1
|
Pérez PA, Toledo J, Picech F, Petiti JP, Mukdsi JH, Diaz-Torga G, Torres AI, De Paul AL, Gutiérrez S. Perinatal DEHP exposure modulates pituitary estrogen receptor α and β expression altering lactotroph and somatotroph cell growth in prepuberal and adult male rats. Food Chem Toxicol 2021; 158:112649. [PMID: 34728246 DOI: 10.1016/j.fct.2021.112649] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 10/05/2021] [Accepted: 10/25/2021] [Indexed: 11/17/2022]
Abstract
Phthalates are synthetic chemicals widely used to make polyvinylchloride (PVC) soft and flexible. Of these, Di-(2-ethylhexyl) phthalate (DEHP) is the most commonly used, with high human exposure occurring as early as the fetal developmental stage and affecting the endocrine system. We focused on the perinatal DEHP effects on pituitary estrogen receptor (ER) expression in male rats, explored their impact on lactotroph and somatotroph cell growth, and evaluated the direct effects of this phthalate on pituitary cell cultures. Our results showed that DEHP perinatal exposure was unable to modify the ERα+ pituitary cell number from prepuberal rats, but increased ERβ+ cells. In adulthood, the pituitary ERα+ cells underwent a slight decrease with ERβ showing the greatest changes, and with a significant increase observed in somatotroph cells. Also, in vitro, DEHP reduced the ERα+ cells, increased the percentage of ERβ+ pituitary cells and modified the Ki67 index, as well as decreasing the lactotrophs and increasing the somatotroph cells. In conclusion, the present study showed that DEHP induced ER expression changes in normal pituitary glands from male rats in in vivo and in vitro conditions, suggesting that DEHP could differentially modulate lactotroph and somatotroph cell growth, possibly as a consequence of ER imbalance.
Collapse
Affiliation(s)
- Pablo A Pérez
- Instituto de Investigaciones en Ciencias de la Salud (INICSA) Consejo Nacional de Investigaciones Científicas y Técnicas, Córdoba, Argentina; Centro de Microscopia Electrónica, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Argentina
| | - Jonathan Toledo
- Instituto de Investigaciones en Ciencias de la Salud (INICSA) Consejo Nacional de Investigaciones Científicas y Técnicas, Córdoba, Argentina; Centro de Microscopia Electrónica, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Argentina
| | - Florencia Picech
- Instituto de Investigaciones en Ciencias de la Salud (INICSA) Consejo Nacional de Investigaciones Científicas y Técnicas, Córdoba, Argentina; Centro de Microscopia Electrónica, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Argentina
| | - Juan P Petiti
- Instituto de Investigaciones en Ciencias de la Salud (INICSA) Consejo Nacional de Investigaciones Científicas y Técnicas, Córdoba, Argentina; Centro de Microscopia Electrónica, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Argentina
| | - Jorge H Mukdsi
- Instituto de Investigaciones en Ciencias de la Salud (INICSA) Consejo Nacional de Investigaciones Científicas y Técnicas, Córdoba, Argentina; Centro de Microscopia Electrónica, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Argentina
| | - Graciela Diaz-Torga
- Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Alicia I Torres
- Instituto de Investigaciones en Ciencias de la Salud (INICSA) Consejo Nacional de Investigaciones Científicas y Técnicas, Córdoba, Argentina; Centro de Microscopia Electrónica, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Argentina
| | - Ana L De Paul
- Instituto de Investigaciones en Ciencias de la Salud (INICSA) Consejo Nacional de Investigaciones Científicas y Técnicas, Córdoba, Argentina; Centro de Microscopia Electrónica, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Argentina
| | - Silvina Gutiérrez
- Instituto de Investigaciones en Ciencias de la Salud (INICSA) Consejo Nacional de Investigaciones Científicas y Técnicas, Córdoba, Argentina; Centro de Microscopia Electrónica, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Argentina.
| |
Collapse
|
2
|
Abstract
A healthy nutritional state is required for all aspects of reproduction and is signaled by the adipokine leptin. Leptin acts in a relatively narrow concentration range: too much or too little will compromise fertility. The leptin signal timing is important to prepubertal development in both sexes. In the brain, leptin acts on ventral premammillary neurons which signal kisspeptin (Kiss1) neurons to stimulate gonadotropin releasing hormone (GnRH) neurons. Suppression of Kiss1 neurons occurs when agouti-related peptide neurons are activated by reduced leptin, because leptin normally suppresses these orexigenic neurons. In the pituitary, leptin stimulates production of GnRH receptors (GnRHRs) and follicle-stimulating hormone at midcycle, by activating pathways that derepress actions of the messenger ribonucleic acid translational regulatory protein Musashi. In females, rising estrogen stimulates a rise in serum leptin, which peaks at midcycle, synchronizing with nocturnal luteinizing hormone pulses. The normal range of serum leptin levels (10-20 ng/mL) along with gonadotropins and growth factors promote ovarian granulosa and theca cell functions and oocyte maturation. In males, the prepubertal rise in leptin promotes testicular development. However, a decline in leptin levels in prepubertal boys reflects inhibition of leptin secretion by rising androgens. In adult males, leptin levels are 10% to 50% of those in females, and high leptin inhibits testicular function. The obesity epidemic has elucidated leptin resistance pathways, with too much leptin in either sex leading to infertility. Under conditions of balanced nutrition, however, the secretion of leptin is timed and regulated within a narrow level range that optimizes its trophic effects.
Collapse
Affiliation(s)
- Gwen V Childs
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas
- Correspondence: Gwen V. Childs, PhD, University of Arkansas for Medical Sciences, Little Rock, AR, USA. E-mail:
| | - Angela K Odle
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Melanie C MacNicol
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Angus M MacNicol
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| |
Collapse
|
3
|
Satué K, Marcilla M, Medica P, Cravana C, Fazio E. Temporal relationships of GH, IGF-I and fructosamine concentrations in pregnant Spanish Purebred mares: A substantial contribution from the hormonal standpoint. Theriogenology 2018; 118:164-171. [PMID: 29909261 DOI: 10.1016/j.theriogenology.2018.05.040] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 05/29/2018] [Accepted: 05/29/2018] [Indexed: 11/17/2022]
Abstract
Growth hormone (GH) and insulin-like growth factor (IGF-I) exert essential functions to maintain an adequate metabolic activity for correct placental and fetal development. Although fructosamine (FRUCT) is used in clinical evaluation to assess the glycaemic state, the relationships between GH, IGF-I and FRUCT remain unknown in the mare. Therefore, the objectives of this study were to evaluate the relationships among these parameters in pregnant mares. A total of 30 healthy Spanish Purebred mares, with an age range of 9.33 ± 3.31 years, were studied during the 11 months of gestation. Serum concentrations of GH, IGF-I and FRUCT were analyzed by EIA, competition ELISA and spectrophotometry, respectively. GH showed a biphasic pattern, the first occurred during 4th and 5th month and the second which was higher than the 1st one during the 7th, 8th and 9th month (P < 0.05). Compared to the 1st month, the IGF-I concentrations increased in the 2nd month (P < 0.05), decreased in the 3rd and 4th month and increased to reach the maximum average value in the 5th month (P < 0.05); after slight oscillations between the 6th and 8th month, IGF-I concentrations increased in the 9th month (P < 0.05), decreasing until the end of gestation. FRUCT increased progressively and significantly from the 6th month of gestation, reaching the maximum values in the last month of gestation (P < 0.05). These results suggest that alternative mechanisms other than GH and IGF-I could be involved in the regulation of glycaemic metabolism in pregnant mare.
