• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 9
  • 5
  • 4
  • 2
  • 1
  • 1
  • Tagged with
  • 25
  • 19
  • 19
  • 5
  • 5
  • 4
  • 4
  • 4
  • 3
  • 3
  • 3
  • 3
  • 3
  • 3
  • 3
  • 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.
1

The biologic activity of 5��-reduced pregnanes in the late gestation mare

Strooband, Jenny A. 29 August 2002 (has links)
Mares have an atypical hormone profile during pregnancy. Systemic progesterone (P4) levels approach zero by day 220 of gestation. Other reduced pregnanes such as 5��-pregnane-3, 20-dione (5��), 5��-pregnane-3��, 20��-diol (����), 3��-hydroxy-5��-pregnan-3-one (3��) and 20��-hydroxy-5��-prenan-3-one (20��), increase to near ��g/mL levels in the peripheral system of the mare until directly before parturition when they decrease. This unusual hormone profile during gestation indicates the possibility that other pregnanes, not P4, are responsible for uterine quiescence and gonadotropin inhibition during pregnancy. Three experiments were conducted to determine if these steroids have biologic activity. Experiment 1 consisted of jugular vein blood samples taken from mares from ten days pre-partum until the foal heat ovulation, approximately 15 days postpartum. Samples were analyzed for luteinizing hormone (LH), follicle stimulating hormone (FSH), and pregnane content. Concentrations of these hormones were analyzed for serial correlations. There was a serial negative correlation with pregnanes and FSH (p=0.0138), which were analyzed on a same day basis, day -5 to day of foaling. There also was a positive correlation with pregnanes and FSH analyzed from day of foaling to 10 days post-foaling (p<0.00l). There was also a significant negative correlation (p=0.0196) between pregnanes and LH, analyzed on a lag basis, day -5 to day of foaling for pregnanes, and day -5 to day of ovulation for LH. There was also a significant negative correlation when pregnanes were analyzed from day of foaling to 10 days post foaling, and LH was analyzed from 10 days before ovulation to day of ovulation (p=0.004). Maximum pre-partum pregnane levels did not affect time to ovulation (p=0.34). In experiment 2 equine anterior pituitary glands were harvested and the cells plated to begin a primary cell culture. After attachment, the cells were divided into treatment groups: P4, 5��, ����, 20��, 3�� or a control and each group subjected to a 1.0nM Gonadotropin Hormone Releasing Hormone (GnRH) challenge. Subsequently cells and medium were collected and analyzed for LH and FSH content using radioimmunoassay (RIA). The cells did exhibit a response to GnRH (p=0.015 between positive and negative controls) and there was a treatment effect for FSH (p=0.0058); only 3�� resulted in significantly more FSH release than the positive control (p=0.043) after stimulation with GnRH. There was no treatment effect on LH (p=0.56). Experiment 3 analyzed the response of equine uterine myometrial tissue to pregnane treatment. Myometrial tissue was harvested and placed in a 37��C Krebs buffered saline bath, connected to a physiograph and repetitive spontaneous smooth muscle contraction was induced with oxytocin. Tissue was then treated with P4, 5��, ����, 20��, 3�� or a control (ethanol). The amplitude and frequency of the spontaneous contractions were measured and compared to the control. There were no differences between post treatment responses of the control and pregnane treated samples in either frequency (p=0.78) or amplitude (p=0.63) of myometrial contractions. From these data we conclude that in vivo there is a significant and differential physiologic relationship between pre-partum pregnanes and gonadotropins. Due to lack of response it is unlikely that pregnanes inhibit pituitary secretion, and thus may exert their effects elsewhere, such as at the hypothalamus. The involvement of pregnanes in modulating myometrial contractions remains unclear. It is likely that P4 does play a role in decreasing myometrial responses to OT, however, that result was inconsistent in this study. / Graduation date: 2003
2

Regulation of cytochrome P450 3A4 gene expression through modulating pregnane X receptor transcriptional activity by NF-ꢬ aryl hydrocarbon receptor and xenobiotics

Gu, Xinsheng 15 May 2009 (has links)
Cytochrome P450 3A4 (CYP3A4) is a key enzyme responsible for the metabolism of drugs and endogenous compounds in human liver and intestine. CYP3A4 gene expression is mainly regulated by Pregnane X receptor (PXR) which is a ligand-dependent nuclear receptor. It is a long-standing observation that inflammatory responses and infections decrease drug metabolism capacity in human and experimental animals. In this study, I reported that NF-κB activation by LPS and TNF-α plays a pivotal role in the suppression of CYP3A4 through interactions of NF-κB with PXR/RXR complex. Inhibition of NF-κB by NF-κB specific suppressor SRIκBα reversed the suppressive effects of LPS and TNF-α. Furthermore, I showed that NF-κB p65 disrupted the association of PXR/RXRα complex with DNA sequences as determined by EMSA and chromatin immunoprecipitation assays. NF-κB p65 directly interacted with DNA binding domain of RXRα and DNA binding domain, hinge domain and ligand-binding domain of PXR and may prevent its binding to the consensus DNA sequences, thus inhibiting the transactivation by PXR/RXRα complex. This mechanism of suppression by NF-κB activation may be extended to other nuclear receptor-regulated systems where RXRα is a dimerization partner. Many genes regulated by PXR and AhR are important for phase I, II and III drug metabolism. In this study I reported a crosstalk between PXR and AhR pathways. AhR physically and functionally interacted with PXR and enhanced the PXR transcriptional activity, and the interaction repressed the AhR transcriptional activity. AhR also physically interacted with RXRα. The synergistic induction of Gsta1 in the liver of mice by PCN and TCDD might assume a different mechanism. The results suggested the metabolism kinetics of mixture drugs was different from and more complicated than that of single compound. Using a HepG2 cell-based PXR-driven CYP3A4-Luciferase assay, I reported that E/F domain of PXR was responsible for ligand-dependant activation. A/B domain was necessary for co-activating the ligand-dependent activation and D domain was suppressive. High doses of Valerian Root extraction were PXR-dependent CYP3A4 inducers. Green tea polyphenols, aflatoxin B1, CuSO4 and MnCl2 enhanced the PXR transcription activity activated by rifampicin. The results suggested PXR-mediated drug metabolism kinetics altered on xenobiotic exposure.
3

Activation of pregnane X receptor by Ginkgo biloba extract

Yeung, Eugene Y. H. 11 1900 (has links)
Pregnane X receptor (PXR) is a ligand-activated transcription factor that plays a role in a broad array of biological processes, including drug metabolism and transport. Ginkgo biloba is an herb commonly used to improve cognitive function. In primary cultures of rat hepatocytes, Ginkgo biloba induces the mRNA expression of CYP3A23, a target gene for rat PXR. The present study tested the hypothesis that Ginkgo biloba activates PXR. Cultured HepG2 human hepatoma cells were transfected with the full-length human PXR (pCR3-hPXR), the full-length mouse PXR (pCR3-mPXR), or an empty vector (pCR3) in addition to a reporter plasmid (XREM-CYP3A4-LUC; firefly luciferase) and an internal control plasmid (phRL-TK; Renilla luciferase). At 24 h after transfection, cells were treated for 24 h with Ginkgo biloba extract and luciferase activity was measured. The extract at 200 µg/ml increased mouse and human PXR activity by 3.0-fold and 9.5-fold, respectively, indicating that Ginkgo biloba more effectively activates human PXR. Dose-response experiments showed that the extract produced a log-linear increase over the range of 200–800 µg/ml. To determine whether Ginkgo biloba extract induces human PXR target gene expression, cultured LS180 human colon adenocarcinoma cells were treated for 72 h with the extract. Total cellular RNA was isolated and reverse transcribed. CYP3A4, CYP3A5, and ABCB1 cDNAs were amplified by real-time PCR. Ginkgo biloba extract at 200, 400, and 800 µg/ml increased CYP3A4 mRNA expression by 1.7-, 2.4-, and 2.5-fold, respectively. The extract at the same concentrations increased the mRNA expression of CYP3A5 (1.3 to 3.6-fold) and ABCB1 (2.7 to 6.4-fold). To determine whether the increased expression involved PXR activation, cells were treated with a PXR antagonist, L-sulforaphane, and Ginkgo biloba extract. L-sulforaphane at 5, 10, and 20 µM decreased CYP3A4 mRNA expression by 54%, 78%, and 93%, respectively, in cells co-treated with the extract. A similar pattern of response was obtained with CYP3A5 and ABCB1. In cells co-treated with the extract, L-sulforaphane (5 and 10 µM) was not cytotoxic and did not decrease PXR mRNA expression. Our data from cell culture experiments indicate that Ginkgo biloba activates PXR and increases CYP3A4, CYP3A5, and ABCB1 mRNA expression.
4

Regulation of cytochrome P450 3A4 gene expression through modulating pregnane X receptor transcriptional activity by NF-ꢬ aryl hydrocarbon receptor and xenobiotics

Gu, Xinsheng 15 May 2009 (has links)
Cytochrome P450 3A4 (CYP3A4) is a key enzyme responsible for the metabolism of drugs and endogenous compounds in human liver and intestine. CYP3A4 gene expression is mainly regulated by Pregnane X receptor (PXR) which is a ligand-dependent nuclear receptor. It is a long-standing observation that inflammatory responses and infections decrease drug metabolism capacity in human and experimental animals. In this study, I reported that NF-κB activation by LPS and TNF-α plays a pivotal role in the suppression of CYP3A4 through interactions of NF-κB with PXR/RXR complex. Inhibition of NF-κB by NF-κB specific suppressor SRIκBα reversed the suppressive effects of LPS and TNF-α. Furthermore, I showed that NF-κB p65 disrupted the association of PXR/RXRα complex with DNA sequences as determined by EMSA and chromatin immunoprecipitation assays. NF-κB p65 directly interacted with DNA binding domain of RXRα and DNA binding domain, hinge domain and ligand-binding domain of PXR and may prevent its binding to the consensus DNA sequences, thus inhibiting the transactivation by PXR/RXRα complex. This mechanism of suppression by NF-κB activation may be extended to other nuclear receptor-regulated systems where RXRα is a dimerization partner. Many genes regulated by PXR and AhR are important for phase I, II and III drug metabolism. In this study I reported a crosstalk between PXR and AhR pathways. AhR physically and functionally interacted with PXR and enhanced the PXR transcriptional activity, and the interaction repressed the AhR transcriptional activity. AhR also physically interacted with RXRα. The synergistic induction of Gsta1 in the liver of mice by PCN and TCDD might assume a different mechanism. The results suggested the metabolism kinetics of mixture drugs was different from and more complicated than that of single compound. Using a HepG2 cell-based PXR-driven CYP3A4-Luciferase assay, I reported that E/F domain of PXR was responsible for ligand-dependant activation. A/B domain was necessary for co-activating the ligand-dependent activation and D domain was suppressive. High doses of Valerian Root extraction were PXR-dependent CYP3A4 inducers. Green tea polyphenols, aflatoxin B1, CuSO4 and MnCl2 enhanced the PXR transcription activity activated by rifampicin. The results suggested PXR-mediated drug metabolism kinetics altered on xenobiotic exposure.
5

Novel Functions for the Pregnane X Receptor include Regulation of mRNA Turnover and Involvement in Colon Cancer Progression

Eagleton, Navada Lorraine 2010 August 1900 (has links)
To understand the mechanisms of transcriptional regulation of PXR, we performed yeast two-hybrid screenings to search for PXR-interacting proteins in a human liver cDNA library using the PXR ligand binding domain as the bait. More than one million independent clones were screened. One positive clone was a partial cDNA of CNOT2 (amino acid 183-540). CNOT2 is a component of CCR4-NOT that is a multi-subunit protein complex highly conserved from yeast to humans. Using a mammalian two-hybrid system in CV-1 cells and GST-pull down assays, we confirmed the direct interaction between PXR and CNOT2 and mapped the specific domains of association. In HepG2 cells, over expression of CNOT2 suppressed the PXR-regulated luciferase reporter gene activity. siRNA knockdown of CNOT2 potentiated PXR-transcriptional activity. These results strongly suggest that the CCR4-NOT complex is significantly involved in transcriptional regulation of PXR. The immuno-precipitated CNOT2 complex contained deadenylase activity as determined by an in vitro RNA decay assay. The presence of transfected PXR inhibited the cNOT2-associated deadenylase activity, as demonstrated by poly(A) tail PCR. Cellular localization of PXR and cNOT2 by immuno-fluorescence microscopy indicates that the interaction might occur within Cajal Bodies. Taken together, these results suggest that PXR regulates the mRNA turnover through direct interaction with the NOT2 component of the CCR4-NOT complex. PXR is also involved in colon cancer progression. Our results indicate that the evolutionarily conserved PXR protects organisms from carcinogenesis by inhibiting tumor growth as well as eliminating carcinogenic substances. Our laboratory proposes that pregnane X receptor has an important role in maintaining the balance of cells progressing through the cell cycle. In vitro and in vivo experiments demonstrate expression of PXR in colon cancer cells slows the progression of tumor formation. Colony growth of the PXR-transfected HT29 cells was suppressed in soft agar assay. In the xenograft assay, the tumor size formed in nude mice was significantly suppressed in HT29 cells stably transfected with PXR (310 mg /- 6.2 vs. 120 mg±6, p<0.01). The number of Ki-67 positive cells were significantly decreased in PXR-transfected HT29 xenograft tumor tissue compared vector-transfected HT29 controls (p<0.01) as determined by immuno-histochemistry suggesting that PXR inhibits proliferation of colon cancer cells. Results of flow cytometry analysis indicated that PXR-transfection in HT29 cells caused G0/G1 arrest. The growth inhibitory effects of PXR are likely mediated through the E2F/Rb-regulated check point since E2F1 nuclear expression was significantly inhibited by PXR over expression.
6

Activation of pregnane X receptor by Ginkgo biloba extract

Yeung, Eugene Y. H. 11 1900 (has links)
Pregnane X receptor (PXR) is a ligand-activated transcription factor that plays a role in a broad array of biological processes, including drug metabolism and transport. Ginkgo biloba is an herb commonly used to improve cognitive function. In primary cultures of rat hepatocytes, Ginkgo biloba induces the mRNA expression of CYP3A23, a target gene for rat PXR. The present study tested the hypothesis that Ginkgo biloba activates PXR. Cultured HepG2 human hepatoma cells were transfected with the full-length human PXR (pCR3-hPXR), the full-length mouse PXR (pCR3-mPXR), or an empty vector (pCR3) in addition to a reporter plasmid (XREM-CYP3A4-LUC; firefly luciferase) and an internal control plasmid (phRL-TK; Renilla luciferase). At 24 h after transfection, cells were treated for 24 h with Ginkgo biloba extract and luciferase activity was measured. The extract at 200 µg/ml increased mouse and human PXR activity by 3.0-fold and 9.5-fold, respectively, indicating that Ginkgo biloba more effectively activates human PXR. Dose-response experiments showed that the extract produced a log-linear increase over the range of 200–800 µg/ml. To determine whether Ginkgo biloba extract induces human PXR target gene expression, cultured LS180 human colon adenocarcinoma cells were treated for 72 h with the extract. Total cellular RNA was isolated and reverse transcribed. CYP3A4, CYP3A5, and ABCB1 cDNAs were amplified by real-time PCR. Ginkgo biloba extract at 200, 400, and 800 µg/ml increased CYP3A4 mRNA expression by 1.7-, 2.4-, and 2.5-fold, respectively. The extract at the same concentrations increased the mRNA expression of CYP3A5 (1.3 to 3.6-fold) and ABCB1 (2.7 to 6.4-fold). To determine whether the increased expression involved PXR activation, cells were treated with a PXR antagonist, L-sulforaphane, and Ginkgo biloba extract. L-sulforaphane at 5, 10, and 20 µM decreased CYP3A4 mRNA expression by 54%, 78%, and 93%, respectively, in cells co-treated with the extract. A similar pattern of response was obtained with CYP3A5 and ABCB1. In cells co-treated with the extract, L-sulforaphane (5 and 10 µM) was not cytotoxic and did not decrease PXR mRNA expression. Our data from cell culture experiments indicate that Ginkgo biloba activates PXR and increases CYP3A4, CYP3A5, and ABCB1 mRNA expression.
7

