• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 4
  • 1
  • 1
  • Tagged with
  • 7
  • 7
  • 4
  • 3
  • 3
  • 3
  • 3
  • 3
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • 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

Novel 2-substituted isoflavones: A privileged structure approach to new agents for hormone-dependent breast cancer

Kim, Young-Woo January 2003 (has links)
No description available.
2

The Epigenetic Silencing of PMP24 During the Progression of Prostate Cancer from an Androgen-Dependent to Androgen-Independent State in the LNCAP Cell Model: a Dissertation

Wu, Mengchu 20 January 2005 (has links)
One important objective of prostate cancer (PCa) research is to understand the molecular basis underlying the progression of these cancers from an androgen dependent to an androgen independent state. Hypermethylation of the promoter CpG islands is associated with the transcriptional silencing of specific gene sets in each tumor type and subtype. Transcriptional silencing of antitumor genes via CpG island hypermethylation could be a mechanism mediating PCa progression from an androgen-dependent to an androgen-independent state. Hypermethylation associated gene silencing has been reported for a great number of genes in PCa with the exception of the genes that undergo methylation associated silencing specifically during cancer development to androgen independence. The first aim of this thesis is to identify novel glenes which undergo DNA hypermethylation associated gene silencing during the cancer progression. The androgen-dependent (AD, as defined as the inability of celill to proliferate in the absence of androgen) PCa cell line LNCaP gives rise to the androgen-independent (AI) subline LNCaPcs generated by maintaining LNCaP in medium with charcoal-stripped (CS) serum for over 30 passages. This LNCaP cell model was used to identify differentially methylated sequences between the two genomes using the Methylation-Sensitive Restriction Fingerprinting (MSRF) technique. One sequence identified is located in a 5' CpG island, which encompasses part of the promoter, exon 1, and part of intron 1, of the Peroxisomal Membrane Protein 24 KD (PMP24) gene. PMP24 is silenced in concert with the hypermethylation of its CpG island in AI LNCaPcsand PC-3 cell lines. The silencing is reactivated by the treatment with a DNA methyltransferase inhibitor, 5-aza-2'-deoxycytidine (5AZAdC). PMP24 is specifically silenced in PCa cancer cell lines and shows potential antitumor properties. These results demonstrate the utility of MSRF in the identification of novel, differentially methylated DNA sequences in the genome and suggest that hypermethylation-mediated silencing of PMP24 is an epigenetic event involved in PCa progression to androgen independence. The next study investigated the molecular mechanism for DNA methylation associated gene silencing of PMP24 in AI LNCaPcs and PC-3 cell lines. We demonstrated that PMP24 transcription is repressed by the disruption of transcription factor binding to a critical cis-element by hypermethylation of its promoter CpG island. We found a CpG containing activator protein 2 (AP-2) cis-element in the intron 1 of PMP24 whose first CpG dinucleotidle is essential for the sequence-specific protein binding and the promoter activity of the gene. We presented first in cellulo evidence that the methylation of AP-2 cis-element alone but not the whole CpG island, using a newly developed methylated oligonucleotides treatment, is sufficient for the downregulation of PMP24. Our study is the first to report that the silencing mechanism for PMP24 in AI LNCaPcs and PC-3 is mediated by the complete methylation of a single GpG site of AP-2 cis-element in the intron 1 part of the CpG island, which interferes with transcription factor binding. Most interestingly, the promoter CpG island of PMP24 is hypermethylated in AD LNCaP cells with the incomplete methylation specifically at the AP-2 cis-element. The silencing of PMP24 in AD LNCaP cells was reactivated not by the 5AZAdC treatment but by the treatment with Trichostatin A (TSA), a histone deacetylase inhibitor. An alternative silencing mechanism for PMP24 other than the interference with transcription factor binding by methylation is therefore likely involved at this androgen-dependent stage. During the androgen ablation process, this mechanism is either evolved by the spread of methylation in the promoter CpG island or selected against, leading to the methylation-dominant silencing mechanism in the AI cells as seen in LNCaPcsand PC-3 cells. Taken together, this thesis emphasized the important role of DNA methylation in the progression of PCa into androgen independence. Particular respect should be paid to the specific CpG dinucleotides in cis-elements critical for the promoter activity, whose complete methylation could dominate the silencing mechanism which is independent of androgen. This thesis also pointed to the importance of monitoring the effects of cell culture on the methylation status of genes. Most importantly, this thesis raised the possibility that the silencing mechanisms for PMP24 could be different in AD LNCaP cells as compared to AI LNCaPcs and PC-3 cells. Either the evolution of such mechanism or the selectivity against it during the androgen ablation process would result in a methylation-dominant silencing mechanism of the genes such as PMP24 in AI cells and may contribute to the overall androgen independence of the cells.
3

