Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 648
Filter
Add more filters

Publication year range
1.
Am J Physiol Regul Integr Comp Physiol ; 318(5): R855-R869, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32186897

ABSTRACT

Angiotensin II (ANG II) Agtr1a receptor (AT1A) is expressed in cells of the arcuate nucleus of the hypothalamus that express the leptin receptor (Lepr) and agouti-related peptide (Agrp). Agtr1a expression in these cells is required to stimulate resting energy expenditure in response to leptin and high-fat diets (HFDs), but the mechanism activating AT1A signaling by leptin remains unclear. To probe the role of local paracrine/autocrine ANG II generation and signaling in this mechanism, we bred mice harboring a conditional allele for angiotensinogen (Agt, encoding AGT) with mice expressing Cre-recombinase via the Lepr or Agrp promoters to cause cell-specific deletions of Agt (AgtLepr-KO and AgtAgrp-KO mice, respectively). AgtLepr-KO mice were phenotypically normal, arguing against a paracrine/autocrine AGT signaling mechanism for metabolic control. In contrast, AgtAgrp-KO mice exhibited reduced preweaning survival, and surviving adults exhibited altered renal structure and steroid flux, paralleling previous reports of animals with whole body Agt deficiency or Agt disruption in albumin (Alb)-expressing cells (thought to cause liver-specific disruption). Surprisingly, adult AgtAgrp-KO mice exhibited normal circulating AGT protein and hepatic Agt mRNA expression but reduced Agt mRNA expression in adrenal glands. Reanalysis of RNA-sequencing data sets describing transcriptomes of normal adrenal glands suggests that Agrp and Alb are both expressed in this tissue, and fluorescent reporter gene expression confirms Cre activity in adrenal gland of both Agrp-Cre and Alb-Cre mice. These findings lead to the iconoclastic conclusion that extrahepatic (i.e., adrenal) expression of Agt is critically required for normal renal development and survival.


Subject(s)
Adrenal Glands/metabolism , Agouti-Related Protein/metabolism , Angiotensinogen/metabolism , Arcuate Nucleus of Hypothalamus/metabolism , Energy Metabolism , Kidney/metabolism , Receptors, Leptin/metabolism , Adrenal Glands/growth & development , Agouti-Related Protein/deficiency , Agouti-Related Protein/genetics , Angiotensinogen/deficiency , Angiotensinogen/genetics , Animals , Arcuate Nucleus of Hypothalamus/growth & development , Autocrine Communication , Female , Gene Expression Regulation, Developmental , Kidney/growth & development , Male , Mice, Knockout , Myocardium/metabolism , Paracrine Communication , Receptors, Leptin/deficiency , Receptors, Leptin/genetics , Serum Albumin/genetics , Serum Albumin/metabolism , Signal Transduction
2.
Pediatr Res ; 87(4): 767-772, 2020 03.
Article in English | MEDLINE | ID: mdl-31645056

ABSTRACT

BACKGROUND: Most neonatal outcomes in neonates are related to normal adrenal gland function. Assessment of adrenal function in a sick preterm neonate remains a challenge, thus we hypothesized that adrenal steroid precursors to their product ratios have a direct relationship with neonatal outcomes. METHODS: We studied demographics of pregnancy and neonatal outcomes in 99 mother-infant pairs (24-41 weeks) and assayed 7 glucocorticoid precursors in the cortisol biosynthesis/degradation pathway. We correlated antenatal factors and short-term neonatal outcomes with these precursors and their ratios to assess maturity of individual enzymes. RESULTS: We found no correlation between cortisol levels with antenatal factors and outcomes. Antenatal steroid use impacted several cortisol precursors. 17-OH pregnenolone-to-cortisol ratio at birth was the best predictor of short-term neonatal outcomes, such as hypotension, RDS, IVH and PDA. A cord blood 17-OH pregnenolone:cortisol ratio of <0.21 predicts which neonate will have a normal outcome with a high sensitivity and specificity. CONCLUSIONS: Maternal factors and antenatal steroids impact neonatal adrenal function and leads to maturation of adrenal function. 17-OH pregnenolone:cortisol ratio and not cortisol is the best predictor of adrenal function. Adrenal function can be assessed by evaluating the profile of adrenal steroids.


Subject(s)
17-alpha-Hydroxypregnenolone/blood , Adrenal Cortex Function Tests , Adrenal Glands/metabolism , Hydrocortisone/blood , Adrenal Glands/growth & development , Age Factors , Biomarkers/blood , Child Development , Female , Humans , Infant, Newborn , Predictive Value of Tests , Pregnancy , Prognosis , Prospective Studies , Time Factors
3.
Curr Opin Pediatr ; 32(4): 574-581, 2020 08.
Article in English | MEDLINE | ID: mdl-32692055

ABSTRACT

PURPOSE OF REVIEW: Adrenarche is the pubertal maturation of the innermost zone of the adrenal cortex, the zona reticularis. The onset of adrenarche occurs between 6 and 8 years of age when dehydroepiandrosterone sulfate (DHEAS) concentrations increase. This review provides an update on adrenal steroidogenesis and the differential diagnosis of premature development of pubic hair. RECENT FINDINGS: The complexity of adrenal steroidogenesis has increased with recognition of the alternative 'backdoor pathway' and the 11-oxo-androgens pathways. Traditionally, sulfated steroids such as DHEAS have been considered to be inactive metabolites. Recent data suggest that intracellular sulfated steroids may function as tissue-specific intracrine hormones particularly in the tissues expressing steroid sulfatases such as ovaries, testes, and placenta. SUMMARY: The physiologic mechanisms governing the onset of adrenarche remain unclear. To date, no validated regulatory feedback mechanism has been identified for adrenal C19 steroid secretion. Available data indicate that for most children, premature adrenarche is a benign variation of development and a diagnosis of exclusion. Patients with premature adrenarche tend to have higher BMI values. Yet, despite greater knowledge about C19 steroids and zona reticularis function, much remains to be learned about adrenarche.


Subject(s)
Adrenal Glands , Adrenarche/metabolism , Adrenarche/physiology , Child Development/physiology , Puberty, Precocious , Puberty/physiology , Zona Reticularis/physiology , Adrenal Glands/growth & development , Adrenal Glands/metabolism , Adrenal Glands/physiology , Androgens , Child , Dehydroepiandrosterone Sulfate/blood , Dehydroepiandrosterone Sulfate/metabolism , Female , Humans , Pregnancy , Steroids/metabolism
4.
Cell Mol Life Sci ; 75(6): 1057-1070, 2018 03.
Article in English | MEDLINE | ID: mdl-29058015

ABSTRACT

Neuroblastoma (NB) is a tumor of the sympathoadrenal system arising in children under 15 years of age. In Germany, NB accounts for 7% of childhood cancer cases, but 11% of cancer deaths. It originates from highly migratory progenitor cells that leave the dorsal neural tube and contribute neurons and glial cells to sympathetic ganglia, and chromaffin and supportive cells to the adrenal medulla and paraganglia. Clinically, histologically and molecularly, NBs present as extremely heterogeneous, ranging from very good to very poor prognosis. The etiology of NB still remains unclear and needs to be elucidated, however, aberrant auto- and paracrine embryonic cell communications seem to be likely candidates to initiate or facilitate the emergence, progression and regression of NB. The wingless-type MMTV integration site (WNT) family of proteins represents an evolutionary highly conserved signaling system that orchestrates embryogenesis. At least 19 ligands in the human, numerous receptors and co-receptors are known, which control not only proliferation, but also cell polarity, migration and differentiation. Here we seek to interconnect aspects of WNT signaling with sympathoadrenal and paraganglionic development to define new WNT signaling cues in the etiology and progression of NB.


Subject(s)
Adrenal Gland Diseases/genetics , Gene Expression Regulation, Neoplastic , Neuroblastoma/genetics , Paraganglioma/genetics , Wnt Proteins/genetics , Wnt Signaling Pathway/genetics , Adolescent , Adrenal Gland Diseases/metabolism , Adrenal Gland Diseases/mortality , Adrenal Gland Diseases/pathology , Adrenal Glands/growth & development , Adrenal Glands/metabolism , Adrenal Glands/pathology , Child , Child, Preschool , Humans , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neural Crest/growth & development , Neural Crest/metabolism , Neural Crest/pathology , Neuroblastoma/metabolism , Neuroblastoma/mortality , Neuroblastoma/pathology , Neurons/metabolism , Neurons/pathology , Paraganglioma/metabolism , Paraganglioma/mortality , Paraganglioma/pathology , Stem Cells/metabolism , Stem Cells/pathology , Survival Analysis , Sympathetic Nervous System/growth & development , Sympathetic Nervous System/metabolism , Sympathetic Nervous System/pathology , Wnt Proteins/metabolism
5.
Biochem J ; 475(1): 75-85, 2018 01 02.
Article in English | MEDLINE | ID: mdl-29127254

ABSTRACT

The translocator protein (TSPO) has been proposed to act as a key component in a complex important for mitochondrial cholesterol importation, which is the rate-limiting step in steroid hormone synthesis. However, TSPO function in steroidogenesis has recently been challenged by the development of TSPO knockout (TSPO-KO) mice, as they exhibit normal baseline gonadal testosterone and adrenal corticosteroid production. Here, we demonstrate that despite normal androgen levels in young male TSPO-KO mice, TSPO deficiency alters steroidogenic flux and results in reduced total steroidogenic output. Specific reductions in the levels of progesterone and corticosterone as well as age-dependent androgen deficiency were observed in both young and aged male TSPO-KO mice. Collectively, these findings indicate that while TSPO is not critical for achieving baseline testicular and adrenal steroidogenesis, either indirect effects of TSPO on steroidogenic processes, or compensatory mechanisms and functional redundancy, lead to subtle steroidogenic abnormalities which become exacerbated with aging.


