111 |
In vitro assessment of fertilization and embryo development with Bovine spermatozoa after scrotal insulationWalters, Anneke H. 01 December 2004 (has links)
Fertilization and cleavage of bovine embryos depend not only on maternal involvement, but also on the paternal contributions that involve more than just providing the haploid male genome. Therefore, the overall objective of this project was to determine the impact of morphologically abnormal spermatozoa on fertilization, subsequent embryonic development, and embryo quality at the cellular level. Four experiments used morphologically abnormal semen samples collected and cryopreserved from four Holstein bulls before (Pre) and after a scrotal insulation (PI) period of 48 h. Zygotes were cultured for 8 d when a developmental score was assigned to each embryo; subpopulations were subjected to either the TUNEL or caspase assays to determine apoptosis. In the final experiment pronuclear decondensation for presumptive zygotes was evaluated by differential interference contrast microscopy at 3 h time intervals from 6 to 18 h post in vitro insemination (hpi). Morphological evaluation of semen samples revealed a decrease (P < 0.01) in the percentages of normal spermatozoa in the PI samples in comparison with the Pre samples for Bulls I and Bull III (74 to 22.3% and 67.7 to 0.5 %, respectively) and the scrotal insulation effects persisted from the time of cleavage through blastocyst formation for Bulls I and III and corresponded with a similar decrease in blastocyst development for PI samples in experiment 1 regardless of which semen separation method was used. Likewise, the overall pronuclear decondensation rate for the PI zygotes of Bull I and III showed no increase over time and remained predominantly at PN1 stage (1.5 ± 0.17; 1.8 ± 0.22, respectively). In contrast, the development for Bull II and Bull IV were unaffected. The embryo quality assessment revealed that the caspase intensity increased significantly for both Bull I (217 ± 147) and Bull III (229 ± 98) for the PI embryo groups compared to those of Bull II (98 ± 115) and Bull IV (90 ± 111). In conclusion, the tested separation methods used seemed inadequate in their ability to provide potentially competent sperm for IVF. The decrease in embryonic development appears to be multifaceted and related to the changes in head shape morphology and we suggest the failure in normal pronuclear formation is associated with an absence of normal decondensation of the penetrating spermatozoon. The inability to consistently measure apoptosis in early stage embryos complicates the assessment of differences in embryo quality. These observations support the hypothesis of uncompensable seminal traits in IVF with abnormal spermatozoa and provide compelling evidence that the effect of morphologically abnormal spermatozoa occurred prior to cleavage, thus is manifested during the early stages of fertilization. / Ph. D.
|
112 |
Investigating the Effect of Ethanol on Wnt7a and its Potential Implications in Fetal Alcohol Spectrum DisorderLytle, Erika 01 January 2020 (has links)
Fetal Alcohol Spectrum Disorders (FASDs), are caused by maternal alcohol consumption during pregnancy [3]. FASD encompasses a wide variety of cardiac and neural anomalies, while also associated with improper limb development, abnormal craniofacial features, problems within the central nervous system (CNS), and disabilities in learning and communication. Gene-regulating FASDs have not been well studied during the crucial phases of early embryonic development. Genes within the Wnt/Beta-catenin pathway control a vast amount of embryonic developmental processes. Among them is the Wnt7a gene, a significant downstream gene regulator which positively controls neural stem cell proliferation and cardiomyocyte differentiation on a large scale during early embryonic development. This project will serve to provide potential insight into the genes involved in FASD. We hypothesize that ethanol administration to early embryonic mice will suppress Wnt7a expression in the heart and brain, leading to FASD development. RNA-sequencing (RNA-Seq) and real-time quantitative PCR (qPCR) were used to measure Wnt7a gene expression within the early embryonic mouse heart and brain. After evaluation of RNA-Seq data and a comparative analysis using the 2-ΔΔCTmethod, it is evident Wnt7a is present in embryonic mouse age E10.5 heart and brain samples, and Wnt7a is suppressed at age E10.5 in embryonic mouse heart, but not brain, when induced with ethanol.
|
113 |
Anatomical and extracellular matrix development of embryonic chick leg muscle in vivo and in vitroDrushel, Richard Frederick January 1993 (has links)
No description available.
