• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 353
  • 315
  • 148
  • 52
  • 36
  • 36
  • 19
  • 19
  • 12
  • 6
  • 4
  • 3
  • 3
  • 3
  • 3
  • Tagged with
  • 1131
  • 744
  • 739
  • 422
  • 243
  • 221
  • 219
  • 209
  • 196
  • 189
  • 168
  • 156
  • 144
  • 129
  • 129
  • 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

The role of cultured chondrocytes and mesenchymal stem cells in the repair of acute articular cartilage injuries

Secretan, Charles Coleman 06 1900 (has links)
Osteoarthritis (OA) is a disease that has significant individual, social, and economic impact worldwide. Although many etiologies lead to the eventual development of OA, one potentially treatable cause is the acute articular cartilage (AC) injury. These injuries are common and have a poor inherent healing capacity, leading to the formation of OA. In an effort to repair AC injuries several treatment strategies have been developed but none have proven completely successful. Studies examining AC tissue-engineering strategies have suggested that those with the most potential for success involve the introduction of autogenous or allogenous cells to the site of injury. These strategies are designed to encourage creation of a matrix with the appropriate characteristics of normal AC. However, development of a completely successful repair method has proven difficult because the biomechanical properties of normal AC are not easy to replicate, a cell source with the appropriate functional characteristics has not been optimized, and the problem of effective incorporation of a repair construct into the host tissue remains unresolved. In an effort to more fully understand the cartilage repair process, this work first focused on the development and utilization of an in vitro human explant model of AC to study the ability of seeded human chondrocytes to integrate into an AC defect. Further work elucidated the gene expression patterns of cultured adult human chondrocytes and human mesenchymal stem cell (MSC)-derived chondrocytes. Results from this work determined that cultured human chondrocytes were able to adhere to articular cartilage defects in a viable in vitro explant model and produce a matrix containing collagen type II. However, further work with the in vitro expanded chondrocytes revealed that these cells have increased expression of collagen type I which promotes the formation of a less durable fibrocartilagenous tissue. This unfavorable expression persisted despite placing the chondrocytes in an environment favoring a chondrocytic phenotype. Further work with MSC-derived chondrocytes demonstrated a similar and unfavorable production of collagen type I. This work represented an important first step towards a treatment for acute AC lesions but it is clear that further work to optimize the culture microenvironment is still required. / Experimental Surgery
82

Cellular approach for the treatment of amyotrophic lateral sclerosis using adult mesenchymal stem cells

Boucherie, Cédric 12 December 2008 (has links)
Amyotrophic lateral sclerosis (ALS) is a progressive, lethal, degenerative disorder of the CNS. The hallmark of this disease is the premature and selective death of upper and lower motor neurons (MNs) in the brain and spinal cord, leading to fatal paralysis. Although the archetypal vision of neurotoxicity in neurodegenerative diseases is based on the idea that a specific neuronal population is particularly vulnerable to a cumulative toxic event (protein aggregation, mitochondria dysfunction, compromised axonal transport etc…), experimental evidence illustrate that ALS possibly does not arise strictly from damage within MNs. There is now convincing data supporting a non-cell autonomous mechanism in which neurodegeneration is influenced by the toxicity of non-neuronal cells in the vicinity of neurons such as astrocytes and microglia. Considering the accumulation of data implicating astrocytes in the pathogenesis of ALS (loss of GLT-1, secretion of toxic factor, enhanced inflammation, etc…), approaches aiming at replacing astrocytes at site of lesions constitute promising therapeutic strategies. Rapid progresses in the characterization of adult stem cell biology have generated considerable enthusiasm for the development of therapeutic strategies for CNS insults. Several observations support the hypothesis that stem cells may display a valuable influence on diseased host tissues by exerting a protective “chaperone” effect to neurons after differentiation in glial cells. Hence, we decided to study the neuroprotective potential of adult mesenchymal stem cells (MSCs) in ALS. In contrast to neural stem cells (NSCs) which localization in the central nervous system complicates their isolation, MSCs are easily isolated from the bone marrow. The relevance of using on MSCs in stem cell therapies of neurodegenerative disorders is also justified by their capacity to (trans)differentiate into neural cells. For this purpose, we exposed MSCs to growth factors involved in the astroglial differentiation of NSCs. The differentiation of MSCs was characterized by the acquisition of astrocyte morphology in addition to an increased expression of gene related to NSCs (nestin) and astrocytes (glutamine synthetase). The astroglial differentiation of MSCs is associated with the acquisition of a glial-like specific regulation of the production of GDNF, a potent neurotrophic factor for neurons. Then, we characterized the glutamate uptake in differentiated MSCs, a critical function of astrocytes. Our data demonstrate that the differentiation of MSCs is associated with an increased expression of the high affinity glutamate transporter, GLT-1. Thus, our in vitro results confirm the astrocytic differentiation potential of MSCs and we decided to use then in stem cell therapy of ALS. Indeed, we demonstrated that mechanism of stem cell recruitment is present in the spinal cord during the development of the disease by the secretion of stem cell factor (SCF). We injected MSCs derived from healthy animals into the cerebrospinal fluid of a transgenic rat model of familial ALS (expressing a mutated form of the human superoxide dismutase-1, hSOD1G93A) at disease onset. MSCs were found to infiltrate the nervous parenchyma and migrate substantially into the ventral grey matter by interacting with the SCF. At the site of lesion, MSCs differentiated massively into astrocytes around MNs. The intrathecal delivery of MSCs preserved motor functions and extended the survival of hSOD1G93A rats. Investigation of the lumbar spinal cord 35 days after graft demonstrated that the generation of healthy astrocytes from MSCs decreased motor neuron loss. However, this beneficial effect is not related to a decreased excitotoxicity by the rescue of GLT-1 expression but rather a decreased inflammation around MNs. Together, the data presented in this thesis highlight the protective capacity of adult MSC-derived astrocytes in the treatment of ALS.
83

Development of Osteoinductive Tissue Engineering Scaffolds with a Bioreactor

Thibault, Richard 24 July 2013 (has links)
The conventional treatments for craniofacial bone defects currently are unsatisfactory due to several drawbacks. Replacement of lost bone by autografts typically causes donor site morbidity while allografts, xenografts, and demineralized bone matrix all have a chance of disease transmission. Current synthetic implants placed within the defect site generally lack osseointegration and biodegradability. There are several methods of generating a hybrid extracellular matrix (ECM) and synthetic material construct. These include coating the synthetic material scaffold with collagen and calcium phosphate, incorporating acellular biological tissue within the scaffold material, and using cells to generate an ECM coating on the synthetic material scaffold. The research performed for this thesis developed and characterized mesenchymal stem cell (MSC)-generated extracellular matrix poly(ε-caprolactone) constructs (PCL/ECM) for the replacement of bone tissue. The osteogenic potential of the PCL/ECM constructs was explored by culturing i) MSCs and ii) whole marrow cells combined with MSCs onto the construct with or without the osteogenic differentiation supplement, dexamethasone. It was established that the osteogenic differentiation of MSCs seeded onto ECM-containing constructs was maintained even in the absence of dexamethasone and that the co-culture of MSCs and whole bone marrow cells without dexamethasone and ECM enhances the proliferation of a cell population (or populations) present in the whole bone marrow. The osteogenicity of the constructs encouraged the characterization of the protein and mineral composition of the ECM coating on the PCL/ECM constructs. Characterization revealed that at short culture durations the MSCs used to generate the ECM deposited cellular adhesion proteins that are a prerequisite protein network for further bone formation. At the later culture durations, it was determined that the ECM was composed of collagen 1, hydroxyapatite, matrix remodeling proteins, and regulatory proteins. The prior studies on the PCL/ECM constructs persuaded exploration of the effect of various devitalization and demineralization processes on the retention of the ECM components within and the osteogenicity of the PCL/ECM constructs. Analysis demonstrated that the freeze-thaw technique is a milder method of devitalization of cell-generated ECM constructs as compared to other methods, but it reduced the osteogenicity of the constructs. In addition, it was elucidated that void spaces in the surface of the constructs are important for allowing access of MSCs into the interior of the constructs.
84

Controlled In Vivo Mechanical Stimulation of Bone Repair Constructs

Duty, Angel Osborne 12 April 2004 (has links)
Bone grafts are used to treat more than 300,000 fracture patients yearly, as well as patients with congenital defects, bone tumors, and those undergoing spinal fusion. Given the established limitations of autograft and allograft bone, there is a substantial need for bone graft substitutes. Tissue engineering strategies employing the addition of osteogenic cells and/or osteoinductive factors to porous scaffolds represent a promising alternative to traditional bone grafts. While many bone defects are in load-bearing sites, very little is known about the response of bone grafts and their substitutes to mechanical loading, despite vast documentation on the ability of normal bone to adapt to its mechanical environment. The goal of this research was to quantify the effects of controlled in vivo mechanical stimulation on bone graft repair and bone graft substitutes and identify the local stress/strain environment associated with load-induced changes in bone formation. The global hypothesis that cyclic in vivo mechanical loading improves mineralized matrix formation within bone grafts and bone graft substitutes was addressed in this work using orthotopic and ectopic models specifically designed to facilitate modeling of local stresses and strains. In the first study, a bone defect repair model utilizing an orthotopic implant capable of supplying a controlled mechanical stimulus to a trabecular allograft showed a significant reduction in new bone formation with controlled in vivo mechanical loading. Although the reason remains unclear, loading conditions may not have been ideal for increased bone formation or potential micromotion may have influenced the results. A second study demonstrated for the first time that controlled in vivo mechanical stimulation enhances mineralized matrix production on a mesenchymal stem cell-seeded polymeric construct using a novel subcutaneous implant system. In addition, the local stresses and strains associated with this adaptive response were predicted. The novel subcutaneous implant represents technology which may be adapted for the preparation of tissue-engineered bone constructs, capitalizing on the benefits of mechanical loading and a vascularized in vivo environment. Such an approach may produce larger, stronger, and more homogeneous constructs than could be developed in a static culture system subject to diffusional limitations.
85

The Role of a Novel Gene ROGDI in Bleomycin-induced Pulmonary Fibrosis

Chang, Ching-Hung 01 August 2012 (has links)
ROGDI, a novel gene, locates on human¡¦s chromosome 16p13.3. According to Gene Ontology Annotation database, ROGDI is related to hemopoiesis and positive regulation of cell proliferation. In order to investigate the function of this novel gene in pulmonary fibrosis, fibrotic models in vivo and in vitro were created. Mice which received single intra-tracheal bleomycin injection were sacrificed on various intervals. Rogdi and other pro-fibrotic mediators, including CCL2 and TGF-£]1, were up-regulated in the early phase(< 10 days). On contrary, the anti-fibrotic mediators IL-10, IFN-£^ and heme oxygenase(HO)-1 were up-regulated in the late phase(> 10 days). The precursor microRNA 21 (miR-21) was up-regulated as the fibrotic severity increased. The human embryonic fibroblasts(WI-38 cells) showed fibrogenic phenotype and up-regulation of precursor miR-21 and ROGDI after bleomycin treatment. Human embryonic fibroblasts transfected by coding sequence of ROGDI showed up-regulated precursor miR-21 and £\-SMA compared to those transfected by empty vectors after bleomycin treatment. Two signaling molecules related to positive regulation of cell proliferation, Akt and Erk, showed over-expressed after ROGDI transfection and bleomycin treatment compared to those with empty vector transfection. Our results imply that ROGDI is up-regulated in pulmonary fibrosis and turns fibroblasts into fibrogenic phenotype through positive regulation of miR-21. The increase of precursor, but not primary miR-21, after ROGDI transfection and bleomycin treatment indicates that ROGDI may regulate the TGF-£] signaling pathway in human embryonic fibroblasts. Our results support that ROGDI is a novel gene for pulmonary fibrosis and warrants for further investigation. £[
86

The Therapeutic Potential and Mechanism of POMC stress Hormone for Metastatic Cancer

Tsai, Han-en 23 August 2012 (has links)
Despite the development of novel target therapy drugs in recent years, metastatic cancer remains refractory to current cancer therapies and accounts for the majority of cancer mortalities worldwide. Metastasis consists of multiple steps including angiogenesis, extravasion, escape from immune surveillance, adhesion, and clonal expansion in different organs that a systemic therapy is required for effective control of metastasis. The pro-inflammatory nuclear factor kappa B (NF£eB) pathway plays an important role during each of these metastatic events and constitutes an excellent target for metastasis control. Stress hormone pro-opiomelanocortin (POMC) and its derived neuropeptides including corticotrophin (ACTH), £\-, £]-, and £^-melanocyte¡Vstimulating hormone (£\-, £]-, and £^-MSH), £]-endorphin are potent inhibitors of NF£eB pathway. Other than the central regulation of stress response and energy homeostasis, POMC also regulates the skin pigmentation, inflammatory processes, and immune reactions in the peripheral system. Since adenovirus¡Vmediated POMC gene delivery leads to hepatic POMC expression, it seems plausible that POMC gene therapy may elicit systemic production of anti-inflammatory POMC-derived peptides and hold promises for control of primary and metastatic cancers. In B16-F10 melanoma models, POMC gene delivery elevated the circulating ACTH levels for more than 8 weeks and suppressed the growth of established melanoma, thereby prolonging the life span of tumor-bearing mice. Moreover, combination of POMC therapy with cisplatin further enhances the survival outcome. Subsequent analysis reveals that POMC gene therapy inhibits the growth and metastasis of melanoma through apoptosis, angiogenesis inhibition, and modulation of epithelial-mesenchymal transition. Besides, £\-MSH/melanortin-1 receptor (MC-1R) pathway is involved in the POMC-mediated melanoma suppression. To investigate whether POMC therapy could be applied to other types of tumor, we evaluated the therapeutic efficacy of POMC gene therapy in Lewis lung carcinoma (LLC) cells which lack MC-1R. Interestingly, POMC gene delivery effectively inhibited the proliferation and colony formation of LLC cells in vitro and the growth of established LLC in mice. Histological analysis indicated that POMC gene delivery attenuated LLC through proliferation inhibition, apoptosis induction, and angiogenesis blockade. Moreover, POMC gene delivery perturbed £]-catenin signaling by reducing protein levels of £]-catenin and its downstream proto-oncogenes, including cyclin D1 and c-myc. These results support the existence of an MC-1R-independent pathway for POMC gene therapy and expand the therapeutic spectrum of POMC therapy for multiple types of cancer. To elucidate the role of host immunity in anti-neoplastic mechanism underlying POMC therapy, we compared the treatment efficacy of POMC gene therapy for B16-F10 melanoma between severe combined immune-deficient (SCID) and immune-competent C57BL/6 mice, and found similar extent of tumor suppression in both strains of mice. In addition, POMC gene therapy reduced the spleen weight and the number of circulating lymphocytes in B6 mice. These findings suggest that POMC therapy was not dependent on host immunity, yet instead induced immune suppression of animals through ACTH/cortisol production. To minimize such side effect of POMC therapy, we generated a series of adenovirus vectors encoding POMC with mutations in ACTH domain (ACTH-K15A/R17A), which fails to stimulate cortisol synthesis in vitro and in vivo. Gene delivery of ACTH (K15A/R17A) remained capable of suppressing the primary and metastatic melanoma, but had no effect on immune functions in mice. In conclusion, we have characterized the anti-neoplastic function and mechanism of POMC therapy for cancer. Furthermore, we have developed improved POMC gene vectors to minimize its adverse effect for future cancer therapy.
87

Generating a Consistent Framework for Evaluating Cell Response to External Stimuli through Epigenetic Assessors

Wang, Bo 2011 May 1900 (has links)
Mesenchymal stem cells are more and more widely used in tissue engineering due to their pluripotency and no relative ethical problems. Traditional characterization techniques to detect mesenchymal stem cell states include flow cytometry, gene expressing profiling and immunohistochemistry. However, these methods can only provide transient and low level information from current RNA or protein levels about mesenchymal stem cells, which may cause problems when predicting the possible downstream lineages they will commit into. We have developed chromatin immunoprecipitation (ChIP)-based epigenetic technique to detect mesenchymal stem cell states. For the systems we tested, this epigenetic assessor successfully characterized cell state changes and gave similar results obtained from gene expression profiling or protein expression assay. This epigenetic technique can provide information about mesenchymal stem cells states from a more fundamental chromatin level, which is promising for predicting future lineages from current states.
88

The Effect Of Mechanical Forces On Adipogenic Differentiation

Sharafi, Parisa 01 January 2008 (has links) (PDF)
Numerous intra and extra cellular factors take role in differentiation of cell towards a given lineage. These factors have crucial role in cell-cell and cell-environment interactions. In this study, the aim is to investigate the effect of mechanical forces on the adipogenic differentiation of preadipocytes and mesenchymal stem cells in an in vitro model. Human preadipocytes and mesenchymal stem cells were embedded in 2 % agarose discs. According to the stress-relaxation test results it was observed that initial mechanical properties of agarose-mesenchymal stem cell (MSC) discs did not change compared to acellular agarose whereas those of preadipocytes decreased significantly. The discs with cells were exposed to compression under different weights (1.4 &plusmn / 0.2 g, 7.5 &plusmn / 0.2 g, and 14.6 &plusmn / 0.3 g.) continuously in differentiation medium for 21 days. The control discs were treated with differentiation medium without any compressive weight on top of them. After 21 days, total ribonucleic acids (RNA) have been isolated. Adipogenic differentiation was investigated via reverse transcription coupled quantitative polymerase chain reaction (PCR). The expression of peroxisome proliferators-activated receptors (PPAR-gamma), CCAAT-enhancer binding protein (C/EBP-Beta), leptin, adiponectin, adipophilin and human stearoyl-CoA desaturase (hSCD) have been assessed as adipogenic markers. Differentiation to adipocytes has been further investigated by histochemical Sudan IV staining and immunochemistry and compared to control group. Decrease in the expression of adipogenic factors, size and number of lipid droplets were observed for both MSCs and preadipocytes subjected to compression in agarose discs. The decreases were correlated with the level of mechanical stress. The highest depletion of gene expression was observed in leptin and C/EBP&amp / #61538 / . From our results, it was shown for the first time that mechanical stress impaired the adipogenic differentiation of MSCs and preadipocytes in agarose discs. However, the differentiation pathways should be further investigated.
89

Attenuated apoptosis as consequence of Epithelial Mesenchymal Transition

Keitel, Ulrike 09 September 2013 (has links)
No description available.
90

Chondroitin sulfate microparticles modulate TGF-B1-induced chondrogenesis in human mesenchymal stem cell spheroids

Goude, Melissa Chou 08 June 2015 (has links)
Due to the limited intrinsic healing ability of mature cartilage tissue, stem cell therapies offer the potential to restore cartilage lost due to trauma or arthritis. Mesenchymal stem cells (MSCs) are a promising cell source due to their ability to differentiate into various adult tissues under specific biochemical and physical cues. Current MSC chondrogenic differentiation strategies employ large pellets, however, we have previously developed a high-throughput technique to form small MSC aggregates (500-1,000 cells) that may reduce diffusion barriers while maintaining a multicellular structure that is analogous to cartilaginous condensations. The objective of this study was to examine the effects on chondrogenesis of incorporating chondroitin sulfate methacrylate (CSMA) microparticles (MPs) within these small MSC spheroids when cultured in the presence of transforming growth factor-β1 (TGF-β1) over 21 days. Spheroids +MP induced earlier increases in collagen II and aggrecan gene expression (chondrogenic markers) than spheroids -MP, although no large differences in immunostaining for these matrix molecules were observed by day 21. Collagen I and X was also detected in the ECM of all spheroids by immunostaining. Interestingly, histology revealed that CSMA MPs clustered together near the center of the MSC spheroids and induced circumferential alignment of cells and ECM around the material core. Because chondrogenesis was not hindered by the presence of CSMA MPs, this study demonstrates the utility of this culture system to further examine the effects of matrix molecules on MSC phenotype, as well as potentially direct differentiation in a more spatially controlled manner that better mimics the architecture of specific target tissues.

Page generated in 0.0777 seconds