Subject(s)
Adrenal Glands/metabolism , Aging/genetics , Gene Expression Regulation, Developmental , Receptors, GABA/genetics , Testis/metabolism , Adrenal Glands/growth & development , Aging/metabolism , Aldosterone/biosynthesis , Androgens/biosynthesis , Animals , Corticosterone/biosynthesis , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Progesterone/biosynthesis , Receptors, GABA/deficiency , Testis/growth & development
6.
PLoS Genet ; 11(10): e1005583, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26492326

ABSTRACT

Remodelling the methylome is a hallmark of mammalian development and cell differentiation. However, current knowledge of DNA methylation dynamics in human tissue specification and organ development largely stems from the extrapolation of studies in vitro and animal models. Here, we report on the DNA methylation landscape using the 450k array of four human tissues (amnion, muscle, adrenal and pancreas) during the first and second trimester of gestation (9,18 and 22 weeks). We show that a tissue-specific signature, constituted by tissue-specific hypomethylated CpG sites, was already present at 9 weeks of gestation (W9). Furthermore, we report large-scale remodelling of DNA methylation from W9 to W22. Gain of DNA methylation preferentially occurred near genes involved in general developmental processes, whereas loss of DNA methylation mapped to genes with tissue-specific functions. Dynamic DNA methylation was associated with enhancers, but not promoters. Comparison of our data with external fetal adrenal, brain and liver revealed striking similarities in the trajectory of DNA methylation during fetal development. The analysis of gene expression data indicated that dynamic DNA methylation was associated with the progressive repression of developmental programs and the activation of genes involved in tissue-specific processes. The DNA methylation landscape of human fetal development provides insight into regulatory elements that guide tissue specification and lead to organ functionality.


Subject(s)
Cell Differentiation/genetics , DNA Methylation/genetics , Epigenesis, Genetic , Fetal Development/genetics , Adrenal Glands/growth & development , Adrenal Glands/metabolism , Amnion/growth & development , Amnion/metabolism , CpG Islands/genetics , Female , Gene Expression Regulation, Developmental , Genome, Human , Humans , Muscle, Skeletal/growth & development , Muscle, Skeletal/metabolism , Organ Specificity/genetics , Pancreas/growth & development , Pancreas/metabolism , Pregnancy , Pregnancy Trimester, First/genetics , Pregnancy Trimester, Second/genetics , Promoter Regions, Genetic , Regulatory Sequences, Nucleic Acid
7.
Int J Mol Sci ; 19(4)2018 Apr 07.
Article in English | MEDLINE | ID: mdl-29642441

ABSTRACT

Compensatory adrenal growth evoked by unilateral adrenalectomy (hemiadrenalectomy) constitutes one of the most frequently studied in vivo models of adrenocortical enlargement. This type of growth has been quite well characterized for its morphological, biochemical, and morphometric parameters. However, the molecular basis of compensatory adrenal growth is poorly understood. Therefore, the aim of this study was to investigate the rat adrenal transcriptome profile during the time of two previously described adrenocortical proliferation waves at 24 and 72 h after unilateral adrenalectomy. Surgical removal of the left adrenal or a sham operation was accomplished via the classic dorsal approach. As expected, the weight of the remaining right adrenal glands collected at 24 and 72 h after hemiadrenalectomy increased significantly. The transcriptome profile was identified by means of Affymetrix® Rat Gene 2.1 ST Array. The general profiles of differentially expressed genes were visualized as volcano plots and heatmaps. Detailed analyzes consisted of identifying significantly enriched gene ontological groups relevant to adrenal physiology, by means of DAVID and GOplot bioinformatics tools. The results of our studies showed that compensatory adrenal growth induced by unilateral adrenalectomy exerts a limited influence on the global transcriptome profile of the rat adrenal gland; nevertheless, it leads to significant changes in the expression of key genes regulating the circadian rhythm. Our results confirm also that regulation of compensatory adrenal growth is under complex and multifactorial control with a pivotal role of neural regulatory mechanisms and a supportive role of other components.