|
114 |
The Role of PEA3 in Mammary Gland Development and TumorigenesisMacNeil, Lesley 09 1900 (has links)
<p> PEA3 is a member of the ets family of transcription factors. It is expressed throughout embryonic development and in mouse mammary adenocarcinomas induced by expression of the receptor tyrosine kinase Neu. Mice lacking PEA3 due to a targeted disruption of the gene, develop normally, however, male mice fail to mate for yet undetermined reasons. To further understand the role of PEA3 in mammary gland development and tumorigenesis, the effects of loss of function of PEA3 were examined in tumor formation and in mammary gland development. </p> <p> Analysis of tumor formation in PEA3 +I+ and PEA3 -/-animals failed to show a statistically significant difference in tumor onset. Loss of PEA3 did not affect the tumor morphology, nor did it inhibit metastasis of these tumors to the lung. These data indicate that PEA3 is not required for tumor formation or metastasis. </p> <p> PEA3 deficient animals displayed defects in branching morphogenesis in the mammary gland. Decreased ductal branching was observed in virgin and pregnant females. Mice with decreased levels of PEA3 expression also exhibited defects in branching morphogenesis, indicating a dosage effect. PEA3 is expressed in the myoepithelial cells during puberty and pregnancy. It is also express in the highly proliferative cap cell layer of the terminal end bud. In the embryonic mammary gland, PEA3 is expressed as early as 10.5 days in the mammary epithelium and continues late in embryogenesis. Expression in the male mammary gland is lost at approximately embryonic day 16. </p> / Thesis / Candidate in Philosophy
|
115 |
Effects of Diethylstilbestrol on Murine Early Embryonic Stem Cell Differentiation Using an Embryoid Body Culture SystemLadd, Sabine Margaret 04 May 2005 (has links)
Objectives: The effects of estrogens on immune system formation and function are well documented. Diethylstilbestrol (DES), a synthetic estrogen, has been linked to neoplasia and immune cell dysfunction in humans and animals exposed in-utero. In-vitro effects of DES exposure of murine embryonic stem (ES) cells on the early embryonic immune system development and the expression of cellular surface markers associated with common hemangioblastic and hematopoietic precursors of the endothelial, lymphoid & myeloid lineages were investigated.
Hypothesis: Early ES cell expression of CD45 a marker common to lymphoid lineage hematopoietic stem cells and differentiation of lymphoid lineage precursors are affected by in-vitro exposure to DES.
Methods: Murine ES cells were cultured using a variety of techniques: an OP9 co-culture system, and formation of embryoid bodies (EBs) in a liquid medium and hanging drop system. The OP9 co-culture system did not appear to give rise to well differentiated lymphoid lineage cells during 12 days of differentiation. The hanging drop EB culture system, previously shown to promote differentiation of endothelial and lymphoid precursor cells, was chosen for further studies of ES cell differentiation. ES cells were harvested at five time points: undifferentiated (day 0), and differentiated (days 3, 8, 12 and 16). Differentiating ES cells were treated with DES beginning on day 3. The synthetic estrogen, DES, was chosen as a treatment because of its similar potency to 17β estradiol and documented association with neoplasia in women exposed in-utero. Surface marker expression, measured by real-time RT-PCR amplification, was recorded using fluorogenic TaqMan(R) probes designed specifically for the surface proteins of interest: oct4, c-Kit, Flk1, ERα, ERβ, CD45, Flt1, & VE-cadherin.
Analysis & Results: Changes in surface marker gene expression between day 0 and day 16 of differentiation were analyzed using the RT-PCR threshold counts (CT) and the comparative threshhold cycle method. The expression of each target mRNA was normalized internally to a housekeeping gene (18s rRNA) and calculated relative to day 0. ANOVA (Type 3 fixed-effects analysis, SAS) was performed using the unexponentiated ΔΔCT values. The effects of DES, time, and the interaction between DES and time were evaluated for days 8, 12 and 16. Additionally, the effects of DES on the expression of each marker were evaluated for day 16. Expression of estrogen receptor receptor α
& β (ERα & β) in the EBs was established, and did not appear to be affected at any time by treatment with DES. ERα was expressed in significant levels on day 16, while ERβ was expressed in low levels throughout the period of differentiation. The expression of the cell surface marker, c-Kit was significantly (P<0.0001) altered by the presence of DES between the three time points sampled. The expression of the VEGF receptor, Flt1, and the adhesion molecule, VE-cadherin, markers of endothelial cells, were also significantly (P<0.026) altered by treatment with DES on day 16 of differentiation. Treatment with DES appeared to have no effect on the expression of CD45, a marker common to lymphoid precursor cells.
Conclusions: These results indicate the presence of estrogen receptors in differentiating ES cells as early as day three in-vitro (ERβ) until day 16 (ERα). DES alters expression of common hemangioblastic and hematopoietic precursor, as well as endothelial lineage markers, but has no effect on expression of the marker of lymphoid lineage development before day 16. These effects coincided with the expression of ERα. The enduring effects of DES exposure in-utero may not be manifest in this ES model, or may occur at later stages of differentiation or in selected subpopulations of CD45+ cells. / Master of Science
|
116 |
Kinase regulation of HOX transcription factorsPrimon, Monika, Hunter, K.D., Pandha, H.S., Morgan, Richard 04 October 2019 (has links)
Yes / The HOX genes are a group of homeodomain-containing transcription factors that play
important regulatory roles in early development, including the establishment of cell and tissue
identity. HOX expression is generally reduced in adult cells but is frequently re-established as an
early event in tumour formation and supports an oncogenic phenotype. HOX transcription factors
are also involved in cell cycle regulation and DNA repair, along with normal adult physiological
process including stem cell renewal. There have been extensive studies on the mechanism by which
HOX proteins regulate transcription, with particular emphasis on their interaction with cofactors
such as Pre-B-cell Leukaemia Homeobox (PBX) and Myeloid Ecotropic Viral Integration Site 1 (MEIS).
However, significantly less is known of how the activity of HOX proteins is regulated. There is
growing evidence that phosphorylation may play an important role in this context, and in this
review, we draw together a number of important studies published over the last 20 years, and discuss
the relevance of phosphorylation in the regulation and function of HOX proteins in development,
evolution, cell cycle regulation, and cancer.
|
117 |
In-vitro induction of embryonic stem cells into neural lineage through stromal cell-derived inducing activity.January 2005 (has links)
Fong Shu Pan. / Thesis (M.Phil.)--Chinese University of Hong Kong, 2005. / Includes bibliographical references (leaves 147-167). / Abstracts in English and Chinese. / ACKNOWLEDGEMENTS --- p.i / LIST OF PUBLICATIONS --- p.ii / ABSTRACT --- p.iii / ABSTRACT [IN CHINESE] --- p.vii / TABLE OF CONTENT --- p.ix / LISTS OF FIGURES --- p.xv / LIST OF TABLES --- p.xxi / LIST OF ABBREVATIONS --- p.xxii / Chapter Chapter 1 --- Introduction --- p.1 / Chapter 1.1 --- Embryonic stem (ES) cells --- p.1 / Chapter 1.2 --- Stem cell plasticity --- p.5 / Chapter 1.2.1 --- Differentiation and trans-differentiation of lineage-restricted stem cells --- p.5 / Chapter 1.2.1.1 --- Multilineage differentiation in-vitro --- p.5 / Chapter 1.2.1.2 --- Trans-differentiation --- p.6 / Chapter 1.2.2 --- Prospective applications of stem cells --- p.7 / Chapter 1.2.2.1 --- Basic research on development --- p.7 / Chapter 1.2.2.2 --- Study of human disease --- p.7 / Chapter 1.2.2.3 --- Cancer research --- p.7 / Chapter 1.2.2.4 --- Drug screening --- p.8 / Chapter 1.2.2.5 --- Cell therapy --- p.8 / Chapter 1.3 --- Neuro-degenerative diseases and cell therapy --- p.9 / Chapter 1.3.1 --- Neuro-degenerative diseases --- p.9 / Chapter 1.3.2 --- Neuro-regeneration --- p.10 / Chapter 1.3.3 --- Cell sources for neuro-regenerative therapy --- p.11 / Chapter 1.3.3.1 --- Comparison of stem cells --- p.11 / Chapter 1.3.3.2 --- Stem cells in neuro-regenerative therapy --- p.12 / Chapter 1.4 --- In-vitro derivation into neural lineage --- p.17 / Chapter 1.4.1 --- In-vitro induction strategies available --- p.17 / Chapter 1.4.1.1 --- Chemical agents --- p.18 / Chapter 1.4.1.1.1 --- Retinoic acid (RA) --- p.18 / Chapter 1.4.1.1.2 --- Ascorbic acid --- p.19 / Chapter 1.4.1.2 --- Growth factors/cytokines --- p.19 / Chapter 1.4.1.2.1 --- Neurotrophins --- p.20 / Chapter 1.4.1.2.2 --- Stimulants --- p.20 / Chapter 1.4.1.2.3 --- Signalling molecules --- p.21 / Chapter 1.4.1.3 --- Culture Selection --- p.23 / Chapter 1.4.1.3.1 --- Conditions --- p.23 / Chapter 1.4.1.3.2 --- Medium --- p.23 / Chapter 1.4.1.4 --- Transfection of regulator genes using viral vector --- p.24 / Chapter 1.4.1.5 --- Stromal cell-derived inducing activity (SDIA) --- p.26 / Chapter Chapter 2 --- Aims --- p.28 / Chapter 2.1 --- Hypothesis and study objectives --- p.28 / Chapter 2.1.1 --- Soliciting an optimal method for ES cell propagation --- p.28 / Chapter 2.1.2 --- Pursuing alternative SDIA --- p.29 / Chapter Chapter 3 --- Materials and Methods --- p.33 / Chapter 3.1 --- Chemicals and Reagents --- p.33 / Chapter 3.1.1 --- Cell Culture --- p.33 / Chapter 3.1.2 --- Immunohistochemistry and staining --- p.35 / Chapter 3.1.3 --- Molecular Biology --- p.36 / Chapter 3.2 --- Consumable --- p.37 / Chapter 3.3 --- Cell lines --- p.39 / Chapter 3.3.1 --- Feeder cells --- p.39 / Chapter 3.3.1.1 --- Primary mouse embryonic fibroblasts --- p.39 / Chapter 3.3.1.2 --- STO --- p.39 / Chapter 3.3.1.3 --- L Cells --- p.40 / Chapter 3.3.1.4 --- L-Wnt-3A Cells --- p.40 / Chapter 3.3.1.5 --- C17.2 --- p.40 / Chapter 3.3.2 --- ES cells --- p.41 / Chapter 3.3.2.1 --- ES-D3 --- p.41 / Chapter 3.3.2.2 --- ES-E14TG2a --- p.41 / Chapter 3.4 --- In-house prepared solutions --- p.42 / Chapter 3.4.1 --- "Stock solution of Insulin, Transferrin, Selentine (ITS) Supplement" --- p.42 / Chapter 3.4.2 --- Enriched Knock-Out Dulbecco's