• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 169
  • 80
  • 71
  • 30
  • 19
  • 16
  • 7
  • 6
  • 5
  • 4
  • 3
  • 3
  • 2
  • 1
  • 1
  • Tagged with
  • 527
  • 128
  • 93
  • 75
  • 64
  • 62
  • 59
  • 55
  • 52
  • 50
  • 47
  • 43
  • 42
  • 41
  • 41
  • 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.
21

The role of p27kip1 in human malignant brain tumors

Tsai, Feng-Lin 08 September 2003 (has links)
Gliomas are the most common human brain tumors and are divided into four stages by WHO classification scheme. Benign gliomas are defined as grades I (Pilocytic astrocytomas) and II (Astrocytomas), whereas grade III (Anaplastic Astrocytomas, AA) and grade IV (Glioblastoma Multiforme, GBM) are malignant. Although both grades III and IV are malignant, the prognoses for these tumors are quite different. The 2-year survival rate for grade III gliomas is 50%, and grade IV is < 20 %. Mechanisms of tumorigenesis are not exactly elucidated in brain tumor cells. The thesis is to study the role of p27 kip1 in human malignant brain tumors. The experimental methods include ribonuclease protection assay (RPA), western blotting, immunohistochemical staining and immunocytochemical staining. mRNAs of p130, p107, Rb, p53 and p27 kip1 in normal brain tissues and brain tumors were overexpressed in most case. The p27kip1 mRNA were expressed in all astrocytomas and GBM, and mRNA quantity of p27kip1 were more in brain tumors than in normal brain tissues. PI3K/Akt pathway regulates several cellular functions such as cell survival and cell proliferation. Active Akt can phosphorylate p27kip1 that may contribute cell cycle from G1 phase to S phase. Skp2 identifies phospho-p27kip1 and promotes p27kip1 degradation. We found p27kip1 overexpression and Akt activation in astrocytomas and GBM. The expression of p-Akt were found in 20 %, 87 % and 71 % in normal brain tissues, astrocytomas and GBM, respectively. Expression of p27kip1 and p-Akt has shown significant correlation in GBM (P = 0.0236). Overexpression of p27kip1 mRNA in brain tumors may be consequence of p-Akt and Skp2.
22

Repetitive Stretching Prevents Muscle Atrophy in Denervated Soleus Muscle via Akt/mTOR/p70S6K Pathways

Agata, Nobuhide, 縣, 信秀 25 March 2009 (has links)
名古屋大学博士学位論文 学位の種類:博士(医療技術学)(課程)学位授与年月日:平成21年3月25日
23

Androgen Promotes Osteoblast Proliferation through Activation of Phosphatidylinositol-3-OH Kinase /Akt Signaling Pathway

Huang, Kai-Lieh 08 July 2003 (has links)
Androgen has been shown to stimulate proliferation of osteoblast-like MC3T3-E1 cells. However, the molecular mechanism responsible for this effect remains to be elucidated. In the present study we demonstrate herein the non-genomic effect of androgen on osteoblast-like MC3T3-E1 cells involving activation of a PI(3)K/Akt signaling pathway and stimulating proliferation. In studies of steroids signaling, 5a-dihydrotestosterone (DHT), testosterone and 17b-estradiol but not dexamethasone or progesterone induced a rapid and transient phosphorylation of Akt in MC3T3-E1 cells. The androgen-induced Akt activation reached to the climax after 15 min and gradually diminished to baseline after 60 min. This induction of androgen was unaffected by actinomycin D and was specifically blocked by androgen receptor (AR) antagonist hydroxyflutamide (HF) or transfection of siRNA-AR. Treatment of MC3T3-E1 cells with PI(3)K inhibitor LY294002 or transfection with kinase-deficient Akt blocked androgen-induced cells proliferation. Moreover, androgen-induced activation of Akt was abolished by inhibitors of Src kinase, Gi-protein and phospholipase C showing the involvement of these effectors in androgen signaling pathway. Further, androgen-induced activation of Akt was dependent on intracellular calcium as shown by the effect of EGTA and intracellular calcium chelator BAPTA/AM. Fluorescence microscopy showed translocation of phospho-Akt from cytosol into nucleus after androgen treatment but no change in the subcellular distribution of phospho-Akt when HF or LY294002 pretreatment was administered to the cells. These results strongly suggest that phosphorylation of Akt in osteoblast cells is mediated by androgen receptor and the androgen-induced translocation of Akt is an important step in the androgen/AR signaling pathway that mediates osteoblast cells proliferation.
24