Etude de la fonction de TFF1 dans le cancer du sein / Study of the TFF1 function in breast cancer

Simoes, David 17 July 2013 (has links)
Afin d’étudier la fonction de TFF1 (Trefoil Factor 1) dans les cancers du sein, de grandes quantités de TFF1 recombinant semblable au TFF1 humain physiologique sont indispensables.Parmi différents systèmes de production étudiés, seul TFF1 recombinant produit dans E.Coli s’est avéré avoir le bon poids moléculaire et être fonctionnel. En effet, la présence de TFF1 produit dans E.Coli a permis d’augmenter la capacité migratoire des cellules épithéliales mammaires MCF10A de 70 % au cours d’un test de reconstitution in vitro.L’étude de l’hétérodimère TFF1-GKN2 (GKN: Gastrokine2) , précédemment mis en évidence dans l’estomac humain sain, suggère que sa formation est peu spécifique. De plus, la co-exprimant TFF1 et GKN2 dans les cellules gastriques n’a pas d’impact, ni sur la prolifération, ni sur la migration cellulaire.En conclusion, seul le système d’expression bactérien permet l’obtention de TFF1 recombinant fonctionnel. Une formation et une éventuelle fonction de l’hétérodimère TFF1-GKN2 semblent peu probables. / In order to study the function of TFF1 (Trefoil Factor 1) in breast cancer, important quantities of recombinant physiologic-like TFF1 peptide is required.Among several production system studied, only E. coli produced TFF1 has a molecular weight identical to the physiological one and is active. Indeed, when compared to the negative control, recombinant TFF1 peptide produced in E. coli can increase the migratory capabilities of MCF10A (mammary epithelial cells) cells by 70% in a wound healing assay.The study of the heterodimer TFF1-GKN2 (GKN: Gastrokine2) , previously shown to form in healthy human stomach, suggest that its formation is not specific. Furthermore the co-expression of TFF1 and GKN2 in gastric cell lines did not impact migration nor proliferation of these cells.In conclusion, only the bacterial expression system (E. coli) has allowed us to obtain functional recombinant TFF1. The specific formation of TFF1-GKN2 heterodimer in vitro seems to be very weak.
4

Effects of a widely conserved AvrE-family effector and the phytotoxin coronatine on host plant defense signaling pathways

Turo, Alexander Joshua January 2021 (has links)
No description available.
5

Growth factor activation of ErbB2/ErbB3 signaling pathways regulate the activity of Estrogen Receptors (ER)

Sanchez, Melanie 04 1900 (has links)
La signalisation par l’estrogène a longtemps été considérée comme jouant un rôle critique dans le développement et la progression des cancers hormono-dépendants tel que le cancer du sein. Deux tiers des cancers du sein expriment le récepteur des estrogènes (ER) qui constitue un élément indiscutable dans cette pathologie. L’acquisition d’une résistance endocrinienne est cependant un obstacle majeur au traitement de cette forme de cancer. L’émergence de cancers hormono-indépendants peut est produite par l’activation de ER en absence d’estrogène, l’hypersensibilité du récepteur aux faibles concentrations plasmique d’estrogène ainsi que l’activation de ER par des modulateurs sélectifs. L’activité du ER est fortement influencée par l’environnement cellulaire tel que l’activation de voie de signalisation des facteurs de croissances, la disponibilité de protéines co-régulatrices et des séquences promotrices ciblées. Présentement, les études ont principalement considérées le rôle de ERα, cependant avec la découverte de ERβ, notre compréhension de la diversité des mécanismes potentiels impliquant des réponses ER-dépendantes s’est améliorée. L’activation des voies des kinases par les facteurs de croissance entraîne le développement d’un phénotype tumoral résistant aux traitements actuels. Nos connaissances des voies impliquées dans l’activation de ER sont restreintes. ERα est considéré comme le sous-type dominant et corrèle avec la plupart des facteurs de pronostic dans le cancer du sein. Le rôle de ERβ reste imprécis. Les résultats présentés dans cette thèse ont pour objectif de mieux comprendre l’implication de ERβ dans la prolifération cellulaire par l’étude du comportement de ERβ et ERα suite à l’activation des voies de signalisation par les facteurs de croissance. Nous démontrons que l’activation des récepteurs de surfaces de la famille ErbB, spécifiquement ErbB2/ErbB3, inhibe l’activité transcriptionnelle de ERβ, malgré la présence du coactivateur CBP, tout en activant ERα. De plus, l’inhibition de ERβ est attribuée à un résidu sérine (Ser-255) situé dans la région charnière, absente dans ERα. Des études supplémentaires de ErbB2/ErbB3 ont révélé qu’ils activent la voie PI3K/Akt ciblant à son tour la Ser-255. En effet, cette phosphorylation de ERβ par PI3K/Akt induit une augmentation de l’ubiquitination du récepteur qui promeut sa dégradation par le système ubiquitine-protéasome. Cette dégradation est spécifique pour ERβ. De façon intéressante, la dégradation par le protéasome requiert la présence du coactivateur CBP normalement requis pour l’activité transcriptionnelle des récepteurs nucléaires. Malgré le fait que l’activation de la voie PI3K/Akt corrèle avec une diminution de l’expression des gènes sous le contrôle de ERβ, on observe une augmentation de la prolifération des cellules cancéreuses. L’inhibition de la dégradation de ERβ réduit cette prolifération excessive causée par le traitement avec Hrgβ1, un ligand de ErbB3. Un nombre croissant d’évidences indique que les voies de signalisations des facteurs de croissance peuvent sélectivement réguler l’activité transcriptionnelle de sous-types de ER. De plus, le ratio ERα/ERβ dans les cancers du sein devient un outil de diagnostique populaire afin de déterminer la sévérité d’une tumeur. En conclusion, la caractérisation moléculaire du couplage entre la signalisation des facteurs de croissance et la fonction des ERs permettra le développement de nouveaux traitements afin de limiter l’apparition de cellules tumorales résistantes aux thérapies endocriniennes actuelles. / It has long been appreciated that estrogenic signaling plays a critical role in the development of hormone-dependent cancers such as breast cancer. Two-thirds of breast cancers express estrogen receptor (ER) which has been demonstrated to play an irrefutable role in tumour development and progression. However the acquisition of endocrine resistance has become a major obstacle in the treatment of hormone-dependent cancers that have acquired a hormone-independent state. Hormone-independent cancers emerge from an array of pathways involving ER activation in the absence of estrogen, hypersensitivity of ER to low serum levels of estrogen and activation by estrogen antagonists. The activity of ER is critically influenced by the cellular environment such as growth factor signaling pathways, availability of coregulatory proteins and the promoter sequence of target genes. The mechanisms studied have mostly considered the role of ERα, however with the discovery of the second subtype, ERβ, the understanding on the diversity of potential mechanisms involving ER-dependent responses have improved. Hormonal-independent activation of ER can occur in estrogen-dependent breast tumours, with concomitant rise in kinase signaling pathways, resulting in the acquisition of a therapeutic resistant phenotype in treated women. Our knowledge is relatively limited on which pathways trigger ER signaling and how these phosphorylation-coupled events affect ER activity. ERα is considered the dominant subtype and correlates with most of the prognostic factors in breast cancers. Conversely the role of ERβ remains unclear. The results presented in this thesis were carried out with the objective of gaining a better understanding of ERβ’s role in cellular proliferation by examining the behavior of ERβ and ERα during the activation of growth factor signaling pathways by cell-surface receptor-tyrosine kinases. We demonstrate here that the activation of cell surface receptors of the ErbB family, specifically ErbB2/ErbB3, inhibits the transcriptional activity of ERβ despite the presence of the coactivator CBP, yet activated ERα. Furthermore the inhibition of ERβ was attributed to a specific serine residue located within the hinge region, not present in ERα. Additional studies of ErbB2/ErbB3-initiated signaling revealed that it triggered the activation of the PI3K/Akt pathway which targeted the serine residue within the hinge region of ERβ. In fact, phosphorylation of ERβ by the PI3K/Akt pathway led to an increase in receptor ubiquitination which promoted its degradation by the ubiquitin-proteasome system which was subtype specific. Interestingly, proteasomal degradation required the presence of the coactivator CBP, which is normally involved in assisting nuclear receptor transcriptional activity. Although the activation of the PI3K/Akt pathway correlated with a decrease in the expression of ERβ target genes it led to an increase in the proliferation of breast cancer cells. Inhibiting the degradation of ERβ reduced the enhanced proliferation of breast cancer cells brought about by the treatment of ErbB3’s ligand, Hrgβ1. Increasing evidence indicates that growth factor signaling pathways can selectively regulate the transcriptional activity of ER subtypes, and the ratio of ERα/ERβ expression in breast tumours is becoming a popular prognostic factor to evaluate the severity of the tumour. Therefore the molecular characterization of the coupling between growth factor signaling and ER function should provide improved therapeutical approaches to overcome or delay the onset of resistance to endocrine therapy in hormone-dependent cancers.
6

Growth factor activation of ErbB2/ErbB3 signaling pathways regulate the activity of Estrogen Receptors (ER)

Sanchez, Melanie 04 1900 (has links)
La signalisation par l’estrogène a longtemps été considérée comme jouant un rôle critique dans le développement et la progression des cancers hormono-dépendants tel que le cancer du sein. Deux tiers des cancers du sein expriment le récepteur des estrogènes (ER) qui constitue un élément indiscutable dans cette pathologie. L’acquisition d’une résistance endocrinienne est cependant un obstacle majeur au traitement de cette forme de cancer. L’émergence de cancers hormono-indépendants peut est produite par l’activation de ER en absence d’estrogène, l’hypersensibilité du récepteur aux faibles concentrations plasmique d’estrogène ainsi que l’activation de ER par des modulateurs sélectifs. L’activité du ER est fortement influencée par l’environnement cellulaire tel que l’activation de voie de signalisation des facteurs de croissances, la disponibilité de protéines co-régulatrices et des séquences promotrices ciblées. Présentement, les études ont principalement considérées le rôle de ERα, cependant avec la découverte de ERβ, notre compréhension de la diversité des mécanismes potentiels impliquant des réponses ER-dépendantes s’est améliorée. L’activation des voies des kinases par les facteurs de croissance entraîne le développement d’un phénotype tumoral résistant aux traitements actuels. Nos connaissances des voies impliquées dans l’activation de ER sont restreintes. ERα est considéré comme le sous-type dominant et corrèle avec la plupart des facteurs de pronostic dans le cancer du sein. Le rôle de ERβ reste imprécis. Les résultats présentés dans cette thèse ont pour objectif de mieux comprendre l’implication de ERβ dans la prolifération cellulaire par l’étude du comportement de ERβ et ERα suite à l’activation des voies de signalisation par les facteurs de croissance. Nous démontrons que l’activation des récepteurs de surfaces de la famille ErbB, spécifiquement ErbB2/ErbB3, inhibe l’activité transcriptionnelle de ERβ, malgré la présence du coactivateur CBP, tout en activant ERα. De plus, l’inhibition de ERβ est attribuée à un résidu sérine (Ser-255) situé dans la région charnière, absente dans ERα. Des études supplémentaires de ErbB2/ErbB3 ont révélé qu’ils activent la voie PI3K/Akt ciblant à son tour la Ser-255. En effet, cette phosphorylation de ERβ par PI3K/Akt induit une augmentation de l’ubiquitination du récepteur qui promeut sa dégradation par le système ubiquitine-protéasome. Cette dégradation est spécifique pour ERβ. De façon intéressante, la dégradation par le protéasome requiert la présence du coactivateur CBP normalement requis pour l’activité transcriptionnelle des récepteurs nucléaires. Malgré le fait que l’activation de la voie PI3K/Akt corrèle avec une diminution de l’expression des gènes sous le contrôle de ERβ, on observe une augmentation de la prolifération des cellules cancéreuses. L’inhibition de la dégradation de ERβ réduit cette prolifération excessive causée par le traitement avec Hrgβ1, un ligand de ErbB3. Un nombre croissant d’évidences indique que les voies de signalisations des facteurs de croissance peuvent sélectivement réguler l’activité transcriptionnelle de sous-types de ER. De plus, le ratio ERα/ERβ dans les cancers du sein devient un outil de diagnostique populaire afin de déterminer la sévérité d’une tumeur. En conclusion, la caractérisation moléculaire du couplage entre la signalisation des facteurs de croissance et la fonction des ERs permettra le développement de nouveaux traitements afin de limiter l’apparition de cellules tumorales résistantes aux thérapies endocriniennes actuelles. / It has long been appreciated that estrogenic signaling plays a critical role in the development of hormone-dependent cancers such as breast cancer. Two-thirds of breast cancers express estrogen receptor (ER) which has been demonstrated to play an irrefutable role in tumour development and progression. However the acquisition of endocrine resistance has become a major obstacle in the treatment of hormone-dependent cancers that have acquired a hormone-independent state. Hormone-independent cancers emerge from an array of pathways involving ER activation in the absence of estrogen, hypersensitivity of ER to low serum levels of estrogen and activation by estrogen antagonists. The activity of ER is critically influenced by the cellular environment such as growth factor signaling pathways, availability of coregulatory proteins and the promoter sequence of target genes. The mechanisms studied have mostly considered the role of ERα, however with the discovery of the second subtype, ERβ, the understanding on the diversity of potential mechanisms involving ER-dependent responses have improved. Hormonal-independent activation of ER can occur in estrogen-dependent breast tumours, with concomitant rise in kinase signaling pathways, resulting in the acquisition of a therapeutic resistant phenotype in treated women. Our knowledge is relatively limited on which pathways trigger ER signaling and how these phosphorylation-coupled events affect ER activity. ERα is considered the dominant subtype and correlates with most of the prognostic factors in breast cancers. Conversely the role of ERβ remains unclear. The results presented in this thesis were carried out with the objective of gaining a better understanding of ERβ’s role in cellular proliferation by examining the behavior of ERβ and ERα during the activation of growth factor signaling pathways by cell-surface receptor-tyrosine kinases. We demonstrate here that the activation of cell surface receptors of the ErbB family, specifically ErbB2/ErbB3, inhibits the transcriptional activity of ERβ despite the presence of the coactivator CBP, yet activated ERα. Furthermore the inhibition of ERβ was attributed to a specific serine residue located within the hinge region, not present in ERα. Additional studies of ErbB2/ErbB3-initiated signaling revealed that it triggered the activation of the PI3K/Akt pathway which targeted the serine residue within the hinge region of ERβ. In fact, phosphorylation of ERβ by the PI3K/Akt pathway led to an increase in receptor ubiquitination which promoted its degradation by the ubiquitin-proteasome system which was subtype specific. Interestingly, proteasomal degradation required the presence of the coactivator CBP, which is normally involved in assisting nuclear receptor transcriptional activity. Although the activation of the PI3K/Akt pathway correlated with a decrease in the expression of ERβ target genes it led to an increase in the proliferation of breast cancer cells. Inhibiting the degradation of ERβ reduced the enhanced proliferation of breast cancer cells brought about by the treatment of ErbB3’s ligand, Hrgβ1. Increasing evidence indicates that growth factor signaling pathways can selectively regulate the transcriptional activity of ER subtypes, and the ratio of ERα/ERβ expression in breast tumours is becoming a popular prognostic factor to evaluate the severity of the tumour. Therefore the molecular characterization of the coupling between growth factor signaling and ER function should provide improved therapeutical approaches to overcome or delay the onset of resistance to endocrine therapy in hormone-dependent cancers.
7

Razvoj bioloških testova za identifikaciju liganada steroidnih receptora i ispitivanje aktivnosti steroidogenog enzima aromataze / Development of biological assays for identification of steroid receptor ligands and determination of activity of steroidogenic enyzme aromatase

Bekić Sofija 07 August 2020 (has links)
<p>U ovoj doktorskoj disertaciji&nbsp; razvijen&nbsp; je fluorescentni test u kvascu za identifikaciju potencijalnih prirodnih ili sintetičkih liganada&nbsp; ER&alpha;, ER&beta; ili AR i kvantifikaciju&nbsp; njihovog&nbsp; afiniteta&nbsp; vezivanja sa mogućno&scaron;ću testiranja čitavih biblioteka modifikovanih steroida i ksenoestrogena. Takođe, opisana&nbsp; je primena optimizovanog biosenzora&nbsp; za&nbsp; procenu&nbsp; estrogenog&nbsp; potencijala sintetskih steroida i odabranih biljnih ekstrakata bogatih jedinjenjima fitoestrogenih osobina. U cilju potpunijeg sagledavanja mehanizma&nbsp; delovanja&nbsp; odabranih&nbsp; modifikovanih&nbsp; steroida&nbsp; ispitana&nbsp; je&nbsp; njihova antiproliferativna aktivnost prema ćelijskim&nbsp; linijama estrogen receptor pozitivnog kancera dojke&nbsp; (MCF-7) i kancera prostate (PC-3), dok su&nbsp; in silico metodom molekularnog&nbsp; dokinga&nbsp; predviđene&nbsp; energije&nbsp; i&nbsp; geometrije&nbsp; vezivanja&nbsp; ovih&nbsp; jedinjenja za ligand-vezujuće&nbsp; domene&nbsp; ER&alpha; i ER&beta;. Drugi deo ovog rada obuhvata razvoj testa za&nbsp; ispitivanje aktivnosti humanog enzima aromataze,&nbsp; heterologno eksprimiranog u ćelijama&nbsp; kvasca&nbsp; Saccharomyces cerevisiae&nbsp; i/ili&nbsp; bakterija Escherichia coli, u prisustvu&nbsp; ili&nbsp; odsustvu&nbsp; inhibitora.&nbsp; Interakcije modifikovanih&nbsp; steroida, odabranih na osnovu strukture,&nbsp; sa&nbsp; aromatazom&nbsp; ispitane&nbsp; su&nbsp; osetljivim spektroskopskim metodama, praćenjem promene spinskog stanja Fe iz hem grupe ili promene fluorescencije ostatka&nbsp; triptofana&nbsp; iz&nbsp; aktivnog&nbsp; centra usled konformacione&nbsp; promene&nbsp; proteina, izazvane interakcijom sa ligandom. Razvijeni in vitro testovi bez upotrebe radioaktivnih izotopa su, osim&nbsp; visoke efikasnosti&nbsp; i&nbsp; bezbednosti&nbsp; po&nbsp; korisnika&nbsp; i&nbsp; okolinu, pokazali&nbsp; specifičnost&nbsp; i&nbsp; ekonomičnost&nbsp; u preliminarnom&nbsp; skriningu&nbsp; liganada&nbsp; steroidnih receptora&nbsp; i inhibitora aromataze. Jedinjenja&nbsp; kod kojih je detektovana&nbsp; značajna biolo&scaron;ka aktivnost mogu potencijalno poslužiti kao osnova za razvoj terapeutika u lečenju hormon-zavisnih bolesti i stanja, koja danas predstavljaju globalni zdravstveni problem.</p> / <p>In&nbsp; this&nbsp; doctoral&nbsp; dissertation,&nbsp; a&nbsp; fluorescent&nbsp; assay&nbsp; in&nbsp; yeast&nbsp; was&nbsp; developed&nbsp; for&nbsp; identification&nbsp; of&nbsp; potential&nbsp; natural or synthetic ligands of ER&alpha;, ER&beta; or AR and<br />quantification&nbsp; of&nbsp; their&nbsp; binding&nbsp; affinity,&nbsp; as&nbsp; well&nbsp; asevaluation&nbsp; of&nbsp; the&nbsp; estrogenic&nbsp; potential&nbsp; of&nbsp; synthetic steroids&nbsp; and&nbsp; selected&nbsp; plant&nbsp; extracts&nbsp; rich&nbsp; in phytoestrogen&nbsp; content.&nbsp; The&nbsp; assay&nbsp; could&nbsp; be&nbsp; used&nbsp; to&nbsp; screen&nbsp; libraries&nbsp; of&nbsp; modified&nbsp; steroids&nbsp; and xenoestrogens.&nbsp; In&nbsp; order&nbsp; to&nbsp; better&nbsp; understand&nbsp; the biomedical&nbsp; potential&nbsp; of&nbsp; selected&nbsp; modified&nbsp; steroids, results&nbsp; were&nbsp; compared&nbsp; to&nbsp; antiproliferative&nbsp; activity against&nbsp; estrogen&nbsp; receptor&nbsp; positive&nbsp; breast&nbsp; cancer (MCF-7)&nbsp; and&nbsp; prostate&nbsp; cancer&nbsp; (PC-3)&nbsp; cell&nbsp; lines. Binding&nbsp; energies&nbsp; and&nbsp; the&nbsp; geometry&nbsp; of&nbsp; binding&nbsp; of these&nbsp; compounds&nbsp; for&nbsp; ER&alpha;&nbsp; and&nbsp; ER&beta;&nbsp; ligand&nbsp; binding domains&nbsp; were&nbsp; predicted&nbsp; in&nbsp; silico&nbsp; by&nbsp; molecular&nbsp; docking&nbsp; methods.&nbsp; The&nbsp; second&nbsp; part&nbsp; of&nbsp; this&nbsp; study includes development&nbsp; of&nbsp; an&nbsp; assay&nbsp; for&nbsp; study&nbsp; of&nbsp; aromatase&nbsp; activity&nbsp; in&nbsp; the&nbsp; presence&nbsp; or&nbsp; absence&nbsp; of inhibitors&nbsp; by&nbsp; heterologous&nbsp; expression&nbsp; of&nbsp; human aromatase&nbsp; in&nbsp; Saccharomyces&nbsp; cerevisiae&nbsp; and/or Escherichia&nbsp; coli&nbsp; cells,&nbsp; as&nbsp; model-organisms. Furthermore, interactions between modified steroids, selected&nbsp; according&nbsp; to&nbsp; their&nbsp; structure,&nbsp; and&nbsp; aromatase were&nbsp; tested&nbsp; using&nbsp; sensitive&nbsp; spectroscopic&nbsp; methods based on ligand-induced changes&nbsp; in&nbsp; the&nbsp; spin state of Fe&nbsp; from&nbsp; the&nbsp; heme&nbsp; group&nbsp; or&nbsp; changes&nbsp; in&nbsp; the fluorescence&nbsp; of&nbsp; a&nbsp; tryptophan&nbsp; residue&nbsp; in&nbsp; the&nbsp; active site.&nbsp; The&nbsp; non-radioactive&nbsp; in&nbsp; vitro&nbsp; assays&nbsp; developed&nbsp; here, besides high efficiency, user and environmental safety,&nbsp; also&nbsp; have&nbsp; greater&nbsp; specificity&nbsp; and&nbsp; are&nbsp; more cost-effective&nbsp; for&nbsp; preliminary&nbsp; screening&nbsp; of&nbsp; steroid receptor&nbsp; ligands&nbsp; and&nbsp; aromatase&nbsp; inhibitors. Additionally,&nbsp; compounds&nbsp; identified&nbsp; to&nbsp; express significant biological activity can serve as a basis for the&nbsp; development&nbsp; of&nbsp; potential&nbsp; therapeutics&nbsp; in&nbsp; the treatment&nbsp; of&nbsp; hormone-dependent&nbsp; diseases&nbsp; and conditions, a global health issue today.</p>

Page generated in 1.0089 seconds