Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Katie J. Turner is active.

Publication


Featured researches published by Katie J. Turner.


Endocrinology | 1999

Permanent Effects of Neonatal Estrogen Exposure in Rats on Reproductive Hormone Levels, Sertoli Cell Number, and the Efficiency of Spermatogenesis in Adulthood*

Nina Atanassova; Chris McKinnell; Marion Walker; Katie J. Turner; Jane S. Fisher; M. Morley; Michael Millar; Nigel P. Groome; Richard M. Sharpe

This study aimed to identify the mechanism(s) for impairment of spermatogenesis in adulthood in rats treated neonatally with estrogens. Rats were treated (days 2-12) with 10, 1, or 0.1 microg diethylstilbestrol (DES), 10 microg ethinyl estradiol (EE), 10 mg/kg of a GnRH antagonist (GnRHa), or vehicle and killed in adulthood. DES/EE caused dose-dependent reductions in testis weight, total germ cell volume per testis, and Sertoli cell volume per testis. Sertoli cell number at 18 days of age in DES-treated rats was reduced dose dependently. GnRHa treatment caused changes in these parameters similar to those in rats treated with 10 microg DES. Plasma FSH levels were elevated (P < 0.001) to similar levels in all treatment groups regardless of differences in Sertoli cell number and levels of inhibin B; the latter reflected Sertoli cell number, but levels were disproportionately reduced in animals treated with high doses of DES/EE. Neonatal estrogen treatment, but not GnRHa, caused dose-dependent reductions (40-80%) in plasma testosterone levels in adulthood, but did not alter LH levels. Preliminary evidence suggests that the decrease in testosterone levels in estrogen-treated rats is not due to reduced Leydig cell volume per testis. GnRHa-treated rats exhibited a significant increase in germ cell volume per Sertoli cell and a reduction in germ cell apoptosis, probably because of the raised FSH levels. Despite similar raised FSH levels, rats treated with DES (10 or 1 microg) or EE (10 microg) had reduced germ cell volume/Sertoli cell and increased germ cell apoptosis, especially when compared with GnRHa-treated animals. The latter changes were associated with an increase in lumen size per testis, indicative of impaired fluid resorption from the efferent ducts, resulting in fluid accumulation in the testis. Rats treated neonatally with 0.1 microg DES showed reduced germ cell apoptosis comparable to that in GnRHa-treated animals. The changes in apoptotic rate among treatment groups occurred across all stages of the spermatogenic cycle. It is concluded that 1) neonatal estrogen treatment results in dose-dependent alterations in Sertoli cell numbers, germ cell volume, efficiency of spermatogenesis, and germ cell apoptosis in adulthood; 2) the relatively poor spermatogenesis in estrogen-treated animals is most likely due to altered testis fluid dynamics and/or altered Sertoli cell function; 3) as indicated by FSH (LH) and testosterone levels, the hypothalamic-pituitary axis and Leydig cells are probably more sensitive than the Sertoli cells to reprogramming by estrogens neonatally; and 4) elevated FSH levels in adulthood may improve the efficiency of spermatogenesis.


Advances in Experimental Medicine and Biology | 1997

Fetal and Perinatal Influence of Xenoestrogens on Testis Gene Expression

Philippa T. K. Saunders; Gregor Majdic; P. Parte; Michael Millar; Jane S. Fisher; Katie J. Turner; Richard M. Sharpe

The incidence of reproductive abnormalities in the male has been reported to have increased during the past 50 years. It has been suggested that these changes may be attributable to the presence of chemicals with oestrogenic activity in our environment. The aim of the experiments described in this chapter was to investigate the effects of acute exposure to high levels of xenoestrogens either indirectly during fetal life, or directly during neonatal life, on gene expression in the testis and pituitary. Fetal treatment involved administration of diethylstilbestrol (DES), 4-octylphenol (OP) or vehicle (oil, control) to pregnant rats on days 11.5 and 15.5 post coitum; fetuses were recovered on day 17.5. There was no difference between fetuses from control and treated mothers in either the overall histology of the testes or numbers of Leydig cells as determined by immunohistochemistry with an antibody directed against 3 beta-HSD. However there was a consistent and striking reduction in the amount of P450 17-a hydroxylase C17, 20 lyase (P450c17) and steroidogenic factor 1 (SF-1) detected by immunocytochemistry in testes from treatment groups given the higher doses of OP and DES. Oestrogen receptors (ER alpha) were present in the fetal leydig cells of all animals. Neonatal treatment involved direct injection of oil (control), DES, OP or Bisphenol A (Bis A) on days 2, 4, 6, 8, 10 and 12; pituitaries and testes were recovered on day 18. Testis weights and seminiferous tubule diameters were significantly reduced in animals treated with DES. In these same animals immunocytochemical localisation revealed that the amounts of FSH beta subunit and inhibin alpha subunit were reduced in their pituitaries and testes respectively. OP did not appear to have an acute, measurable effect on testis gene expression but a reduction in testis weight was noted in adult animals given the same treatment regime. The effects observed are consistent with negative feedback by oestrogens on pituitary production of FSH resulting in retarded maturation of seminiferous tubules and reduced Sertoli cell numbers. These studies have demonstrated that administration of high levels of oestrogens can affect gene expression in the testis early in life. However, the relevance of these findings to observations in man await a) a greater understanding of the physiological role(s) of oestrogens in normal males, b) an evaluation of the sources, routes of exposure, concentrations in vivo and bioavailability of xenoestrogens.


Molecular and Cellular Endocrinology | 2001

Modulation of gene expression by androgen and oestrogens in the testis and prostate of the adult rat following androgen withdrawal

Katie J. Turner; Michelle Morley; Sheila Macpherson; Michael Millar; Julie Wilson; Richard M. Sharpe; Philippa T. K. Saunders

Androgens are important for the structural and functional integrity of the testis and the prostate and this may in part be mediated by the aromatisation of testosterone to oestradiol. The aim of the present study was to establish an in vivo model that would allow the identification of genes, the expression of which was regulated acutely by androgen and/or oestrogen in the male reproductive system. In rats in which the Leydig cells were ablated by administration of ethane dimethane sulfonate (EDS) 6 days earlier, testosterone esters (T) were administered from day 0 (To), and additional animals were administered either T, 17beta-oestradiol benzoate (EB) or diethylstilbestrol (DES) for 1 or 4 h on day 6 after EDS-treatment. Nuclear immunoexpression of the androgen receptor (AR) was reduced or absent from the testis but unaffected in the ventral prostate following these treatments. ERbeta immunoexpression in these tissues was unchanged. Northern blot analysis showed that EB and DES as well as T administration 4 h earlier could modulate mRNA expression of two androgen-responsive genes, C3 and SGP-2, in the prostate. The co-administration of T or EB with the AR antagonist, flutamide, or with the ER antagonist, ICI 182,780 (ICI), did not block the suppression of SGP-2 mRNA expression by T or EB. In contrast, the upregulation of C3 mRNA expression by T was successfully antagonised by both flutamide and by ICI. A preliminary evaluation of the expression of three Sertoli cell and five germ cell mRNAs revealed that their expression was not steroid regulated. Our results support the hypothesis that the action of testosterone in the male reproductive system may in part be mediated by its conversion to oestradiol. This in vivo model should prove of value in future studies to identify androgen and oestrogen regulated genes in the male reproductive system.


Molecular and Cellular Endocrinology | 1995

Phosphatidylethanolamine binding protein is an abundant secretory product of haploid testicular germ cells in the rat

Philippa T. K. Saunders; Chris McKinnell; Michael Millar; J. Gaughan; Katie J. Turner; Bernard Jégou; Viqar Syed; Richard M. Sharpe

An abundant cellular and secretory product of isolated seminiferous tubules from adult rats was identified as having an apparent molecular weight of approximately 24,000 and a pI of 5.3 on autoradiographs of two-dimensional polyacrylamide gels. A protein with identical migration characteristics was identified as a major secretory product of isolated round spermatids. Microsequencing revealed that the protein had homology to phosphatidylethanolamine binding protein (PEBP) identified in rat brain. Primers were used in conjunction with RTPCR to amplify a partial cDNA which was used to probe a rat testis library to obtain full length clones. On Northern blots, PEBP mRNA was abundant in adult rat testis and epididymis and fractions enriched in germ cells but was very low/absent from fetal or immature rat testis or adult rat Sertoli cells. In situ hybridisation identified that abundant mRNA was first detectable in pachytene spermatocytes at stage VII and thereafter at particularly high levels in round and elongating spermatids until step 14. Proteins with significant sequence homology to the rat testis PEBP have been identified previously in mouse testis and epididymis, in rat germ cell cultures and coating the surface of mature rat sperm. Differences in the timing of expression of the PEBP mRNA (first expressed in pachytene spermatocytes) and secretion of the PEBP protein (not a major secretory product until round spermatids) is consistent with PEBP mRNA undergoing delayed translation. The role(s) of secreted lipid binding proteins in spermatogenesis are discussed.


Apmis | 2001

Neonatal exposure to potent and environmental oestrogens and abnormalities of the male reproductive system in the rat: evidence for importance of the androgen-oestrogen balance and assessment of the relevance to man

K. Williams; Chris McKinnell; Philippa T. K. Saunders; Marion Walker; Jane S. Fisher; Katie J. Turner; Nina Atanassova; Richard M. Sharpe

The effects on reproductive tract development in male rats, of neonatal exposure to potent (reference) oestrogens, diethylstilboestrol (DES) and ethinyl oestradiol (EE), with those of two environmental oestrogens, octylphenol and bisphenol A were systematically compared. Other treatments, such as administration of a gonadotrophin‐releasing hormone antagonist (GnRHa) or the anti‐oestrogen tamoxifen or the anti‐androgen flutamide, were used to aid interpretation of the pathways involved. All treatments were administered in the neonatal period before onset of puberty. The cellular sites of expression of androgen receptors (AR) and of oestrogen receptor‐α (ERα) and ERβ were also established throughout development of the reproductive system. The main findings were as follows: (i) all cell types that express AR also express one or both ERs at all stages of development; (ii) Sertoli cell expression of ERβ occurs considerably earlier in development than does expression of AR; (iii) most germ cells, including fetal gonocytes, express ERβ but not AR; (iv) treatment with high, but not low, doses of potent oestrogens such as DES and EE, induces widespread structural and cellular abnormalities of the testis and reproductive tract before puberty; (v) the latter changes are associated with loss of immunoexpression of AR in all affected tissues and a reduction in Leydig cell volume per testis; (vi) none of the effects in (iv) and (v) can be duplicated by treating with high‐dose octylphenol or bisphenol A; (vi) none of the reproductive tract changes in (iv) and (v) can be induced by simply suppressing androgen production (GnRHa treatment) or action (flutamide treatment); and (vii) the adverse changes induced by high‐dose DES (iv and v) can be largely prevented by co‐administration of testosterone. Thus, it is suggested that many of the adverse changes to the testis and reproductive tract induced by exposure to oestrogens result from a combination of high oestrogen and low androgen action. High oestrogen action or low androgen action on their own are unable to induce the same changes.


Archive | 1998

Estrogen Effects on Development and Function of the Testis

Richard Sharpe; Jane S. Fisher; Philippa Saunders; Gregor Majdic; Michael Millar; Priyanke Parte; J. B. Kerr; Katie J. Turner

It is well established that exposure of the developing or adult male to even small amounts of exogenous estrogens can cause a range of adverse changes in reproductive development or function (1, 2). Such effects have been reported in laboratory and domestic animals as well as in man (1, 3). It has been generally presumed that these effects occur primarily as a consequence of the suppression of gonadotropin secretion (4), although there are suggestions in the literature that effects of estrogens within the testis or elsewhere in the male reproductive tract are possible (1, 2, 5).


Endocrinology | 2000

Comparative effects of neonatal exposure of male rats to potent and weak (environmental) estrogens on spermatogenesis at puberty and the relationship to adult testis size and fertility: evidence for stimulatory effects of low estrogen levels.

Nina Atanassova; Chris McKinnell; Katie J. Turner; Marion Walker; Jane S. Fisher; M. Morley; Michael Millar; Nigel P. Groome; Richard M. Sharpe


Environmental Health Perspectives | 1999

Effect of neonatal exposure to estrogenic compounds on development of the excurrent ducts of the rat testis through puberty to adulthood

Jane S. Fisher; Katie J. Turner; Dennis Brown; Richard M. Sharpe


Biology of Reproduction | 1998

Abnormalities in Functional Development of the Sertoli Cells in Rats Treated Neonatally with Diethylstilbestrol: A Possible Role for Estrogens in Sertoli Cell Development

Richard M. Sharpe; Nina Atanassova; Chris McKinnell; P. Parte; Katie J. Turner; Jane S. Fisher; J. B. Kerr; Nigel P. Groome; Sheila Macpherson; Michael Millar; Philippa T. K. Saunders


Human Reproduction Update | 2001

Neonatal exposure to potent and environmental oestrogens and abnormalities of the male reproductive system in the rat: evidence for importance of the androgen–oestrogen balance and assessment of the relevance to man

K. Williams; Chris McKinnell; Philippa T. K. Saunders; Marion Walker; Jane S. Fisher; Katie J. Turner; Nina Atanassova; Richard M. Sharpe

Collaboration


Dive into the Katie J. Turner's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jane S. Fisher

Medical Research Council

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Nina Atanassova

Bulgarian Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

K. Williams

University of Edinburgh

View shared research outputs
Top Co-Authors

Avatar

Marion Walker

University of Nottingham

View shared research outputs
Top Co-Authors

Avatar

Nigel P. Groome

Oxford Brookes University

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge