• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 759
  • 286
  • 82
  • 66
  • 52
  • 40
  • 28
  • 27
  • 20
  • 20
  • 20
  • 20
  • 20
  • 20
  • 17
  • Tagged with
  • 1640
  • 1640
  • 372
  • 368
  • 279
  • 277
  • 264
  • 261
  • 249
  • 248
  • 234
  • 231
  • 218
  • 211
  • 203
  • 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.
81

Characteristics of cells under different tumor microenvironmental conditions.

January 2002 (has links)
by Ng Mei Yu. / Thesis (M.Phil.)--Chinese University of Hong Kong, 2002. / Includes bibliographical references (leaves 173-183). / Abstracts in English and Chinese. / Acknowledgements --- p.2 / Abbreviations --- p.3 / Abstracts --- p.4 / List of figures and tables --- p.7 / Contents Page No / General Introduction --- p.13 / Chapter CHAPTER ONE --- Biological Characterization of A431 Cells & SiHa Cells Under Different Microenvironments --- p.16 / Chapter 1.1 --- Introduction --- p.17 / Chapter 1.1.1 --- Microenvironment Surrounding Tumor Cells --- p.18 / Chapter 1.1.2 --- Hypoxic Environment --- p.20 / Chapter 1.1.2.1 --- The Hypoxic Chamber --- p.20 / Chapter 1.1.3 --- Reoxygenation --- p.22 / Chapter 1.1.4 --- Acidic Environment --- p.23 / Chapter 1.1.5 --- Glucose Depletion --- p.24 / Chapter 1.1.6 --- Irradiation --- p.25 / Chapter 1.2 --- Objectives --- p.26 / Chapter 1.3 --- Materials and Methods --- p.27 / Chapter 1.3.1 --- Materials --- p.27 / Chapter 1.3.2 --- Methods --- p.29 / Chapter 1.3.2.1 --- Cell Lines --- p.29 / Chapter 1.3.2.2 --- The Working Procedure for the Hypoxic Chamber --- p.30 / Chapter 1.3.2.3 --- "Aerobic, Hypoxic and Reoxygenated Conditions" --- p.33 / Chapter 1.3.2.4 --- Acidic Condition --- p.35 / Chapter 1.3.2.5 --- Glucose Depleted Condition --- p.36 / Chapter 1.3.2.6 --- Gamma-Irradiation --- p.37 / Chapter 1.3.2.7 --- Analysis of the Growth Pattern by MTT Assay and Cell Counting --- p.38 / Chapter 1.3.2.8 --- Cell Cycle Analysis --- p.39 / Chapter 1.3.2.9 --- Western Blot Analysis --- p.40 / Chapter 1.3.2.10 --- DNA Fragmentation Analysis --- p.42 / Chapter 1.4 --- Results --- p.43 / Chapter 1.4.1 --- Cell Proliferation Profile by MTT Assay --- p.43 / Chapter 1.4.1.1 --- Proliferation of cells under hypoxia --- p.43 / Chapter 1.4.1.2 --- Proliferation of cells under acidic pH environments --- p.49 / Chapter 1.4.1.3 --- Proliferation of cells under glucose depleted environment --- p.52 / Chapter 1.4.2 --- Distribution of cell cycles under different micro environments --- p.54 / Chapter 1.4.3 --- General Protein Expression Pattern by Western Blot Analysis --- p.57 / Chapter 1.4.4 --- Detection of Apoptosis by DNA Fragmentation Assay --- p.59 / Chapter 1.5 --- Discussion --- p.58 / Chapter CHAPTER TWO --- REACTION KINETICS OF A431 CELLS AND SiHa CELLS INDUCED BY EGF --- p.71 / Chapter 2.1 --- Introduction --- p.72 / Chapter 2.1.1 --- Structure of EGF and EGFR --- p.74 / Chapter 2.1.2 --- EGF Signaling Pathway --- p.76 / Chapter 2.2 --- Objectives --- p.79 / Chapter 2.3 --- Materials and Methods --- p.80 / Chapter 2.3.1 --- Materials --- p.80 / Chapter 2.3.2 --- Methods --- p.82 / Chapter 2.3.2.1 --- Cell Lines --- p.82 / Chapter 2.3.2.2 --- EGF Sensitivity Assay --- p.83 / Chapter 2.3.2.3 --- Combination Effect of Hypoxia and EGF --- p.83 / Chapter 2.3.2.4 --- Early Kinetics Analysis by Low EGF Concentration Treatment --- p.84 / Chapter 2.3.2.5 --- Late Kinetics Analysis by High EGF Concentration Treatment --- p.85 / Chapter 2.4 --- Results --- p.86 / Chapter 2.4.1 --- Sensitivity of A431 cells and SiHa cells to EGF by MTT Assay --- p.86 / Chapter 2.4.2 --- Early/Late Kinetics of EGF induced protein tyrosine phosphorylation Pattern --- p.90 / Chapter 2.4.3 --- Raf protein expression --- p.96 / Chapter 2.4.4 --- EGFR expression level --- p.100 / Chapter 2.5 --- Discussions --- p.104 / Chapter CHAPTER THREE --- IDENTIFICATION OF DIFFERENTIALLY EXPRESSED GENES IN A431 CELLS BY DIFFERENTIAL DISPLAY UNDER DIFFERENT TUMOR MICROENVIRONMENTS --- p.107 / Chapter 3.1 --- Introduction --- p.108 / Chapter 3.2 --- Materials and Methods --- p.112 / Chapter 3.2.1 --- Materials --- p.112 / Chapter 3.2.2 --- Methods --- p.114 / Chapter 3.2.2.1 --- Spheroid Cells --- p.114 / Chapter 3.2.2.2 --- Identification of Differentally Expressed Genes by RT-PCR --- p.117 / Chapter 3.2.2.3 --- Ligation and Cloning of Differentially Expressed cDNA --- p.120 / Chapter 3.2.2.4 --- Screening and Sequencing of the cDNA Inserts --- p.121 / Chapter 3.2.2.5 --- Northern Blot Analysis --- p.123 / Chapter 3.3 --- Results --- p.124 / Chapter 3.4 --- Discussions --- p.161 / GENERAL CONCLUSION --- p.165 / REFERENCES --- p.167
82

