Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters











Database
Language
Publication year range
1.
Reprod Fertil Dev ; 25(2): 439-45, 2013.
Article in English | MEDLINE | ID: mdl-23445819

ABSTRACT

This study compared maternal plasma amino acid concentrations, placental protein secretion in vitro and fetal body composition and plasma amino acid and hormone concentrations in feto-placental units from the smallest and a normally-sized fetus carried by Large White × Landrace or Meishan gilts on Day 100 of pregnancy. Compared with Large White × Landrace, Meishan placental tissue secreted more protein and Meishan fetuses contained relatively more fat and protein, but less moisture. Fetal plasma concentrations of insulin, triiodothryonine, thyroxine and insulin-like growth factor (IGF)-II were higher in Meishan than Large White × Landrace fetuses. In both breeds, fetal cortisol concentrations were inversely related to fetal size, whereas concentrations of IGF-I were higher in average-sized fetuses. Concentrations of 10 amino acids were higher in Large White × Landrace than Meishan gilts, while glutamine concentrations were higher in Meishan gilts. Concentrations of alanine, aspartic acid, glutamic acid and threonine were higher in Meishan than Large White × Landrace fetuses. Average-sized fetuses had higher concentrations of asparagine, leucine, lysine, phenylalanine, threonine, tyrosine and valine than the smallest fetus. This study revealed novel genotype and fetal size differences in porcine maternal-fetal amino acid status and fetal hormone and metabolite concentrations.


Subject(s)
Amino Acids/blood , Fetal Development/physiology , Maternal-Fetal Exchange/physiology , Pregnancy Proteins/metabolism , Sus scrofa/genetics , Animals , Breeding/methods , Chromatography, Ion Exchange/veterinary , Crosses, Genetic , Female , Fetus/anatomy & histology , Fetus/metabolism , Genotype , Hydrocortisone/blood , Pregnancy , Radioimmunoassay/veterinary , Species Specificity , Sus scrofa/physiology
2.
Reprod Biol Endocrinol ; 6: 4, 2008 Jan 29.
Article in English | MEDLINE | ID: mdl-18230131

ABSTRACT

BACKGROUND: The hypogonadal (hpg) mouse is widely used as an animal model with which to investigate the endocrine regulation of spermatogenesis. Chronic treatment of these GnRH-deficient mice with estradiol is known to induce testicular maturation and restore qualitatively normal spermatogenesis. The aim of the current studies was to investigate whether these effects of estradiol are direct effects in the testis, or indirect actions via paradoxical stimulation of FSH secretion from the pituitary gland. METHODS: Initially, Western blot and immunohistochemistry were used to analyse tissues from hpg mice to identify potential sites of action of estradiol. In the main study, hpg mice were treated for 50 days with either an estradiol implant or daily injections of recombinant human FSH, or a combination of both, to determine whether estradiol would have an additive or synergistic effect with FSH on testis development, as assessed by histological analysis and stereological quantification of Leydig, Sertoli and germ cell proliferation. RESULTS: Western blot analysis revealed ERalpha immunoreactive bands of appropriate molecular weight in extracts of testis and pituitary glands from hpg mice, and immunohistochemical studies confirmed ERalpha in nuclei of anterior pituitary cells and Leydig and peritubular cells in hpg mice. Histological and morphometric analyses revealed that estradiol treatment alone was as effective as FSH in promoting Sertoli cell production and proliferation of the seminiferous epithelium, resulting in the production of elongating spermatids. Combined estradiol and FSH treatment did not produce a greater effect than either treatment alone, though an increased dose of FSH significantly increased seminiferous tubule volume and testis weight and increase Sertoli cell numbers further within the same time frame. In contrast, estradiol caused substantial increases in the wet weight of the seminal vesicles, whereas FSH was without effect on this tissue, and did not augment the actions of estradiol. CONCLUSION: As ERalpha receptor is abundantly expressed in the pituitary gland of hpg mice, and estradiol did not exert effects on testis development over and above those of FSH, we conclude that the action of estradiol on testis development in hpg mice is predominantly via the stimulation of pituitary FSH release.