Activation of pregnane X receptor by Ginkgo biloba extract

Yeung, Eugene Y. H. 11 1900 (has links)
Pregnane X receptor (PXR) is a ligand-activated transcription factor that plays a role in a broad array of biological processes, including drug metabolism and transport. Ginkgo biloba is an herb commonly used to improve cognitive function. In primary cultures of rat hepatocytes, Ginkgo biloba induces the mRNA expression of CYP3A23, a target gene for rat PXR. The present study tested the hypothesis that Ginkgo biloba activates PXR. Cultured HepG2 human hepatoma cells were transfected with the full-length human PXR (pCR3-hPXR), the full-length mouse PXR (pCR3-mPXR), or an empty vector (pCR3) in addition to a reporter plasmid (XREM-CYP3A4-LUC; firefly luciferase) and an internal control plasmid (phRL-TK; Renilla luciferase). At 24 h after transfection, cells were treated for 24 h with Ginkgo biloba extract and luciferase activity was measured. The extract at 200 µg/ml increased mouse and human PXR activity by 3.0-fold and 9.5-fold, respectively, indicating that Ginkgo biloba more effectively activates human PXR. Dose-response experiments showed that the extract produced a log-linear increase over the range of 200–800 µg/ml. To determine whether Ginkgo biloba extract induces human PXR target gene expression, cultured LS180 human colon adenocarcinoma cells were treated for 72 h with the extract. Total cellular RNA was isolated and reverse transcribed. CYP3A4, CYP3A5, and ABCB1 cDNAs were amplified by real-time PCR. Ginkgo biloba extract at 200, 400, and 800 µg/ml increased CYP3A4 mRNA expression by 1.7-, 2.4-, and 2.5-fold, respectively. The extract at the same concentrations increased the mRNA expression of CYP3A5 (1.3 to 3.6-fold) and ABCB1 (2.7 to 6.4-fold). To determine whether the increased expression involved PXR activation, cells were treated with a PXR antagonist, L-sulforaphane, and Ginkgo biloba extract. L-sulforaphane at 5, 10, and 20 µM decreased CYP3A4 mRNA expression by 54%, 78%, and 93%, respectively, in cells co-treated with the extract. A similar pattern of response was obtained with CYP3A5 and ABCB1. In cells co-treated with the extract, L-sulforaphane (5 and 10 µM) was not cytotoxic and did not decrease PXR mRNA expression. Our data from cell culture experiments indicate that Ginkgo biloba activates PXR and increases CYP3A4, CYP3A5, and ABCB1 mRNA expression. / Pharmaceutical Sciences, Faculty of / Graduate
8

Regulation of cytochrome P450-3A (CYP3A) and pregnane X receptor (PXR) : implications to drug-drug interactions /

Sachdeva, Karuna. January 2005 (has links)
Thesis (Ph. D.)--University of Rhode Island, 2005. / Typescript. Includes bibliographical references (leaves 129-140).
9

Etude des xénorécepteurs CAR (NR1I3) et PXR (NR1I2) : identification d’un nouveau gène cible de CAR (SPOT14) et d’une nouvelle isoforme de PXR (PXR-small) dans l'hépatocyte humain / Study of the CAR (NR1I3) and PXR (NR1I2) : identification of a new CAR target gene (SPOT14) and a new PXR isoform (PXR-small) in human hepatocyte

Breuker, Cyril 16 December 2010 (has links)
CAR (Constitutive Androstane Receptor, NR1I3) et PXR (Pregnane X Receptor, NR1I2) sont deux récepteurs nucléaires dédiés à la reconna issance et à l'élimination de molécules lipophiles potentiellement toxiques pour l'organisme. Ces facteurs de transcription peuvent être activés par des ligands d'origines et de structures diverses (médicaments, polluants environnementaux, produits de l'alimentation et de phytothérapies). L'activation de ces récepteurs entraîne l'expression des gènes majeurs de la fonction de détoxication entéro-hépatique (CYP450, transférases, transporteurs) permettant l'élimination de ces toxiques. Dans ce travail, nous avons dans un premier temps 1) montré que CAR contrôle l'expression de Spot14, une protéine pro-lipogénique, et 2) nous avons identifié une nouvelle isoforme de PXR (PXR-small) codant uniquement pour le domaine de liaison des ligands de PXR. Nous avons pu déterminer les origines de transcription par 5'-RACE PCR et montrer que PXR-small représente environ 10% de l'ensemble des transcrits de PXR dans le tissu hépatique sain par une approche de PCR qua ntitative. Nous avons pu détecter sa présence par western-blot sur des extraits de protéines nucléaires issus de tissus hépatiques et de lignées cellulaires hépatiques. Par des expériences de gel retard, nous avons observé que cette nouvelle isoforme tronquée, qui ne code que pour le LBD de PXR, ne peut pas se lier à l'ADN. Des expériences de gènes rapporteurs suggèrent que cette isoforme se comporte comme un dominant négatif de PXR. Enfin, la présence d'un ilot CpG situé juste en amont de PXR-small suggère que cette nouvelle isoforme pourrait être régulée épigénétiquement par méthylation, notamment dans les cellules tumorales. / CAR (Constitutive Androstane Receptor, NR1I3) and PXR (Pregnane X Receptor, NR1I2) are two nuclear receptors devoted to the recognition and elimination of lipohilic molecules potentially toxic to the body.These transcription factors can be activated by ligands of different origins and structures (drugs, environmental pollutants, food products and herbal medicine...). The activation of these receptors leads to the expression of major genes of the detoxification process (CYP450, transferases, transporters) leading to the elimination of these toxics. In this work, we 1) showed that Spot14, a pro-lipogenic protein, is a target gene of CAR, then 2) we identified a novel isoform of PXR (PXR-small), coding only the ligand binding domain of PXR. By using 5'-RACE PXR, we established the origins of transcription of PXR-small and by quantitative PCR we observed that PXR-small represents about 10% of all PXR transcripts in human liver. By using western blo t, we detect its presence on nuclear protein extracts from liver tissues and hepatic cell lines. In Electromobility shift essays experiments, we observed that PXR-small cannot bind to DNA, while reporter essay experiments suggest that this isoform acts as a dominant negative of PXR. Finally, the presence of a CpG island just upstream of PXR-small suggests that this novel isoform might be regulated epigenetically by methylation, more particularly in tumor cells.
10

Engineering the pregnane X receptor and estrogen receptor alpha to bind novel small molecules using negative chemical complementation

Shaffer, Hally A. 05 April 2011 (has links)
Nuclear receptors are ligand-activated transcription factors that play significant roles in various biological processes within the body, such as cell development, hormone metabolism, reproduction, and cardiac function. As transcription factors, nuclear receptors are involved in many diseases, such as diabetes, cancer, and arthritis, resulting in approximately 10-15% of the pharmaceutical drugs presently on the market being targeted toward nuclear receptors. Structurally, nuclear receptors consist of a DNA-binding domain (DBD), responsible for binding specific sequences of DNA called response elements, fused to a ligand-binding domain (LBD) through a hinge region. The LBD binds a small molecule ligand. Upon ligand binding, the LBD changes to an active conformation leading to the recruitment of coactivator (CoAC) proteins and initiation of transcription. As a result of their involvement in disease, there is an emphasis on engineering nuclear receptors for applications in gene therapy, drug discovery and metabolic engineering.

Page generated in 0.0343 seconds