The bioinformatics of the novel genes revealed by sequencing of human heart cDNA and the molecular characterization of one such gene that codes for a human fibroblast growth factor. / CUHK electronic theses & dissertations collection / Digital dissertation consortium

January 1997 (has links)
Kok Dick Shun , Louis. / Thesis (Ph.D.)--Chinese University of Honbg Kong, 1997. / Includes bibliographical references (p. i-xiii). / Electronic reproduction. Hong Kong : Chinese University of Hong Kong, [2012] System requirements: Adobe Acrobat Reader. Available via World Wide Web. / Electronic reproduction. Ann Arbor, MI : ProQuest Information and Learning Company, [200-] System requirements: Adobe Acrobat Reader. Available via World Wide Web. / Mode of access: World Wide Web.
83

Functional characterization of IGF2BP2, a diabetes-susceptibility gene

Le, Hang Thi Thu January 2011 (has links)
No description available.
84

Mechanisms through which nuclear estrogen receptors remain transcriptionally active in the mouse hippocampus in absence of ovarian estrogens.

January 2017 (has links)
acase@tulane.edu / The goal of the following experiments was to determine the cellular mechanisms through which estrogen receptor activity is maintained in hippocampal cells following termination of ovarian function. Aim 1 determined that kinase signaling contributes to the maintenance of estrogen receptor activity in the hippocampus of ovariectomized mice in addition to local synthesis of brain derived “neuroestrogens”. Inhibition of both the mitogen activated protein kinase (MAPK) and phosphoinositide-3 kinase (PI3K) cascades with intracerebroventricular infusion of specific kinase inhibitors reduced estrogen response element (ERE)-dependent gene expression in the hippocampus of ovariectomized mice. Aim 2 determined that neuroestrogen synthesis, MAPK signaling, and PI3K signaling interact to regulate the transcriptional output of estrogen receptors in response to insulin like growth factor-1 receptor (IGF-1R) activation in the Neuro-2A cell culture model. Rapid IGF-1R-dependent MAPK signaling promotes, while PI3K signaling inhibits, IGF-1R-dependent activation of endogenous estrogen receptors in Neuro-2A cells. Long-term IGF-1R stimulation reduces ERE-dependent gene expression in part through phosphorylation of estrogen receptor alpha (ERα). Rapid IGF-1R-dependent activation but not long-term repression of estrogen receptor activity in Neuro-2A cells requires neuroestrogen synthesis. Aim 3 determined that exposure to 40 days of continuous unopposed estradiol at the time of ovariectomy results in lasting enhancement of estrogen receptor activity in the hippocampus and lasting enhancement of hippocampus dependent memory in female mice beyond the period of short-term estradiol exposure. Together these three aims determine that neuroestrogen synthesis and kinase signaling interact to actively maintain estrogen receptor signaling in neuronal cells and these autonomous neuronal mechanisms of estrogen receptor activation have functional consequences on cognition long after cessation of ovarian function. / 1 / Kevin J Pollard
85

Therapeutic systems for Insulin-like growth factor-1 / Therapeutische Systeme für Insulin-like growth factor-1

Schultz, Isabel January 2015 (has links) (PDF)
SUMMARY Insulin-like growth factor I (IGF-I) is a polypeptide with a molecular weight of 7.649 kDa and an anabolic potential. Thereby, IGF-I has a promising therapeutic value e.g. in muscle wasting diseases such as sarcopenia. IGF-I is mainly secreted by the liver in response to growth hormone (GH) stimulation and is rather ubiquitously found within all tissues. The effects of IGF-I are mediated by its respective IGF-I transmembrane tyrosine kinase receptor triggering the stimulation of protein synthesis, glucose uptake and the regulation of cell growth. The actions of IGF-I are modulated by six IGF binding proteins binding and transporting IGF-I in a binary or ternary complex to tissues and receptors and modulating the binding of IGF-I to its receptor. The nature of the formed complexes impacts IGF-I`s half-life, modulating the half-life between 10 minutes (free IGF-I) to 12 - 15 hours when presented in a ternary complex with IGF binding protein 3 and an acid labile subunit (ALS). Therefore, sustained drug delivery systems of free IGF-I are superficially seen as interesting for the development of controlled release profiles, as the rate of absorption is apparently and easily set slower by simple formulation as compared to the rapid rate of elimination. Thereby, one would conclude, the formulation scientist can rapidly develop systems for which the pharmacokinetics of IGF-I are dominated by the formulation release kinetics. However, the in vivo situation is more complex and as mentioned (vide supra), the half-life may easily be prolonged up to hours providing proper IGF-I complexation takes place upon systemic uptake. These and other aspects are reviewed in Chapter I, within which we introduce IGF-I as a promising therapeutic agent detailing its structure and involved receptors along with the resulting signaling pathways. We summarize the control of IGF-I pharmacokinetics in nature within the context of its complex system of 6 binding proteins to control half-life and tissue distribution. Furthermore, we describe IGF-I variants with modulated properties in vivo and originated from alternative splicing. These insights were translated into sophisticated IGF-I delivery systems for therapeutic use. Aside from safety aspects, the challenges and requirements of an effective IGF-I therapy are discussed. Localized and systemic IGF-I delivery strategies, different routes of administration as well as liquid and solid IGF-I formulations are reviewed. Effective targeting of IGF-I by protein decoration is outlined and consequently this chapter provides an interesting guidance for successful IGF-I-delivery. In Chapter II, we firstly outline the stability of IGF-I in liquid formulations with the intention to deliver the biologic through the lung and the impact of buffer type, sodium chloride concentration and pH value on IGF-I stability is presented. IGF-I integrity was preserved in histidine buffer over 4 months at room temperature, but methionine 59 oxidation (Met(o)) along with reducible dimer and trimer formation was observed in an acidic environment (pH 4.5) and using acetate buffer. Strong aggregation resulted in a complete loss of IGF-I bioactivity, whereas the potency was partly maintained in samples showing a slight aggregation and complete IGF-I oxidation. Atomization by air-jet or vibrating-mesh nebulizers yielded in limited Met(o) formation and no aggregation. The results of IGF-I nebulization experiments regarding aerosol output rate, mass median aerodynamic diameter and fine particle fraction were comparable with 0.9% sodium chloride reference, approving the applicability of liquid IGF-I formulations for pulmonary delivery. In Chapter III we escalated the development to solid delivery systems designed for alveolar landing upon inhalation and by deploying trehalose and the newly introduced for pulmonary application silk-fibroin as carriers. Microparticles were produced using nano spray drying following analyses including IGF-I integrity, IGF-I release profiles and aerodynamic properties. In vitro transport kinetics of IGF-I across pulmonary Calu-3 epithelia were suggesting similar permeability as compared to IGF-I’s cognate protein, insulin that has already been successfully administered pulmonary in clinical settings. These in vivo results were translated to an ex vivo human lung lobe model. This work showed the feasibility of pulmonary IGF-I delivery and the advantageous diversification of excipients for pulmonary formulations using silk-fibroin. Chapter IV focuses on an innovative strategy for safe and controllable IGF-I delivery. In that chapter we escalated the development to novel IGF-I analogues. The intention was to provide a versatile biologic into which galenical properties can be engineered through chemical synthesis, e.g. by site directed coupling of polymers to IGF-I. For this purpose we genetically engineered two IGF-I variants containing an unnatural amino acid at two positions, respectively, thereby integrating alkyne functions into the primary sequence of the protein. These allowed linking IGF-I with other molecules in a site specific manner, i.e. via a copper catalyzed azide-alkyne Huisgen cycloaddition (click reaction). In this chapter we mainly introduce the two IGF-I variants, detail the delivery concept and describe the optimization of the expression conditions of the IGF-I variants. In conclusion, we span from simple liquid formulations for aerolization through solid systems for tailored for maximal alveolar landing to novel engineered IGF-I analogues. Thereby, three strategies for advanced IGF-I delivery were addressed and opportunities and limitations of each were outlined. Evidence was provided that sufficiently stable and easy to manufacture formulations can be developed as typically required for first in man studies. Interestingly, solid systems – typically introduced in later stages of pharmaceutical development – were quite promising. By use of silk-fibroin as a new IGF-I carrier for pulmonary administration, a new application was established for this excipient. The demonstrated success using the ex vivo human lung lobe model provided substantial confidence that pulmonary IGF-I delivery is possible in man. Finally, this work describes the expression of two IGF-I variants containing two unnatural amino acids to implement an innovative strategy for IGF-I delivery. This genetic engineering approach was providing the fundament for novel IGF-I analogues. Ideally, the biologic is structurally modified by covalently linked moieties for the control of pharmacokinetics or for targeted delivery, e.g. into sarcopenic muscles. One future scenario is dicussed in the ‘conclusion and outlook’ section for which IGF-I is tagged to a protease sensitive linker peptide and this linker peptide in return is coupled to a polyethylenglykole (PEG) polymer (required to prolong the half-life). Some proteases may serve as proxy for sarcopenia such that protease upregulation in compromised muscle tissues drives cleavage of IGF-I from the PEG. Thereby, IGF-I is released at the seat of the disease while systemic side effects are minimized. / ZUSAMMEMFASSUNG Insulin-like growth factor I (IGF-I) ist ein 7.6 kDa großes Polypeptid, das eine anabole Wirkung besitzt und dadurch ein vielversprechendes Therapeutikum in Muskelerkrankungen wie z.B. Sarkopenie darstellt. IGF-I wird hauptsächlich von der Leber gebildet und infolge der Stimulation des Wachstumshormons Somatropin sezerniert. In fast jedem Gewebe des Körpers kommt IGF-I vor. Die Wirkungen von IGF-I werden über eigene Rezeptoren, die an die Zellmembran gebunden sind, die Rezeptor-Tyrosinkinasen, ausgeführt. Zu den Wirkungen gehören unter anderem die Stimulation der Proteinsynthese, die Aufnahme von Glucose in die Zellen und die Regulierung des Zellwachstums. Die Effekte von IGF-I werden von 6 IGF- Bindungsproteinen (IGFBP 1-6) gesteuert, indem IGF-I in einem binären oder ternären Komplex zu den Geweben transportiert oder auch die Bindung von IGF-I an den Rezeptor verhindert werden kann. Die sich bildenden Komplexe haben auch einen Einfluss auf die Halbwertszeit (HWZ) von IGF-I, da für ungebundenes IGF-I eine HWZ von ca. 10 Minuten festgestellt werden konnte, aber IGF-I, gebunden in einem ternären Komplex mit dem Bindungsprotein 3 und der säurelabilen Untereinheit (ALS) eine erhöhte HWZ von 12 – 15 Stunden aufweist. Deswegen sind „sustained drug delivery“ Systeme von ungebundenem IGF-I auf den ersten Blick interessant für die Entwicklung von kontrollierten Freisetungsprofilen, da die Absorptionsgeschwindigkeit offensichtlich und problemlos durch triviale Formulierung verlangsamt werden kann im Vergleich zu der schnellen Eliminationsgeschwindigkeit. Deshalb könnte man daraus schließen, dass ein Formulierungsexperte recht schnell Systeme entwickeln kann, in denen die Freisetzungskinetik der Formulierung über die pharmakokinetischen Eigenschaften von IGF-I dominiert. Jedoch ist die in vivo Situation wesentlich komplexer und wie oben bereits erwähnt, könnte die Halbwertszeit problemlos bis zu mehreren Stunden verlängert werden, sofern geeignete Komplexbildung von IGF-I nach systemischer Aufnahme erfolgt. Diese und weitere Aspekte werden in Kapitel I beschrieben. Außerdem stellen wir IGF-I als wertvolles Therapeutikum vor, beschreiben dessen Struktur, die beteiligten Rezeptoren und die daraus resultierenden Signalwege. Wir fassen die Kontrolle der Pharmakokinetik von IGF-I in der Natur zusammen, im Rahmen von einem komplexen System aus 6 Bindungsproteinen, die die Halbwertszeit und die Gewebeverteilung steuern. Außerdem beschreiben wir IGF-I Varianten, die veränderte Eigenschaften in vivo aufweisen und durch alternatives Spleißen entstanden sind. Diese Erkenntnisse werden in hochentwickelte „IGF-I delivery“ Systeme für den therapeutischen Gebrauch umgesetzt. Neben Sicherheitsaspekten werden die Herausforderungen und Anforderungen einer effektiven IGF-I Therapie diskutiert. Darüber hinaus wird über lokale und systemische „IGF-I delivery“ Strategien, verschiedene Verabreichungswege sowie flüssige und feste IGF-I Formulierungen berichtet. Ebenso wird die wirkungsvolle IGF-I Freisetzung am Zielort durch Ausschmückung des Proteins beschrieben und dementsprechend liefert dieses Kapitel eine interessante Orientierungshilfe für eine erfolgreiche IGF-I Therapie. Im Kapitel II untersuchen wir die Stabilität von IGF-I in flüssigen Formulierungen zur pulmonalen Anwendung bezüglich Puffersystem, Natriumchlorid Konzentration und pH Wert. Die IGF-I Integrität wurde im Histidin Puffer über 4 Monate bei Raumtemperatur aufrechterhalten. Allerdings wurde bei Verwendung eines Acetat Puffers pH 4.5, Oxidation am Methionin 59 (Met(o)) sowie die Entstehung von reduzierbaren Dimeren und Trimeren beobachtet. Starke Aggregation führte zum vollständigen Verlust der IGF-I Bioaktivität, während die Wirkung in Proben aufrechterhalten werden konnte, in denen eine geringe Aggregation, aber deutliche Oxidation festgestellt wurde. Nach der Verneblung der flüssigen IGF-I Formulierung im Histidin-Puffer pH 6.5 mit einem Druckluftvernebler und einem Schwingmembranvernebler wurde jeweils eine leichte Bildung von Met(o), aber keine Aggregatbildung ermittelt. Die Ergebnisse der IGF-I Verneblungsexperimente waren vergleichbar mit den Referenzwerten einer isotonischen Kochsalzlösung bezüglich der Abgabeleistung, dem massenbezogenen medianen aerodynamischen Durchmesser und dem Feinpartikel Anteil. Hierdurch wurde gezeigt, dass sich flüssige IGF-I Formulierungen zur pulmonalen Anwendung eignen. Im Kapitel III berichten wir von einer Weiterentwicklung zu festen IGF-I Formulierungen für die pulmonale Route unter Verwendung von Trehalose und Seidenfibroin als neues Trägermaterial für die pulmonale Applikation. Mikropartikel wurden durch Nanosprühtrocknung hergestellt und anschließend auf IGF-I Integrität, IGF-I Freisetzung und dem aerodynamischen Durchmesser untersucht. Die Kinetik des in vitro Transportes von IGF-I durch Calu-3 Lungenepithelzellen war vergleichbar zur Durchgängigkeit von Insulin, das bereits erfolgreich pulmonal verabreicht wurde. Dieser Erfolg wurden auch ex vivo in einem menschlichen Lungenlappen Model bestätigt. In der Arbeit wird somit gezeigt, dass IGF-I zur pulmonalen Anwendung geeignet ist und die Verwendung von Seidenfibroin eine nützliche Erweiterung zu den bisher eingesetzten Trägermaterialien darstellt. Das Kapitel IV konzentriert sich auf eine innovative Strategie, um IGF-I sicher und kontrollierbar zu verabreichen. In diesem Kapitel erweitern wir die Entwicklung zu neuartigen IGF-I Varianten. Wir streben damit an ein vielseitiges Biologikum zu entwickeln, dessen Eigenschaften durch chemische Reaktionen verändert werden können wie zum Beispiel die spezifische Verknüpfung mit Polymeren. Zu diesem Zweck erzeugten wir gentechnisch zwei IGF-I Varianten, die jeweils an zwei Positionen eine unnatürliche Aminosäure aufweisen und führten dadurch Alkine Gruppen in die Primärstruktur der Proteine ein. Diese Vorgehensweise ermöglicht es nun IGF-I mit anderen Molekülen positionsspezifisch zu verbinden wie zum Beispiel durch die kupferkatalysierte Azid-Alkin-Cycloaddition (Click – Reaktion). In diesem Kapitel stellen wir hauptsächlich die zwei IGF-I Varianten vor, beschreiben ausführlich das Konzept der IGF-I Zustellung und erklären die Vorgehensweise zur Optimierung der Expressionsbedingungen der IGF-I Varianten. Abschließend lässt sich sagen, dass sich diese Arbeit über einfach flüssige Formulierungen zur Verneblung, feste Formulierung mit guten aerodynamischen Eigenschaften zur Erreichung der Alveolen und neuartig entwickelte IGF-I Varianten erstreckt. Hierzu werden drei Strategien zur modernen IGF-I Gabe thematisiert und sowohl die Möglichkeiten als auch die Grenzen der jeweiligen Therapie erörtert. Wir haben den Nachweis erbracht, dass ausreichend stabile und leicht herzustellende Formulierungen entwickelt werden können, die üblicherweise für „First-In-Man“ Studien benötigt werden. Interessanterweise stellten sich die festen Formulierungen, die eigentlich in den späteren Phasen der pharmazeutischen Entwicklung eingeführt werden, als sehr vielversprechend heraus. Durch den Einsatz von Seidenfibroin als neuen Träger zur pulmonalen Anwendung haben wir einen neuen Verwendungszweck für Seidenfibroin etabliert. Der erfolgreiche Versuch ex vivo am menschlichen Lungenlappen Model liefert die feste Überzeugung, dass es möglich ist, IGF-I im Menschen pulmonal anzuwenden. Letztendlich, beschreibt die Arbeit die Expression von zwei IGF-I Varianten, die zwei unnatürliche Aminosäuren aufweisen, um eine neuartige Strategie zur IGF-I Verabreichung umzusetzen. Dieser gentechnische Ansatz liefert die Grundlage für neue IGF-I Varianten. Idealerweise, wird das Biopharmazeutikum strukturell durch kovalent gebundene Reste verändert, um die pharmakokinetischen Eigenschaften zu steuern oder um zielgenaue Wirkstoffabgabe zu erreichen zum Beispiel in den sarkopenischen Muskeln. Ein Zukunftsszenarium wird im Abschnitt „Conclusion and Outlook“ diskutiert, in dem IGF-I mit einem Protease empfindlichen Linker versehen wird, der wiederum mit einem Polyethylenglykol (PEG) Polymer verknüpft ist. Der PEG Rest wird benötigt, um die Hablbwertszeit von IGF-I zu erhöhen. Einige Proteasen könnten als Stellvertreter für Sarkopenie dienen, so dass die Hochregulierung der Proteasen in gefährdeten Muskelgeweben zur Spaltung von IGF-I und dem PEG Rest führt. Dadurch wird IGF-I am Ursprung der Erkrankungen freigesetzt, während die systemischen Nebenwirkungen weitgehend vermindert sind.
86

Studies of bladder cancer progression

Hung, Tzong Tyng, Clinical School - Prince of Wales Hospital, Faculty of Medicine, UNSW January 2009 (has links)
Bladder cancer (BlCa) is the second most common genitourinary cancer, affecting both men and women. Most (70%) cases present at the superficial stage; 20% of these recur with muscle-invasive disease. Major genetic alterations associated with BlCa include: loss/gain in expression or mutations in Retinoblastoma (RB) gene, human epidermal growth factor receptors (HERs), H-ras, p53 and FGFR3. Only p53 mutations are well correlated with invasive BlCa; other changes show variable correlations with disease status. To understand the progression of BlCa, a model of nine human BlCa cell sublines derived from a single parent but differing in in vivo characteristics, has been developed previously. These cells represent a heterogenous population from a single tumour and a model of different stages of BlCa progression, from non-tumourigenic to invasive. Two sublines were selected for further investigation: C3 (non-tumourigenic) and B8 (invasive). These were transfected with green (C3-GSP-2) and red fluorescent reporters (B8-RSP-gck) respectively to investigate the effects of their co-injection in vivo, specifically, promotion of C3 tumour growth by B8 cells. Surprisingly, B8 tumour growth was inhibited by C3 cells in vivo at different cell numbers and proportions of cells injected. Microarray analysis of C3 and B8 cells revealed differential expression of 1367 genes with dramatic differences in the transforming growth factor-?? and integrin-mediated pathways. Gene expression of BMP2, INHBB, FST, NOG, ID4 and TGF- ??1, in the TGF- ?? pathway was further analysed with qRT-PCR in all nine sublines. Expression of BMP2 was significantly related to tumourigenic potential (p=0.0238, Mann-Whitney) and INHBB to invasive ability (p=0.0476, Mann-Whitney). The BlCa model did not include a metastatic component. To broaden the model, cell lines were established from an invaded lymph-node (B8-RSP-LN) and a bone-metastasis (B8-RSP-BN) after subcutaneous and intra-cardiac injection of B8-RSP-gck cells. No significant differences were observed in the migratory capability and anchorage-independent colony formation of these metastatic cells compared with B8 cells. Evaluation of expression of the panel of TGF-beta genes (BMP2, INHBB, FST, NOG, ID4 and TGF- ??1) and metastasis-related genes (MMP9, MMP2 and KAI1) indicated that expression of BMP2, FST, ID4 and MMP9 was decreased or lost in the metastatic sublines.
87

Characterization of bovine insulin like growth factor binding protein-2 : structure and function

Carrick, Francine Ellen. January 2001 (has links) (PDF)
Includes bibliographical references (leaves 291-311)
88

Characterization of bovine insulin like growth factor binding protein-2 : structure and function / by Francine Ellen Carrick. / Characterization of bovine insulin like growth factor binding protein two

Carrick, Francine Ellen January 2001 (has links)
Includes bibliographical references (leaves 291-311) / xxiii, 313 leaves : ill. (chiefy col.) ; 30 cm. / Title page, contents and abstract only. The complete thesis in print form is available from the University Library. / Thesis (Ph.D.)--University of Adelaide, Dept. of Molecular Biosciences, 2002
89

Studies of complexes formed in blood in vivo between an insulin-like growth factor analog and binding proteins

Gajanandana, Oraprapai. January 1997 (has links) (PDF)
Includes bibliographical references (43 leaves) This study shows that when LR3IGF-I is administered to animals in pharmacologically active doses, it may be present in either the free form or bound to IGF-binding protein(s) in the circulation. Age and nutrition which are factors that regulate synthesis of endogenous IGF-I and IGF-binding proteins, affect the in vivo formation of complexes between the analog and IGFBP(s). This study also suggests that IGFBP-1 inhibits the pharmacological activity of circulating LR3IGF-I on thymus whereas it appears to stimulate the pharmacological activity of LR3IGF-I in kidneys.
90

The Regulation of Growth Factor Signaling in Drosophila Development and Disease

Lindner, Jonathan Ryan 2010 December 1900 (has links)
Developmental signaling pathways have many diverse roles throughout the life of an organism. The proper regulation of these pathways is essential for normal development, and misregulation can lead to diseases such as cancer. Heparan sulfate proteoglycans function to modulate growth factor signaling in many biological processes by acting as co-receptors, or by influencing ligand distribution. The heparan sulfate proteoglycan Trol, the Drosophila Perlecan homolog, is known to modulate signaling in a population of neuroblasts in the developing Drosophila central nervous system. My studies aim to determine the function Trol has in regulating signaling pathways during development. trol mutants are examined to determine how various mutant alleles impact signaling in several different developmental contexts. The role growth factor pathways play during induction of a Drosophila prostate cancer model is also examined. Gene expression profiles are determined for two types of prostate model overproliferation. Trol is shown to be able to differentially regulate multiple signaling pathways during several developmental processes. The Drosophila prostate cancer model is also shown to have many characteristics similar to those of human prostate cancer, and that signaling and proteoglycan expression are impacted by aberrant overgrowth in the model. My results indicate that Trol is able to specifically modulate different signaling pathways depending on the tissue and developmental context.

Page generated in 0.0365 seconds