Subject(s)
Adrenal Glands/growth & development , Adrenalectomy/methods , Gene Expression Profiling/methods , Oligonucleotide Array Sequence Analysis/methods , Adrenal Glands/chemistry , Adrenal Glands/surgery , Animals , Circadian Rhythm , Disease Models, Animal , Gene Expression Regulation , Gene Ontology , Gene Regulatory Networks , Male , Organ Size , Rats
8.
Clin Endocrinol (Oxf) ; 86(3): 325-331, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27748964

ABSTRACT

OBJECTIVE: To evaluate volumetric and densitometric properties of the adrenal glands in patients with unilateral and bilateral disease in comparison with normal controls. DESIGN: A total of 77 patients (56 males and 21 females) diagnosed with primary aldosteronism (PA) with a mean age of 53 ± 10 years were prospectively enrolled. Unenhanced and contrast-enhanced computed tomography scans were analysed for adrenal volumes and mean densities. These values were compared with normal controls and between PA subtypes. RESULTS: Adrenals containing an aldosterone-producing adenoma (APA, n = 56) had on average higher attenuation values as compared to adrenals with bilateral adrenal hyperplasia (n = 21). Mean adrenal gland volume in PA patients was not significantly different between PA subtypes. In comparison with normal adrenal glands, volumes were significantly higher in PA patients (P < 0·0001) including adrenals contralateral to APAs, which were significantly larger in comparison with controls. CONCLUSION: Independent of subtype differentiation, adrenal volumetry reveals higher adrenal volumes in PA patients in comparison with normal controls. These findings provide indirect evidence for a general adrenal growth dysregulation in the context of PA.


Subject(s)
Adrenal Glands/pathology , Hyperaldosteronism/diagnosis , Adenoma/metabolism , Adenoma/pathology , Adrenal Glands/growth & development , Adult , Aged , Aldosterone/metabolism , Case-Control Studies , Diagnosis, Differential , Female , Germany , Humans , Hyperplasia , Male , Middle Aged , Organ Size , Tomography, X-Ray Computed , Young Adult
9.
Biochemistry (Mosc) ; 82(3): 373-379, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28320279

ABSTRACT

The morphogenesis of individual organs and the whole organism occurs under the control of intercellular chemical signals mainly during the perinatal period of ontogenesis in rodents. In this study, we tested our hypothesis that the biologically active concentration of noradrenaline (NA) in blood in perinatal ontogenesis of rats is maintained due to humoral interaction between its central and peripheral sources based on their plasticity. As one of the mechanisms of plasticity, we examined changes in the secretory activity (spontaneous and stimulated release of NA) of NA-producing organs under deficiency of its synthesis in the brain. The destruction of NA-ergic neurons was provoked by administration of a hybrid molecular complex - antibodies against dopamine-ß-hydroxylase associated with the cytotoxin saporin - into the lateral cerebral ventricles of neonatal rats. We found that 72 h after the inhibition of NA synthesis in the brain, its spontaneous release from hypothalamus increased, which was most likely due to a compensatory increase of NA secretion from surviving neurons and can be considered as one of the mechanisms of neuroplasticity aimed at the maintenance of its physiological concentration in peripheral blood. Noradrenaline secretion from peripheral sources (adrenal glands and the organ of Zuckerkandl) also showed a compensatory increase in this model. Thus, during the critical period of morphogenesis, the brain is integrated into the system of NA-producing organs and participates in their reciprocal humoral regulation as manifested in compensatory enhancement of NA secretion in each of the studied sources of NA under specific inhibition of NA production in the brain.


Subject(s)
Adrenal Glands , Adrenergic Neurons/metabolism , Cerebral Ventricles , Hypothalamus , Norepinephrine/metabolism , Para-Aortic Bodies/metabolism , Adrenal Glands/growth & development , Adrenal Glands/metabolism , Animals , Cerebral Ventricles/growth & development , Cerebral Ventricles/metabolism , Dopamine beta-Hydroxylase/metabolism , Hypothalamus/growth & development , Hypothalamus/metabolism , Male , Rats , Rats, Wistar , Ribosome Inactivating Proteins, Type 1/toxicity , Saporins
10.
Horm Metab Res ; 48(4): 242-6, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27065461

ABSTRACT

Primary pigmented nodular adrenocortical disease (PPNAD) is a rare type of bilateral adrenal hyperplasia leading to hypercortisolemia. Adrenal nodularity is often appreciable with computed tomography (CT); however, accurate radiologic characterization of adrenal size in PPNAD has not been studied well. We used 3-dimensional (3D) volumetric analysis to characterize and compare adrenal size in PPNAD patients, with and without Cushing's syndrome (CS). Patients diagnosed with PPNAD and their family members with known mutations in PRKAR1A were screened. CT scans were used to create 3D models of each adrenal. Criteria for biochemical diagnosis of CS included loss of diurnal variation and/or elevated midnight cortisol levels, and paradoxical increase in urinary free cortisol and/or urinary 17-hydroxysteroids after dexamethasone administration. Forty-five patients with PPNAD (24 females, 27.8±17.6 years) and 8 controls (19±3 years) were evaluated. 3D volumetric modeling of adrenal glands was performed in all. Thirty-eight patients out of 45 (84.4%) had CS. Their mean adrenal volume was 8.1 cc±4.1, 7.2 cc±4.5 (p=0.643) for non-CS, and 8.0cc±1.6 for controls. Mean values were corrected for body surface area; 4.7 cc/kg/m(2)±2.2 for CS, and 3.9 cc/kg/m(2)±1.3 for non-CS (p=0.189). Adrenal volume and midnight cortisol in both groups was positively correlated, r=0.35, p=0.03. We conclude that adrenal volume measured by 3D CT in patients with PPNAD and CS was similar to those without CS, confirming empirical CT imaging-based observations. However, the association between adrenal volume and midnight cortisol levels may be used as a marker of who among patients with PPNAD may develop CS, something that routine CT cannot do.


Subject(s)
Adrenal Glands/growth & development , Adrenal Hyperplasia, Congenital/diagnostic imaging , Cone-Beam Computed Tomography , Cushing Syndrome/diagnostic imaging , Adolescent , Adrenal Glands/anatomy & histology , Adrenal Hyperplasia, Congenital/genetics , Adrenal Hyperplasia, Congenital/metabolism , Adrenal Hyperplasia, Congenital/physiopathology , Adult , Child , Child, Preschool , Cushing Syndrome/genetics , Cushing Syndrome/metabolism , Cushing Syndrome/physiopathology , Cyclic AMP-Dependent Protein Kinase RIalpha Subunit/genetics , Cyclic AMP-Dependent Protein Kinase RIalpha Subunit/metabolism , Female , Humans , Hydrocortisone/metabolism , Male , Middle Aged , Organ Size , Young Adult
11.
PLoS Genet ; 9(1): e1003160, 2013.
Article in English | MEDLINE | ID: mdl-23300479

ABSTRACT

Mouse sex determination provides an attractive model to study how regulatory genetic networks and signaling pathways control cell specification and cell fate decisions. This study characterizes in detail the essential role played by the insulin receptor (INSR) and the IGF type I receptor (IGF1R) in adrenogenital development and primary sex determination. Constitutive ablation of insulin/IGF signaling pathway led to reduced proliferation rate of somatic progenitor cells in both XX and XY gonads prior to sex determination together with the downregulation of hundreds of genes associated with the adrenal, testicular, and ovarian genetic programs. These findings indicate that prior to sex determination somatic progenitors in Insr;Igf1r mutant gonads are not lineage primed and thus incapable of upregulating/repressing the male and female genetic programs required for cell fate restriction. In consequence, embryos lacking functional insulin/IGF signaling exhibit (i) complete agenesis of the adrenal cortex, (ii) embryonic XY gonadal sex reversal, with a delay of Sry upregulation and the subsequent failure of the testicular genetic program, and (iii) a delay in ovarian differentiation so that Insr;Igf1r mutant gonads, irrespective of genetic sex, remained in an extended undifferentiated state, before the ovarian differentiation program ultimately is initiated at around E16.5.


Subject(s)
Gonads , Insulin , Receptor, IGF Type 1 , Receptor, Insulin , Sex Determination Processes/genetics , Adrenal Cortex/growth & development , Adrenal Cortex/pathology , Adrenal Glands/growth & development , Adrenal Glands/metabolism , Animals , Cell Differentiation/genetics , Cell Lineage , Cell Proliferation , Disorders of Sex Development/genetics , Female , Gonads/growth & development , Gonads/metabolism , Gonads/pathology , Humans , Insulin/genetics , Insulin/metabolism , Male , Mice , Ovary/growth & development , Ovary/metabolism , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/metabolism , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , Sex Chromosomes , Signal Transduction , Testis/growth & development , Testis/metabolism
12.
Genet Mol Res ; 15(2)2016 May 06.
Article in English | MEDLINE | ID: mdl-27173313

ABSTRACT

The effect of weaning age on the adrenal cortex, which plays a vital role in the stress response, is currently unknown. Therefore, plasma adrenocorticotropic hormone (ACTH) and cortisol levels, weights and relative weights of adrenal glands, and steroidogenesis-related protein and enzyme expression levels in piglets weaned on different days were determined. Piglets weaned at 35 days had significantly lower ACTH levels than those weaned at 14 or 21 days, and cortisol levels of piglets weaned at 21, 28, and 35 days were significantly lower than those of piglets weaned on day 14. Adrenal gland weights of piglets weaned at 28 and 35 days and relative adrenal gland weights of piglets weaned at 35 days were significantly lower than those of piglets weaned at 14 days. However, no significant difference was detected in the expression of melanocortin-type 2 receptor mRNA, which is associated with weaning age. Steroidogenic acute-regulatory (StAR) mRNA and cholesterol side-chain cleavage cytochrome P450 mRNA expression levels in piglets weaned at 28 and 35 days were significantly lower than in those weaned at 14 or 21 days, and P450 11ß mRNA expression levels in piglets weaned at 28 and 35 days were significantly lower than in those weaned at 14 days. Therefore, early-weaned piglets exhibited increased adrenal gland weights and StAR and steroidogenic enzyme expression, all of which contributed to high cortisol levels. The high plasma ACTH and cortisol levels in early-weaned piglets indicate that these animals would be greatly affected by stress.


Subject(s)
Hydrocortisone/blood , Weaning , Adrenal Glands/growth & development , Adrenal Glands/metabolism , Adrenocorticotropic Hormone/blood , Animals , Cytochrome P450 Family 2/genetics , Cytochrome P450 Family 2/metabolism , Female , Male , Phosphoproteins/genetics , Phosphoproteins/metabolism , Receptor, Melanocortin, Type 2/genetics , Receptor, Melanocortin, Type 2/metabolism , Swine
13.
Dokl Biochem Biophys ; 466: 74-6, 2016.
Article in English | MEDLINE | ID: mdl-27025493

ABSTRACT

Using the method of high performance liquid chromatography with electrochemical detection, the age dynamics of the content of noradrenaline (NA) in the brain, adrenal gland, and the organ of Zuckerkandl in prenatal (18th and 21st days of embryogenesis) and early postnatal (3, 7, 15, and 30th days) periods of development was studied. The potential contribution of these organs to the formation of physiologically active concentration of noradrenalin in the blood was also assessed. The results suggest that, during the development of the organism, the activity of the sources of noradrenaline in the general circulation changes, which gives a reason to assume the existence of humoral interaction between NA-producing organs in the perinatal period of ontogenesis.


Subject(s)
Adrenal Glands/growth & development , Brain/growth & development , Norepinephrine/metabolism , Signal Transduction , Adrenal Glands/embryology , Adrenal Glands/metabolism , Animals , Brain/embryology , Brain/metabolism , Growth , Male , Norepinephrine/blood , Para-Aortic Bodies/metabolism , Rats , Rats, Wistar
14.
Dokl Biochem Biophys ; 467(1): 153-6, 2016 Mar.
Article in English | MEDLINE | ID: mdl-27193722

ABSTRACT

Spontaneous and K(+)-stimulated release of noradrenaline from the hypothalamus, adrenal gland, and organ of Zuckerkandl under their flowing incubation was investigated in the perinatal period of ontogenesis of rats. The results suggest that, during the investigated period of ontogenesis, adrenal glands are the main source of noradrenaline in the blood, whereas the contributions of the organ of Zuckerkandl and the brain are not as significant and change during this period.


Subject(s)
Adrenal Glands/metabolism , Hypothalamus/metabolism , Norepinephrine/metabolism , Para-Aortic Bodies/metabolism , Adrenal Glands/drug effects , Adrenal Glands/growth & development , Adrenergic Agents/administration & dosage , Animals , Blood Chemical Analysis , Cations, Monovalent/administration & dosage , Chromatography, High Pressure Liquid , Hypothalamus/drug effects , Hypothalamus/growth & development , Male , Para-Aortic Bodies/drug effects , Potassium/administration & dosage , Rats, Wistar
15.
Development ; 139(24): 4561-70, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23136395

ABSTRACT

Adrenal and gonadal steroids are essential for life and reproduction. The orphan nuclear receptor SF1 (NR5A1) has been shown to regulate the expression of enzymes involved in steroid production in vitro. However, the in vivo role of this transcription factor in steroidogenesis has not been elucidated. In this study, we have generated steroidogenic-specific Cre-expressing mice to lineage mark and delete Sf1 in differentiated steroid-producing cells of the testis, the ovary and the adrenal gland. Our data show that SF1 is a regulator of the expression of steroidogenic genes in all three organs. In addition, Sf1 deletion leads to a radical change in cell morphology and loss of identity. Surprisingly, sexual development and reproduction in mutant animals were not compromised owing, in part, to the presence of a small proportion of SF1-positive cells. In contrast to the testis and ovary, the mutant adult adrenal gland showed a lack of Sf1-deleted cells and our studies suggest that steroidogenic adrenal cells during foetal stages require Sf1 to give rise to the adult adrenal population. This study is the first to show the in vivo requirements of SF1 in steroidogenesis and provides novel data on the cellular consequences of the loss of this protein specifically within steroid-producing cells.


Subject(s)
Adrenal Glands/metabolism , DNA-Binding Proteins/physiology , Gonadal Steroid Hormones/metabolism , Ovary/metabolism , Testis/metabolism , Transcription Factors/physiology , Adrenal Glands/cytology , Adrenal Glands/embryology , Adrenal Glands/growth & development , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cholesterol Side-Chain Cleavage Enzyme/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Embryo, Mammalian , Female , Gene Deletion , Integrases/genetics , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Male , Mice , Mice, Transgenic , Ovary/cytology , Ovary/embryology , Ovary/growth & development , RNA Splicing Factors , Testis/cytology , Testis/embryology , Testis/growth & development , Transcription Factors/genetics , Transcription Factors/metabolism , Transgenes/genetics
16.
Pediatr Res ; 77(2): 298-309, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25470028

ABSTRACT

BACKGROUND: At birth, the large fetal adrenal involutes rapidly, and the patterns of steroidogenesis change dramatically; the event(s) triggering these changes remain largely unexplored. Fetal abdominal viscera receive hypoxic blood having a partial pressure of oxygen of only ~2 kPa (20-23 mm Hg); perinatal circulatory changes change this to adult values (~20 kPa). We hypothesized that transition from fetal hypoxia to postnatal normoxia participates in altering perinatal steroidogenesis. METHODS: We grew midgestation human fetal adrenal cells and human NCI-H295A adrenocortical carcinoma cells in 2% O2, then transitioned them to 20% O2 and quantitated steroidogenic mRNAs by quantitative PCR and microarrays. RESULTS: Transitioning fetal adrenal cells from hypoxia to normoxia increased mRNAs for 17α-hydroxylase/17,20 lyase (P450c17), 3ß-hydroxysteroid dehydrogenase (3ßHSD2), and steroidogenic acute regulatory protein (StAR). We repeated the protocol with NCI-H295A cells acclimated to hypoxia for 15 d, quantitating 31,255 transcripts by microarray. Using an arbitrary 1.5-fold difference, 1 d of normoxia increased 4 transcripts and decreased 56, whereas 2 d of normoxia increased 62 transcripts and decreased 105. P450c17, 3ßHSD2, and StAR were ranked among the top eight increased transcripts. CONCLUSION: These data suggest that the hypoxic/normoxic transition at birth contributes to perinatal changes in adrenal steroidogenesis.


Subject(s)
Adrenal Glands/growth & development , Adrenal Glands/metabolism , Culture Media/chemistry , Fetus/metabolism , Oxygen/metabolism , Steroids/biosynthesis , 3-Hydroxysteroid Dehydrogenases/metabolism , Cell Line, Tumor , Humans , Infant, Newborn , Microarray Analysis , Phosphoproteins/metabolism , Polymerase Chain Reaction , Steroid 17-alpha-Hydroxylase/metabolism
17.
Br J Nutr ; 113(10): 1539-49, 2015 May 28.
Article in English | MEDLINE | ID: mdl-25865869

ABSTRACT

Vitamin C (VitC) deficiency is surprisingly common in humans even in developed parts of the world. The micronutrient has several established functions in the brain; however, the consequences of its deficiency are not well characterised. To elucidate the effects of VitC deficiency on the brain, increased knowledge about the distribution of VitC to the brain and within different brain regions after varying dietary concentrations is needed. In the present study, guinea pigs (like humans lacking the ability to synthesise VitC) were randomly divided into six groups (n 10) that received different concentrations of VitC ranging from 100 to 1500 mg/kg feed for 8 weeks, after which VitC concentrations in biological fluids and tissues were measured using HPLC. The distribution of VitC was found to be dynamic and dependent on dietary availability. Brain saturation was region specific, occurred at low dietary doses, and the dose-concentration relationship could be approximated with a three-parameter Hill equation. The correlation between plasma and brain concentrations of VitC was moderate compared with other organs, and during non-scorbutic VitC deficiency, the brain was able to maintain concentrations from about one-quarter to half of sufficient levels depending on the region, whereas concentrations in other tissues decreased to one-sixth or less. The adrenal glands have similar characteristics to the brain. The observed distribution kinetics with a low dietary dose needed for saturation and exceptional retention ability suggest that the brain and adrenal glands are high priority tissues with regard to the distribution of VitC.


Subject(s)
Adrenal Glands/metabolism , Ascorbic Acid Deficiency/prevention & control , Ascorbic Acid/metabolism , Brain/metabolism , Dietary Supplements , Neurons/metabolism , Adrenal Glands/growth & development , Animals , Animals, Outbred Strains , Ascorbic Acid/administration & dosage , Ascorbic Acid/cerebrospinal fluid , Ascorbic Acid/therapeutic use , Ascorbic Acid Deficiency/blood , Ascorbic Acid Deficiency/cerebrospinal fluid , Ascorbic Acid Deficiency/metabolism , Brain/growth & development , Cerebellum/growth & development , Cerebellum/metabolism , Female , Frontal Lobe/growth & development , Frontal Lobe/metabolism , Guinea Pigs , Hippocampus/growth & development , Hippocampus/metabolism , Kidney/growth & development , Kidney/metabolism , Kinetics , Liver/growth & development , Liver/metabolism , Organ Specificity , Phosphorylation , Random Allocation , Tissue Distribution
18.
Gen Comp Endocrinol ; 217-218: 62-70, 2015.
Article in English | MEDLINE | ID: mdl-25776460