Collapse
Affiliation(s)
- K Satué
- Department of Animal Medicine and Surgery, Faculty of Veterinary, Cardenal Herrera-CEU University, Valencia, Spain.
| | - M Marcilla
- Department of Animal Medicine and Surgery, Faculty of Veterinary, Cardenal Herrera-CEU University, Valencia, Spain
| | - P Medica
- Department of Veterinary Sciences, Veterinary Physiology Unit, Polo Universitario Annunziata, Messina University, Italy
| | - C Cravana
- Department of Veterinary Sciences, Veterinary Physiology Unit, Polo Universitario Annunziata, Messina University, Italy
| | - E Fazio
- Department of Veterinary Sciences, Veterinary Physiology Unit, Polo Universitario Annunziata, Messina University, Italy
| |
Collapse
|
4
|
Bentz AB, Sirman AE, Wada H, Navara KJ, Hood WR. Relationship between maternal environment and DNA methylation patterns of estrogen receptor alpha in wild Eastern Bluebird (Sialia sialis) nestlings: a pilot study. Ecol Evol 2016; 6:4741-52. [PMID: 27547309 PMCID: PMC4979703 DOI: 10.1002/ece3.2162] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 04/11/2016] [Accepted: 04/13/2016] [Indexed: 12/18/2022] Open
Abstract
There is mounting evidence that, across taxa, females breeding in competitive environments tend to allocate more testosterone to their offspring prenatally and these offspring typically have more aggressive and faster‐growing phenotypes. To date, no study has determined the mechanisms mediating this maternal effect's influence on offspring phenotype. However, levels of estrogen receptor alpha (ERα) gene expression are linked to differences in early growth and aggression; thus, maternal hormones may alter gene regulation, perhaps via DNA methylation, of ERα in offspring during prenatal development. We performed a pilot study to examine natural variation in testosterone allocation to offspring through egg yolks in wild Eastern Bluebirds (Sialia sialis) in varying breeding densities and percent DNA methylation of CG dinucleotides in the ERα promoter in offspring brain regions associated with growth and behavior. We hypothesized that breeding density would be positively correlated with yolk testosterone, and prenatal exposure to maternal‐derived yolk testosterone would be associated with greater offspring growth and decreased ERα promoter methylation. Yolk testosterone concentration was positively correlated with breeding density, nestling growth rate, and percent DNA methylation of one out of five investigated CpG sites (site 3) in the diencephalon ERα promoter, but none in the telencephalon (n = 10). Percent DNA methylation of diencephalon CpG site 3 was positively correlated with growth rate. These data suggest a possible role for epigenetics in mediating the effects of the maternal environment on offspring phenotype. Experimentally examining this mechanism with a larger sample size in future studies may help elucidate a prominent way in which animals respond to their environment. Further, by determining the mechanisms that mediate maternal effects, we can begin to understand the potential for the heritability of these mechanisms and the impact that maternal effects are capable of producing at an evolutionary scale.
Collapse
Affiliation(s)
- Alexandra B Bentz
- Poultry Science Department University of Georgia 203 Poultry Science Bldg. Athens Georigia 30602
| | - Aubrey E Sirman
- Department of Biological Sciences Auburn University 101 Life Science Building Auburn Alabama 36849
| | - Haruka Wada
- Department of Biological Sciences Auburn University 101 Life Science Building Auburn Alabama 36849
| | - Kristen J Navara
- Poultry Science Department University of Georgia 203 Poultry Science Bldg. Athens Georigia 30602
| | - Wendy R Hood
- Department of Biological Sciences Auburn University 101 Life Science Building Auburn Alabama 36849
| |
Collapse
|
5
|
Selek A, Cetinarslan B, Gurbuz Y, Tarkun I, Canturk Z, Cabuk B. Aromatase enzyme expression in acromegaly and its possible relationship with disease prognosis. Endocrine 2015; 49:250-7. [PMID: 25300784 DOI: 10.1007/s12020-014-0445-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2014] [Accepted: 10/01/2014] [Indexed: 10/24/2022]
Abstract
The purpose of this study was to evaluate aromatase enzyme expression in growth hormone (GH) secreting adenomas and comparison with prolactinomas, nonfunctional adenomas, and normal pituitary tissues. Also the impact of its expression on clinical and prognostic features was evaluated. 38 acromegaly, 26 prolactinoma, and 31 nonfunctional pituitary adenoma and 11 normal pituitary gland samples from autopsies were included. Aromatase and estrogen receptor-alpha (ERα) were evaluated by Immunohistochemical method; demographic, pre- and postoperative features of the patients were noted. Aromatase was expressed in varying degrees in all cases in study including controls. Aromatase expression in patients with acromegaly was significantly higher than patients with prolactinoma, nonfunctional adenoma, and controls (p = 0.04, p = 0.01 and p < 0.001, respectively). Taken together two functional adenoma groups, prolactinoma and acromegaly, aromatase expression was negatively correlated with ER-alpha (p = 0.02, r = -0.34). Also, Ki-67 immunohistochemical results were negatively correlated with aromatase expression (p = 0.03, r = -0.27) while positively correlated with ER expression (p < 0.01). Consistent with the growing evidence about testosterone effect on pituitary functions, aromatase expression was found to be higher in GH-secreting pituitary adenoma. Aromatase was expressed in all pituitary tissues including autopsy samples; however, it was highest in patients with acromegaly. In patients with acromegaly and prolactinoma, aromatase expression was negatively correlated with Ki-67 score, and also it was higher in patients with complete postoperative remission than without remission. Therefore, aromatase expression may be a good prognostic marker predominantly in acromegaly.
Collapse
Affiliation(s)
- Alev Selek
- Department of Endocrinology and Methabolism, Faculty of Medicine, Kocaeli University, Kocaeli, 41380, Turkey,
| | | | | | | | | | | |
Collapse
|
6
|
Syed M, Cozart M, Haney AC, Akhter N, Odle AK, Allensworth-James M, Crane C, Syed FM, Childs GV. Ghrelin restoration of function in vitro in somatotropes from male mice lacking the Janus kinase (JAK)-binding site of the leptin receptor. Endocrinology 2013; 154:1565-76. [PMID: 23417423 PMCID: PMC3602631 DOI: 10.1210/en.2012-2254] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Deletion of the signaling domain of leptin receptors selectively in somatotropes, with Cre-loxP technology, reduced the percentage of immunolabeled GH cells and serum GH. We hypothesized that the deficit occurred when leptin's postnatal surge failed to stimulate an expansion in the cell population. To learn more about the deficiency in GH cells, we tested their expression of GHRH receptors and GH mRNA and the restorative potential of secretagogue stimulation in vitro. In freshly plated dissociated pituitary cells from control male mice, GHRH alone (0.3 nM) increased the percentage of immunolabeled GH cells from 27 ± 0.05% (vehicle) to 42 ± 1.8% (P < .002) and the secretion of GH 1.8-3×. Deletion mutant pituitary cells showed a 40% reduction in percentages of immunolabeled GH cells (16.7 ± 0.4%), which correlated with a 47% reduction in basal GH levels (50 ng/mL control; 26.7 ng/mL mutants P = .01). A 50% reduction in the percentage of mutant cells expressing GHRH receptors (to 12%) correlated with no or reduced responses to GHRH. Ghrelin alone (10 nM) stimulated more GH cells in mutants (from 16.7-23%). When added with 1-3 nM GHRH, ghrelin restored GH cell percentages and GH secretion to levels similar to those of stimulated controls. Counts of somatotropes labeled for GH mRNA confirmed normal percentages of somatotropes in the population. These discoveries suggest that leptin may optimize somatotrope function by facilitating expression of membrane GHRH receptors and the production or maintenance of GH stores.