Characterizing ErbB2-induced mammary tumourigenesis

Oliver, Joseph James 18 September 2007 (has links)
Approximately 30% of human breast cancers demonstrate overexpression of the receptor tyrosine kinase ErbB2/HER2/Neu, with these cases correlating with recurrence and poor prognosis. While therapy targeting ErbB2 has met with some success, particularly in early-stage breast cancers, transformation and progression towards a later-stage metastatic phenotype is likely sustained by aberrant signaling from additional players, for instance that downstream of integrins. In fact, treatment with integrin-blocking antibodies and ß1-integrin ablation leads to reversion of the malignant phenotype of human breast cells in three-dimensional culture and in vivo. Moreover, ErbB2 has recently been found to interact with the ß4-integrin subunit to promote tumour formation and progression, as deletion of the ß4-integrin signaling domain led to suppression of mammary tumour onset and invasive growth, coupled with decreases in ErbB2-dependent signaling. ErbB2 may interact with integrin subunits by direct binding or via the intracellular kinases Src and focal adhesion kinase (FAK), both known to be activated downstream of ErbB2 and integrins and having well-established roles in cell adhesion, migration, and invasion. Using an inducible model of ErbB2 activation, we have demonstrated that controlled ErbB2 activation in human mammary epithelial cells leads to phosphorylation of Src Tyr215 and FAK Tyr861, consistent with a recently published clinical study examining phosphorylated forms of Src and FAK in ErbB2-positive human breast tumour samples. We have also confirmed that ErbB2 activation increases the capacity of cells for survival: Normally, MCF10A human mammary epithelial cells cultured in three-dimensional, laminin-rich extracellular matrix gel form mammary acini-like spheroids with hollow lumen surrounded by a single layer of polarized epithelial cells. However, ErbB2 activation prevents luminal clearance and induces luminal filling in acini formed in three-dimensional culture, and leads to activation of Akt, a known survival signal. Taken together these data indicate a potential role for ErbB2 at the apex of cell survival signaling via Src, FAK, and Akt, contributing to luminal cell survival in three-dimensional culture. We have thus confirmed Src, FAK, and Akt as potential players in early onset of breast cancer, and targeting these signaling players concurrently with ErbB2 may prove effective, especially in early stage breast cancers. / Thesis (Master, Pathology & Molecular Medicine) -- Queen's University, 2007-08-31 16:25:59.917
25

Herpes Simplex Virus Requires VP11/12 to Activate Src Family Kinase-PI3 Kinase-Akt Signalling

Wagner, Melany Unknown Date
No description available.
26

The Role and Regulation of the Phosphatase PPM1D in Chemoresistant Gynecological Cancers

Ali, Ahmed Y. 24 January 2014 (has links)
Cisplatin (CDDP; cis-diamminedichloroplatinum) resistance presents a major impediment in the treatment of several gynecologic solid tumors, including ovarian and cervical tumors. p53, a critical regulator of cellular apoptosis, is a determinant of CDDP sensitivity. In our study, we have observed that the dysregulation of p53 regulators, checkpoint kinase 1 (Chk1) and protein phosphatase magnesium-dependent 1 (PPM1D), significantly reduced CDDP responsiveness in human cancer cells. Isogenic wt-p53 CDDP-sensitive (OV2008) and -resistant (C13*) cervical cancer cells, and isogenic wt-p53 CDDP-sensitive (A2780s) and p53 mutant resistant (A2780cp) ovarian cancer cells, along with CDDP-resistant ovarian cancer cell lines (OCC-1 and OVCAR-3, mutant p53; SKOV-3, p53 null) were used to elucidate the mechanisms of p53 regulation in human gynecologic cancer cells. We have complemented our study with a xenograft model (A2780s) and a tissue microarray of human ovarian tumors to validate our in vitro observations. We have demonstrated that CDDP differentially regulated the p53 activator Chk1 in sensitive and resistant cancer cells; it enhances Chk1 activation in sensitive but not resistant cells. This differential regulation also extended to PPM1D, whereby CDDP enhanced PPM1D content in resistant but not sensitive cells. PPM1D knockdown sensitized resistant cells to CDDP, which was associated with up-regulation of Chk1 and p53 activations, while PPM1D over-expression had the opposite effect. We have also shown that CDDP sensitivity in response to PPM1D down-regulation was p53-dependent. Moreover, CDDP promotes PPM1D nuclear localization in resistant cells and nuclear exclusion in sensitive cells and xenograft tumors. Enhanced PPM1D expression in human ovarian tumors is significantly associated with tumor aggression. Dysregulation of the oncogene Akt has been implicated in a variety of human malignancies, including ovarian cancer. We have demonstrated that Akt regulates PPM1D stability, since activated Akt over-expression in sensitive cells rescued PPM1D from CDDP-induced proteasomal degradation and Akt down-regulation in resistant cells lead to PPM1D de-stabilization and down-regulation. We have shown for the first time that PPM1D is downstream of Akt through which it can modulate CDDP sensitivity in human cancer cells. These findings extend the current knowledge on the molecular basis of CDDP resistance in gynecological cancers and may help in developing effective therapeutic strategies.
27

Sprachliches Handeln und Transaktionsanalyse die Psychologie im Sprechakt

Hansen, Hans Harald January 1987 (has links)
Zugl.: Paderborn, Univ., Staatsexamensarbeit, 1987
28

Sprechintentionen in deutschen und japanischen Zeitungskommentaren Illokutionstypologie und kontrastive Analysen von empirischen Texten

Dillmann, Gerhard January 2008 (has links)
Zugl.: Freiburg (Breisgau), Univ., Diss., 2008
29

Essai du traitement pré-clinique du carcinome hépatocellulaire sur la cirrhose dans le modèle de rat / Pre-trial of hepatocellular carcinoma on cirrhosis in a rat model

Zeybek, Ayça 22 December 2016 (has links)
Hepatocellular carcinoma (HCC) is the second most common cause of cancerrelated mortality worldwide. AKT pathway has been found activated in 50% of HCC cases, making it promising target. Therefore we assess efficacy of the allosteric AKT inhibitor or the combination of Sorafenib with AKT inhibitor compared to untreated control and to standard treatment, Sorafenib, in vitro and in vivo. AKT inhibitor blocked phosphorylation of AKT in vitro and strongly inhibited cell growth with significantly higher potency than Sorafenib. Similarly, apoptosis and cell migration were strongly reduced by AKT inhibitor in vitro. To mimic human advanced HCC, we used diethylnitrosamine-induced cirrhotic rat model with fully developed HCC. MRI analyses showed that AKT inhibitor significantly reduced overall tumor size. Furthermore, number of tumors was decreased by AKT inhibitor, which was associated with increased apoptosis and decreased proliferation. Tumor contrast enhancement was significantly decreased in the AKT inhibitor group. Moreover, on tumor tissue sections, we observed a vascular normalization and a significant decrease in fibrosis in surrounding liver of animals treated with AKT inhibitor. Finally, pAKT/AKT levels in AKT inhibitor treated tumors were reduced, followed by down regulation of actors of AKT downstream signalling pathway: pmTOR, pPRAS40, pPLCγ1 and pS6K1. In conclusion, we demonstrated that AKT inhibitor blocks AKT phosphorylation in vitro and in vivo. In HCC-rat model, AKT inhibitor was well tolerated, showed anti-fibrotic effect and had stronger antitumor effect than Sorafenib. Our results confirm the importance of targeting AKT in HCC. / Hepatocellular carcinoma (HCC) is the second most common cause of cancerrelated mortality worldwide. AKT pathway has been found activated in 50% of HCC cases, making it promising target. Therefore we assess efficacy of the allosteric AKT inhibitor or the combination of Sorafenib with AKT inhibitor compared to untreated control and to standard treatment, Sorafenib, in vitro and in vivo. AKT inhibitor blocked phosphorylation of AKT in vitro and strongly inhibited cell growth with significantly higher potency than Sorafenib. Similarly, apoptosis and cell migration were strongly reduced by AKT inhibitor in vitro. To mimic human advanced HCC, we used diethylnitrosamine-induced cirrhotic rat model with fully developed HCC. MRI analyses showed that AKT inhibitor significantly reduced overall tumor size. Furthermore, number of tumors was decreased by AKT inhibitor, which was associated with increased apoptosis and decreased proliferation. Tumor contrast enhancement was significantly decreased in the AKT inhibitor group. Moreover, on tumor tissue sections, we observed a vascular normalization and a significant decrease in fibrosis in surrounding liver of animals treated with AKT inhibitor. Finally, pAKT/AKT levels in AKT inhibitor treated tumors were reduced, followed by down regulation of actors of AKT downstream signalling pathway: pmTOR, pPRAS40, pPLCγ1 and pS6K1. In conclusion, we demonstrated that AKT inhibitor blocks AKT phosphorylation in vitro and in vivo. In HCC-rat model, AKT inhibitor was well tolerated, showed anti-fibrotic effect and had stronger antitumor effect than Sorafenib. Our results confirm the importance of targeting AKT in HCC.
30