Subject(s)
Estradiol/pharmacology , Follicle Stimulating Hormone/pharmacology , Hypogonadism/physiopathology , Testis/drug effects , Testis/growth & development , Animals , Blotting, Western , Cell Proliferation/drug effects , Drug Synergism , Estrogen Receptor alpha/metabolism , Follicle Stimulating Hormone/blood , Humans , Immunohistochemistry , Leydig Cells/metabolism , Male , Mice , Organ Size/drug effects , Pituitary Gland/metabolism , Pituitary Gland, Anterior/metabolism , Pituitary Gland, Anterior/pathology , Recombinant Proteins/pharmacology , Seminal Vesicles/pathology , Seminiferous Epithelium/metabolism , Seminiferous Epithelium/pathology , Sertoli Cells/metabolism , Spermatids/pathology , Spermatogenesis/drug effects , Testis/metabolism , Testis/pathology
3.
Reproduction ; 133(3): 653-61, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17379659

ABSTRACT

Glucocorticoids play a critical role in fetal development, but inappropriate exposure is associated with reduced fetal growth. We investigated cortisol exposure and supply in a porcine model of differential fetal growth. This model compares the smallest fetus of a litter with an average-sized sibling at three stages of gestation. At day 45, small fetuses had reduced plasma cortisol (16.8 +/- 3.4 ng/ml) relative to average fetuses (34.4 +/- 3.4 ng/ml, P < 0.001). At day 65 levels had reduced in small and average fetuses to similar concentrations (5.7 +/- 1.0 vs 4.8 +/- 0.5 ng/ml, P = 0.128). By day 100, elevated levels were found in small fetuses (10.7 +/- 1.5 vs 7.6 +/- 0.7 ng/ml, P < 0.001). Maternal plasma cortisol was unchanged over gestation (day 45, 56.7 +/- 21.6 ng/ml; day 65, 57.8 +/- 14.4 ng/ml; day 100, 55.7 +/- 6.5 ng/ml). We examined the cause of altered cortisol by investigating the fetal hypothalamic-pituitary-adrenal axis through the measurement of adrenocorticotropic hormone and assessing exposure to maternal cortisol by quantifying placental 11beta-hydroxysteroid dehydrogenase-isoform 2 (11beta HSD-2) gene expression. These data suggest that altered cortisol supply was of fetal origin. We examined organ glucocorticoid (GC) metabolism by the measurement of GC receptor (GR) and 11beta-hydroxysteroid dehydrogenase-isoform 1 (11beta HSD-1) gene expression. We found that fetal organs have different temporal patterns of 11beta HSD-1 and GR expression, with the liver particularly sensitive to cortisol in late gestation. This study examines GC exposure in naturally occurring differential growth and simultaneously explores tissue GC sensitivity and handling, at three key stages of gestation.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenases/analysis , Hydrocortisone/blood , Maternal-Fetal Exchange , Pituitary-Adrenal System/embryology , Receptors, Glucocorticoid/analysis , Swine/embryology , 11-beta-Hydroxysteroid Dehydrogenase Type 1/analysis , 11-beta-Hydroxysteroid Dehydrogenase Type 1/genetics , 11-beta-Hydroxysteroid Dehydrogenase Type 2/analysis , 11-beta-Hydroxysteroid Dehydrogenase Type 2/genetics , 11-beta-Hydroxysteroid Dehydrogenases/genetics , Adrenocorticotropic Hormone/blood , Animals , Biomarkers/analysis , Blotting, Northern/methods , Body Weight , Female , Fetal Blood/chemistry , Fetal Development/physiology , Gene Expression , Gestational Age , Liver/chemistry , Models, Biological , Pregnancy , RNA, Messenger/analysis , Receptors, Glucocorticoid/metabolism , Reverse Transcriptase Polymerase Chain Reaction
4.
Hum Fertil (Camb) ; 9(3): 127-35, 2006 Sep.
Article in English | MEDLINE | ID: mdl-17008264

ABSTRACT

The hypogonadal (hpg) mouse is an excellent animal model in which to investigate the mechanism of action of estrogens on spermatogenesis because it has arrested reproductive development without the need for surgical, endocrine, pharmacological or immunological intervention. Hpg mice are hypogonadotrophic and fail to show normal postnatal testicular development due to the congenital inability to synthesize gonadotropin-releasing hormone in the hypothalamus. The hpg testis remains responsive to gonadotropins and androgens in that fertility can be induced by treatment with these hormones. Surprisingly, chronic treatment with low concentrations of estradiol alone induces qualitatively normal spermatogenesis. The induction of testicular development by estradiol in hpg mice is accompanied by a paradoxical increase in FSH production. The actions of estradiol in hpg mice appear to be via genomic estrogen receptors, as concurrent treatment with estrogen-receptor antagonist ICI182,780 completely blocks these pituitary and testis responses. Concurrent treatment with the androgen receptor antagonist bicalutamide does not affect the estradiol-induced increase in pituitary FSH content, but markedly attenuates the estradiol-induced increase in testicular weight. Western blot analyses and immunohistochemistry provide evidence for estrogen-receptor alpha and beta expression in both pituitary gland and testis of the hpg mouse. Estradiol may therefore exert direct actions within the testes and/or indirect neuroendocrine actions via the release of FSH or other hormones from the pituitary gland, but its actions are dependent upon the availability of low levels of androgen within the testis.


Subject(s)
Estrogens/physiology , Hypogonadism , Models, Animal , Spermatogenesis/physiology , Animals , Estradiol/pharmacology , Follicle Stimulating Hormone/biosynthesis , Follicle Stimulating Hormone/metabolism , Humans , Hypogonadism/genetics , Male , Mice , Mice, Mutant Strains , Pituitary Gland/drug effects , Pituitary Gland/metabolism , Receptors, Estrogen/physiology , Sertoli Cells/physiology , Spermatogenesis/drug effects , Testis/drug effects , Testis/growth & development
5.
Reproduction ; 130(5): 643-54, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16264094

ABSTRACT

Testicular development is arrested in the hypogonadal (hpg) mouse due to a congenital deficiency of hypothalamic gonadotropin-releasing hormone synthesis. Previous studies have demonstrated that chronic treatment of these mice with estradiol induces testicular maturation and qualitatively normal spermatogenesis, but it is not known whether these are direct effects via estrogen receptors expressed in the testis, or indirect actions via the pituitary gland. The aim of the current studies was to determine whether the actions of estradiol require the presence of androgens. Sensitive assays revealed that chronic estradiol treatment produced time-dependent increases in pituitary FSH production but no increases in pituitary LH or testicular testosterone content could be detected. As a functional test of androgen dependence, hpg mice were treated for 70 days with estradiol plus Casodex (bicalutamide), an androgen receptor antagonist. Casodex treatment markedly attenuated both the estradiol-induced increase in testicular weight and the proliferation of the seminiferous epithelium, as revealed by morphometric analysis. However, it did not affect the estradiol-induced increase in pituitary FSH content, nor did it affect estradiol-induced increases in the weight of the seminal vesicles and epididymides. We conclude that increased FSH production is not sufficient to explain the increase in testicular development induced by estradiol in hpg mice; there is a requirement for functional androgen receptors for induction of testicular growth.


Subject(s)
Androgens/metabolism , Estradiol/pharmacology , Hypogonadism/metabolism , Spermatogenesis/drug effects , Testis/metabolism , Androgen Antagonists/pharmacology , Anilides/pharmacology , Animals , Estradiol/analogs & derivatives , Female , Follicle Stimulating Hormone/blood , Fulvestrant , Male , Mice , Mice, Inbred C3H , Mice, Mutant Strains , Nitriles , Pituitary Gland/drug effects , Pituitary Gland/metabolism , Receptors, Androgen/metabolism , Seminiferous Epithelium/drug effects , Seminiferous Epithelium/growth & development , Seminiferous Epithelium/metabolism , Testis/drug effects , Testis/growth & development , Tosyl Compounds , Uterus/drug effects
6.
Reprod Biol Endocrinol ; 3: 48, 2005 Sep 21.
Article in English | MEDLINE | ID: mdl-16176578

ABSTRACT

BACKGROUND: Testicular development is arrested in the hypogonadal (hpg) mouse due to a congenital deficiency in hypothalamic gonadotropin-releasing hormone (GnRH) synthesis. Chronic treatment of male hpg mice with estradiol induces FSH synthesis and secretion, and causes testicular maturation and qualitatively normal spermatogenesis. As estradiol negative feedback normally inhibits FSH production in the male, this study tested whether this paradoxical response to estradiol in the male hpg mouse might be due to inadequate masculinisation or incomplete defeminization in the neonatal period. Previous studies have demonstrated that treatment of hpg mice with testosterone propionate in the immediate neonatal period is necessary to allow full reproductive behaviors to be expressed following suitable endocrine stimulation at adult ages. METHODS: Hpg mice were treated with 100 mug testosterone propionate or vehicle on postnatal day 2. At 35 days of age, subgroups of these mice were treated with silastic implants containing estradiol or cholesterol. Reproductive behavior was scored in tests with steroid-primed female mice, then testicular development was assessed histologically, and measures of pituitary FSH content made at 85 days of age. RESULTS: The neonatal testosterone propionate treatment successfully defeminized female litter mates, as revealed by impaired vaginal opening and deficiencies in lordosis behavior, and it allowed appropriate male reproductive behavior to be expressed in a proportion of the hpg males when tested at an adult age. However, neonatal androgen supplementation did not block or even reduce the subsequent actions of estradiol in increasing pituitary FSH content, nor did it affect the ability of estradiol to induce qualitatively normal spermatogenesis. CONCLUSION: The ability of the hpg male to show a "female" neuroendocrine response to estradiol is not a result of inadequate androgenization during neonatal development, and thus the actions of estradiol revealed in this rodent model are not an artefact of incomplete sexual differentiation, but reflect a physiological role of estradiol occurring during a specific early temporal window of male reproductive development.


Subject(s)
Androgens/pharmacology , Estradiol/pharmacology , Follicle Stimulating Hormone/biosynthesis , Hypogonadism/physiopathology , Spermatogenesis/drug effects , Testosterone Propionate/pharmacology , Animals , Animals, Newborn , Cholesterol/pharmacology , Female , Male , Mice , Mice, Inbred C3H , Mice, Mutant Strains , Organ Size/drug effects , Sexual Behavior, Animal , Testis/anatomy & histology , Testis/drug effects
7.
Reproduction ; 128(2): 229-35, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15280562

ABSTRACT

Low birth weight is a major factor in neonatal morbidity and mortality in humans and domestic species and is a predictor of physiological disorders in adulthood. This study utilised the naturally occurring variation in pig fetal size within a uterus to test the hypothesis that placental amino acid transport capability is associated with fetal growth. Leucine uptake by trophoblast vesicles prepared from placentas supplying an average-sized fetus and the smallest fetus in the uterus was assessed. On days 45 and 65 of gestation, uptake of leucine by the porcine placenta was predominantly sodium independent and was inhibited by the non-metabolised leucine analogue 2-amino-2-norbornane-carboxylic acid, indicating that uptake occurs via system L. By day 100 the uptake of leucine by placentas supplying average-sized fetuses had changed from being predominantly sodium independent to involving both sodium-dependent (system B0) and -independent (system L) pathways. This change was not seen in placentas supplying the smallest fetus, which continued to display predominantly sodium-independent uptake. In conclusion, these data show gestational- and fetal size-dependent changes in the transport of leucine across the porcine placenta.


Subject(s)
Fetal Growth Retardation/metabolism , Leucine/metabolism , Placenta/metabolism , Signal Transduction/physiology , Animals , Biological Transport/physiology , Birth Weight , Female , Gestational Age , Models, Animal , Pregnancy , Sodium/metabolism , Swine
SELECTION OF CITATIONS
SEARCH DETAIL