Academic literature on the topic 'SGTA; androgen receptor; ovary'

Create a spot-on reference in APA, MLA, Chicago, Harvard, and other styles

Select a source type:

Consult the lists of relevant articles, books, theses, conference reports, and other scholarly sources on the topic 'SGTA; androgen receptor; ovary.'

Next to every source in the list of references, there is an 'Add to bibliography' button. Press on it, and we will generate automatically the bibliographic reference to the chosen work in the citation style you need: APA, MLA, Harvard, Chicago, Vancouver, etc.

You can also download the full text of the academic publication as pdf and read online its abstract whenever available in the metadata.

Journal articles on the topic "SGTA; androgen receptor; ovary"

1

Trotta, Andrew P., Eleanor F. Need, Lisa M. Butler, Luke A. Selth, Melissa A. O'Loughlin, Gerhard A. Coetzee, Wayne D. Tilley, and Grant Buchanan. "Subdomain structure of the co-chaperone SGTA and activity of its androgen receptor client." Journal of Molecular Endocrinology 49, no. 2 (June 12, 2012): 57–68. http://dx.doi.org/10.1530/jme-11-0152.

Full text
Abstract:
Ligand-dependent activity of steroid receptors is affected by tetratricopeptide repeat (TPR)-containing co-chaperones, such as small glutamine-rich tetratricopeptide repeat-containing alpha (SGTA). However, the precise mechanisms by which the predominantly cytoplasmic TPR proteins affect downstream transcriptional outcomes of steroid signaling remain unclear. In this study, we assessed how SGTA affects ligand sensitivity and action of the androgen receptor (AR) using a transactivation profiling approach. Deletion mapping coupled with structural prediction, transcriptional assays, andin vivoregulation of AR-responsive promoters were used to assess the role of SGTA domains in AR responses. At subsaturating ligand concentrations of ≤0.1 nM 5α-dihydrotestosterone, SGTA overexpression constricted AR activity by an average of 32% (P<0.002) across the majority of androgen-responsive loci tested, as well as on endogenous promotersin vivo. The strength of the SGTA effect was associated with the presence or absence of bioinformatically predicated transcription factor motifs at each site. Homodimerizaion of SGTA, which is thought to be necessary for chaperone complex formation, was found to be dependent on the structural integrity of amino acids 1–80, and a core evolutionary conserved peptide within this region (amino acids 21–40) necessary for an effect of SGTA on the activity of both exogenous and endogenous AR. This study provides new insights into the subdomain structure of SGTA and how SGTA acts as a regulator of AR ligand sensitivity. A change in AR:SGTA ratio will impact the cellular and molecular response of prostate cancer cells to maintain androgenic signals, which may influence tumor progression.
APA, Harvard, Vancouver, ISO, and other styles
2

Tetsuka, M., P. F. Whitelaw, W. J. Bremner, M. R. Millar, C. D. Smyth, and S. G. Hillier. "Developmental regulation of androgen receptor in rat ovary." Journal of Endocrinology 145, no. 3 (June 1995): 535–43. http://dx.doi.org/10.1677/joe.0.1450535.

Full text
Abstract:
Abstract Androgen receptor (AR) distribution and developmental regulation in the rat ovary were examined by semiquantitative immunohistochemistry. Ovarian AR mRNA levels were also determined by Northern analysis of total RNA and compared with the levels of cytochrome P450aromatase (P450arom), an established marker of preovulatory follicular maturity. Hypophysectomized immature female rats were treated with recombinant human (rh)-FSH and/or rh-LH, or human menopausal gonadotrophin (HMG). AR was predominately located in granulosa cells. There was no indication of specific AR immunoreactivity in thecal cells, but scattered stromal cells did stain positively. In control and LH-treated ovaries, only small preantral/early antral follicles were present. Granulosa cells in these follicles showed intense AR immunostaining. Treatment with FSH, FSH and LH or HMG stimulated varying degrees of preovulatory follicular development. In these follicles, the intensity of AR immunostaining progressively declined as follicular development progressed. In intact immature rats treated with FSH, the abundance of ovarian AR mRNA was significantly decreased to 35% of the control value while combined treatment of FSH and LH resulted in further down-regulation of AR mRNA expression to 17% of the control value. A decrease in the abundance of AR mRNA was accompanied by a simultaneous increase in the abundance of P450arom mRNA. Similar results were obtained in hypophysectomized immature rats treated with FSH and LH, suggesting an inverse relationship between AR mRNA expression and granulosa cell maturity. These results suggest that (1) the AR is most abundant in the granulosa cells of rat ovaries and (2) the expression of AR and its mRNA are developmentally regulated, being down-regulated during FSH-stimulated preovulatory follicular development. Journal of Endocrinology (1995) 145, 535–543
APA, Harvard, Vancouver, ISO, and other styles
3

Lin, Lawrence H., Maria C. P. Baracat, Gustavo A. R. Maciel, José M. Soares, and Edmund C. Baracat. "Androgen receptor gene polymorphism and polycystic ovary syndrome." International Journal of Gynecology & Obstetrics 120, no. 2 (November 24, 2012): 115–18. http://dx.doi.org/10.1016/j.ijgo.2012.08.016.

Full text
APA, Harvard, Vancouver, ISO, and other styles
4

Hampton, J. H., M. Manikkam, D. B. Lubahn, M. F. Smith, and H. A. Garverick. "Androgen receptor mRNA expression in the bovine ovary." Domestic Animal Endocrinology 27, no. 1 (July 2004): 81–88. http://dx.doi.org/10.1016/j.domaniend.2004.01.005.

Full text
APA, Harvard, Vancouver, ISO, and other styles
5

Salinas, Irving, Niharika Sinha, and Aritro Sen. "Androgen-induced epigenetic modulations in the ovary." Journal of Endocrinology 249, no. 3 (June 2021): R53—R64. http://dx.doi.org/10.1530/joe-20-0578.

Full text
Abstract:
In recent years, androgens have emerged as critical regulators of female reproduction and women’s health in general. While high levels of androgens in women are associated with polycystic ovary syndrome (PCOS), recent evidence suggests that a certain amount of direct androgen action through androgen receptor is also essential for normal ovarian function. Moreover, prenatal androgen exposure has been reported to cause developmental reprogramming of the fetus that manifests into adult pathologies, supporting the Developmental Origins of Health and Disease (DOHaD) hypothesis. Therefore, it has become imperative to understand the underlying mechanism of androgen actions and its downstream effects under normal and pathophysiological conditions. Over the years, there has been a lot of studies on androgen receptor function as a transcriptional regulator in the nucleus as well as androgen-induced rapid extra-nuclear signaling. Conversely, new evidence suggests that androgen actions may also be mediated through epigenetic modulation involving both the nuclear and extra-nuclear androgen signaling. This review focuses on androgen-induced epigenetic modifications in female reproduction, specifically in the ovary, and discusses emerging concepts, latest perceptions, and highlight the areas that need further investigation.
APA, Harvard, Vancouver, ISO, and other styles
6

Abbott, David H. "Neuronal androgen receptor: Molecular gateway to polycystic ovary syndrome?" Proceedings of the National Academy of Sciences 114, no. 16 (April 4, 2017): 4045–47. http://dx.doi.org/10.1073/pnas.1703436114.

Full text
APA, Harvard, Vancouver, ISO, and other styles
7

Walters, Kirsty A. "Polycystic ovary syndrome: Is it androgen or estrogen receptor?" Current Opinion in Endocrine and Metabolic Research 12 (June 2020): 1–7. http://dx.doi.org/10.1016/j.coemr.2020.01.003.

Full text
APA, Harvard, Vancouver, ISO, and other styles
8

Duffy, Diane M., Salah E. Abdelgadir, Kenneth R. Stott, John A. Resko, Richard L. Stouffer, and Mary B. Zelinski-Wooten. "Androgen Receptor mRNA Expression in the Rhesus Monkey Ovary." Endocrine 11, no. 1 (1999): 23–30. http://dx.doi.org/10.1385/endo:11:1:23.

Full text
APA, Harvard, Vancouver, ISO, and other styles
9

Hirai, Mitsuo, Shuji Hirata, Takaaki Osada, Kazuki Hagihara, and Junzo Kato. "Androgen receptor mRNA in the rat ovary and uterus." Journal of Steroid Biochemistry and Molecular Biology 49, no. 1 (May 1994): 1–7. http://dx.doi.org/10.1016/0960-0760(94)90294-1.

Full text
APA, Harvard, Vancouver, ISO, and other styles
10

Wang, Fangfang, Jiexue Pan, Ye Liu, Qing Meng, Pingping Lv, Fan Qu, Guo-Lian Ding, et al. "Alternative splicing of the androgen receptor in polycystic ovary syndrome." Proceedings of the National Academy of Sciences 112, no. 15 (March 30, 2015): 4743–48. http://dx.doi.org/10.1073/pnas.1418216112.

Full text
Abstract:
Polycystic ovary syndrome (PCOS) is one of the most common female endocrine disorders and a leading cause of female subfertility. The mechanism underlying the pathophysiology of PCOS remains to be illustrated. Here, we identify two alternative splice variants (ASVs) of the androgen receptor (AR), insertion and deletion isoforms, in granulosa cells (GCs) in ∼62% of patients with PCOS. AR ASVs are strongly associated with remarkable hyperandrogenism and abnormalities in folliculogenesis, and are absent from all control subjects without PCOS. Alternative splicing dramatically alters genome-wide AR recruitment and androgen-induced expression of genes related to androgen metabolism and folliculogenesis in human GCs. These findings establish alternative splicing of AR in GCs as the major pathogenic mechanism for hyperandrogenism and abnormal folliculogenesis in PCOS.
APA, Harvard, Vancouver, ISO, and other styles

Dissertations / Theses on the topic "SGTA; androgen receptor; ovary"

1

Martínez, Pinto Jonathan Eloy. "Neonatal exposure to estradiol reprograms the expression of androgen receptor and anti-müllerian hormone [recurso electrónico]: short and long term effects and their relation to the polycystic ovary phenotype." Tesis, Universidad de Chile, 2014. http://www.repositorio.uchile.cl/handle/2250/116937.

Full text
Abstract:
Doctor en Bioquímica
Reproduction is regulated through the integration of information that comes from the hypothalamus, hypophysis, and the ovary. There are critical hormone sensitive periods during development during which they are especially vulnerable to exposure to abnormal hormonal levels resulting from metabolic problems or environmental sources. These exposures may permanently alter the differentiation and function of reproductive organs. Several studies performed in humans and animal models have suggested that polycystic ovary syndrome (PCOS) originates during early development due to exposure to abnormal steroidal hormone levels. Supporting this, our group recently reported that the administration of a single dose of estradiol valerate (EV) to newborn rats can irreversibly program the polycystic ovary condition during adulthood. However, there is no information about how this neonatal exposure to a single dose of EV can determine the functional and structural changes seen in the adult ovary. The aim of this research was to determine if there are genes—primarily those most reported to be related to PCOS in humans—with permanently altered expression and if these expression patterns are due to modifications in the methylation pattern in their DNA sequence. A single dose (10 mg/kg) of EV was administered to neonatal rats, and PCR array and real-time PCR analyses were conducted to examine the subsequent gene expression patterns of growth factors, nuclear receptors, and coregulators. Twenty-four hours after the exposure, the EV-exposed rat ovaries expressed more androgen receptor (Ar) than did the control ovaries; in the 60-day old rats, the Ar mRNA levels decreased 6.4-fold relative to the controls. The interstitial tissue and antral follicles from the adult EV-treated rats expressed more Ar than did the control preantral follicles, suggesting a failure of the control mechanism for Ar expression in antral follicles. Using the mass-array technique, the methylation patterns of different transcription factor binding sites were found to be associated with the Ar gene. NBRE, the response element of nerve growth factor-induced B (Ngfi-b), was hypomethylated in the Ar gene from the EV-treated ovaries. There was also an increase in anti-Müllerian hormone (AMH) expression in adult ovaries (mRNA and protein) that was induced by the neonatal exposure to estradiol. The EV-treated rat ovaries had a higher level of AMH immunoreactivity in the antral follicles than did the controls; however, no significant differences were seen between the preantral follicles of the treated and control groups. The methylation pattern of the Amh gene from the ovaries of EV-treated adult rats showed differential methylation in the CpGs related to the estradiol response element (ERE) in the Amh gene. The neonatal ovary samples had hypermethylated CpGs in comparison to the 30- and 60-day old rats; the samples (granulosa cells) from the 60-day old rats had more methylation of the CpGs than did those from the 30-day old rats; thus, the methylation pattern depended on the stage of development. When we compared the EV-exposed and control rats, we found more methylation of the CpGs in the samples from the 60-day old EV-exposed rats than in those from the controls; there were no differences between the groups for the 2- and 30-day old rats. This work demonstrated that the neonatal exposure to estradiol induces an overexpression of AMH and AR in the rat ovary. The mechanism by which these changes are induced may involve an increase in the methylation of the ERE associated with the Amh gene, suggesting that the change in methylation allows ESR1 to induce Amh expression. The hypomethylation of NBRE associated with the AR gene suggests that AR expression may be induced in response to nerve growth factor or luteinizing hormone. These epigenetic modifications found in the current rat model provide a new framework for understanding the genesis of the polycystic ovary and its maintenance in humans, allowing more focus on the effects due to estrogen exposure
Die Fortpflanzung in Säugetieren wird durch Signale aus dem Hypothalamus, der Hypophyse und der Gebärmutter gesteuert. Während der Anlage der Gebärmutter in der Embryonalentwicklung gibt es ein kritisches Zeitfenster bei dem die Entwicklung durch veränderte Hormonspiegel oder andere äußere Einflüsse gestört werden kann. Dabei kann es zur permanenten Schädigung der reproduktiven Organe kommen. Studien am Menschen und in Tiermodellen legen nahe, dass das sogenannte Polyzystische Ovarialsyndrom durch erhöhte Steroidhormonspiegel verursacht wird. So konnte unsere Arbeitsgruppe kürzlich zeigen, dass bei neugeborenen Ratten die Gabe einer einzigen Dosis Estradiolvalerat (EV) ausreicht um ein Polyzystisches Ovarialsyndrom in den erwachsenen Tieren zu erzeugen. Die zugrundeliegenden Mechanismen sind allerdings bis heute nicht bekannt. Ziel dieser Arbeit war es zu untersuchen, ob die Expression bestimmter Gene in erwachsenen Ratten durch die Estradiolvaleratgabe im neonatalen Stadium permanent verändert wird und ob die veränderte Genexpression mit epigenetischen Veränderungen auf der Ebene der DNA Methylierung einhergeht. Hierzu wurden neugeborene Ratten mit einer einzigen Dosis EV (10 mg/Kg) behandelt um anschließend die Genexpression von Wachstumsfaktoren, nukleären Hormonrezeptoren und ihre Kofaktoren mittels PCR Arrays und quantitativer RT-PCR zu untersuchen. Wir konnten zeigen, dass die Ovarien von EV-behandelten Ratten das Androgenrezeptorgen (Ar) nach 24h stärker exprimierten als die Kontrollen. Nach 60 Tagen ging die ovariale Ar Expression allerdings um das 4-6-fache gegenüber unbehandelten Tieren zurück. Histologisch war die Ar Expression in antralen Follikeln und im Interstitium vom EV-behandelten adulten Tieren stärker als in preantralen Follikeln, was auf eine Störung von Regulationsmechanismen in antralen Follikeln hindeutet. Mittels MassArray-Technologie konnte außerdem gezeigt werden, dass die veränderte Ar Expression in EV-behandelten Tieren mit der verringerten DNA Methylierung einer potentiellen Transkriptionsfaktorbindestelle (NBRE: nerve growth factor-induced B response element) korreliert. Daneben war die Expression des Anti-Müller-Hormons (AMH) in Ovarien von EV-behandelten adulten Tieren sowohl auf mRNA als auch auf Proteinebene erhöht. Immunhistologisch zeigten insbesondere die antralen Follikeln von EV-behandelten adulten Tieren (nicht aber Interstitium und preantrale Follikeln) eine stärkere Färbung für AMH. Auch im Amh Gen konnten entwicklungs- und behandlungsabhängige Veränderungen der DNA Methylierung gemessen werden. Eine Bindungsstelle für den Estrogenrezeptor (ERE: estradiol response element) im Amh Gen zeigte Methylierungsunterschiede in Ovarien von adulten EV-behandelten Ratten. In normalen Tieren war der Methylierungsstatus dieser Region abhängig vom Entwicklungsstadium: im Vergleich zu 30 und 60 Tage alten Ratten war das Element in neugeborenen Ratten hypermethyliert und in Granulosazellpräperaten von 30 Tage alten Ratten weniger methyliert als in Präparaten von 60 Tage alten Tieren. Im Vergleich mit EV-behandelten Tieren zeigten Proben von 60 Tage alten Tieren eine signifikant erhöhte DNA Methylierung, während EV-behandelte neugeborene und 30 Tage alte Tiere sich nicht signifikant von den Kontrollen unterschieden. In dieser Arbeit konnte also gezeigt werden, dass bei Ratten die Gabe von Estradiol im neonatalen Stadium zu einer ovarialen Überproduktion von AMH und AR führt. Die Mechanismen die zu diesen Veränderungen führen könnten mit dem Verlust der DNA Methylierung im ERE des Amh Gens zusammenhängen, was vermutlich zu einer erhöhten ESR1-Bindung und AMH Expression führt. Die Hypomethylierung des NBRE im Ar Gen deutet darauf hin, dass hier NGF (nerve growth factor) oder LH (luteinisierende Hormon) an der veränderten Regulation beteiligt sind. Die im Rattenmodel gefundenen epigenetischen Veränderungen bieten neue Anhaltspunkte um die Entstehung und die Persistenz des Polyzystische Ovarialsyndroms im Menschen besser zu verstehen und zukünftige Forschung wird sich noch gründlicher mit den Effekten der Estrogenexposition auseinandersetzen müssen
Conicyt Mecesup Fondecyt
APA, Harvard, Vancouver, ISO, and other styles
2

Braun, Alyssa Maria. "The effects of androgens on steroidogenesis in the ovary of Atlantic croaker (Micropogonias undulatus) mechanism of action, the biochemical characterization of a membrane androgen receptor, and the chronic effects of exposure to an environmental antiandrogen /." Access restricted to users with UT Austin EID Full text (PDF) from UMI/Dissertation Abstracts International, 2002. http://wwwlib.umi.com/cr/utexas/fullcit?p3077433.

Full text
APA, Harvard, Vancouver, ISO, and other styles
3

Braun, Alyssa Maria. "The effects of androgens on steroidogensis in the ovary of Atlantic croaker (Micropogonias undulatus) : mechanism of action, the biochemical characterization of a membrane androgen receptor, and the chronic effects of exposure to an environmental antiandrogen /." Full text (PDF) from UMI/Dissertation Abstracts International, 2002. http://wwwlib.umi.com/cr/utexas/fullcit?p3077433.

Full text
APA, Harvard, Vancouver, ISO, and other styles
4

Martínez, Pinto Jonathan Eloy [Verfasser], Hernan [Akademischer Betreuer] Lara, and Michael [Akademischer Betreuer] Rehli. "“Neonatal exposure to estradiol reprograms the expression of androgen receptor and anti-Müllerian hormone: short and long term effects and their relation to the polycystic ovary phenotype” / Jonathan Eloy Martínez Pinto. Betreuer: Hernan Lara ; Michael Rehli." Regensburg : Universitätsbibliothek Regensburg, 2016. http://d-nb.info/1083250930/34.

Full text
APA, Harvard, Vancouver, ISO, and other styles
5

Butler, Miriam Simone. "The role of small glutamine-rich tetratricopeptide repeat containing protein alpha in female reproductive tissues." Thesis, 2012. http://hdl.handle.net/2440/80189.

Full text
Abstract:
Small glutamine-rich tetratricopeptide repeat containing protein alpha (SGTA) is an evolutionarily conserved protein that functions as a mediator of intracellular protein transport. In particular, SGTA has been shown to act as a co-chaperone protein that promotes cytoplasmic retention of the androgen receptor (AR) and thus acts to modulate AR transcriptional activity. Androgens and the AR are known to be essential for the developmental and physiological processes of male reproductive tissues and the potential importance of SGTA in male tissues has been highlighted by the reduced expression of SGTA relative to AR in metastatic prostate cancer. Although the direct actions of androgens in female reproductive tissues are not well understood, there is increasing interest due to the association of androgen levels with several aspects of female health. In females, the AR is expressed in all reproductive tissues, including the ovary and uterus. Therefore, if present in these female tissues, SGTA could feasibly have implications for the regulation of AR action. In support of this concept, recent genetic studies have identified polymorphisms in the SGTA gene that are associated with increased risk of developing polycystic ovary syndrome (PCOS), a disorder characterised by hyperandrogenism. To date, the expression and function of SGTA in female reproductive tissues has not previously been investigated. Therefore, the overall objective of this thesis was to examine the potential role of SGTA in female reproductive tissues with the following specific aims: a) compare expression and localisation of SGTA and AR, b) ascertain whether SGTA is altered in select female reproductive diseases, and c) determine whether SGTA is able to influence AR subcellular localisation and subsequent action. The results demonstrated that SGTA was evident in the cytoplasm of the epithelial cells of the ovary, Fallopian tube, endometrium, and placenta. Moreover, AR expression was evident in those cells that expressed SGTA, suggesting that the two proteins may interact to influence androgen action as in male tissues. A novel finding of this study, that contravenes the theory of SGTA as an epithelial-specific protein in reproductive tissues, was the detection of SGTA protein in the mesenchymal-derived thecal cells of the ovary. This was further supported by similar expression in the Leydig cells of the testes, which are derived from a common mesenchymal origin. Quantitative analysis of SGTA protein levels did not reveal any difference between normal and pathologic states of the ovary. However, AR protein levels were significantly reduced in ovarian cancer tissues. As a result, the AR:SGTA ratio was significantly reduced, suggesting that genomic AR activity would be negatively impacted in the ovarian epithelial cancer cells. Using several ovarian cancer cell lines in this thesis it was demonstrated that knockdown of SGTA resulted in nuclear localisation of the AR in the absence of androgen hormone. Knockdown of AR resulted in an inhibition of ovarian cancer cell viability, which suggests AR is important in the growth of ovarian cancer cells. However, SGTA knockdown per se had no effect on ovarian cancer cell viability. Furthermore, in this thesis it was demonstrated that knockdown of either AR or SGTA affected expression levels of two genes implicated in ovarian cancer proliferation: distal-less homeobox 5 and calcium-dependent cytosolic phospholipase A₂, raising the possibility of non-genomic actions that remain to be elucidated. In conclusion, the results of this thesis support a role for SGTA in regulating AR action in female reproductive tissues and show that SGTA may influence the pathology of ovarian cancer, but the precise mechanisms remain to be resolved. The information presented in this thesis provides the basis for further investigations to evaluate the role of SGTA in female reproductive tissues.
Thesis (Ph.D.) -- University of Adelaide, School of Medicine, 2012
APA, Harvard, Vancouver, ISO, and other styles
6

Hickey, Theresa E. "Androgen receptor mediated activity in the ovary : implications for Polycystic Ovary Syndrome." 2006. http://hdl.handle.net/2440/37839.

Full text
Abstract:
Context : The expression of androgen receptors ( AR ) in follicular granulosa cells ( GC ) of mammals suggests a role for direct AR - mediated androgen activity in the regulation of folliculogenesis, however this role and the mechanistic pathways involved have not been fully characterised. In women, excess androgen is a characteristic feature of polycystic ovary syndrome ( PCOS ), but AR - mediated activity has not been widely investigated in relation to the pathophysiology of this disorder. Hypotheses : The current thesis tested two general hypotheses related to AR activity in PCOS : 1 ) The polymorphic ( CAG ) n repeat region in the AR gene, which has functional implications for receptor activity, influences the manifestation of PCOS and 2 ) AR signalling is disrupted in GC from women with PCOS. Results : In a cross - sectional population analysis, this thesis reports an association between PCOS and long CAG repeat tracts in the AR gene, which functionally represent reduced androgen sensitivity. The association was further enhanced by compensating for the influence of X chromosome inactivation ( XCI ) on expression of specific AR alleles. Preferential expression of long CAG repeat tracts positively correlated with serum testosterone levels in PCOS patients. In an analysis of sister pairs with the same CAG repeat genotype at the AR locus, different patterns of XCI were evident when sisters had a different clinical manifestation of PCOS. Collectively, these results provide evidence that supports the hypothesis that the ( CAG ) n polymorphism in the AR influences the manifestation of PCOS, the effects of which are modulated by variable allele expression via a mechanism involving XCI. These findings accord with the concept that both genetic and environmental factors are determinants of this disorder. At the level of the ovary, AR - mediated signalling in follicular GC was influenced by proximity to the oocyte in both pigs and humans. In particular, the ability of androgen to directly induce porcine GC proliferation in vitro was dependent upon presence of the oocyte or the oocyte mitogen, growth differentiation 9 ( GDF9 ). This finding provides a potential mechanism to explain how androgens may enhance early follicle growth. Granulosa cells from women with PCOS had normal mRNA expression for AR signalling molecules, but GC surrounding the oocyte in vivo had reduced AR protein content and diminished responses to androgen in culture as compared to those from normal ovaries. GC from women with PCOS also expressed mRNA for an androgen - regulated serine protease ( hKLK3 ), which did not occur in normal GC. Therefore, follicular GC from women with PCOS have evidence of perturbed AR - mediated signalling which is likely to contribute to the pathophysiology of this disorder. As AR - mediated signalling is influenced by the oocyte, the differences in AR - mediated signalling in GC from women with PCOS may be indicative of dysregulated signals emanating from the oocyte. Conclusion : The results of this thesis indicate that abnormal AR action occurs in PCOS, but further investigation is required to determine whether this phenomenon represents a primary disruption or a secondary consequence of another primary disruption in the sequence of events that leads to aberrant folliculogenesis in this disorder.
Thesis (Ph.D.)--School of Paediatrics and Reproductive Health, 2006.
APA, Harvard, Vancouver, ISO, and other styles
7

Hickey, Theresa E. "Androgen receptor mediated activity in the ovary : implications for Polycystic Ovary Syndrome." Thesis, 2006. http://hdl.handle.net/2440/37839.

Full text
Abstract:
Context : The expression of androgen receptors ( AR ) in follicular granulosa cells ( GC ) of mammals suggests a role for direct AR - mediated androgen activity in the regulation of folliculogenesis, however this role and the mechanistic pathways involved have not been fully characterised. In women, excess androgen is a characteristic feature of polycystic ovary syndrome ( PCOS ), but AR - mediated activity has not been widely investigated in relation to the pathophysiology of this disorder. Hypotheses : The current thesis tested two general hypotheses related to AR activity in PCOS : 1 ) The polymorphic ( CAG ) n repeat region in the AR gene, which has functional implications for receptor activity, influences the manifestation of PCOS and 2 ) AR signalling is disrupted in GC from women with PCOS. Results : In a cross - sectional population analysis, this thesis reports an association between PCOS and long CAG repeat tracts in the AR gene, which functionally represent reduced androgen sensitivity. The association was further enhanced by compensating for the influence of X chromosome inactivation ( XCI ) on expression of specific AR alleles. Preferential expression of long CAG repeat tracts positively correlated with serum testosterone levels in PCOS patients. In an analysis of sister pairs with the same CAG repeat genotype at the AR locus, different patterns of XCI were evident when sisters had a different clinical manifestation of PCOS. Collectively, these results provide evidence that supports the hypothesis that the ( CAG ) n polymorphism in the AR influences the manifestation of PCOS, the effects of which are modulated by variable allele expression via a mechanism involving XCI. These findings accord with the concept that both genetic and environmental factors are determinants of this disorder. At the level of the ovary, AR - mediated signalling in follicular GC was influenced by proximity to the oocyte in both pigs and humans. In particular, the ability of androgen to directly induce porcine GC proliferation in vitro was dependent upon presence of the oocyte or the oocyte mitogen, growth differentiation 9 ( GDF9 ). This finding provides a potential mechanism to explain how androgens may enhance early follicle growth. Granulosa cells from women with PCOS had normal mRNA expression for AR signalling molecules, but GC surrounding the oocyte in vivo had reduced AR protein content and diminished responses to androgen in culture as compared to those from normal ovaries. GC from women with PCOS also expressed mRNA for an androgen - regulated serine protease ( hKLK3 ), which did not occur in normal GC. Therefore, follicular GC from women with PCOS have evidence of perturbed AR - mediated signalling which is likely to contribute to the pathophysiology of this disorder. As AR - mediated signalling is influenced by the oocyte, the differences in AR - mediated signalling in GC from women with PCOS may be indicative of dysregulated signals emanating from the oocyte. Conclusion : The results of this thesis indicate that abnormal AR action occurs in PCOS, but further investigation is required to determine whether this phenomenon represents a primary disruption or a secondary consequence of another primary disruption in the sequence of events that leads to aberrant folliculogenesis in this disorder.
Thesis (Ph.D.)--School of Paediatrics and Reproductive Health, 2006.
APA, Harvard, Vancouver, ISO, and other styles
8

Zhang, Chenan. "Testosterone acts at the cell surface to induce granulosa/theca cell death via an apoptotic pathway in Atlantic croaker (Micropogonias undulatus)." Thesis, 2011. http://hdl.handle.net/2152/23902.

Full text
Abstract:
The teleost ovarian follicle undergoes extensive remodeling and regression during the reproductive cycle—a process involving apoptosis and cell death. However, the hormonal regulation of these processes remains unclear. In the current study the role of testosterone in regulating regression of Atlantic croaker (Micropogonias undulatus) ovarian follicles was investigated in co-cultured granulosa/theca (G/T) cells. Testosterone (T) treatment enhanced serum starvation-induced cell death and apoptosis of G/T cells during the mature stage of oocyte maturation. This effect was mimicked by a cell-impermeable T conjugate, T-bovine serum albumin, indicating that this androgen action is initiated at the cell surface. Mibolerone, a nuclear androgen receptor agonist, was ineffective in promoting apoptosis and cell death, which suggests that T actions are independent from the nuclear receptor. Together, the data suggests that T-induction of apoptosis and cell death are through a novel membrane androgen receptor in the croaker ovary. T treatment also increased expression of a pro-apoptotic member of the Bcl-2 gene family, Bax, and two Bax upstream regulators, JNK and p53. These results suggest that T induces cell death of G/T cells in croaker through the apoptotic pathway involving JNK, p53 and Bax. An opposite response of cell death protection by T was also observed in G/T cells cultured from late-stage ovaries. This response was accompanied by a rapid increased ERK-1/-2 phosphorylation not seen in Mibolerone treatment. By examining the role of T in croaker follicle cell death and elucidating the corresponding basic mechanisms of androgen action, we are learning more about the regulatory components involved in the breakdown and remodeling stages of the teleost reproductive cycle.
text
APA, Harvard, Vancouver, ISO, and other styles
9

Braun, Alyssa Maria 1974. "The effects of androgens on steroidogenesis in the ovary of Atlantic croaker (Micropogonias undulatus) : mechanism of action, the biochemical characterization of a membrane androgen receptor, and the chronic effects of exposure to an environmental antiandrogen." 2002. http://hdl.handle.net/2152/10926.

Full text
APA, Harvard, Vancouver, ISO, and other styles

Book chapters on the topic "SGTA; androgen receptor; ovary"

1

L. Yanes Cardozo, Licy, Alexandra M. Huffman, Jacob E. Pruett, and Damian G. Romero. "Androgens and Cardiovascular Risk Factors in Polycystic Ovary Syndrome." In Reproductive Hormones. IntechOpen, 2021. http://dx.doi.org/10.5772/intechopen.96005.

Full text
Abstract:
Polycystic Ovary Syndrome (PCOS) is the most common endocrine disorder in reproductive-aged women. Clinical or biochemical signs of androgen excess is a cardinal feature of the syndrome and are present in approximately 80% of women with PCOS. Increased blood pressure and insulin resistance, two major cardiovascular risk factors, are frequently present in women with PCOS. This chapter aims to highlight the fundamental role of androgens in mediating the increased blood pressure and insulin resistance in women with PCOS. This chapter is also a call for action to develop new pharmacological therapies that target the androgen synthesis and androgen receptor activation dysregulation present in women with PCOS. These novel therapies will allow to prevent or mitigate the excess androgen-mediated cardiovascular risk factors that affect women with PCOS.
APA, Harvard, Vancouver, ISO, and other styles
2

Sacerdote, Alan. "Rare and Underappreciated Causes of Polycystic Ovarian Syndrome." In Polycystic Ovary Syndrome [Working Title]. IntechOpen, 2022. http://dx.doi.org/10.5772/intechopen.101946.

Full text
Abstract:
While hyperinsulinemia is a common contributing mechanism in the pathogenesis of polycystic ovarian syndrome (PCOS), other mechanisms may give rise to or add to the effects of hyperinsulinemia, as well as other causes of hyperandrogenism, in the pathogenesis of PCOS. Such underappreciated causes may include autoimmune, insulin receptor mutations, mutations of post-receptor insulin signaling response elements, polymorphisms of LH, androgen, and estrogen signaling pathways, epigenetic alterations in hormonal signaling cascade response elements, infestations and infections with organisms capable of endocrine disruption by various mechanisms, as well as drugs and other chemicals which may be endocrine disruptors. In addition, alterations in the gut, oral, or vaginal biome may be associated with PCOS and insulin resistance and may, in some instances, have a role to play in its pathogenesis. In this chapter I plan to review what is known about these lesser-known causes of PCOS, in the hopes of alerting clinicians to consider them and stimulating investigators to better understand PCOS pathogenesis in general and, hopefully, develop more individualized, precision treatment and prevention strategies for the people in our care.
APA, Harvard, Vancouver, ISO, and other styles
3

Butler, Gary, and Jeremy Kirk. "Differences of sex development (DSD)." In Paediatric Endocrinology and Diabetes, 335–50. Oxford University Press, 2020. http://dx.doi.org/10.1093/med/9780198786337.003.0011.

Full text
Abstract:
• Embryology: the gonad is initially bipotential. • The testes develop under active control of SRY and other genes. Disorders of sex development (DSDs) are classified according to the karyotype: • 46,XY DSD (incomplete masculinization of a male fetus): ◦ The commonest cause is androgen insensitivity syndrome (AIS): ■ mutations in androgen receptor (AR) gene on X chromosome in complete forms ■ alterations in androgen binding in partial forms. ◦ Abnormalities of testosterone synthesis and conversion, may be: ■ isolated, e.g. 17β‎HSD, 5α‎RD ■ occur in association with defects in steroid biosynthesis, e.g. StAR, 3β‎HSD. • Pure 46,XY gonadal dysgenesis (Swyer syndrome): ◦ phenotype unambiguously female; may present with delayed puberty ◦ Müllerian structures are present but only streak gonads are seen. • Mixed gonadal dysgenesis: ◦ usually asymmetrical, e.g. ovary/streak gonad or ovotestis ◦ karyotype is 45,X/46,XY or 46,XX/46,XY. • Pure 46,XX gonadal dysgenesis: ◦ absent puberty in a phenotypically normal female ◦ intact Müllerian structures but streak ovaries; normal genitalia. • 46,XX DSD (masculinization of a female fetus): ◦ the commonest cause is congenital adrenal hyperplasia, with the vast majority (>90%) due to 21-hydroxylase deficiency (21OHD). • Ovotesticular DSD is rare: ◦ aetiology is unknown, and karyotype usually 46,XX ◦ asymmetrical gonad development; ovary and testis or ovotestis. • DSD may also be part of other genetic syndromes, e.g. Antley–Bixler, Smith–Lemli–Opitz, trisomy 13. • Management requires careful evaluation and counselling, working as part of a multidisciplinary team.
APA, Harvard, Vancouver, ISO, and other styles

Conference papers on the topic "SGTA; androgen receptor; ovary"

1

Trotta, Andrew P., Eleanor F. Need, Melissa O'Loughlin, Grant Buchanan, and Wayne D. Tilley. "Abstract 1704: The co-chaperone, SGTA, acts as a regulator of androgen receptor activity in prostate cancer." In Proceedings: AACR 101st Annual Meeting 2010‐‐ Apr 17‐21, 2010; Washington, DC. American Association for Cancer Research, 2010. http://dx.doi.org/10.1158/1538-7445.am10-1704.

Full text
APA, Harvard, Vancouver, ISO, and other styles
We offer discounts on all premium plans for authors whose works are included in thematic literature selections. Contact us to get a unique promo code!

To the bibliography