HUMAN CLCA2 IS A P53-DEPENDENT ZINC METALLOPROTEASE AND ITS INTERACTION WITH EVA1 MAINTAINS DIFFERENTIATION OF HUMAN MAMMARY EPITHELIAL CELLS

Ramena, Grace Theresa Nicholas 01 May 2015 (has links)
CLCA2 is a p53-inducible transmembrane protein that is frequently downregulated in breast cancer. CLCA2 is a 943 amino acid type I transmembrane protein that is cleaved near amino acid 700 to produce a diffusible 100kD product. The N-terminus contains a hydrolase-like domain with well-conserved HEXXH zinc binding amino acid motif that was proposed to cleave the precursor auto-proteolytically. We investigate the auto-proteolysis of CLCA2 precursor. Using membrane extracts or purified protein from CLCA2-transfected cells, we show here that CLCA2 cleavage is catalyzed by zinc and inhibited by metal chelator EDTA. Moreover, an E165Q mutation in the metal binding site abolished processing without affecting stability or trafficking. The mutant could be cleaved by co-transfected wild type CLCA2, showing that the mutation had not caused an un-cleavable conformation and suggesting that it occurs in trans. Wild type CLCA2 was able to cleave CLCA2 E165Q mutant in vitro only after denaturation and renaturation, suggesting that a conformational shift is required for cleavage. The efficiency of cleavage increased steeply with increasing concentration of precursor, consistent with trans proteolysis but not cis or cleavage by another agent. Accordingly, CLCA2 molecules bearing different epitope tags formed a stable complex that could be co-immunoprecipitated. Cleavage appears to be specific within isoforms; CLCA1 was unable to neither cleave CLCA2 nor form a stable complex with it. Furthermore, cleavage causes a conformational shift: an N-terminal antibody that immunoprecipitates the precursor fails to precipitate the N-terminal product unless it is first denatured with ionic detergent. We found that cleavage is enhanced by p53 induction due to DNA damage, implying that the cleavage has functional consequences for stress response. Moreover, we found that HEK and MCF10A cells expressing the E165Q mutant had a higher proliferation rate than cells expressing wild type CLCA2, suggesting that the metalloprotease activity contributes to the anti-proliferative effect of CLCA2. Physiologically, CLCA2 plays an essential role in epithelial differentiation. It is induced during epithelial differentiation in immortalized human mammary epithelial cells (HMEC), and its knockdown causes epithelial to mesenchymal transition (EMT). To determine how CLCA2 promotes epithelial differentiation, we searched for interactors using membrane dihybrid screening. We discovered a strong interaction with Epithelial V-like Antigen 1 (EVA1) and confirmed by co-immunoprecipitation. Like CLCA2, EVA1 is a type I transmembrane protein that is regulated by p53 family. EVA1 resembles tight junction proteins called Junctional Adhesion Molecules (JAMs) by structure but we found by confocal analysis that EVA1 is localized the lateral interface at cell-cell junctions. Analysis of transcriptional profiles revealed that EVA1 is frequently downregulated in breast tumors and breast cancer cell lines, especially those of mesenchymal phenotype, and upregulated during epithelial differentiation. Like CLCA2, knockdown of EVA1 resulted in rapid EMT in immortalized HMEC. The interacting domains were delimited by deletion analysis, revealing that both the proteins interact via their transmembrane segments (TMS). The interaction was specific, as other transmembrane proteins did not interact with CLCA2 or EVA1. We also found that CLCA2 binds to ZO-1 and beta-catenin at its c-terminus but EVA1 does not. Interestingly, we found that EVA1 does interact with ZO-1 in the presence of CLCA2, indicating that these three form a complex at the cell-cell junctions that allows stabilization of belt-like adherens junctions (AJ). On the other hand CLCA2 may also stabilize adherens junctions by sequestering beta-catenin at the cell-cell junctions. These results indicate that CLCA2 plays a key role in maintaining epithelial differentiation via multiple ways. Either by binding to beta-catenin or forming a complex with EVA1 and ZO-1, it plays a pivotal role in maintaining epithelial differentiation. This explains the downregulation of both CLCA2 and EVA1 during tumor progression.

Page generated in 0.0407 seconds