• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 17
  • 16
  • 7
  • 3
  • Tagged with
  • 42
  • 17
  • 9
  • 9
  • 8
  • 7
  • 7
  • 7
  • 6
  • 6
  • 6
  • 6
  • 6
  • 4
  • 4
  • 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.
41

Bone marrow niche-mimetics modulate hematopoietic stem cell function via adhesion signaling in vitro

Kräter, Martin 26 October 2017 (has links)
As graft source for lymphoma or leukemia treatment, hematopoietic stem and progenitor cells (HSPCs) have been the focus of translational medicine for decades. HSPCs are defined by their self-renewing capacity and their ability to give rise to all mature blood cells. They are found anchored to a specialized microenvironment in the bone marrow (BM) called the hematopoietic niche. HSPCs can be enriched by sorting them based on the presence of the surface antigen CD34 before clinical or tissue engineering use. As these cells represent a minority in most graft sources and the amount of applicable cells is limited, ex vivo expansion-cultures were established using cytokine cocktails or small molecules. However, in vitro culture of HSPCs as suspension-cultures result in heterogeneous cell populations with undefined cellular identities. In the BM niche, HSPCs are not exclusively maintained by cytokines but also by cell-matrix adhesions mediated by integrins (ITGs). Thus, β1 and β2 ITGs were found to promote initial contact of HSPCs with mesenchymal stromal cells (MSCs) and ITGβ3 expression was shown to be a marker for long-term repopulating HSPCs in vivo. Consequently, ex vivo remodeling of the BM niche using co-cultures of HSPCs and niche cells like MSCs came into spotlight and was proven to be a promising tool for stem cell expansion. However, in clinical and research applications, direct contact of two cell populations necessitates HSPC post-culture purification. To address these problems, we established a novel culture method for remodeling the BM extra cellular stroma in vitro wherein we used decellularized extracellular matrix (ECM) scaffolds derived from immortalized mesenchymal stromal cells (SCP-1). Such scaffolds were found to be highly reproducible and served as in vitro niche for HSPCs by being more effective for the expansion of CD34+ cells, compared to classical suspension cultures. ECMs were shown to consist of multiple proteins including fibronectins, collagens, and a major niche chemokine responsible for BM homing and retention of HSPCs in vivo, namely, stromal derived factor 1 (SDF-1). SDF-1 is known to be secreted by MSCs and is anchored to matrix proteins. This reveals that ECM scaffolds produced by SCP-1 cells are a naïve reconstructed microenvironment. When CD34+ cells were seeded, only around 20% of the cells adhered to the provided ECM scaffold. These cells recognized SDF-1 via C-X-C chemokine receptor type 4 (CXCR-4), as shown by laser scanning confocal microscopy. Thus, adhesive sides as they are present in the BM niche are provided. However, CD34+ cells isolated from G-CSF mobilized peripheral blood of healthy donors were found to be heterogenous with respect to adhesion capacity. Nonetheless, it was similar to HSPC co-cultures with SCP-1 cells as feeder layer. Therefore, we separated and analyzed two cell fractions, the adherent (AT-cells) and the non- adherent supernatant (SN-cells) cells. Other signals provided by the BM extracellular stroma to HSPCs are physical cues that control HSPC fate. HSPCs sense these physical features through focal contacts and accordingly remodel their morphological and biomechanical properties. Using real-time deformability cytometry (RT-DC) to uncover biomechanical phenotypes of freshly isolated HSPCs, SN-cells, AT-cells, and classical suspension cultured HSPCs in plastic culture dishes (PCD) were analyzed. We found freshly isolated cells to be less deformable and small. AT-cells displayed actin polymerization to stress fibers, and exhibited a stiffer mechanical phenotype compared to PCD-cultured or SN-cells. This might constitute the first hint of functional adaptation. Integrins are known to establish mechanosensing focal contacts. Thus, we analyzed ITG surface expression and identified ITGαIIb, ITGαV, and ITGβ3 to be enriched on AT-cells compared to freshly isolated cells or SN-cells. Active integrins need to form heterodimers consisting of one α- and one β subunit. Interestingly, the identified ITGs exclusively interact with each other to form RGD peptide receptors. RGD is a tripeptide consisting of the amino acids arginine, glycine, and aspartic acid and was identified as an adhesion sequence within fibronectin and other extracellular proteins. Consequently, we could confirm an important role for ITGαVβ3 in HSPC- ECM interaction with respect to adhesion and migration. However, we also identified ITGβ3 expression on a subset of CD34+ cells either freshly isolated or ECM cultured cells, as a marker for erythrocyte differentiation. These findings demonstrate that, in vitro, the ECM compartment acts as a regulator of HSPC fate and portray mechanical recognition as a potent driver of differentiation. In this context, targeted modulation of ECM scaffolds could enhance cell-ECM interactions and accelerate stem cell expansion or differentiation. These modulations could also provide further insights into HSPC-niche regulation. We demonstrate that ECMs derived from osteogenic differentiated SCP-1 cells increase HSPC expansion but do not lead to increased cell adhesion. As ECM adhesion preliminary alters HSPC function, we aimed at developing ECM scaffolds with increased adhesion capacity. Using lentiviral transduction, we generated a stable knock down of fibulin-1 in SCP-1 cells. Fibulin-1 is an ECM protein known to form anti-adhesion sites with fibronectin. However, we failed to increase adherent cell numbers or enhance HSPC expansion in the fibulin-1 knock down ECMs. Taken together, SCP-1 cell-derived ECM protein scaffolds provide an in vitro niche for HSPCs capable of stem cell expansion. Integrin mediated signaling altered the biomechanical and functional properties of HSPCs and hints at suspension cultures as being inappropriate to study the physiological aspects of HSPCs. Targeted modulation of ECM scaffolds could theoretically generate suitable ex vivo environments with the capacity to gain detailed insight into HSPC regulation within their niche. This will enhance the functionality of new biomaterials and will lead to improved regenerative therapies like BM transplantation.:List of contents I List of figures IV List of tables VI Abbreviations VII 1 Introduction 1 1.1 The stem cell microenvironment 3 1.1.1 The cellular endosteal bone marrow microenvironment 6 1.1.1.1 Mesenchymal stem/stromal cells 7 1.1.1.2 Hematopoietic stem and progenitor cells 8 1.1.2 Extracellular bone marrow microenvironment 10 1.1.2.1 Extracellular matrix 11 Chemokines and Cytokines 12 Cell adhesion to ECM 13 1.2 Native ex vivo ECM scaffolds 16 2 Aim of the study 19 3 Materials and methods 21 3.1 Materials 21 3.1.1 Chemicals and reagents 21 3.1.2 Kits 23 3.1.3 Media 24 3.1.4 Antibodies 24 3.1.5 Primers, sh-RNA sequences, and vectors 25 3.1.6 Equipment 26 3.1.7 Software 27 3.2 Methods 27 3.2.1 Cell preparation and culture 27 3.2.1.1 Mesenchymal stromal cells 27 3.2.1.2 Hematopoietic stem cells 28 3.2.1.3 Single cell picked clone 1 (SCP-1) cells 28 3.2.2 Generation of surface immobilized ECM preparations 29 3.2.2.1 Surface functionalization 29 3.2.2.2 ECM preparation 29 3.2.3 Flow cytometry and fluorescent activated cell sorting 30 3.2.4 Cell cycle analyses 30 3.2.5 Proliferation analyses 31 3.2.6 Colony forming unit cell assay (CFU-GEMM) 31 3.2.7 Migration assays 31 3.2.7.1 Transwell migration 31 3.2.7.2 Live cell migration 32 3.2.8 Confocal laser scanning microscopy 32 3.2.9 Real-time deformability cytometry (RT-DC) 32 3.2.10 Molecular biological methods 33 3.2.10.1 RNA isolation, reverse transcription, and PCR 33 3.2.10.2 Lentiviral shRNA transduction 34 3.2.10.3 Western blot 35 3.2.10.4 ELISA 36 3.2.11 Statistical analysis 37 4 Results 38 4.1 Extracellular matrix scaffolds for HSPCs 38 4.1.1 ECM properties 39 4.1.2 HSPC survival in ECM and PCD cultures 40 4.1.3 HSPC expansion in ECM and PCD cultures 41 4.2 HSPC morphological and mechanical adaptation to ECM 44 4.2.1 Actin polymerization and polarization 45 4.2.2 Biomechanical phenotype 46 4.3 Bioactive SDF-1 is incorporated in ECM scaffolds 49 4.3.1 CXCR4 polarization towards ECM 50 4.4 HSPC integrin expression and migration 52 4.4.1 Integrin surface expression on HSPC subsets 52 4.4.2 Focal contact formation 53 4.4.3 Integrin activation via ECM adhesion 55 4.4.4 Clonogenicity of ECM cultured HSPCs 57 4.4.5 HSPC migration when attached to ECM scaffolds 60 4.4.5.1 Reduced migratory behavior via ITGαVβ3 inhibition 61 4.4.5.2 SDF-1 induces migration but not adhesion 64 4.5 Targeted modulation of ECM scaffolds 65 4.5.1 Fibulin-1 knock down in SCP-1 cells 66 4.5.2 HSPC support of fibulin-1 reduced ECM scaffolds 70 5 Discussion 73 5.1 SCP-1 cells as a source for ECM scaffold production 74 5.2 Cell adhesion and focal contact formation 75 5.3 HSPC multilineage potential 78 5.4 ECM scaffold modulation 79 6 Summary 83 7 Zusammenfassung 86 Bibliography 89 Danksagung 108 Anlagen 110 Erklärung zur Eröffnung des Promotionsverfahrens [Formblatt 1.2.1] 110 Erklärung zur Einhaltung rechtlicher Vorschriften [Formblatt 1.1] 110
42

Quantifying adhesive interactions between cells and extracellular matrix by single-cell force spectroscopy

Taubenberger, Anna Verena 07 October 2009 (has links)
Interactions of cells with their environment regulate important cellular functions and are required for the organization of cells into tissues and complex organisms. These interactions involve different types of adhesion receptors. Interactions with extracellular matrix (ECM) proteins are mainly mediated by the integrin family of adhesion molecules. Situations in which integrin-ECM interactions are deregulated cause diseases and play a crucial role in cancer cell invasion. Thus, the mechanisms underlying integrin-binding and regulation are of high interest, particularly at the molecular level. How can cell-ECM interactions be studied? While there are several methods to analyze cell adhesion, few provide quantitative data on adhesion forces. One group, single-cell force spectroscopy (SCFS), quantifies adhesion at the single-cell level and can therefore differentiate the adhesive properties of individual cells. One implementation of SCFS is based on atomic force microscopy (AFM); this technique has been employed in the presented work. Advantageously AFM-SCFS combines high temporal and spatial cell manipulation, the ability to measure a large range of adhesion forces and sufficiently high-force resolution to allow the study of single-molecule binding events in the context of a living cell. Since individual adhesion receptors can be analyzed within their physiological environment, AFM-SCFS is a powerful tool to study the mechanisms underlying integrin-regulation. The presented work is split into six chapters. Chapter one gives background information about cell-ECM interactions. In chapter two, different adhesion assays are compared and contrasted. The theoretical Bell-Evans model which is used to interpret integrin-mediated cell adhesion is discussed in chapter three. Thereafter, the three projects that form the core of the thesis are detailed in chapters four through six. In the first project (chapter 4), α2β1-integrin mediated cell adhesion to collagen type I, the most abundant structural protein in vertebrates, was quantified using CHO cells. Firstly, α2β1-collagen interactions were investigated at the single-molecule level. Dynamic force spectroscopy permitted calculation of bond specific parameters, such as the bond dissociation rate koff (1.3 ± 1.3 sec-1) and the barrier width xu (2.3 ± 0.3 Å). Next, α2β1-integrin mediated cell adhesion to collagen type I was monitored over contact times between 0 and 600 sec. Thereby the kinetics of α2β1-integrin mediated interactions was explored and insights into the underlying binding mechanisms were gained. In the second project (chapter five), effects of cryptic integrin binding sites within collagen type I exerted on pre-osteoblasts were investigated. Collagen type I matrices were thermally denatured which lead to exposure of cryptic RGD (Arg-Gly-Asp)-motifs. As a consequence pre-osteoblasts enhanced their adhesion to denatured collagen. Compared to native collagen type I, adhesion to denatured collagen was mediated by a different set of integrins, including αv- and α5β1-integrins. Cells grown on denatured collagen showed enhanced spreading and motility, which correlated with increased focal adhesion kinase phosphorylation levels. Moreover, osteogenic differentiation kinetics and differentiation potential were increased on denatured collagen. The findings of this project open new perspectives for optimization of tissue engineering substrates. In the third part (chapter six), the effect of the fusion protein BCR/ABL, a hallmark of chronic myeloid leukemia, on adhesion of myeloid progenitor cells was studied. Adhesion between BCR/ABL transformed progenitor cells to bone marrow derived stromal cells and to different ECM proteins was quantitatively compared to that of control cells. The tyrosine kinase activity of BCR/ABL enhanced cell adhesion, which was blocked by imatinib mesylate, a drug interfering with BCR/ABL activity. BCR/ABL-enhanced adhesion correlated with increased β1-integrin cell surface concentrations. Since adhesion of leukemic cells to the bone marrow compartment is critical for the development of drug resistance, the reported results may provide a basis for optimized target therapies. In the three described projects AFM-based SCFS was applied to investigate early steps of integrin-mediated adhesion at the molecular level. Taken together, the results demonstrate that AFM-SCFS is a versatile tool that permits monitoring of cell adhesion from single-molecule interactions to the formation of more complex adhesion sites at the force level. / Interaktionen zwischen Zellen und ihrer Umgebung sind maßgeblich an der Regulierung zellulärer Funktionen beteiligt und daher notwendig für die Organisation von Zellen in Geweben und komplexen Organismen. Zellinteraktionen mit der extrazellulären Matrix (EZM) werden hauptsächlich durch Integrine vermittelt. Situationen, in denen Integrin- EZM Interaktionen verändert sind, können Krankheiten verursachen und spielen zudem eine wichtige Rolle bei der Invasion von Krebszellen. Daher besteht ein großes Interesse darin, die molekularen Mechanismen, die Integrin-EZM Interaktionen regulieren, besser zu verstehen. Wie können Zell-EZM Interaktionen untersucht werden? Obwohl es mehrere Methoden gibt, mit denen Zelladhäsion untersucht werden kann, sind die wenigsten dazu geeignet, Zelladhäsionskräfte zu quantifizieren. Einzelzellspektroskopie erfasst die Adhäsionskräfte einzelner Zellen quantitativ und ermöglicht dadurch eine differenzierte Betrachtung der Adhäsion individueller Zellen. Eine Variante der Einzelzellspektroskopie basiert auf der Rasterkraftmikroskopie (AFM); diese Technik wurde in der vorliegenden Arbeit verwendet. Ein Vorteil von AFM- Einzelzellspektroskopie besteht darin, dass Zellen mit hoher zeitlicher und räumlicher Präzision manipuliert werden können. Zelladhäsionskräfte können zudem über einen großen Kraftbereich hinweg untersucht werden. Dabei ermöglicht es die hohe Kraftauflösung, einzelne Integrin-Ligandenbindungen in lebenden Zellen zu untersuchen. Die vorliegende Arbeit gliedert sich in sechs Kapitel. Kapitel eins gibt Hintergrundinformationen über Zell-EZM Wechselwirkungen. In Kapitel zwei werden verschiedene Adhäsionsassays einander gegenüber gestellt. Das theoretische Bell-Evans Modell, mit dessen Hilfe die gewonnenen Daten interpretiert wurden, wird in Kapitel drei diskutiert. Im Anschluss werden drei Projekte, welche das Herzstück dieser Doktorarbeit bilden, in Kapiteln vier bis sechs näher ausgeführt. Im ersten Projekt (Kapitel vier) wurde die Adhäsion von α2β1-Integrin exprimierenden CHO Zellen zu Kollagen I, dem häufigsten strukturellen Protein in Wirbeltieren, quantitativ untersucht. Zunächst wurden α2β1-Kollagen-Interaktionen auf Einzelmolekülebene analysiert. Mithilfe der dynamischen Kraftspektroskopie wurden für diese Bindung Dissoziationsrate koff (1.3 ± 1.3 sec-1) und Potentialbarrierenbreite xu (2.3 ± 0.3 Å) bestimmt. Daraufhin wurde die α2β1-vermittelte Adhäsion über einen Zeitraum von zehn Minuten untersucht. Dadurch konnten Einblicke in die Kinetik von α2β1-integrin vermittelter Zelladhäsion sowie in die zugrunde liegenden Regulationsmechanismen gewonnen werden. Im zweiten Projekt (Kapitel fünf) wurde die Rolle von kryptischen Integrin-Bindungsstellen in Kollagen I untersucht. Die zuvor verwendeten Kollagenoberflächen wurden thermisch denaturiert, wodurch versteckte RGD (Arg-Gly-Asp)-Sequenzen freigelegt wurden. Die partielle Denaturierung hatte- verglichen mit nativem Kollagen I- eine erhöhte Adhäsion von Präosteoblasten (MC3T3-E1) zur Folge, was auf das Binden zusätzlicher Integrine zurückgeführt wurde. Im Unterschied zu nativem Kollagen wurde die Zelladhäsion zu denaturiertem Kollagen I u.a. durch αv- and α5β1-Integrine vermittelt. Präosteoblasten zeigten verstärktes Zellspreiten sowie höhere Motilität auf denaturiertem Kollagen I; zudem wurde ein erhöhtes Differenzierungpotential der Präosteoblasten festgestellt. Die in diesem Projekt erhaltenen Einblicke bilden eine hilfreiche Basis für die Entwicklung optimierter Oberflächen für diverse Zell- und Gewebekulturanwendungen. Im dritten Projekt (Kapitel sechs) wurde der Einfluss des Fusionproteins BCR/ABL, charakteristisch für chronische myeloische Leukämie, auf die Adhäsion von myeloischen Vorläuferzellen untersucht. Dazu wurde die Adhäsion von BCR/ABL transformierten Vorläuferzellen (32D Zellen) bzw. Kontrollzellen zu Stromazellen (M2-10B4) sowie verschiedenen EZM Proteinen untersucht. BCR/ABL erhöhte die Zelladhäsion der myeloischen Vorläuferzellen signifikant. Dieser Effekt wurde durch die Zugabe von Imatinib, welches die Tyrosinkinaseaktivität von BCR/ABL inhibiert, aufgehoben. Die BCR/ABL-verstärkte Zelladhäsion korrelierte mit erhöhten β1-Integrin-konzentrationen. Da die Adhäsion von Leukämiezellen im Knockenmark bekanntermaßen kritisch für die Entwicklung von Resistenzen gegenüber verschiedenen Wirkstoffen ist, könnten die Ergebnisse dieser Studie eine Grundlage für die Entwicklung optimierter Target-Therapien sein. In den drei beschriebenen Projekten wurde AFM Einzelzellspektroskopie verwendet, um Integrin- vermittelte Adhäsion auf molekularer Ebene zu untersuchen. Die Ergebnisse zeigen, dass AFM-Einzelzellspektroskopie ein vielseitiges Werkzeug darstellt, das überaus geeignet dazu ist, Zelladhäsion- ausgehend von Einzelmolekülinteraktionen bis hin zur Entstehung komplexerer Adhäsionsstellen- auf der Kraftebene zu verfolgen.

Page generated in 0.1429 seconds