Modified Eagle's Medium (KO DMEM) --- p.42 / Chapter 3.4.3 --- Mitomycin C solution --- p.42 / Chapter 3.4.4 --- Gelatin solution 0.1% --- p.42 / Chapter 3.4.5 --- p-mercaptoethanol solution --- p.43 / Chapter 3.4.5.1 --- (3-mercaptoethanol solution 0.1M --- p.43 / Chapter 3.4.5.2 --- P-mercaptoethanol solution 0.1M --- p.43 / Chapter 3.4.5.3 --- p-mercaptoethanol solution 0.1M for preparation of culture medium --- p.43 / Chapter 3.4.6 --- ALL-trans retinoic acid --- p.43 / Chapter 3.4.6.1 --- ALL-trans retinoic acid stock solution 0.01M --- p.43 / Chapter 3.4.6.2 --- ALL-trans retinoic acid working solution lμM --- p.43 / Chapter 3.4.7 --- Paraformaldehyde solution 4% (PFA) --- p.44 / Chapter 3.4.8 --- TritoxX-100 solution --- p.44 / Chapter 3.4.8.1 --- Tritox X-100 solution 3% --- p.44 / Chapter 3.4.8.2 --- Tritox X-100 solution 0.3% --- p.44 / Chapter 3.4.9 --- Popidium iodide solution lug/mL (PI) --- p.44 / Chapter 3.4.10 --- Geneticin solution --- p.45 / Chapter 3.4.10.1 --- Geneticin solution 50mg/mL --- p.45 / Chapter 3.4.10.2 --- Geneticin solution 5mg/mL --- p.45 / Chapter 3.4.11 --- Poly-L-ornithine solution --- p.45 / Chapter 3.4.12 --- Laminin solution --- p.45 / Chapter 3.4.13 --- Maintenance medium for cell feeders --- p.46 / Chapter 3.4.14 --- Mitomycin C inactivation medium --- p.46 / Chapter 3.4.15 --- Freezing medium --- p.46 / Chapter 3.4.16 --- Propagation medium for ES cells --- p.47 / Chapter 3.4.16.1 --- Serum-based propagation medium for ES cells --- p.47 / Chapter 3.4.16.2 --- Serum-free propagation medium for ES cells --- p.47 / Chapter 3.4.16.3 --- Serum-free induction medium for ES cells --- p.48 / Chapter 3.4.16.3.1 --- Serum-free induction medium 1 --- p.48 / Chapter 3.4.16.3.2 --- Serum-free induction medium II --- p.48 / Chapter 3.4.16.3.3 --- Serum-free induction medium III --- p.48 / Chapter 3.5 --- Equipments --- p.49 / Chapter 3.6 --- Methods --- p.50 / Chapter 3.6.1 --- Cell Culture --- p.50 / Chapter 3.6.1.1 --- Preparation of round cover-slips --- p.50 / Chapter 3.6.1.2 --- Gelatinization of tissue culture wares --- p.51 / Chapter 3.6.1.3 --- Poly-L-ornithine and laminin coating --- p.51 / Chapter 3.6.1.4 --- Thawing frozen cells --- p.51 / Chapter 3.6.1.5 --- Passage of adherent culture --- p.52 / Chapter 3.6.1.6 --- Cell count --- p.52 / Chapter 3.6.1.7 --- Cytospin --- p.53 / Chapter 3.6.1.8 --- Cell viability test --- p.53 / Chapter 3.6.1.9 --- Cryopreservation --- p.53 / Chapter 3.6.1.10 --- Preparation of primary mouse embryonic fibroblast (PMEF) --- p.54 / Chapter 3.6.1.11 --- Mitomycin C inactivation of feeder cells --- p.55 / Chapter 3.6.1.12 --- Gamma irradiation of various feeders --- p.55 / Chapter 3.6.1.13 --- Preparation of CM from feeder cells --- p.56 / Chapter 3.6.1.14 --- Propagation of ES cells in serum-based medium --- p.56 / Chapter 3.6.1.15 --- Propagation of ES cell in serum-free medium --- p.56 / Chapter 3.6.1.16 --- Neural differentiation using all-trans retinoic acid --- p.57 / Chapter 3.6.1.17 --- Stromal cells-derived inducing activity --- p.58 / Chapter 3.6.1.18 --- BrdU labeling of the cell products --- p.59 / Chapter 3.6.2 --- Molecular analysis --- p.60 / Chapter 3.6.2.1 --- RNA extraction --- p.60 / Chapter 3.6.2.2 --- RNA quantitation --- p.60 / Chapter 3.6.2.3 --- Reverse Transcription of the First Strand complementary DNA --- p.61 / Chapter 3.6.2.4 --- Polymerase chain reaction --- p.61 / Chapter 3.6.2.5 --- RNA Integrity Check --- p.66 / Chapter 3.6.2.6 --- Electrophoresis and visualization of gene products --- p.66 / Chapter 3.6.3 --- Immunofluoresent staining --- p.66 / Chapter 3.6.4 --- In-vivo studies --- p.69 / Chapter 3.6.4.1 --- Induction of cerebral ischaemia in mice --- p.69 / Chapter 3.6.4.2 --- Transplantation --- p.69 / Chapter 3.6.4.3 --- Assessment of learning ability and memory --- p.70 / Chapter 3.6.5 --- Histological analysis --- p.70 / Chapter 3.6.5.1 --- Animal sacrifice for brain harvest --- p.70 / Chapter 3.6.5.2 --- Cryosectioning --- p.71 / Chapter 3.6.5.3 --- Paraffin sectioning --- p.71 / Chapter 3.6.5.4 --- Haematoxylin and eosin staining --- p.72 / Chapter 3.7 --- Data analysis --- p.73 / Chapter Chapter 4 --- Results --- p.74 / Chapter 4.1 --- ES cell maintenance --- p.74 / Chapter 4.1.1 --- Serum effect --- p.74 / Chapter 4.1.2 --- Feeder effect --- p.79 / Chapter 4.1.3 --- Serum-free and feeder-free condition --- p.86 / Chapter 4.1.4 --- Overall effect --- p.89 / Chapter 4.2 --- ES cell Induction --- p.91 / Chapter 4.2.1 --- Retinoic acid --- p.91 / Chapter 4.2.2 --- Stromal cell-derived inducing activity --- p.96 / Chapter 4.2.2.1 --- Molecular characterization of candidate stromal cells --- p.96 / Chapter 4.2.2.2 --- Direct contact co-culture --- p.98 / Chapter 4.2.2.3 --- Non-contact co-culture --- p.100 / Chapter 4.2.2.4 --- Cultures in CM --- p.109 / Chapter 4.3. --- ES cell Differentiation --- p.115 / Chapter 4.4 --- In vivo study of ES cell-derived cell products --- p.117 / Chapter 4.4.1 --- Animal preparation --- p.117 / Chapter 4.4.2 --- Cell preparation --- p.117 / Chapter 4.4.3 --- Cell implantation --- p.117 / Chapter 4.4.4 --- Behaviour Monitoring --- p.121 / Chapter 4.4.5 --- Histology of cell-implanted brain --- p.125 / Chapter Chapter 5 --- Discussion --- p.129 / Chapter Chapter 6 --- Conclusion --- p.144 / References --- p.147
|
118 |
Targeted differentiation of embryonic stem cells towards the neural fate. / CUHK electronic theses & dissertations collectionJanuary 2009 (has links)
Embryonic stem (ES) cells, which possess proliferating and differentiating abilities, are a potential source of cells for regenerative medicine. Nowadays, the challenge in using ES cells for developmental biology and regenerative medicine has been to direct the wide differentiation potential towards the derivation of a specific cell fate. This study is aimed to establish a simple and efficient method to derive ES cells into neural lineage cells and examine the safety and efficacy of derived cells in a mouse ischemic stroke model. To explore the underlying mechanisms responsible for lineage commitment of stem cells, Notch signaling and serotonin responses are also studied. / In a non-contact coculture system, mouse ES cells (D3 and E14TG2a) were cocultured with the stromal cells MS5 for eight days. On the other hand, human ES cells (H9 and H14) were directly cocultured with MS5 in a contact manner for two weeks. Derived cells were further propagated in a serum-free medium and selected subsequently in a differentiating medium. The cell viability, numbers, phenotypes and lineage-specific gene expression profile were evaluated at stages of induction, propagation and selection. / In vivo, behavioral assessments of ischemic mice after transplantation of mouse ES cell derivatives revealed a significant improvement in spatial learning and memory ability as compared to ischemic mice without cell therapy. Histology of brain sections of transplanted mice demonstrated the migration of BrdU+ cells to the CA1 region of the hippocampus, which was evident of both an increase of pyramidal neuron density and normalized morphology. Teratoma development was found in one out of 17 transplanted mice. / MS5 was noted to express genes encoding neurotrophins and neuroprotective factors. Functional tests showed that MS5 exerted neurotrophism on neuroblastoma cell lines (SK-N-AS, SH-SY5Y, and SK-N-MC) and ES cells. The numbers of viable cells and the proportion of neural subtypes derived from ES cells at three stages of the culture system were significantly higher than those of the control cultures without MS5 induction, respectively. MS5 cocultures generate a relatively higher yield of neural lineage cells but select against the mesodermal and endodermal lineage derivatives. Together with non-contact MS5 coculture, serotonin could further increase the proportion of neural precursors and accelerate maturation of neural progenitor cells in a synergistic manner. During the induction phase with non-contact MS5 coculture, the Notch inhibitor could significantly decrease the number of derived neural precursors and instigate non-neural differentiation. With the supplement of the Notch inhibitor, serotonin could neither promote the expression of neuroectodermal genes nor enhance the proportion of neural precursors in MS5-cocultured ES cells. Notably, in the propagation of undifferentiated human ES cells, Notch signaling was also found to play an active role in maintaining cell survival. / The Notch inhibitor (gamma-secretase inhibitor) and serotonin were supplemented into induction cultures to investigate the roles of Notch signaling and the neurotransmitter serotonin in neural differentiation. For in vivo study, mouse ES cell-derived cells were labeled with BrdU and implanted onto the caudate putamens of mice having undergone transient occlusion of bilateral common carotid arteries and reperfusion to induce cerebral ischemia. Spatial learning and memory ability of transplanted mice were assessed in a water maze system. Histological assessment was also conducted on brain sections of mice three weeks post transplant to examine the migration and homing of implanted cells. / This study describes a simple and efficient differentiation protocol to derive mouse ES cells and human ES cells into neural lineage cells. Derived cells appear to significantly improve cognitive functions in a mouse ischemic stroke model. Data of the study suggest that MS5 cells may exert a neurotrophic effect on ES cells. With MS5 coculture, serotonin synergistically promotes neural commitment and facilitates maturation of derived neural precursors in ES cell cultures. In contact coculture with MS5, Notch signaling is shown to play a role in the directed neural differentiation of human ES cells, whereas in maintenance culture, Notch signaling is also important to cell survival of human ES cells. Thus, Notch signaling through cell-cell interaction may explain, at least partially, the difference between mouse ES cells and human ES cells in cell growth ability when seeded at low cell densities. / Yang Tao. / Adviser: Ho Keung Ng. / Source: Dissertation Abstracts International, Volume: 70-09, Section: B, page: . / Thesis submitted in: November 2008. / Thesis (Ph.D.)--Chinese University of Hong Kong, 2009. / Includes bibliographical references (leaves 161-194). / 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, [200-] System requirements: Adobe Acrobat Reader. Available via World Wide Web. / Abstracts in English and Chinese. / School code: 1307.
|
119 |
Purification of cardiomyocytes derived from differentiated embryonic stem cells and study of the cytokines' effect on embryonic stem cell differentiation.January 2008 (has links)
Leung, Sze Lee Cecilia. / Thesis (M.Phil.)--Chinese University of Hong Kong, 2008. / Includes bibliographical references (leaves 144-153). / Abstracts in English and Chinese. / Abstract --- p.i / Abstract in Chinese (摘要) --- p.iii / Acknowledgements --- p.v / Table of Content --- p.vi / Abbreviations --- p.xv / Chapter CHAPTER 1 --- INTRODUCTION / Chapter 1.1 --- Stem cells --- p.1 / Chapter 1.1.1 --- Adult stem cells --- p.2 / Chapter 1.1.2 --- Embryonic stem cells --- p.2 / Chapter 1.1.3 --- Pros and cons of embryonic and adult stem cells --- p.5 / Chapter 1.1.4 --- Human embryonic stem cells (hESCs) --- p.6 / Chapter 1.1.5 --- Mouse embryonic stem cells (mESCs) --- p.7 / Chapter 1.1.6 --- Characteristics of ESC-derived cardiomyocytes --- p.7 / Chapter 1.2 --- Cardiovascular Diseases (CVD) --- p.9 / Chapter 1.2.1 --- Causes and statistics of CVD --- p.9 / Chapter 1.2.2 --- Current treatment for CVD --- p.10 / Chapter 1.2.3 --- Current hurdles of putting hESC-CMs into clinical use --- p.11 / Chapter 1.3 --- Myosin light chain2v --- p.13 / Chapter 1.4 --- Genetic-engineering of hESCs & their cardiac derivatives by lentiviral-mediate gene transfer --- p.14 / Chapter 1.5 --- Cytokines secretion during myocardial infarction --- p.15 / Chapter 1.6 --- Aims of the Project --- p.19 / Chapter 1.7 --- Significance of the Project --- p.19 / Chapter CHAPTER 2 --- MATERIALS AND METHODS / Chapter 2.1 --- Subcloning --- p.20 / Chapter 2.1.1 --- Amplification of MLC-2v --- p.20 / Chapter 2.1.2 --- Purification of DNA product --- p.21 / Chapter 2.1.3 --- Restriction enzyme digestion --- p.21 / Chapter 2.1.4 --- Ligation of MLC-2v promoter with DuetO 11 vector --- p.22 / Chapter 2.1.5 --- Transformation of ligation product into competent cells --- p.22 / Chapter 2.1.6 --- PCR confirmation of successful ligation --- p.23 / Chapter 2.1.7 --- Small-scale preparation of bacterial plasmid DNA --- p.23 / Chapter 2.1.8 --- Restriction enzyme digestions to reconfirm positive clones --- p.24 / Chapter 2.1.9 --- DNA sequencing of the cloned plasmid DNA --- p.25 / Chapter 2.1.10 --- Large-scale preparation of target recombinant expression vector --- p.25 / Chapter 2.2 --- Mouse Embryonic Fibroblast (MEF) Culture --- p.26 / Chapter 2.2.1 --- Derivation of MEF --- p.26 / Chapter 2.2.2 --- Mouse embryonic fibroblast cells culture --- p.27 / Chapter 2.2.3 --- Irradiation of mouse embryonic fibroblast --- p.28 / Chapter 2.3 --- HESC culture --- p.29 / Chapter 2.3.1 --- Thawing and Plating hESCs --- p.29 / Chapter 2.3.2 --- Splitting hESCs --- p.30 / Chapter 2.3.3 --- "Culture maintainence, selection and colony removal" --- p.31 / Chapter a) --- Distinguish differentiated and undifferentiated cells and colonies / Chapter b) --- "Remove differentiated cells by ""Picking to Remove""" / Chapter c) --- "Remove undifferentiated cells by ""Picking to Keep""" / Chapter 2.3.4 --- Freezing hESCs --- p.31 / Chapter 2.3.5 --- Differentiation of hESCs --- p.32 / Chapter 2.3.6 --- "HESC culture on feeder free system, mTeSR TM1" --- p.34 / Chapter a) --- Preparation of mTeSRTMl / Chapter b) --- Preparation of BD MatrigelTM hESC-qualified Matrix aliquots / Chapter c) --- Coating plates with BD MatrigelTM hESC-qualified Matrix / Chapter d) --- Human Embryonic stem cells culture in mTeSRTMl / Chapter 2.4 --- ES Cell Characterization (Chemicon Cat# SCR001) --- p.36 / Chapter 2.4.1 --- Alkaline Phosphatase Staining --- p.36 / Chapter 2.4.2 --- Immunofluorescence staining --- p.37 / Chapter 2.5 --- MESC culture --- p.38 / Chapter 2.5.1 --- Thawing and Plating mESCs --- p.38 / Chapter 2.5.2 --- Splitting mESCs --- p.38 / Chapter 2.5.3 --- Differentiation of mESCs --- p.39 / Chapter 2.5.4 --- To study the effects of cytokines on mESC differentiation --- p.40 / Chapter 2.6 --- Lentivirus (LV) Packaging --- p.41 / Chapter 2.6.1 --- Transfection of lentiviral vectors into HEK293FT cells --- p.41 / Chapter 2.6.2 --- LV titering --- p.42 / Chapter 2.7 --- MultipleTransduction --- p.43 / Chapter 2.8 --- Selection of transduced cells by hygromycin --- p.43 / Chapter 2.8.1 --- Determination of hygromycin selection dosage --- p.43 / Chapter 2.8.2 --- Selection of stable clones --- p.44 / Chapter 2.9 --- Isolation of green fluorescent cardiomyocytes derived from differentiated hESCs --- p.45 / Chapter 2.9.1 --- Collagenase digestion of embryoid bodies into single cells --- p.45 / Chapter 2.9.2 --- FACS --- p.46 / Chapter 2.10 --- Gene expression study / Chapter 2.10.1 --- Primer design --- p.46 / Chapter 2.10.2 --- RNA extraction --- p.46 / Chapter 2.10.3 --- DNase Treatment --- p.47 / Chapter 2.10.4 --- Synthesis of Double-stranded cDNA from Total RNA --- p.47 / Chapter 2.10.5 --- Quantitative real-time PCR --- p.48 / Chapter 2.10.6 --- Quantification of mRNA expression --- p.49 / Chapter 2.11 --- Protein Expression study --- p.49 / Chapter 2.11.1 --- Crude protein extraction --- p.49 / Chapter 2.11.2 --- Quantitation of protein samples --- p.50 / Chapter 2.11.3 --- SDS-PAGE --- p.50 / Chapter 2.11.4 --- Western Blot --- p.51 / Chapter 2.11.5 --- Western blot luminal detection --- p.52 / Chapter 2.11.6 --- Quantification of protein expression --- p.52 / Chapter CHAPTER 3 --- PURIFICATION OF CARDIOMYOCYTES DERIVED FROM DIFFERENTIATED HESCs / Chapter 3.1 --- Subcloning --- p.57 / Chapter 3.1.1 --- Linearization of DuetO11 and excision of UBC promoter --- p.58 / Chapter 3.1.2 --- PCR cloning of MLC-2V --- p.59 / Chapter 3.1.3 --- Ligation of MLC-2v promoter to linearized DuetO11 --- p.60 / Chapter 3.1.3.1 --- Colony PCR to screen for positive clones --- p.61 / Chapter 3.1.3.2 --- Restriction digestion to confirm the success of ligation --- p.61 / Chapter 3.2 --- Lentivirus (LV) packaging --- p.62 / Chapter 3.2.1 --- Transfection --- p.63 / Chapter 3.2.2 --- LV titering --- p.64 / Chapter 3.3 --- HESC culture --- p.66 / Chapter 3.4 --- Multi-transduction of hESCs with LVs --- p.67 / Chapter 3.5 --- Differentiation after transduction --- p.69 / Chapter 3.6 --- Antibiotic selection --- p.71 / Chapter 3.6.1 --- Characterization of hESCs on feeder free system --- p.72 / Chapter 3.6.1.1 --- Alkaline Phosphatase (AP) staining --- p.72 / Chapter 3.6.1.2 --- Immunostaining with pluripotency marker --- p.73 / Chapter 3.6.2 --- Determination of hygromycin dosage by MTT assay --- p.74 / Chapter 3.6.3 --- HESCs after selection in feeder free system --- p.75 / Chapter 3.7 --- Differentiation of hESCs after selection --- p.76 / Chapter 3.8 --- FACS --- p.77 / Chapter 3.9 --- QPCR of cells after FACS --- p.80 / Chapter 3.9.1 --- Gene expression of Nkx2.5 --- p.81 / Chapter 3.9.2 --- Gene expression of c-Tnl --- p.82 / Chapter 3.9.3 --- Gene expression of c-TnT --- p.83 / Chapter 3.9.3 --- Gene expression of MLC-2v --- p.84 / Chapter CHAPTER 4 --- THE STUDY OF CYTOKINES' EFFECT ON MESC DIFFERENTIATION / Chapter 4.1 --- mESC culture --- p.85 / Chapter 4.2 --- The effect of cytokines on the differentiation of mESCs --- p.86 / Chapter 4.2.1 --- Beating curves of mESCs treated with different concentrations of cytokines at differentiation day 2 to 6 before attachment --- p.88 / Chapter 4.2.2 --- qPCR to determine the cytokines' effect on the differentiation of mESCs --- p.94 / Chapter 4.2.2.1 --- The effect of IL-1α on the expression of cardiac specific genes --- p.95 / Chapter 4.2.2.2 --- The effect of IL-1β on the expression of cardiac specific genes --- p.98 / Chapter 4.2.2.3 --- The effect of IL-6 on the expression of cardiac specific genes --- p.101 / Chapter 4.2.2.4 --- The effect of IL-10 on the expression of cardiac specific genes --- p.104 / Chapter 4.2.2.5 --- The effect of IL-18 on the expression of cardiac specific genes --- p.107 / Chapter 4.2.2.6 --- The effect of TNF-α on the expression of cardiac specific genes --- p.110 / Chapter 4.2.3 --- Western blot analysis of the cytokines' effect on the differentiation of mESCs --- p.113 / Chapter 4.2.3.1 --- The effect of IL-lα on the abundance of cardiac specific proteins --- p.114 / Chapter 4.2.3.2 --- The effect of IL-1β on the abundance of cardiac specific proteins --- p.116 / Chapter 4.2.3.3 --- The effect of IL-6 on the abundance of cardiac specific proteins --- p.118 / Chapter 4.2.3.4 --- The effect of IL-10 on the abundance of cardiac specific proteins --- p.120 / Chapter 4.2.3.5 --- The effect of IL-18 on the abundance of cardiac specific proteins --- p.122 / Chapter 4.2.3.6 --- The effect of TNF-α on the abundance of cardiac specific proteins --- p.124 / Chapter CHAPTER 5 --- DISCUSSION / Chapter 5.1 --- Purification of cardiomyocytes derived from differentiated hESCs --- p.127 / Chapter 5.2 --- Study on the effect of cytokines on mESC differentiation --- p.135 / Chapter 5.3 --- Conclusion --- p.142 / REFERENCES --- p.144
|
120 |
Verfassungs- und europarechtliche Probleme im Stammzellgesetz (StZG) /Chong, Mun-sik. January 2005 (has links)
Thesis (doctoral)--Humboldt-Universiẗat, Berlin, 2005. / Includes bibliographical references (p. 231-260) and index.
|
Page generated in 0.063 seconds