Collapse
Affiliation(s)
- Mohsin Syed
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, College of Medicine, 4301 West Markham, Slot 510, Little Rock, Arkansas 72205, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Avtanski D, Novaira HJ, Wu S, Romero CJ, Kineman R, Luque RM, Wondisford F, Radovick S. Both estrogen receptor α and β stimulate pituitary GH gene expression. Mol Endocrinol 2013; 28:40-52. [PMID: 24284820 DOI: 10.1210/me.2013-1245] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Although sex steroids have been implicated in the control of mammalian growth, their direct effect on GH synthesis is less clear. The aim of this study was to establish whether estradiol (E2) directly affects GH synthesis in somatotrophs. Somatotroph GH3 and MtT/S cells were used as in vitro models. At physiological doses of E2 stimulation, GH mRNA levels were increased and the ER antagonist ICI 182,780 completely abolished this effect. Estrogen receptor (ER) α- and ERβ-selective agonists, propylpyrazole triol (PPT), and 2,3-bis(4-hydroxyphenyl) propionitrile (DPN), respectively, augmented GH mRNA expression and secretion, whereas E2 and PPT, but not DPN increased prolactin (PRL) mRNA levels. E2, PPT, and DPN stimulated expression of the pituitary transcription factor Pou1f1 and increased its binding to the GH promoter. In vivo evidence of E2 effects on GH synthesis was obtained from the generation of the somatotroph-specific ERα knockout (sERα-KO) mouse model. Basal pituitary GH, PRL, POU1F1, and ERα mRNA expression levels were lower in sERα-KO mice compared with those in controls; whereas ERβ mRNA levels remained unchanged. E2 and DPN stimulated pituitary GH mRNA expression and serum GH levels in control and sERα-KO ovariectomized mice; however, serum GH levels were unchanged in PPT-treated ovariectomized sERα-KO mice. In these animal models, PRL mRNA levels increased after either E2 or PPT, but an increase was not seen after DPN treatment. Thus, we propose a mechanism by which estrogen directly regulates somatotroph GH synthesis at a pretranslational level. In contrast to the predominant effect of ERα in the lactotroph, these results support a role for both ERα and ERβ in the transcriptional control of Gh in the somatotroph and illustrate important differences in ER isoform specificity in the anterior pituitary gland.
Collapse
Affiliation(s)
- Dimiter Avtanski
- Department of Pediatrics (D.A., H.J.N., S.W., C.J.R., S.R.), Division of Endocrinology, and Department of Pediatrics (F.W.), Division of Metabolism, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287; and Research and Development Division (R.K., R.M.L.), Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois 60612
| | | | | | | | | | | | | | | |
Collapse
|
8
|
Akhter N, Odle AK, Allensworth-James ML, Haney AC, Syed MM, Cozart MA, Chua S, Kineman R, Childs GV. Ablation of leptin signaling to somatotropes: changes in metabolic factors that cause obesity. Endocrinology 2012; 153:4705-15. [PMID: 22865370 PMCID: PMC3512011 DOI: 10.1210/en.2012-1331] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2012] [Accepted: 07/12/2012] [Indexed: 01/22/2023]
Abstract
Mice with somatotrope-specific deletion of the Janus kinase binding site in leptin receptors are GH deficient as young adults and become obese by 6 months of age. This study focused on the metabolic status of young (3-4.5 month old) preobese mutant mice. These mutants had normal body weights, lean body mass, serum leptin, glucose, and triglycerides. Mutant males and females showed significantly higher respiratory quotients (RQ) and lower energy output, resulting from a higher volume of CO(2) output and lower volume of O(2) consumption. Deletion mutant females were significantly less active than controls; they had higher levels of total serum ghrelin and ate more food. Mutant females also had lower serum insulin and higher glucagon. In contrast, deletion mutant males were not hyperphagic, but they were more active and spent less time sleeping. Adiponectin and resistin, both products of adipocytes, were increased in male and female mutant mice. In addition, mutant males showed an increase in circulating levels of the potent lipogenic hormone, glucose-dependent insulinotropic peptide. Taken together, these results indicate that mutant mice may become obese due to a reduction in lipid oxidation and energy expenditure. This may stem from GH deficiency. Reduced fat oxidation and enhanced insulin sensitivity (in females) are directly related to GH deficiency in mutant mice because GH has been shown by others to increase insulin sensitivity and fat oxidation and reduce carbohydrate oxidation. Gender-dependent alterations in metabolic signals may further exacerbate the future obese phenotype and affect the timing of its onset. Females show a delay in onset of obesity, perhaps because of their low serum insulin, which is lipogenic, whereas young males already have higher levels of the lipogenic hormone, glucose-dependent insulinotropic peptide. These findings signify that leptin signals to somatotropes are vital for the normal metabolic activity needed to optimize body composition.
Collapse
Affiliation(s)
- Noor Akhter
- Department of Neurobiology and Developmental Sciences, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Effect of estradiol-17β on follicle-stimulating hormone secretion and egg-laying performance of Japanese quail. Animal 2012; 6:1955-60. [PMID: 23031602 DOI: 10.1017/s1751731112000997] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The aim of this study was to measure the effect of estradiol-17β (E2) injection on follicle-stimulating hormone (FSH) secretion and egg-laying performance of Japanese quail. Female Japanese quail were housed in cages and fed ad libitum. After a 7-day adaptation period, the birds were randomly assigned to three groups, that is, one control group and two test groups. The birds were weighed, before every injection. The control group was subcutaneously injected with 0.2 ml sesame oil-ethanol mixture, whereas test groups were injected, twice in a week, with 0.2 ml sesame oil-ethanol mixture containing 0.1 or 0.2 mg E2 along the study. One day after the first injection, egg number, egg weight, eggshell strength and food conception were daily recorded. On the last day of the experiment, the birds were injected and 3 h later seven birds from each group were randomly selected for bleeding. Blood samples (2 ml/bird) were collected from the jugular vein for the measurements of serum concentrations of E2, FSH, calcium (Ca) and phosphorus (P). E2 injection did not cause any significant changes in serum FSH concentrations, daily egg laid/bird, food conception/bird, serum concentrations of the Ca and the P. Egg weight was significantly increased in the 0.1 mg E2-injected group as compared with the control and 0.2 mg E2-injected groups. Eggshell strength in the 0.2 mg E2-injected group was significantly high as compared with the control, whereas the difference between the 0.1 mg E2- and 0.2 mg E2-injected groups was not statistically important. These results show that serum FSH concentration was not increased even when slightly suppressed by subcutaneous injection of 0.1 or 0.2 mg E2. Different doses of E2 have different functions. The increase in BWs in the 0.1 mg E2-injected group was a result of the dose effect, which probably increased growth hormone secretion from the pituitary or IGF-1 synthesis from the liver or both. The dose, 0.2 mg E2, was ineffective in increasing the BW, but it significantly increased eggshell strength probably via the increase in Ca and P utilizations.
Collapse
|
10
|
Cheng S, Grasso L, Martinez-Orozco JA, Al-Agha R, Pivonello R, Colao A, Ezzat S. Pregnancy in acromegaly: experience from two referral centers and systematic review of the literature. Clin Endocrinol (Oxf) 2012; 76:264-71. [PMID: 21777265 DOI: 10.1111/j.1365-2265.2011.04180.x] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND Acromegaly results from increased growth hormone and its target insulin-like growth factor-1, most commonly due to a pituitary tumour. As it is frequently accompanied by infertility, little is known about the course of this disease in pregnancy. OBJECTIVE We describe 13 new pregnancies in acromegalic women and compare their outcomes in a systematic review of the literature. METHODS We collected clinical, biochemical, imaging, and outcomes data during and following pregnancy and performed a systematic review for a total of 47 pregnancies. An extended analysis of 106 pregnancies was also performed. RESULTS In 13 newly described cases, pregnancy was un-complicated without need for additional surgical intervention. In these pregnancies, adjunctive medical therapy was required in three patients. This was in the form of somatostatin analogs (SA) (3/13) as well as pegvisomant in 1/13 to control symptomatic and biochemical progression. One 37-year-old female succeeded in having two separate pregnancies 2 years apart both without need for any form of medical therapy. Review of an additional 34 published reports allowed for an analysis of outcomes in 47 pregnancies. Adjunctive medical therapy during pregnancy was required in 15 of these cases where 12 received SA and an additional three received dopamine agonists. None of these patients developed endocrine or neurologic complications during pregnancy. In an extended analysis of 106 pregnancies, treatment during pregnancy appears to be associated with good disease control but increased risk of microsomic or macrosomic newborns depending on the medical agent used. CONCLUSIONS In 13 newly described pregnancies along with systematic review of an additional 34 cases indicate that pregnancy in treated acromegalic women can proceed without significant complications or teratogenicity. Medical treatment during pregnancy with DA or SA appears to be associated with altered neonatal weight. Nevertheless, gestation may have a beneficial impact on acromegaly control both during and following pregnancy.
Collapse
Affiliation(s)
- Sonia Cheng
- Department of Medicine, University Health Network, Toronto, ON, Canada
| | | | | | | | | | | | | |
Collapse
|
11
|
Childs GV, Akhter N, Haney A, Syed M, Odle A, Cozart M, Brodrick Z, Gaddy D, Suva LJ, Akel N, Crane C, Benes H, Charlesworth A, Luque R, Chua S, Kineman RD. The somatotrope as a metabolic sensor: deletion of leptin receptors causes obesity. Endocrinology 2011; 152:69-81. [PMID: 21084451 PMCID: PMC3033057 DOI: 10.1210/en.2010-0498] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2010] [Accepted: 10/07/2010] [Indexed: 01/01/2023]
Abstract
Leptin, the product of the Lep gene, reports levels of adiposity to the hypothalamus and other regulatory cells, including pituitary somatotropes, which secrete GH. Leptin deficiency is associated with a decline in somatotrope numbers and function, suggesting that leptin may be important in their maintenance. This hypothesis was tested in a new animal model in which exon 17 of the leptin receptor (Lepr) protein was selectively deleted in somatotropes by Cre-loxP technology. Organ genotyping confirmed the recombination of the floxed LepR allele only in the pituitary. Deletion mutant mice showed a 72% reduction in pituitary cells bearing leptin receptor (LEPR)-b, a 43% reduction in LEPR proteins and a 60% reduction in percentages of immunopositive GH cells, which correlated with reduced serum GH. In mutants, LEPR expression by other pituitary cells was like that of normal animals. Leptin stimulated phosphorylated Signal transducer and activator of transcription 3 expression in somatotropes from normal animals but not from mutants. Pituitary weights, cell numbers, IGF-I, and the timing of puberty were not different from control values. Growth curves were normal during the first 3 months. Deletion mutant mice became approximately 30-46% heavier than controls with age, which was attributed to an increase in fat mass. Serum leptin levels were either normal in younger animals or reflected the level of obesity in older animals. The specific ablation of the Lepr exon 17 gene in somatotropes resulted in GH deficiency with a consequential reduction in lipolytic activity normally maintained by GH and increased adiposity.
Collapse
Affiliation(s)
- Gwen V Childs
- Professor and Chair, Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Hong H, Kim WJ, Yoo SH, Abanto OD, Kim TJ, Kim YJ, Jung BM, Yun SS, Hwang SG. Dietary ungerminated and germinated soybean supplementation improves bone mineralization and strength in growing male rats. J Med Food 2010; 13:640-9. [PMID: 20438320 DOI: 10.1089/jmf.2009.1092] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
It has been thought that soybean products rich in phytoestrogenic isoflavones are effective in promoting bone metabolism in ovariectomized rats. However, it is not clear if such an effect can be changed by germination of soybean and can be observed in growing males. The present study was undertaken to determine the effects of different concentrations of dietary ungerminated and germinated soybean (UGS and GS, respectively) on bone metabolism in young male rats. Forty-nine 6-week-old male Sprague-Dawley rats were assigned to seven dietary treatment groups: AIN 93G (control); control with UGS powder at 3% (UGS-L), 15% (UGS-M), or 30% (UGS-H); and control with GS powder at 3% (GS-L), 15% (GS-M), or 30% (GS-H). The biomarkers of bone metabolism (plasma and urinary Ca and P) and the physical properties and mineral content of bones were measured. Plasma osteocalcin and urinary deoxypyridinoline levels were not affected by soybean diets. The plasma P level was significantly elevated by dietary soybeans (P < .001). However, there was a negative correlation (r = -0.555) between plasma P level and dietary UGS level (P < .01). Between the UGS and GS groups, the P level of the GS-M group was significantly higher than that of the UGS-M group (P < .001). The tibial weight was decreased with low to medium doses of soybeans, but not with higher levels. Femoral bone strength was improved in the UGS-H, GS-M, and GS-H groups. The P contents of femoral and tibial bones were increased in all UGS groups as well as in GS-M and GS-H groups. In conclusion, soybean supplementation in young growing male rats improved bone mineralization and bone strength.
Collapse
Affiliation(s)
- Heeok Hong
- Department of Food Service Management, Sangmyung University, Seoul, Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Hao L, Li M, Dai J, Wu Q, Liu Y, Liu S, Zhang Y. Reduced somatostatin in hypothalamus of young male mouse increases local but not circulatory GH. Neurochem Res 2010; 35:143-51. [PMID: 19771516 DOI: 10.1007/s11064-009-0039-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/28/2009] [Indexed: 12/16/2022]
Abstract
The release of growth hormone (GH) from the pituitary gland is primarily inhibited by somatostatin (SRIF) from the hypothalamus via interactions with five types of SRIF receptors (SSTRs). However, the inhibition mechanism of SRIF on GH has not been fully examined. In this study, we repressed the hypothalamic SRIF in young male mice by stereotaxic injection of the lentiviral-shRNA against SRIF to investigate the role of hypothalamic SRIF on hormone secretion in the GH/IGF-1 axis. We found that the reduction of SRIF in hypothalamus was associated with an increase in the protein, but not the mRNA level, of the GH in the pituitary where SSTR 2 and SSTR 5 act importantly. Interestingly, the level of blood circulatory SRIF, GH, IGF-1 and the body weight were not significantly influenced by the downregulation of hypothalamic SRIF. Our findings provide insights into the mechanisms underlying the inhibition of SRIF on GH secretion.
Collapse
Affiliation(s)
- Linlin Hao
- Department of Biochemistry, Veterinary College, Jilin University, No 5333 Xi'an Road, Changchun 130062, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
14
|
Miyakoshi T, Kajiya H, Miyajima K, Takei M, Tobita M, Takekoshi S, Osamura RY. The expression of Wnt4 is regulated by estrogen via an estrogen receptor alpha-dependent pathway in rat pituitary growth hormone-producing cells. Acta Histochem Cytochem 2009; 42:205-13. [PMID: 20126574 PMCID: PMC2808504 DOI: 10.1267/ahc.09033] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2009] [Accepted: 10/25/2009] [Indexed: 11/22/2022] Open
Abstract
Wnt signaling is important in many aspects of cell biology and development. In the mouse female reproductive tract, Wnt4, Wnt5a, and Wnt7a show differential expression during the estrus cycle, suggesting that they participate in female reproductive physiology. Although the pituitary is a major gland regulating reproduction, the molecular mechanism of Wnt signaling here is unclear. We elucidated the subcellular distribution of Wnt4 in the pituitary of estrogen-treated ovariectomized female rats. Expression of Wnt4 mRNA increased dramatically, particularly in proestrus compared with estrus and metestrus. Wnt4 protein was observed in the cytoplasm of almost all growth hormone (GH)-producing cells and in only a few thyroid-stimulating hormone β (TSHβ)-producing cells. In rat GH-producing pituitary tumor (MtT/S) cells, estrogen-induced expression of Wnt4 mRNA was completely inhibited by estrogen receptor antagonist ICI 182,780 in vitro. Thus, rat pituitary GH cells synthesize Wnt4 and this is induced by estrogen mediated via an estrogen receptor alpha-dependent pathway.
Collapse
Affiliation(s)
| | - Hanako Kajiya
- Department of Pathology, Tokai University School of Medicine
| | | | - Mao Takei
- Department of Pathology, Tokai University School of Medicine
- Department of Neurosurgery, Nippon Medical School
| | - Maya Tobita
- Department of Pathology, Tokai University School of Medicine
- Division of Diabetes, Metabolism and Endocrinology, Jikei University School of Medicine
| | | | | |
Collapse
|
15
|
Díaz L, Ceja-Ochoa I, Restrepo-Angulo I, Larrea F, Avila-Chávez E, García-Becerra R, Borja-Cacho E, Barrera D, Ahumada E, Gariglio P, Alvarez-Rios E, Ocadiz-Delgado R, Garcia-Villa E, Hernández-Gallegos E, Camacho-Arroyo I, Morales A, Ordaz-Rosado D, García-Latorre E, Escamilla J, Sánchez-Peña LC, Saqui-Salces M, Gamboa-Dominguez A, Vera E, Uribe-Ramírez M, Murbartián J, Ortiz CS, Rivera-Guevara C, De Vizcaya-Ruiz A, Camacho J. Estrogens and human papilloma virus oncogenes regulate human ether-à-go-go-1 potassium channel expression. Cancer Res 2009; 69:3300-7. [PMID: 19351862 DOI: 10.1158/0008-5472.can-08-2036] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Ether-à-go-go-1 (Eag1) potassium channels are potential tools for detection and therapy of numerous cancers. Here, we show human Eag1 (hEag1) regulation by cancer-associated factors. We studied hEag1 gene expression and its regulation by estradiol, antiestrogens, and human papillomavirus (HPV) oncogenes (E6/E7). Primary cultures from normal placentas and cervical cancer tissues; tumor cell lines from cervix, choriocarcinoma, keratinocytes, and lung; and normal cell lines from vascular endothelium, keratinocytes, and lung were used. Reverse transcription-PCR (RT-PCR) experiments and Southern blot analysis showed Eag1 expression in all of the cancer cell types, normal trophoblasts, and vascular endothelium, in contrast to normal keratinocytes and lung cells. Estradiol and antiestrogens regulated Eag1 in a cell type-dependent manner. Real-time RT-PCR experiments in HeLa cells showed that Eag1 estrogenic regulation was strongly associated with the expression of estrogen receptor-alpha. Eag1 protein was detected by monoclonal antibodies in normal placenta and placental blood vessels. Patch-clamp recordings in normal trophoblasts treated with estradiol exhibited potassium currents resembling Eag1 channel activity. Eag1 gene expression in keratinocytes depended either on cellular immortalization or the presence of HPV oncogenes. Eag1 protein was found in keratinocytes transfected with E6/E7 HPV oncogenes. Cell proliferation of E6/E7 keratinocytes was decreased by Eag1 antibodies inhibiting channel activity and by the nonspecific Eag1 inhibitors imipramine and astemizole; the latter also increased apoptosis. Our results propose novel oncogenic mechanisms of estrogen/antiestrogen use and HPV infection. We also suggest Eag1 as an early indicator of cell proliferation leading to malignancies and a therapeutic target at early stages of cellular hyperproliferation.
Collapse
Affiliation(s)
- Lorenza Díaz
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Zárate S, Jaita G, Zaldivar V, Radl DB, Eijo G, Ferraris J, Pisera D, Seilicovich A. Estrogens exert a rapid apoptotic action in anterior pituitary cells. Am J Physiol Endocrinol Metab 2009; 296:E664-71. [PMID: 19158323 DOI: 10.1152/ajpendo.90785.2008] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
It is now accepted that estrogens not only stimulate lactotrope proliferation but also sensitize anterior pituitary cells to proapoptotic stimuli. In addition to their classical mechanism of action through binding to intracellular estrogen receptors (ERs), there is increasing evidence that estrogens exert rapid actions mediated by cell membrane-localized ERs (mERs). In the present study, we examined the involvement of membrane-initiated steroid signaling in the proapoptotic action of estradiol in primary cultures of anterior pituitary cells from ovariectomized rats by using estren, a synthetic estrogen with no effect on classical transcription and a cell-impermeable 17beta-estradiol conjugate (E2-BSA). Both compounds induced cell death of anterior pituitary cells after 60 min of incubation as assessed by flow cytometry and the [3-(4,5-dimethylthiazol-2-yl)]-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium assay. Estren, E2, and E2-BSA induced apoptosis of lactotropes and somatotropes as evaluated by the deoxynucleotidyltransferase-mediated dUTP nick end-labeling assay and immunodetection of prolactin (PRL) and growth hormone (GH). The proapoptotic effect of E2-BSA was abrogated by ICI-182,780, an antagonist of ERs. The expression of membrane-associated ERalpha was observed in PRL- and GH-bearing cells. Our results indicate that estradiol is able to exert a rapid apoptotic action in anterior pituitary cells, especially lactotropes and somatotropes, by a mechanism triggered by mERs. This mechanism could be involved in anterior pituitary cell turnover.
Collapse
Affiliation(s)
- S Zárate
- Instituto de Investigaciones en Reproducción, Facultad de Medicina, Universidad de Buenos Aires, Paraguay 2155, piso 10, Buenos Aires, Argentina
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Veldhuis JD, Mielke KL, Cosma M, Soares-Welch C, Paulo R, Miles JM, Bowers CY. Aromatase and 5alpha-reductase inhibition during an exogenous testosterone clamp unveils selective sex steroid modulation of somatostatin and growth hormone secretagogue actions in healthy older men. J Clin Endocrinol Metab 2009; 94:973-81. [PMID: 19088159 PMCID: PMC2681279 DOI: 10.1210/jc.2008-2108] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND How endogenous testosterone (Te), 5alpha-dihydrotestosterone (DHT), and estradiol (E(2)) regulate pulsatile GH secretion is not understood. HYPOTHESIS Conversion of Te to androgenic (Te-->DHT) or estrogenic (Te-->E(2)) products directs GH secretion. SUBJECTS AND LOCATION: Healthy older men (N = 42, ages 50-79 yr) participated at an academic medical center. METHODS We inhibited 5alpha-reduction with dutasteride and aromatization with anastrozole during a pharmacological Te clamp and infused somatostatin (SS), GHRH, GH-releasing peptide-2 (GHRP-2), and L-arginine/GHRH/GHRP-2 (triple stimulus) to modulate GH secretion. ENDPOINTS Deconvolution-estimated basal and pulsatile GH secretion was assessed. RESULTS Administration of Te/placebo elevated Te by 2.8-fold, DHT by 2.6-fold, and E(2) concentrations by 1.9-fold above placebo/placebo. Te/dutasteride and Te/anastrozole reduced stimulated DHT and E(2) by 89 and 86%, respectively. Stepwise forward-selection regression analysis revealed that 1) Te positively determines mean (P = 0.017) and peak (P < 0.001) GH concentrations, basal GH secretion (P = 0.015), and pulsatile GH secretion stimulated by GHRP-2 (P < 0.001); 2) Te and E(2) jointly predict GH responses to the triple stimulus (positively for Te, P = 0.006, and negatively for E(2), P = 0.031); and 3) DHT correlates positively with pulsatile GH secretion during SS infusion (P = 0.011). These effects persisted when abdominal visceral fat was included in the regression. CONCLUSION The present outcomes suggest a tetrapartite model of GH regulation in men, in which systemic concentrations of Te, DHT, and E(2) along with abdominal visceral fat determine the selective actions of GH secretagogues and SS.
Collapse
Affiliation(s)
- Johannes D Veldhuis
- Department of Internal Medicine and Pediatrics, Endocrine Research Unit, Clinical Translational Research Center, Mayo Medical and Graduate Schools, Mayo Clinic, Rochester Minnesota 55901, USA.
| | | | | | | | | | | | | |
Collapse
|
18
|
Cosma M, Bailey J, Miles JM, Bowers CY, Veldhuis JD. Pituitary and/or peripheral estrogen-receptor alpha regulates follicle-stimulating hormone secretion, whereas central estrogenic pathways direct growth hormone and prolactin secretion in postmenopausal women. J Clin Endocrinol Metab 2008; 93:951-8. [PMID: 18089703 PMCID: PMC2266945 DOI: 10.1210/jc.2007-1322] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND Estradiol (E(2)) stimulates GH and prolactin secretion and suppresses FSH secretion in postmenopausal women. Whether central nervous system (CNS) or pituitary mechanisms (or both) mediate such actions is not known. OBJECTIVE Our objective was to distinguish between hypothalamic and pituitary or peripheral (hepatic) actions of E2. SETTING This study was performed in an academic medical center. DESIGN This was a double-blind, prospectively randomized, placebo (Pl)-controlled study. METHODS The capability of a selective, noncompetitive, non-CNS permeant estrogen receptor (ER)-alpha antagonist, fulvestrant (FUL) to antagonize the effects of transdermal E2 and Pl on GH, prolactin, and FSH secretion was assessed in 43 women (ages 50-80 yr) in a four parallel-cohort study. Each woman received four secretagogue infusions to stimulate GH secretion. IGF-I and its binding proteins were measured secondarily. RESULTS Administration of Pl/E2 increased GH and prolactin concentrations by 100%, and suppressed FSH concentrations by more than 50% (each P<or=0.004 compared with Pl/Pl). Treatment with FUL/E2 compared with Pl/E2 partially relieved estrogen's inhibition of FSH secretion (P=0.041), without altering E2's stimulation of prolactin secretion. ANOVA further revealed that: 1) estrogen milieu (P=0.014) and secretagogue type (P<0.001) each determined GH concentrations; 2) FUL/Pl suppressed IGF-I concentrations (P<0.001); 3) FUL abrogated estrogen's elevation of IGF binding protein-1 concentrations (P<0.001); and 4) FUL did not oppose estrogen's suppression of IGF binding protein-3 concentrations (P<0.001). SUMMARY AND CONCLUSIONS Responses to a non-CNS permeant ERalpha antagonist indicate that E2 inhibits FSH secretion in part via pituitary/peripheral ERalpha, drives prolactin output via nonpituitary/nonperipheral-ERalpha effects, and directs GH secretion and IGF-I-binding proteins by complex mechanisms.
Collapse
Affiliation(s)
- Mihaela Cosma
- Endocrine Research Unit, Department of Internal Medicine, Mayo Medical and Graduate Schools of Medicine, Mayo Clinic, 200 First Street S.W., Rochester, Minnesota 55905, USA
| | | | | | | | | |
Collapse
|
19
|
Luque RM, Kineman RD. Gender-dependent role of endogenous somatostatin in regulating growth hormone-axis function in mice. Endocrinology 2007; 148:5998-6006. [PMID: 17761762 DOI: 10.1210/en.2007-0946] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
It has been previously reported that male and female somatostatin (SST) knockout mice (Sst-/-) release more GH, compared with Sst+/+ mice, due to enhanced GH-secretory vesicle release. Endogenous SST may also regulate GH secretion by directly inhibiting GHRH-stimulated GH gene expression and/or by modulating hypothalamic GHRH input. To begin to explore these possibilities and to learn more about the gender-dependent role of SST in modulating GH-axis function, hypothalamic, pituitary, and liver components of the GH-axis were compared in male and female Sst+/+ and Sst-/- mice. Pituitary mRNA levels for GH and receptors for GHRH and ghrelin were increased in female Sst-/- mice, compared with Sst+/+ controls, and these changes were reflected by an increase in circulating GH and IGF-I. Elevated levels of IGF-I in female Sst-/- mice were associated with elevated hepatic mRNA levels for IGF-I, as well as for GH and prolactin receptors. Consistent with the role of GH/IGF-I in negative feedback regulation of hypothalamic function, GHRH mRNA levels were reduced in female Sst-/- mice, whereas cortistatin (CST) mRNA levels were unaltered. In contrast to the widespread impact of SST loss on GH-axis function in females, only circulating GH, hypothalamic CST, and hepatic prolactin receptor expression were up-regulated in Sst-/- male mice, compared with Sst+/+ controls. These results confirm and extend the sexually dimorphic role of SST on GH-axis regulation, and suggest that CST, a neuropeptide that acts through SST receptors to inhibit GH secretion, may serve a compensatory role in maintaining GH-axis function in Sst-/- male mice.
Collapse
MESH Headings
- Animals
- Blotting, Northern
- Cells, Cultured
- Enzyme-Linked Immunosorbent Assay
- Female
- Gene Deletion
- Growth Hormone/blood
- Growth Hormone/genetics
- Growth Hormone/metabolism
- Insulin-Like Growth Factor I/genetics
- Insulin-Like Growth Factor I/metabolism
- Liver/metabolism
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Pituitary Gland/cytology
- Pituitary Gland/metabolism
- Radioimmunoassay
- Receptors, Ghrelin/blood
- Receptors, Ghrelin/genetics
- Receptors, Ghrelin/metabolism
- Receptors, Pituitary Hormone-Regulating Hormone/blood
- Receptors, Pituitary Hormone-Regulating Hormone/genetics
- Receptors, Pituitary Hormone-Regulating Hormone/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Sex Factors
- Somatostatin/genetics
- Somatostatin/physiology
Collapse
Affiliation(s)
- Raul M Luque
- Jesse Brown Veterans Affairs Medical Center, Research and Development Division, M.P 151, West Side, 820 South Damen Avenue, Chicago, IL 60612, USA
| | | |
Collapse
|
20
|
Veldhuis JD, Keenan DM, Bowers CY. Peripheral estrogen receptor-alpha selectively modulates the waveform of GH secretory bursts in healthy women. Am J Physiol Regul Integr Comp Physiol 2007; 293:R1514-21. [PMID: 17686882 DOI: 10.1152/ajpregu.00438.2007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Estradiol (E(2)) drives growth hormone (GH) secretion via estrogen receptors (ER) located in the hypothalamus and pituitary gland. ERalpha is expressed in GH releasing hormone (GHRH) neurons and GH-secreting cells (somatotropes). Moreover, estrogen regulates receptors for somatostatin, GHR peptide (GHRP, ghrelin), and GH itself, while potentiating signaling by IGF-I. Given this complex network, one cannot a priori predict the selective roles of hypothalamic compared with pituitary ER pathways. To make such a distinction, we introduce an investigative model comprising 1) specific ERalpha blockade with a pure antiestrogen, fulvestrant, that does not penetrate the blood-brain barrier; 2) graded transdermal E(2) administration, which doubles GH concentrations in postmenopausal women; 3) stimulation of fasting GH secretion by pairs of GHRH, GHRP-2 (a ghrelin analog), and l-arginine (to putatively limit somatostatin outflow); and 4) implementation of a flexible waveform deconvolution model to estimate the shape of secretory bursts independently of their size. The combined strategy unveiled that 1) E(2) prolongs GH secretory bursts via fulvestrant-antagonizable mechanisms; 2) fulvestrant extends GHRH/GHRP-2-stimulated secretory bursts; 3) l-arginine/GHRP-2 stimulation lengthens GH secretory bursts whether or not E(2) is present; 4) E(2) limits the capability of l-arginine/GHRP-2 to expand GH secretory bursts, and fulvestrant does not inhibit this effect; and 5) E(2) and/or fulvestrant do not alter the time evolution of l-arginine/GHRH-induced GH secretory bursts. The collective data indicate that peripheral ERalpha-dependent mechanisms determine the shape (waveform) of in vivo GH secretory bursts and that such mechanisms operate with secretagogue selectivity.
Collapse
Affiliation(s)
- Johannes D Veldhuis
- Endocrine Research Unit, Mayo Medical and Graduate Schools, Clinical Translational Research Unit, Mayo Clinic, Rochester, MN 55905, USA.
| | | | | |
Collapse
|
21
|
Crane C, Akhter N, Johnson BW, Iruthayanathan M, Syed F, Kudo A, Zhou YH, Childs GV. Fasting and glucose effects on pituitary leptin expression: is leptin a local signal for nutrient status? J Histochem Cytochem 2007; 55:1059-73. [PMID: 17595338 PMCID: PMC2085236 DOI: 10.1369/jhc.7a7214.2007] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Leptin, a potent anorexigenic hormone, is found in the anterior pituitary (AP). The aim of this study was to determine whether and how pituitary leptin-bearing cells are regulated by nutritional status. Male rats showed 64% reductions in pituitary leptin mRNA 24 hr after fasting, accompanied by significant (30-50%) reductions in growth hormone (GH), prolactin, and luteinizing hormone (LH), and 70-80% reductions in target cells for gonadotropin-releasing hormone or growth hormone-releasing hormone. There was a 2-fold increase in corticotropes. Subsets (22%) of pituitary cells coexpressed leptin and GH, and <5% coexpressed leptin and LH, prolactin, thyroid-stimulating hormone, or adrenocorticotropic hormone. Fasting resulted in significant (55-75%) losses in cells with leptin proteins or mRNA, and GH or LH. To determine whether restoration of serum glucose could rescue leptin, LH, and GH, additional fasted rats were given 10% glucose water for 24 hr. Restoring serum glucose in fasted rats resulted in pituitary cell populations with normal levels of leptin and GH and LH cells. Similarly, LH and GH cells were restored in vitro after populations from fasted rats were treated for as little as 1 hr in 10-100 pg/ml leptin. These correlative changes in pituitary leptin, LH, and GH, coupled with leptin's rapid restoration of GH and LH in vitro, suggest that pituitary leptin may signal nutritional changes. Collectively, the findings suggest that pituitary leptin expression could be coupled to glucose sensors like glucokinase to facilitate rapid responses by the neuroendocrine system to nutritional cues.
Collapse
Affiliation(s)
- Christopher Crane
- Department of Neurobiology and Developmental Sciences, College of Medicine, University of Arkansas for Medical Sciences, 4301 W. Markham St. Slot 510, Little Rock, AR 72205
| | - Noor Akhter
- Department of Neurobiology and Developmental Sciences, College of Medicine, University of Arkansas for Medical Sciences, 4301 W. Markham St. Slot 510, Little Rock, AR 72205
| | - Brandy W. Johnson
- Department of Neurobiology and Developmental Sciences, College of Medicine, University of Arkansas for Medical Sciences, 4301 W. Markham St. Slot 510, Little Rock, AR 72205
| | - Mary Iruthayanathan
- Division of Endocrinology, Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa; Bldg 40 VA, Iowa City, Iowa 52242
| | - Farhan Syed
- Department of Neurobiology and Developmental Sciences, College of Medicine, University of Arkansas for Medical Sciences, 4301 W. Markham St. Slot 510, Little Rock, AR 72205
| | - Akihiko Kudo
- Department of Anatomy, Kyorin University School of Medicine, Shinkawa, Mitaka, Tokyo, 1818611, Japan
| | - Yi-Hong Zhou
- Department of Neurological Surgery, University of California Irvine, 101 The City Drive, Building 36, Suite 400 Zot 5397, Orange, CA 92868
| | - Gwen V. Childs
- Department of Neurobiology and Developmental Sciences, College of Medicine, University of Arkansas for Medical Sciences, 4301 W. Markham St. Slot 510, Little Rock, AR 72205
| |
Collapse
|
22
|
Veldhuis JD, Keenan DM, Bowers CY. Estimation of the size and shape of GH secretory bursts in healthy women using a physiological estradiol clamp and variable-waveform deconvolution model. Am J Physiol Regul Integr Comp Physiol 2007; 293:R1013-21. [PMID: 17537842 DOI: 10.1152/ajpregu.00159.2007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Because estrogen production and age are strong covariates, distinguishing their individual impact on hypothalamo-pituitary regulation of growth hormone (GH) output is difficult. In addition, at fixed elimination kinetics, systemic GH concentration patterns are controlled by three major signal types [GH-releasing hormone (GHRH), GH-releasing peptide (GHRP, ghrelin), and somatostatin (SS)] and by four dynamic mechanisms [the number, mass (size), and shape (waveform) of secretory bursts and basal (time invariant) GH secretion]. The present study introduces an investigative strategy comprising 1) imposition of an experimental estradiol clamp in pre- (PRE) and postmenopausal (POST) women; 2) stimulation of fasting GH secretion by each of GHRH, GHRP-2 (a ghrelin analog), and l-arginine (to putatively limit SSergic restraint); and 3) implementation of a flexible-waveform deconvolution model to estimate basal GH secretion simultaneously with the size and shape of secretory bursts, conditional on pulse number. The combined approach unveiled the following salient percent POST/PRE contrasts: 1) only 27% as much GH secreted in bursts during fasting (P < 0.001); 2) markedly attenuated burstlike GH secretion in response to bolus GHRP-2 (29%), bolus GHRH (30%), l-arginine (37%), constant GHRP-2 (38%), and constant GHRH (42%) (age contrasts, 0.0016 </= P </= 0.027); and 3) a 160% prolongation and 32% abbreviation of the time required to achieve maximal GH secretion after injection of l-arginine and bolus GHRP-2, respectively (both, P < 0.001). Accordingly, age selectively determines both the size (amount) and shape (waveform) of GH secretory bursts in healthy women independently of the short-term estrogen milieu.
Collapse
|
23
|
Akhter N, Johnson BW, Crane C, Iruthayanathan M, Zhou YH, Kudo A, Childs GV. Anterior pituitary leptin expression changes in different reproductive states: in vitro stimulation by gonadotropin-releasing hormone. J Histochem Cytochem 2007; 55:151-66. [PMID: 17046838 PMCID: PMC1780073 DOI: 10.1369/jhc.6a7072.2006] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
This study was designed to learn more about the changes in expression of rat anterior pituitary (AP) leptin during the estrous cycle. QRT-PCR assays of cycling rat AP leptin mRNA showed 2-fold increases from metestrus to diestrus followed by an 86% decrease on the morning of proestrus. Percentages of leptin cells increased in proestrus and pregnancy to 55-60% of AP cells. Dual labeling for leptin proteins and growth hormone (GH) or gonadotropins showed that the rise in leptin protein-bearing cells from diestrus to proestrus was mainly in GH cells. Only 10-20% of leptin cells in male or cycling female rats coexpress gonadotropins. In contrast, 50-73% of leptin cells from pregnant or lactating females coexpress gonadotropins and only 19% coexpress GH, indicating plasticity in the distribution of leptin. Leptin cells expressed GnRH receptors, and estrogen and GnRH together increased the coexpression of leptin mRNA and gonadotropins. GnRH increased cellular leptin proteins three to four times and mRNA 9.8 times in proestrous rats and stimulated leptin secretion in cultures from diestrous, proestrous, and pregnant rats. These regulatory influences, and the high expression of AP leptin during proestrus and pregnancy, suggest a supportive role for leptin during key events involved with reproduction.
Collapse
Affiliation(s)
- Noor Akhter
- Department of Neurobiology and Developmental Sciences, College of Medicine, University of Arkansas for Medical Sciences, 4301 W. Markham St., Slot 510, Little Rock, AR 72205, USA.
| | | | | | | | | | | | | |
Collapse
|
24
|
Ramos-Vara JA, Miller MA. Comparison of two polymer-based immunohistochemical detection systems: ENVISION+? and ImmPRESS? J Microsc 2006; 224:135-9. [PMID: 17204059 DOI: 10.1111/j.1365-2818.2006.01679.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The non-specific background reaction produced in avidin-biotin-based immunohistochemistry, particularly after harsh antigen retrieval procedures, has promoted the use of non-avidin-biotin systems, yet there are few reports comparing the performance of non-avidin-biotin, polymer-based methods. In this study we compare two of these methods, ENVISION+trade mark and ImmPRESS, in animal tissues. We examined the immunoreactivity of 18 antigens in formalin-fixed, paraffin-embedded tissues. Antigens were located in the cytoplasmic membrane (CD11d, CD18 and CD79a), cytoplasm (calretinin, COX-1, COX-2, Glut-1, HepPar 1, KIT, Melan A, tryptase and uroplakin III) or nucleus (MUM-1, PGP 9.5 and thyroid transcription factor 1). We also evaluated three infectious agents (Aspergillus, calicivirus and West Nile virus). The staining with ENVISION+ or ImmPRESS was performed simultaneously for each antigen. The intensity of the reaction and background staining were scored. ImmPRESS yielded similar or higher reaction intensity than ENVISION+trade mark in 16/18 antigens. ImmPRESS produced abundant background with the other two antigens (calretinin and COX-2), which hindered interpretation of the specific reaction. The cost of ImmPRESS was 25% lower than for ENVISION+trade mark. Based on these results, ImmPRESS is a good polymer-based detection system for routine immunohistochemistry.
Collapse
Affiliation(s)
- José A Ramos-Vara
- Purdue University, Animal Disease Diagnostic Laboratory, School of Veterinary Medicine, 406 South University, West Lafayette, IN 47905, USA.
| | | |
Collapse
|
25
|
Childs GV, Iruthayanathan M, Akhter N, Johnson BW. Estrogen mediated cross talk between the ovary and pituitary somatotrope. Pre-ovulatory support for reproductive activity. Mol Cell Endocrinol 2006; 247:60-3. [PMID: 16443322 PMCID: PMC1751516 DOI: 10.1016/j.mce.2005.12.049] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2005] [Revised: 12/13/2005] [Accepted: 12/14/2005] [Indexed: 11/16/2022]
Affiliation(s)
- Gwen V Childs
- Department of Neurobiology and Developmental Sciences, College of Medicine, 4301 W. Markham, Slot 510, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
| | | | | | | |
Collapse
|
26
|
Iruthayanathan M, Zhou YH, Childs GV. Dehydroepiandrosterone restoration of growth hormone gene expression in aging female rats, in vivo and in vitro: evidence for actions via estrogen receptors. Endocrinology 2005; 146:5176-87. [PMID: 16150906 PMCID: PMC1868401 DOI: 10.1210/en.2005-0811] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
A decline in dehydroepiandrosterone (DHEA) and GH levels with aging may be associated with frailty and morbidity. Little is known about the direct effects of DHEA on somatotropes. We recently reported that 17beta-estradiol (E2), a DHEA metabolite, stimulates the expression of GH in vitro in young female rats. To test the hypothesis that DHEA restores function in aging somatotropes, dispersed anterior pituitary (AP) cells from middle-aged (12-14 months) or young (3-4 months) female rats were cultured in vitro with or without DHEA or E2 and fixed for immunolabeling or in situ hybridization. E2 increased the percentage of AP cells with GH protein or mRNA in the aged rats to young levels. DHEA increased the percentages of somatotropes (detected by GH protein or mRNA) from 14-16 +/- 2% to 29-31 +/- 3% (P < or = 0.05) and of GH mRNA (detected by quantitative RT-PCR) only in aging rats. To test DHEA's in vivo effects, 18-month-old female rats were injected with DHEA or vehicle for 2.5 d, followed by a bolus of GHRH 1 h before death. DHEA treatment increased serum GH 1.8-fold (7 +/- 0.5 to 12 +/- 1.3 ng/ml; P = 0.02, by RIA) along with a similar increase (P = 0.02) in GH immunolabel. GHRH target cells also increased from 11 +/- 1% to 19 +/- 2% (P = 0.03). Neither GH nor GHRH receptor mRNAs levels were changed. To test the mechanisms behind DHEA's actions, AP cells from aging rats were treated with DHEA with or without inhibitors of DHEA metabolism. Trilostane, aminogluthemide, or ICI 182,780 completely blocked the stimulatory effects of DHEA, suggesting that DHEA metabolites may stimulate aging somatotropes via estrogen receptors.
Collapse
Key Words
- ap, anterior pituitary
- dhea, dehydroepiandrosterone
- e2, 17β-estradiol
- er, estrogen receptor
- ghrh r, ghrh receptor
- hprt, hypoxanthine guanine phosphoribosyltransferase
- 3β-hsd, 3β-hydroxysteroid dehydrogenase
- iod, integrated optical density
- its, insulin, transferrin, sodium selenite, and bsa
- qrt-pcr, quantitative rt-pcr
Collapse
Affiliation(s)
| | | | - Gwen V. Childs
- Address all correspondence and requests for reprints to: Dr. Gwen V. Childs, Department of Neurobiology and Developmental Sciences, College of Medicine, 4301 W. Markham, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72212. E-mail:
| |
Collapse
|