A, especially under the pathological conditions of extended matrix degradation and dysregulated angiogenesis.
172

Polyhydroxybutyrate als Scaffoldmaterial für das Tissue Engineering von Knochen

Wollenweber, Marcus 10 May 2012 (has links)
In drei inhaltlich abgeschlossen Teilen werden Fragestellungen bearbeitet, die sich mit dem Einsatz von Polyhydroxybutyraten als Scaffoldmaterialien für das Tissue Engioneering von Knochen beschäftigen. Zunächst wird ein Prozess optimiert, in dem mittels Verpressen und Auslösen von Platzhaltern (Porogen) poröse Träger (Scaffolds) aus Poly-3-hydroxybuttersäure (P3HB) sowie aus P3co4HB hergestellt werden. Diese Scaffolds werden in der Folge mechanisch und strukturell charakterisiert, wobei Druckfestigkeit, Dauerfestigkeit und Viskoelastizität untersucht werden. Im Ergebnis finden sich mehrere Kandidaten, die für die weitere Testung im Tierversuch in Frage kommen. Weiter wird das Abbauverhalten von schmelzgeponnenen P3HB-Fäden untersucht. Dabei wird ein beschleunigtes Modellsystem gewählt, das noch möglichst nahe am physiologischen Fall aber ohne biologisch aktive Komponente (zB. Enzyme) definiert wurde. Die Charakterisierung bedient sich hier der Gelpermeationschromatographie (GPC), des gasgestützten Elektronenrastermikroskops (ESEM), der differentiellen Thermoanalyse (DSC) und der Rasterkraftmikroskopie. Als Ergebnis zeichnete sich ab, dass neben der hydrolytischen Degradation im Gegensatz zu PHB mit kleinerer spezifischer Oberfläche bei den Fäden auch Erosion zum Abbau beiträgt. Eine partikuläre Freisetzung wird nicht beobachtet. Im dritten Teil werden textile Scaffolds aus P3HB mit einer künstlichen extrazellulären Matrix aus Chondroitinsulfaten (CS) und Kollagen versehen. Dem CS kann hier ein positiver Einfluss auf die osteogene Differenzierung von humanen mesenchymalen Stammzellen (hMSC) nachgewiesen werden. Dies wird zum einen durch die verstärkte Expression der alkalischen Phosphatase (ALP) sowie durch die Hochregulation von Proteinen ersichtlich, die bei der osteogenen Differenzierung essentiell sind. In wenigen Gene-Arrays lässt sich ebenfalls erkennen, dass die osteogene Differenzierung durch CS positiv beeinflusst wird. Insbesondere frühe Marker wie ZBTB16 und IGFBPs werden hier identifiziert. Basierend auf den Teilergebnissen wird am Ende ein Beitrag geliefert, der das Tissue Engineering insbesondere für überkritische Röhrenknochendefekte als Methode interessant erscheinen lässt. Dabei werden mechanische Lasten durch konventionelle Fixateure aufgenommen und der Defektraum durch den mehrfachen Einsatz von bio-funktionalisierten flachen Scaffolds gefüllt.:1. Vorwort 3 2. Allgemeine Einführung 5 2.1 Der Knochen 5 2.1.1 Die Knochenbildung 5 2.1.2 Zur Anatomie und Physiologie des Knochens 7 2.2 Tissue Engineering 11 2.2.1 Zelltypen für das Tissue Engineering von Knochen 12 2.2.2 Scaffold Design im Tissue Engineering von Knochen 13 2.3 Polyhydroxyalkanoate 13 2.4 Tissue Engineering am Röhrenknochen 16 2.4.1 Poly(3-hydroxybutyrat)-Scaffolds für das Tissue Engineering von Knochenersatz 17 2.4.2 Matrix Engineering 18 2.5 Ziel der Arbeit 19 3. Mechanik poröser PHB-Scaffolds 21 3.1 Einleitung 21 3.2 Materialien und Methoden 23 3.2.1 Polyhydroxybutyrate und Porogene 23 3.2.2 Uniaxiales Heißpressen 24 3.2.3 Mikrographie 26 3.2.4 Dynamische Differenzkalorimetrie (DSC) 26 3.2.5 Mechanische Druckversuche 26 3.2.6 Mikrocomputertomographie (μCT) 27 3.2.7 Zellviabilität auf den Scaffolds 28 3.3 Ergebnisse 29 3.3.1 Mikrographie 29 3.3.2 Mikrocomputertomographie (μCT) 33 3.3.3 Druckversuche 37 3.3.4 Dynamische Differenzkalorimetrie (DSC) 40 3.3.5 Zellviabilität 40 3.4 Diskussion 40 3.5 Schlussfolgernde Zusammenfassung 46 4. Degradation von P3HB-Fasern 47 4.1 Degradation von Polyhydroxyalkanoaten 47 4.2 Materialien und Methoden 49 4.2.1 Herstellung und Vorbehandlung textiler P3HB-Konstrukte 49 4.2.2 Mechanische Prüfung 50 4.2.3 Beschleunigte Degradation 50 4.2.4 Untersuchung der Oberfläche 50 4.2.5 Dynamische Differenzkalorimetrie (DSC) 51 4.2.6 Gel-Permeations-Chromatographie (GPC) 51 4.3 Ergebnisse 52 4.3.1 Mechanische Tests 52 4.3.2 Die Charakterisierung der Oberfläche 52 4.3.3 Thermische Fasereigenschaften.55 4.3.4 Untersuchung der Molekulargewichte in der GPC 58 4.4 Diskussion 60 4.5 Schlussfolgernde Zusammenfassung 64 5. hMSC auf textilen Scaffolds 67 5.1 Einleitung 67 5.2 Material und Methoden 68 5.2.1 Erzeugung der P3HB-Scaffolds 68 5.2.2 Die Immobilisierung der EZM-Komponenten auf den Scaffolds 69 5.2.3 Isolation, Vorkultur, Besiedlung und Kultur der humanen mesenchymalen Vorläuferzellen 69 5.2.4 Kombinierte Bestimmung von ALP, MTT und Proteingehalt 71 5.2.5 Mikroskopische Untersuchungen 72 5.2.6 Nachweis der Kalziummineralisierung 73 5.2.7 Quantitative real time reverse transcribing polymerase chain reaction (rt-PCR) 73 5.2.8 cRNA Microarray-Untersuchung 74 5.2.9 Zusätzliche Experimente 75 5.3 Ergebnisse 76 5.3.1 Vorhergehende Untersuchung 76 5.3.2 Rasterelektronen-Mikroskopie 77 5.3.3 Konfokale Laser-Scanning-Mikroskopie 79 5.3.4 ALP-Aktivität, SDH-Aktivität und Proteingehalt 82 5.3.5 Mineralisierende Kalziumabscheidung 86 5.3.6 rt-PCR 87 5.3.7 cRNA Microarray-Untersuchung 90 5.3.8 Kulturen von hMSC mit Chondroitinsulfat als gelöstem Zusatz 93 5.4 Diskussion 93 5.5 Schlussfolgernde Zusammenfassung 98 6. Zusammenfassung 101
173

Screening a chemically defined extracellular matrix mimetic substrate library to identify substrates that enhance substratemediated transfection

Hamann, Andrew, Thomas, Alvin K., Kozisek, Tyler, Farris, Eric, Lück, Steffen, Zhang, Yixin, Pannier, Angela K. 19 May 2022 (has links)
Nonviral gene delivery, though limited by inefficiency, has extensive utility in cell therapy, tissue engineering, and diagnostics. Substrate-mediated gene delivery (SMD) increases efficiency and allows transfection at a cell-biomaterial interface, by immobilizing and concentrating nucleic acid complexes on a surface. Efficient SMD generally requires substrates to be coated with serum or other protein coatings to mediate nucleic acid complex immobilization, as well as cell adhesion and growth; however, this strategy limits reproducibility and may be difficult to translate for clinical applications. As an alternative, we screened a chemically defined combinatorial library of 20 different extracellular matrix mimetic substrates containing combinations of (1) different sulfated polysaccharides that are essential extracellular matrix glycosaminoglycans (GAGs), with (2) mimetic peptides derived from adhesion proteins, growth factors, and cell-penetrating domains, for use as SMD coatings. We identified optimal substrates for DNA lipoplex and polyplex SMD transfection of fibroblasts and human mesenchymal stem cells. Optimal extracellular matrix mimetic substrates varied between cell type, donor source, and transfection reagent, but typically contained Heparin GAG and an adhesion peptide. Multiple substrates significantly increased transgene expression (i.e. 2- to 20-fold) over standard protein coatings. Considering previous research of similar ligands, we hypothesize extracellular matrix mimetic substrates modulate cell adhesion, proliferation, and survival, as well as plasmid internalization and trafficking. Our results demonstrate the utility of screening combinatorial extracellular matrix mimetic substrates for optimal SMD transfection towards application- and patient-specific technologies.
174

A Comparative Analysis of the Biomechanics and Biochemistry of Cell-Derived and Cell-Remodeled Matrices: Implications for Wound Healing and Regenerative Medicine

Ahlfors, Jan-Eric Wilhelm 03 May 2004 (has links)
The purpose of this research was to study the synthesis and remodeling of extracellular matrix (ECM) by fibroblasts with special emphasis on the culture environment (media composition and initial ECM composition) and the resulting mechanical integrity of the ECM. This was investigated by culturing fibroblasts for 3 weeks in a variety of culture conditions consisting of collagen gels, fibrin gels, or media permissive to the self-production of ECM (Cell-Derived Matrix), and quantifying the mechanics of the resulting ECM. The mechanical characteristics were related to the biochemistry of the resulting ECM, notably in terms of collagen accumulation and collagen fibril diameters. The ultimate tensile strength (UTS) of the collagen gels and fibrin gels at the end of the 3-week period was 168.5 ± 43.1 kPa and 133.2 ± 10.6 kPa, respectively. The ultimate tensile strength of the cell-derived matrices was 223.2 ± 9 kPa, and up to 697.1 ± 36.1 kPa when cultured in a chemically-defined medium that was developed for the rapid growth of matrix in a more defined environment. Normalizing the strength to collagen density resulted in a UTS / Collagen Density in these groups of 6.4 ± 1.9 kPa/mg/cm3, 25.9 ± 2.4 kPa/mg/cm3, 14.5 ± 1.1 kPa/mg/cm3, and 40.0 ± 1.9 kPa/mg/cm3, respectively. Cells were synthetically more active when they produced their own matrix than when they were placed within gels. The resulting matrix was also significantly stronger when it was self-produced than when the cells rearranged the matrix within gels that corresponded to a significantly larger fraction of non-acid and pepsin extractable collagen. These studies indicate that cell-derived matrices have potential both as in vitro wound healing models and as soft connective tissue substitutes.
175

Inhibition of Cancer Stem Cells by Glycosaminoglycan Mimetics

O'Hara, Connor P 01 January 2019 (has links)
Connor O’Hara July 29, 2019 Inhibition of Cancer Stem Cells by Glycosaminoglycan Mimetics In the United States cancer is the second leading cause of death, with colorectal cancer (CRC) being the third deadliest cancer and expected to cause over 51,000 fatalities in 2019 alone.1 The current standard of care for CRC depends largely on the staging, location, and presence of metastasis.2 As the tumor grows and invades nearby lymph tissue and blood vessels, CRC has the opportunity to invade not only nearby tissue but also metastasize into the liver and lung (most commonly).3 The 5-year survival rate for metastasized CRC is <15%, and standard of care chemotherapy regimens utilizing combination treatments only marginally improve survival.3-5 Additionally, patients who have gone into remission from late-stage CRC have a high risk of recurrence despite advances in treatment.6-7 The Cancer Stem-like Cell (CSC) paradigm has grown over the last 20 years to become a unifying hypothesis to support the growth and relapse of tumors previously regressed from chemotherapy (Figure 1).8 The paradigm emphasizes the heterogeneity of a tumor and its microenvironment, proposing that a small subset of cells in the tumor are the source of tumorigenesis with features akin to normal stem cells.9 The CSCs normally in a quiescent state survive this chemotherapy and “seed” tumor redevelopment.10 First observed in acute myeloid lymphoma models, CSCs have since been identified in various other cancers (to include CRC) by their cell surface antigens and unique properties characterizing them from normal cancer cells.11-12 These include tumor initiation, limitless self-renewal capacity to generate clonal daughter cells, as well as phenotypically diverse, mature, and highly differentiated progeny.13-14 Previously our lab has identified a novel molecule called G2.2 (Figure 2) from a unique library of sulfated compounds showing selective and potent inhibition of colorectal CSCs in-vitro.15 G2.2 is a mimetic of glycosaminoglycans (GAGs) and belongs to a class of molecules called non-saccharide GAG mimetics (NSGMs). Using a novel dual-screening platform, comparisons were made on the potency of G2.2 in bulk monolayer cells, primary 3D tumor spheroids of the same cell line, and subsequent generations of tumor spheroids. This work has shown in-vitro the fold-enhancement of CSCs when culturing as 3D tumor spheroids. Spheroid culture serves as a more accurate model for the physiological conditions of a tumor, as well as the functional importance of upregulating CSCs. Evaluation of G2.2 and other NSGMs was performed in only a few cell lines, developing a need to better understand the ability of G2.2 to inhibit spheroids from a more diverse panel of cancer cells to better understand G2.2’s mechanism. The last few decades have seen the advancement in fundamental biological and biochemical knowledge of tumor cell biology and genetics.16 CRC, in particular, has served as a useful preclinical model in recapitulating patient tumor heterogeneity in-vitro.17 Recent work has characterized the molecular phenotypes of CRC cell lines in a multi-omics analysis, stratifying them into 4 clinically robust and relevant consensus molecular subtypes (CMS).18-19 Our work was directed to screen a panel of cells from each of the molecular subtypes and characterize the action of G2.2 and 2nd generation lipid-modified analogs, synthesized to improve the pharmacokinetic properties of the parent compound. Four NSGMs, namely G2.2, G2C, G5C, and G8C (Figure 2) were studied for their ability to inhibit the growth of primary spheroids across a phenotypically diverse panel. Compound HT-29 IC50 (μM) Panel Average IC50 (μM) G2.2 28 ± 1 185 ± 55 G2C 5 ± 2 16 ± 15 G5C 8 ± 2 63 ± 19 G8C 0.7 ± 0.2 6 ± 3 Primary spheroid inhibition assays were performed comparing the potency of new NSGMs to G2.2. Fifteen cell lines were evaluated in a panel of colorectal adenocarcinoma cell lines with several cell lines representing each CMS. Primary spheroid inhibition assays revealed 3 distinct response with regard to G2.2’s ability to inhibit spheroid growth. Cells from CMS 3 and 4, which display poor clinical prognosis, metabolic dysregulation, and enhanced activation of CSC pathways, showed the most sensitivity to G2.2 (mean IC50 = 89 ± 55 μM). Mesenchymal CMS 4 cell lines were over 3-fold more sensitive to treatment with G2.2 when compared to CMS 1 cell lines. Resistant cell lines were composed entirely of CMS 1 and 2 (mean IC50 = 267 ± 105 μM). In contrast, all lipid-modified analogs showed greater potency than the parent NSGM in almost every CRC cell line. Of the three analogs, G8C showed the greatest potency with a mean IC50 of less than 15 μM. Of the CRC spheroids studied, HT-29 (CMS 3) was most sensitive to G8C (IC50 = 0.73 μM). To evaluate the selectivity of NSGMs for CSC spheroid inhibition, MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium) cytotoxicity assays were performed on monolayer cell culture, and the fold-selectivity of NSGM for spheroids was analyzed. Data shows that NSGMs preferentially target CSC-rich spheroids compared with monolayer cellular growth, with G2.2 having over 7-fold selectivity for spheroid conditions. This fold selectivity was enhanced in CMS 3/4, supporting the idea that G2.2 targets a mesenchymal and stem-like phenotype. To further validate this selectivity, limiting dilution assays were performed across the panel to determine the tumor-initiating capacity of each cell line. Cell lines which showed a sensitive response to G2.2 were over 2-fold more likely to develop into spheroids, validating the previous hypothesis. Further characterization was performed analyzing the changes G2.2 induced on CSC markers, as well as the basal expression of a unique pair of cancer cells. Western blots showed a reduction in self-renewal marker across all CMS after treatment with G2.2, and that cell lines sensitive to G2.2-treatment overexpress mesenchymal and stem-like markers. G2.2-resistant cell lines show an epithelial phenotype, lacking this expression. The positive results observed in these studies enhance the understanding of G2.2 and analogs, and further evaluation with additional cell lines of various tissues would improve the knowledge thus far gained. However, all experiments described take valuable time to perform and analyze. Thus, there became a need to develop a high-throughput screening (HTS) platform for our assays that standardized analysis and enhanced productivity. Initial development of the method for this assay are underway, and recent evidence from these evaluations of breast cancer spheroids suggests that G2.2 and analogs may be tissue-specific compounds for the treatment of cancer. Future work entails refining the application of this method for evaluation of the NCI-60 (National Cancer Institute) tumor cell panel. Overall, these results make several suggestions concerning the NSGMs evaluated against the panel. First, G2.2 selectively targets CSCs with limited toxicity to monolayer cells of the same cell line. Further, G2.2 has the greatest potency with CMS 3/4, whose mesenchymal phenotypes are associated with poor clinical prognosis and enrichment of CSCs. Supporting evidence include that sensitive cell lines are highly tumorigenic and show enhanced expression of mesenchymal/CSC markers compared to resistant cell lines. Lipid-modification of G2.2 enhances in-vitro potency against spheroid growth, with nM potency reached in the most sensitive cell lines. Evidence in the development of a HTS platform also suggests these NSGMs show tissue specificity to cancers of the intestine. Further work characterizing the mechanism of NSGMs in a broader multi-tissue panel will enhance our understanding of the compounds as a potential therapy to dramatically improve patient survival through specific targeting of tumorigenesis. References 1. Colorectal Cancer Facts & Figures 2017-2019. American Cancer Society 2017. 2. Compton, C. C.; Byrd, D. R.; Garcia-Aguilar, J.; Kurtzman, S. H.; Olawaiye, A.; Washington, M. K. Colon and rectum. In AJCC Cancer Staging Atlas, 2nd ed.; Ed. Springer Science: New York, 2012; pp 185–201. 3. Van Cutsem, E.; Cervantes, A.; Adam, R.; Sobrero, A.; Van Krieken, J. H.; Aderka, D.; Aranda Aguilar, E.; Bardelli, A.; Benson, A.; Bodoky, G.; et al. ESMO consensus guidelines for the management of patients with metastatic colorectal cancer. Ann. Oncol. 2016, 27, 1386–422. 4. Siegel, R. L.; Miller, K. D.; Fedewa, S. A.; Ahnen, D. J.; Meester, R. G. S.; Barzi, A.; Jemal, A. Colorectal cancer statistics, 2017. CA Cancer J. Clin. 2017, 67, 177–193. 5. Moriarity, A.; O'Sullivan, J.; Kennedy, J.; Mehigan, B.; McCormick, P. Current targeted therapies in the treatment of advanced colorectal cancer: a review. Ther. Adv. Med. Oncol. 2016, 8, 276–293. 6. Seidel, J.; Farber, E.; Baumbach, R.; Cordruwisch, W.; Bohmler, U.; Feyerabend, B.; Faiss, S. Complication and local recurrence rate after endoscopic resection of large high-risk colorectal adenomas of >/=3 cm in size. Int. J. Colorectal Dis. 2016, 31, 603–611. 7. Pugh, S. A.; Shinkins, B.; Fuller, A.; Mellor, J.; Mant, D.; Primrose, J. N. Site and stage of colorectal cancer influence the likelihood and distribution of disease recurrence and postrecurrence survival: data from the FACS randomized controlled trial. Ann. Surg. 2016, 263, 1143–1147. 8. Batlle, E.; Clevers, H. Cancer stem cells revisited. Nat. Med. 2017, 23, 1124–1134. 9. Hanahan, D.; Weinberg, R. A. Hallmarks of cancer: the next generation. Cell 2011, 144, 646–674. 10. Tirino, V.; Desiderio, V.; Paino, F.; De Rosa, A.; Papaccio, F.; La Noce, M.; Laino, L.; De Francesco, F.; Papaccio, G. Cancer stem cells in solid tumors: an overview and new approaches for their isolation and characterization. FASEB J. 2013, 27, 13–24. 11. Bonnet, D.; Dick, J. E. Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell. Nat. Med. 1997, 3, 730–737. 12. Desai, A.; Yan, Y.; Gerson, S. L. Concise reviews: cancer stem cell targeted therapies: toward clinical success. Stem Cells Transl. Med. 2019, 8, 75–81. 13. Munro, M. J.; Wickremesekera, S. K.; Peng, L.; Tan, S. T.; Itinteang, T. Cancer stem cells in colorectal cancer: a review. J. Clin. Pathol. 2018, 71, 110–116. 14. Zhou, Y.; Xia, L.; Wang, H.; Oyang, L.; Su, M.; Liu, Q.; Lin, J.; Tan, S.; Tian, Y.; Liao, Q.; Cao, D. Cancer stem cells in progression of colorectal cancer. Oncotarget 2018, 9, 33403–33415. 15. Patel, N. J.; Karuturi, R.; Al-Horani, R. A.; Baranwal, S.; Patel, J.; Desai, U. R.; Patel, B. B. Synthetic, non-saccharide, glycosaminoglycan mimetics selectively target colon cancer stem cells. ACS Chem. Biol. 2014, 9, 1826–1833. 16. Punt, C. J.; Koopman, M.; Vermeulen, L. From tumour heterogeneity to advances in precision treatment of colorectal cancer. Nat. Rev. Clin. Oncol. 2017, 14, 235–246. 17. Mouradov, D.; Sloggett, C.; Jorissen, R. N.; Love, C. G.; Li, S.; Burgess, A. W.; Arango, D.; Strausberg, R. L.; Buchanan, D.; Wormald, S.; et al. Colorectal cancer cell lines are representative models of the main molecular subtypes of primary cancer. Cancer Res. 2014, 74, 3238–3247. 18. Guinney, J.; Dienstmann, R.; Wang, X.; de Reynies, A.; Schlicker, A.; Soneson, C.; Marisa, L.; Roepman, P.; Nyamundanda, G.; Angelino, P.; et al. The consensus molecular subtypes of colorectal cancer. Nat. Med. 2015, 21, 1350–1356. 19. Berg, K. C. G.; Eide, P. W.; Eilertsen, I. A.; Johannessen, B.; Bruun, J.; Danielsen, S. A.; Bjornslett, M.; Meza-Zepeda, L. A.; Eknaes, M.; Lind, G. E.; et al. Multi-omics of 34 colorectal cancer cell lines - a resource for biomedical studies. Mol. Cancer 2017, 16, 116–132.
176

Understanding the Functional Group-dependent Self-assembly and Cellular Entry of Cationic Conjugated Polymer Nanoparticles

Manandhar, Prakash 26 March 2018 (has links)
Highly fluorescent conjugated polymers (CPs) are an important class of biomaterials used for various biological applications including labelling, sensing, and delivery of biological substances. Synthetic versatility and tunable emission make CPs a superior class of biomaterials. Understanding the structure-function relationship of CPs plays a vital role in designing high performing biomaterials. The cationic CPs are self-assembled to conjugated polymer nanoparticles (CPNs) in an aqueous environment due to their amphiphilicity. The physical and biophysical properties of CPNs are highly dependent on the chemical functionality and backbone structure of CPs. Modulation of the surface property and backbone structure of CPNs play an important role for efficient internalization of CPNs into cells. The goal of this dissertation is to understand the structure function relationship of CPNs in an aqueous environment and the change in their photo physical properties upon the self-assembly of CPNs with different backbone structure upon complexation with biologically significant polysaccharides and cell membrane. This work presents the self-assembly of a set of four cationic CPs with different connectivity and backbone structure upon complexation with a linear polyanion hyaluronic acid (HA). The study of photo physical properties changes upon the complexation with series of Glycosaminoglycans (GAGs) provides more insight about how the self-assembly behavior of cationic CPs changes upon the exposure to negatively charged polysaccharides. The understanding of the self-assembly of CPNs with negatively charged biologically important macromolecules under in vitro conditions can give us an idea of photophysical property changes of CPNs during the treatment of CPNs in the cellular environment. The study of the interaction of CPNs with cell membranes using scanning ion conductance microscopy (SICM)-based topography, potential mapping, and confocal microscopy imaging is presented. CPNs are able to induce transient pore like feature formation on the cell membrane during the cellular internalization process. A comparative study of cellular labelling and delivery of siRNA of five CPNs with guanidine motif is presented. The subcellular localization and delivery of siRNA were dependent on the side chain hydrophilicity. The CPNs fabricated with hydrophilic aminoethoxyethanol possesses excellent cellular imaging with higher siRNA delivery.
177

Untersuchungen zum Einfluss von artifiziellen extrazellulären Matrizes und elektrischen Feldern auf humane mesenchymale Stammzellen / Influence of artificial extracellular matrices and electric fields on human mesenchymal stem cells

Heß, Ricarda 31 July 2013 (has links) (PDF)
Eine bevorzugte Zellquelle für den Einsatz im Tissue Engineering sind mesenchymale Stammzellen (MSZ). Diese besitzen, neben einer hohen Proliferationsrate, die Fähigkeit, sich in verschiedene Zellen des mesodermen Ursprungs und in die entsprechenden Gewebetypen zu entwickeln. Um ein funktionales Gewebe zu erhalten ist es Ziel, sich bereits in vitro den in vivo Bedingungen anzunähern. Hierbei spielen neben der dreidimensionalen Struktur der Scaffolds auch die biochemische Mikroumgebung der Zellen in Form der unlöslichen extrazellulären Matrix (EZM) und den löslichen Mediatorproteinen wie Wachstums- und Differenzierungsfaktoren, sowie die physikalische Stimulation der Zellen eine wichtige Rolle. Während sich gegenwärtige Untersuchungen im TE vorwiegend mit den alleinigen Einflussfaktoren beschäftigen, verfolgt die vorliegende Arbeit das Ziel, die Auswirkungen kombinierter Stimuli durch Verwendung einer artifiziellen EZM, bestehend aus definierten Komponenten der nativen EZM, und physikalischer Stimuli durch elektrische Felder zu untersuchen. Letzteres erfolgte mit einem innerhalb der Arbeitsgruppe neu entwickelten System, dass die Stimulation von Zellen mit ausschließlich elektrischen Feldern, ohne störende Nebeneinflüsse, erlaubt.
178

Glycosaminoglycans and their sulfate derivatives differentially regulate the viability and gene expression of osteocyte-like cell lines

Tsourdi, Elena, Salbach-Hirsch, Juliane, Rauner, Martina, Rachner, Tilman D., Möller, Stephanie, Schnabelrauch, Matthias, Scharnweber, Dieter, Hofbauer, Lorenz C. 11 October 2019 (has links)
Collagen and glycosaminoglycans, such as hyaluronan and chondroitin sulfate, are the major components of bone extracellular matrix, and extracellular matrix composites are being evaluated for a wide range of clinical applications. The molecular and cellular effects of native and sulfatemodified glycosaminoglycans on osteocytes were investigated as critical regulators of bone remodeling. The effects of glycosaminoglycans on viability, necrosis, apoptosis, and regulation of gene expression were tested in two osteocyte-like cell lines, the murine MLO-Y4 and the rat UMR 106-01 cells. Glycosaminoglycans were non-toxic and incorporated by osteocytic cells. In MLO-Y4 cells, sulfation of glycosaminoglycans led to a significant inhibition of osteocyte apoptosis, 42% inhibition for highly sulfated chondroitin sulfate and 58% for highly sulfated hyaluronan, respectively. Cell proliferation was not affected. While treatment with highly sulfated chondroitin sulfate increased cell viability by 20% compared to the native chondroitin sulfate. In UMR 106- 01 cells, treatment with highly sulfated hyaluronan reduced the receptor activator of nuclear factor-κB ligand/osteoprotegerin ratio by 58% compared to the non-sulfated form, whereas highly sulfated chondroitin sulfate led to 60% reduction in the receptor activator of nuclear factor-κB ligand/osteoprotegerin ratio in comparison to the native chondroitin sulfate. The expression of SOST, the gene encoding sclerostin, was reduced by 50% and 45% by highly sulfated hyaluronan and chondroitin sulfate, respectively, compared to their native forms. The expression of BMP- 2, a marker of osteoblast differentiation, was doubled after treatment with the highly sulfated hyaluronan in comparison to its native form. In conclusion, highly sulfated glycosaminoglycans inhibit osteocyte apoptosis in vitro and promote an osteoblast-supporting gene expression profile.
179

Untersuchungen zum Einfluss von artifiziellen extrazellulären Matrizes und elektrischen Feldern auf humane mesenchymale Stammzellen

Heß, Ricarda 20 June 2013 (has links)
Eine bevorzugte Zellquelle für den Einsatz im Tissue Engineering sind mesenchymale Stammzellen (MSZ). Diese besitzen, neben einer hohen Proliferationsrate, die Fähigkeit, sich in verschiedene Zellen des mesodermen Ursprungs und in die entsprechenden Gewebetypen zu entwickeln. Um ein funktionales Gewebe zu erhalten ist es Ziel, sich bereits in vitro den in vivo Bedingungen anzunähern. Hierbei spielen neben der dreidimensionalen Struktur der Scaffolds auch die biochemische Mikroumgebung der Zellen in Form der unlöslichen extrazellulären Matrix (EZM) und den löslichen Mediatorproteinen wie Wachstums- und Differenzierungsfaktoren, sowie die physikalische Stimulation der Zellen eine wichtige Rolle. Während sich gegenwärtige Untersuchungen im TE vorwiegend mit den alleinigen Einflussfaktoren beschäftigen, verfolgt die vorliegende Arbeit das Ziel, die Auswirkungen kombinierter Stimuli durch Verwendung einer artifiziellen EZM, bestehend aus definierten Komponenten der nativen EZM, und physikalischer Stimuli durch elektrische Felder zu untersuchen. Letzteres erfolgte mit einem innerhalb der Arbeitsgruppe neu entwickelten System, dass die Stimulation von Zellen mit ausschließlich elektrischen Feldern, ohne störende Nebeneinflüsse, erlaubt.:1 Einleitung und Zielstellung 2 Theoretische Grundlagen 2.1 Der Knochen 2.1.1 Allgemeine Biologie und Physiologie des Knochengewebes 2.1.2 Knochenersatzmaterialien 2.2 Tissue Engineering von Knochengewebe 2.2.1 Trägermaterialien für das TE von Knochen 2.2.2 Zellen für das TE von Knochen 2.2.3 Artifizielle extrazelluläre Matrizes für das TE von Knochen 2.3 Einfluss elektrischer Felder auf Knochenumbauprozesse 2.3.1 Methoden zur Applikation von elektrischen Feldern 2.3.2 In vitro Untersuchungen zum Einfluss elektrischer Felder 2.3.3 Methode der Transformator-ähnlichen Einkopplung (TC) 3 Materialien 3.1 Technische Hilfsmittel und Geräte 3.2 Verbrauchsmaterialien 3.3 Chemikalien, Reagenzien und Kits 3.4 Antikörper 3.5 Oligonukleotide 3.6 Puffer-, Medien- und Lösungszusammensetzungen 3.7 Zellen 4 Methoden 4.1 Polycaprolacton-Co-Lactid (PCL)-Scaffolds 4.1.1 Präparation und Hydrophilisierung der PCL-Scaffolds 4.1.2 Beschichtung der PCL-Scaffolds 4.1.3 Charakterisierung der Beschichtung auf den PCL-Scaffolds 4.2 Zellkulturtechniken 4.2.1 Auftauen und Subkultivierung 4.2.2 Einfrieren 4.2.3 Induktion der osteogenen Differenzierung 4.2.4 Induktion der adipogenen Differenzierung 4.2.5 Induktion der chondrogenen Differenzierung 4.2.6 Besiedlung und Kultivierung der Zell-Matrix-Konstrukte 4.2.7 Elektrische Stimulation der Zell-Matrix-Konstrukte 4.2.8 Blockierung definierter Signaltransduktionswege 4.3 Mikroskopische Analytik der Zellen 4.3.1 Darstellung der Zellverteilung mittels Rasterelektronenmikroskopie (REM) 4.3.2 Qualitative Bestimmung von Fetttröpfchen mittels Oil-Red-O Färbung 4.3.3 Qualitative Bestimmung der Mineralisierung mittels vonKossa- Färbung 4.4 Durchflusszytometrie 4.5 Biochemische Analytik der Zellen 4.5.1 Bestimmung der Zellzahl mittels Lactatdehydrogenase (LDH)- Aktivität 4.5.2 Bestimmung der alkalische Phosphatase (ALP)-Aktivität 4.5.3 Quantitative Bestimmung des Kalziumgehaltes 4.6 Molekularbiologische Analytik / Genexpressionsanalyse 4.6.1 RNA Extraktion 4.6.2 cDNA-Synthese / Reverse Transkriptase PCR (RT-PCR) 4.6.3 Amplifikation von cDNA mittels quantitativer Real-Time PCR (qPCR) 4.7 Statistische Auswertung 5 Weiterentwicklung der Kammer zur TC-Einkopplung 5.1 Grundlegende theoretische Betrachtungen zur TC-Einkopplung 5.1.1 Ersatzschaltbild der TC-Einkopplung 5.1.2 Abschätzung des Eisenkernquerschnitts 5.1.3 Einfluss der Primärwindungszahl 5.2 Neudimensionierung und Aufbau der Stimulationseinrichtung 5.3 Verlauf der elektrischen Größen 5.3.1 Simulation 5.3.2 Messung 5.3.3 Abschätzung des magnetischen Feldes in der Kammer 5.4 Zusammenfassung 6 Zellexperimentelle Ergebnisse 6.1 Charakterisierung der humanen MSZ nach in vitro Kultivierung 6.1.1 Morphologie 6.1.2 Phänotypische Charakterisierung mittels Durchflusszytometrie 6.1.3 Multipotentes Differenzierungspotential 6.2 Zellverhalten auf den unbeschichteten PCL-Scaffolds 6.2.1 Ermittlung eines geeigneten Besiedlungsregimes 6.2.2 Zellverteilung und Proliferation der MSZ 6.2.3 Osteogene Differenzierung der MSZ 6.3 Einfluss der aEZM auf das Zellverhalten von MSZ 6.3.1 Quantitative Bestimmung der aEZM-Komponenten 6.3.2 Einfluss der aEZM auf die Adhärenz und Proliferation von MSZ 6.3.3 Einfluss der aEZM auf die osteogene Differenzierung von MSZ 6.4 Einfluss elektrischer Felder auf das Zellverhalten von MSZ 6.4.1 Einfluss der elektrischen Felder auf die Proliferation und osteogene Differenzierung von MSZ 6.4.2 Einfluss elektrischer Felder in Kombination mit Koll/sHya enthaltenden aEZM auf die Proliferation und osteogene Differenzierung von MSZ 6.4.3 Untersuchungen zu möglichen Signaltransduktionswegen 7 Diskussion der Ergebnisse 7.1 Charakterisierung der humanen MSZ nach in vitro Kultivierung 7.2 Zellverhalten auf den unbeschichteten PCL-Scaffolds 7.3 Einfluss der aEZM auf das Zellverhalten von MSZ 7.4 Einfluss elektrischer Felder auf das Zellverhalten von MSZ 8 Zusammenfassung und Ausblick Literaturverzeichnis Danksagung Eigene Publikationen und Mitautorschaften A Zusatzinformationen für die quantitative RT-PCR A.1 Versuchsdesign der Genexpressionsanalysen A.2 Qualitätskontrolle der isolierten RNA

Page generated in 0.0554 seconds