ABSTRACT

Androgens take part in the regulation of puberty and promote growth and development. They play their biological role by binding to a specific androgen receptor (AR). The aim of this study was to evaluate the expression of AR mRNA and protein in the pituitary and adrenal glands, to localize AR protein in luteinizing hormone (LH)-producing pituitary and adrenocortical cells, to determine plasma concentrations of adrenocorticotropic hormone (ACTH) and corticosterone and the concentrations of corticosterone, testosterone (T), androstenedione (A4) and oestradiol (E2) in the adrenal glands of male turkeys at the age of 4, 8, 12, 16, 20, 24 and 28weeks. The concentrations of hormones and the expression of AR varied during development. The expression of AR mRNA and protein in pituitary increased during the growth. The increase of AR mRNA levels in pituitary occurred earlier than increase of AR protein. The percentage of pituitary cells expressing ARs in the population of LH-secreting cells increased in week 20. It suggests that AR expression in LH-producing pituitary cells is determined by the phase of development. The drop in adrenal AR mRNA and protein expression was accompanied by an increase in the concentrations of adrenal androgens. Those results could point to the presence of a compensatory mechanism that enables turkeys to avoid the potentially detrimental effects of high androgen concentrations. Our results will expand our knowledge of the role of steroids in the development of the reproductive system of turkeys from the first month of age until maturity.


Subject(s)
Adrenal Glands/metabolism , Adrenocorticotropic Hormone/blood , Corticosterone/blood , Gonadal Steroid Hormones/blood , Pituitary Gland/metabolism , Receptors, Androgen/metabolism , Turkeys/metabolism , Adrenal Glands/growth & development , Animals , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Immunoenzyme Techniques , Male , Pituitary Gland/growth & development , RNA, Messenger/genetics , Radioimmunoassay , Real-Time Polymerase Chain Reaction , Receptors, Androgen/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sexual Maturation/drug effects , Turkeys/growth & development
19.
Ontogenez ; 46(4): 277-82, 2015.
Article in Russian | MEDLINE | ID: mdl-26480487

ABSTRACT

Histological structures and morphometric and some histochemical indicators of elk's adrenal gland development as subspecies of red deer in prenatal and postnatal ontogenies stages was studied. It was found that the growth of the fetus adrenal glands weight and the thickness of the structures adrenal glands fragments continue throughout the prenatal period of ontogeny. The cells of androgenic zone with single wandering sympathogoniae are differentiated in the adrenal glands in the second month of development. The androgenic and definite zone and the adrenal medulla are differentiated by the third month of development. At the 4 months, adrenal gland cortex zona glomerulosa and zona fasciculate-reticularis are differentiated; zona reticularis is differentiated only by the seventh month. By the eighth month, the structure of adrenal glands corresponds to the adrenal glands of a newborn. Full structural formation of the adrenal glands takes place in young animals by age 1.5. Obvious structural changes were not found late in the postnatal stages of development.


Subject(s)
Adrenal Glands , Aging/physiology , Deer , Embryonic Development/physiology , Fetal Development/physiology , Organogenesis/physiology , Adrenal Glands/cytology , Adrenal Glands/embryology , Adrenal Glands/growth & development , Animals , Animals, Newborn , Cell Differentiation/physiology , Deer/embryology , Deer/growth & development , Male
20.
Am J Physiol Regul Integr Comp Physiol ; 306(1): R67-74, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24133102

ABSTRACT

To explore developmental changes in circadian organization of central and peripheral oscillators, circadian rhythms in clock gene expression were examined in 12 organs in transgenic rats carrying a bioluminescence reporter for Per2. Organ slices were obtained from different developmental stages starting at postnatal day 5 and tissue was cultured for more than 6 days. In addition, four organs were examined from embryonic day 20. Robust circadian rhythms in bioluminescence were detected in all organs examined. The circadian period in vitro was specific to each organ and remained essentially the same during development. The circadian peak phase on the first day of culture was significantly different not only among organs but also in the same organ. Three patterns in circadian phase were detected during development. Thus, during development, circadian phase did not change in the suprachiasmatic nucleus, adrenal gland, and liver, whereas delay shifts were seen in the pineal, lung, heart, kidney, spleen, thymus, and testis. Finally, circadian phase advanced at postnatal day 10-15 and subsequently delayed in skeletal muscle and stomach.Circadian amplitude also showed developmental changes in several organs. These findings indicate that the temporal orders of physiological functions of various organs change during development. Such age-dependent and organ-specific changes in the phase relationship among circadian clocks most likely reflect entrainment to organ-specific time cues at different developmental stages.


Subject(s)
Circadian Rhythm/genetics , Gene Expression Regulation, Developmental , Period Circadian Proteins/genetics , Adrenal Glands/growth & development , Adrenal Glands/metabolism , Age Factors , Animals , Animals, Genetically Modified , Circadian Rhythm/physiology , Liver/growth & development , Liver/metabolism , Organ Specificity , Period Circadian Proteins/metabolism , Rats , Rats, Transgenic , Rats, Wistar , Spleen/growth & development , Spleen/metabolism , Suprachiasmatic Nucleus/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL