Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.138
Filter
1.
J Pregnancy ; 2024: 9968509, 2024.
Article in English | MEDLINE | ID: mdl-39238897

ABSTRACT

Background: Uteroplacental insufficiency in fetuses with growth restriction (FGR) leads to chronic hypoxia and stress, predominantly affecting the adrenal glands. However, the mechanisms of impact remain unclear. Objectives: This study is aimed at comparing the Doppler indices of the adrenal artery and the adrenal gland sizes between FGR and those with normal growth. Materials and Methods: A multicenter, cross-sectional study was conducted from February to December 2023. We compared 34 FGR to 34 with normal growth in terms of inferior adrenal artery (IAA) Doppler indices and adrenal gland volumes. Results: The IAA peak systolic velocity (PSV) in the FGR group was 14.9 ± 2.9 cm/s compared to 13.5 ± 2.0 cm/s in the normal group, with a mean difference of 1.4 cm/s (95% confidence interval [CI]: 0.27-2.65; p value = 0.017). There were no significant differences between groups in terms of IAA pulsatility index (PI), resistance index (RI), or systolic/diastolic (S/D), with p values of 0.438, 0.441, and 0.658, respectively. The volumes of the corrected whole adrenal gland and the corrected neocortex were significantly larger in the FGR group, with p values of 0.031 and 0.020, respectively. Conclusion: Both increased IAA PSV and enlarged volumes of the corrected whole adrenal gland and neocortex were found in fetuses with FGR, suggesting significant adrenal gland adaptation in response to chronic intrauterine stress.


Subject(s)
Adrenal Glands , Fetal Growth Retardation , Ultrasonography, Doppler , Ultrasonography, Prenatal , Humans , Fetal Growth Retardation/diagnostic imaging , Fetal Growth Retardation/physiopathology , Female , Pregnancy , Cross-Sectional Studies , Adrenal Glands/diagnostic imaging , Adrenal Glands/blood supply , Adrenal Glands/embryology , Ultrasonography, Prenatal/methods , Adult
2.
J Steroid Biochem Mol Biol ; 244: 106593, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39094666

ABSTRACT

The distribution of CYP11B2-positive or aldosterone producing adrenocortical cells in human fetuses and children and their age-dependent changes has not been studied. We aimed to explore the changes of aldosterone biosynthesis and age-related histological alterations of the zona glomerulosa in human adrenal gland during fetal and pediatric periods. We first reviewed 125 fetal and pediatric autopsy cases and retrieved 78 adrenals from 70 cases. CYP11B2 immunohistochemistry and quantitative image analysis of its results were performed in all adrenal glands. The ratio of the definitive zone (DZ) or zona glomerulosa (ZG) / the whole adrenocortical areas started to increase in the 2nd trimester, subsequently decreased in the 3rd, increased after birth, peaked in infancy, and then gradually decreased. The ratio of CYP11B2-positive / whole adrenocortical areas remained low during the fetal period but increased after birth, peaked at infancy, and then decreased. The ratio of CYP11B2-positive / DZ or ZG areas and CYP11B2-positive areas / depth of DZ or ZG demonstrated a distinctive bimodal pattern, with one peak in the fetal period and another in the neonatal period to infancy. This is the first study to perform quantitative analysis of the distribution of CYP11B2-positive cells, the histological DZ or ZG, and the development of aldosterone biosynthesis in human adrenal glands during fetal and pediatric periods.


Subject(s)
Aldosterone , Cytochrome P-450 CYP11B2 , Fetus , Zona Glomerulosa , Humans , Cytochrome P-450 CYP11B2/metabolism , Cytochrome P-450 CYP11B2/genetics , Aldosterone/biosynthesis , Aldosterone/metabolism , Infant , Zona Glomerulosa/metabolism , Female , Child, Preschool , Fetus/metabolism , Male , Infant, Newborn , Child , Adrenal Glands/metabolism , Adrenal Glands/embryology , Adolescent , Pregnancy
3.
Sci Rep ; 14(1): 12297, 2024 05 29.
Article in English | MEDLINE | ID: mdl-38811798

ABSTRACT

The current study aimed to investigate the effect of Sox9-Cre-directed Nr5a1-conditional knockout (Sox9-Cre;Nr5a1flox/flox) on adrenal development. We showed that SOX9 is expressed by adrenocortical cells at E10.5-E11.5 but is extinguished no later than E12.5. The number of adrenocortical cells significantly reduced in Sox9-Cre;Nr5a1flox/flox mice while the number of cleaved caspase 3-positive cells increased compared to that in the controls at E11.5-E12.5, when the adrenal primordium (AP) is about to expand. This indicated that fetal adrenocortical cells are lost via apoptosis due to Nr5a1 ablation by E12.5. Both medulla formation and encapsulation were perturbed, accompanied by a smaller AP size, in Sox9-Cre;Nr5a1flox/flox mice during embryonic development. Adult Sox9-Cre;Nr5a1flox/flox adrenals were hypoplastic and exhibited irregular organization of the medulla with aberrant sex differentiation in the X zone. Additionally, there were histologically eosin-negative vacuolated cells, which were negative for both the X-zone marker 20αHSD and the steroidogenesis marker 3ßHSD at the innermost cortex of Sox9-Cre;Nr5a1flox/flox adrenals. Although Nr5a1+/- adrenals were hypoplastic, a small number of chromaffin cells were properly located in the center, having normal sex differences in the X-zone. The results collectively provided in-vivo evidence that Nr5a1 plays a critical role in AP expansion and subsequent adrenal development.


Subject(s)
Adrenal Glands , SOX9 Transcription Factor , Steroidogenic Factor 1 , Animals , SOX9 Transcription Factor/metabolism , SOX9 Transcription Factor/genetics , Mice , Steroidogenic Factor 1/metabolism , Steroidogenic Factor 1/genetics , Adrenal Glands/metabolism , Adrenal Glands/embryology , Integrases/metabolism , Integrases/genetics , Mice, Knockout , Female , Male
4.
J Clin Invest ; 132(4)2022 02 15.
Article in English | MEDLINE | ID: mdl-34905510

ABSTRACT

Through their ability to regulate gene expression in most organs, glucocorticoid (GC) hormones influence numerous physiological processes and are therefore key regulators of organismal homeostasis. In bone, GC hormones inhibit expression of the hormone Osteocalcin for poorly understood reasons. Here, we show that in a classical endocrine feedback loop, osteocalcin in return enhanced the biosynthesis of GC as well as mineralocorticoid hormones (adrenal steroidogenesis) in rodents and primates. Conversely, inactivation of osteocalcin signaling in adrenal glands significantly impaired adrenal growth and steroidogenesis in mice. Embryo-made osteocalcin was necessary for normal Sf1 expression in fetal adrenal cells and adrenal cell steroidogenic differentiation and therefore determined the number of steroidogenic cells present in the adrenal glands of adult animals. Embryonic, not postnatal, osteocalcin also governed adrenal growth, adrenal steroidogenesis, blood pressure, electrolyte equilibrium, and the rise in circulating corticosterone levels during the acute stress response in adult offspring. This osteocalcin-dependent regulation of adrenal development and steroidogenesis occurred even in the absence of a functional hypothalamus/pituitary/adrenal axis and explains why osteocalcin administration during pregnancy promoted adrenal growth and steroidogenesis and improved the survival of adrenocorticotropic hormone signaling-deficient animals. This study reveals that a bone-derived embryonic hormone influences lifelong adrenal functions and organismal homeostasis in the mouse.


Subject(s)
Adrenal Glands/embryology , Homeostasis , Hypothalamo-Hypophyseal System/embryology , Osteocalcin/metabolism , Pituitary-Adrenal System/embryology , Signal Transduction , Animals , Female , Glucocorticoids/genetics , Glucocorticoids/metabolism , Macaca mulatta , Mice , Mice, Knockout , Osteocalcin/genetics
5.
Toxicology ; 462: 152932, 2021 10.
Article in English | MEDLINE | ID: mdl-34508824

ABSTRACT

Triadimefon is a broad-spectrum antifungal agent, which is widely used in agriculture to control mold and fungal infections. It is considered an endocrine disruptor. Whether triadimefon exposure can inhibit the development of fetal adrenal glands and the underlying mechanism remain unclear. Thirty-two pregnant female Sprague-Dawley rats were randomly divided into four groups. Dams were gavaged triadimefon (0, 25, 50, and 100 mg/kg/day) daily for 10 days from gestational day (GD) 12 to GD 21. Triadimefon significantly reduced the thickness of the zona fasciculata of male fetuses at 100 mg/kg, although it did not change the thickness of the zona glomerulosa. It significantly reduced the serum aldosterone levels of male fetuses at a dose of 100 mg/kg, and significantly reduced serum corticosterone and adrenocorticotropic hormone levels at doses of 50 and 100 mg/kg. Triadimefon significantly down-regulated the expression of Agtr1, Mc2r, Star, Cyp11b1, Cyp11b2, Igf1, Nr5a1, Sod2, Gpx1, and Cat, but did not affect the mRNA levels of Scarb1, Cyp11a1, Cyp21, Hsd3b1, and Hsd11b2. Triadimefon markedly reduced AT1R, CYP11B2, IGF1, NR5A1, and MC2R protein levels. Triadimefon significantly reduced the phosphorylation of AKT1 and ERK1/2 at 100 mg/kg without affecting the phosphorylation of AKT2. In contrast, it significantly increased AMPK phosphorylation at 100 mg/kg. In conclusion, exposure to triadimefon during gestation inhibits the development of fetal adrenal cortex in male fetuses. This inhibition is possibly due to the reduction of several proteins required for the synthesis of steroid hormones, and may be involved in changes in antioxidant contents and the phosphorylation of AKT1, ERK1/2, and AMPK.


Subject(s)
Adrenal Glands/drug effects , Fungicides, Industrial/toxicity , Maternal Exposure/adverse effects , Triazoles/toxicity , Adrenal Cortex/drug effects , Adrenal Cortex/embryology , Adrenal Glands/embryology , Animals , Antioxidants/metabolism , Dose-Response Relationship, Drug , Endocrine Disruptors/administration & dosage , Endocrine Disruptors/toxicity , Female , Fungicides, Industrial/administration & dosage , Male , Phosphorylation/drug effects , Pregnancy , Rats , Rats, Sprague-Dawley , Triazoles/administration & dosage
6.
Genetics ; 217(2)2021 02 09.
Article in English | MEDLINE | ID: mdl-33724412

ABSTRACT

People with NR5A1 mutations experience testicular dysgenesis, ovotestes, or adrenal insufficiency, but we do not completely understand the origin of this phenotypic diversity. NR5A1 is expressed in gonadal soma precursor cells before expression of the sex-determining gene SRY. Many fish have two co-orthologs of NR5A1 that likely partitioned ancestral gene subfunctions between them. To explore ancestral roles of NR5A1, we knocked out nr5a1a and nr5a1b in zebrafish. Single-cell RNA-seq identified nr5a1a-expressing cells that co-expressed genes for steroid biosynthesis and the chemokine receptor Cxcl12a in 1-day postfertilization (dpf) embryos, as does the mammalian adrenal-gonadal (interrenal-gonadal) primordium. In 2dpf embryos, nr5a1a was expressed stronger in the interrenal-gonadal primordium than in the early hypothalamus but nr5a1b showed the reverse. Adult Leydig cells expressed both ohnologs and granulosa cells expressed nr5a1a stronger than nr5a1b. Mutants for nr5a1a lacked the interrenal, formed incompletely differentiated testes, had no Leydig cells, and grew far larger than normal fish. Mutants for nr5a1b formed a disorganized interrenal and their gonads completely disappeared. All homozygous mutant genotypes lacked secondary sex characteristics, including male breeding tubercles and female sex papillae, and had exceedingly low levels of estradiol, 11-ketotestosterone, and cortisol. RNA-seq showed that at 21dpf, some animals were developing as females and others were not, independent of nr5a1 genotype. By 35dpf, all mutant genotypes greatly under-expressed ovary-biased genes. Because adult nr5a1a mutants form gonads but lack an interrenal and conversely, adult nr5a1b mutants lack a gonad but have an interrenal, the adrenal, and gonadal functions of the ancestral nr5a1 gene partitioned between ohnologs after the teleost genome duplication, likely owing to reciprocal loss of ancestral tissue-specific regulatory elements. Identifying such elements could provide hints to otherwise unexplained cases of Differences in Sex Development.


Subject(s)
Adrenal Glands/metabolism , DNA-Binding Proteins/genetics , Gonadal Dysgenesis/genetics , Gonads/metabolism , Transcription Factors/genetics , Zebrafish Proteins/genetics , Adrenal Glands/embryology , Animals , DNA-Binding Proteins/metabolism , Female , Gonads/embryology , Male , Phenotype , Sex Determination Processes , Transcription Factors/metabolism , Zebrafish , Zebrafish Proteins/metabolism
7.
Nat Genet ; 53(5): 683-693, 2021 05.
Article in English | MEDLINE | ID: mdl-33767450

ABSTRACT

Neuroblastoma is a pediatric tumor of the developing sympathetic nervous system. However, the cellular origin of neuroblastoma has yet to be defined. Here we studied the single-cell transcriptomes of neuroblastomas and normal human developing adrenal glands at various stages of embryonic and fetal development. We defined normal differentiation trajectories from Schwann cell precursors over intermediate states to neuroblasts or chromaffin cells and showed that neuroblastomas transcriptionally resemble normal fetal adrenal neuroblasts. Importantly, neuroblastomas with varying clinical phenotypes matched different temporal states along normal neuroblast differentiation trajectories, with the degree of differentiation corresponding to clinical prognosis. Our work highlights the roles of oncogenic MYCN and loss of TFAP2B in blocking differentiation and may provide the basis for designing therapeutic interventions to overcome differentiation blocks.


Subject(s)
Gene Expression Profiling , Neuroblastoma/genetics , Neuroblastoma/pathology , Single-Cell Analysis , Adrenal Glands/embryology , Adrenal Glands/pathology , Cell Differentiation , Cell Line, Tumor , Cohort Studies , Gene Expression Regulation, Neoplastic , Humans , Transcriptome/genetics , Treatment Outcome
8.
J Clin Endocrinol Metab ; 106(3): 843-857, 2021 03 08.
Article in English | MEDLINE | ID: mdl-33212489

ABSTRACT

CONTEXT: Disorders affecting adrenal steroidogenesis promote an imbalance in the normally tightly controlled secretion of mineralocorticoids, glucocorticoids, and androgens. This may lead to differences/disorders of sex development in the fetus, as seen in virilized girls with congenital adrenal hyperplasia (CAH). Despite the important endocrine function of human fetal adrenals, neither normal nor dysregulated adrenal steroidogenesis is understood in detail. OBJECTIVE: Due to significant differences in adrenal steroidogenesis between human and model species (except higher primates), we aimed to establish a human fetal adrenal model that enables examination of both de novo and manipulated adrenal steroidogenesis. DESIGN AND SETTING: Human adrenal tissue from 54 1st trimester fetuses were cultured ex vivo as intact tissue fragments for 7 or 14 days. MAIN OUTCOME MEASURES: Model validation included examination of postculture tissue morphology, viability, apoptosis, and quantification of steroid hormones secreted to the culture media measured by liquid chromatography-tandem mass spectrometry. RESULTS: The culture approach maintained cell viability, preserved cell populations of all fetal adrenal zones, and recapitulated de novo adrenal steroidogenesis based on continued secretion of steroidogenic intermediates, glucocorticoids, and androgens. Adrenocorticotropic hormone and ketoconazole treatment of ex vivo cultured human fetal adrenal tissue resulted in the stimulation of steroidogenesis and inhibition of androgen secretion, respectively, demonstrating a treatment-specific response. CONCLUSIONS: Together, these data indicate that ex vivo culture of human fetal adrenal tissue constitutes a novel approach to investigate local effects of pharmaceutical exposures or emerging therapeutic options targeting imbalanced steroidogenesis in adrenal disorders, including CAH.


Subject(s)
Adrenal Glands/cytology , Drug Evaluation, Preclinical/methods , Fetus/cytology , Primary Cell Culture/methods , Steroids/biosynthesis , Adrenal Glands/drug effects , Adrenal Glands/embryology , Adrenal Glands/metabolism , Adrenal Hyperplasia, Congenital/drug therapy , Adrenal Hyperplasia, Congenital/metabolism , Adrenal Hyperplasia, Congenital/pathology , Adrenocorticotropic Hormone/pharmacology , Androgens/metabolism , Cell Survival , Culture Media/chemistry , Female , Glucocorticoids/pharmacology , Humans , Ketoconazole/pharmacology , Metabolic Networks and Pathways/drug effects , Models, Biological , Pregnancy , Steroids/analysis , Steroids/metabolism
9.
BMC Pregnancy Childbirth ; 20(1): 774, 2020 Dec 11.
Article in English | MEDLINE | ID: mdl-33308174

ABSTRACT

BACKGROUND: The fetal adrenal gland is a highly vascularized organs and develops two recognizable distinct zones in uetro, inner fetal zone (FZ) and outer definitive zone (DZ). Based on the region supplied, middle adrenal artery (MAA) mainly contribute to FZ while inferior adrenal artery (IAA) mainly to the inferior part of DZ. The purpose of this study was to establish reference ranges of adrenal artery Doppler indices of IAA and MAA, and assess zonal difference of blood supply to fetal adrenal gland. METHODS: The pulsatility index (PI), resistance index (RI), and systolic:diastolic ratio (S/D) of the IAA and MAA were obtained serially at 4-week intervals in normal fetuses. The MAA and IAA were referred based on the course and location in the gland: IAA referring the artery that mainly branches from the renal artery and walks along the renal upper pole, distributing the inferoposterior part of DZ in the adrenal gland while MAA as arterial blood flowing along the single central adrenal vein in the medial part of the gland. Multilevel modeling was performed to establish the gestational age-associated reference ranges for IAA and MAA. Differences in Doppler indices between the IAA and MAA were assessed. RESULTS: One hundred sixty-eight fetuses with 843 observations were included. The IAA had a higher detection rate than the MAA (100% vs 89.2%, p < 0.05). The resistance of IAA had a reduction around 35 weeks of gestation and that of MAA remained unchanged throughout the second half of pregnancy. Lower PI, RI and S/D were observed in the MAA than in the IAA (p < 0.05) from 752 paired measurements. CONCLUSION: There is a zonal difference in blood supply in favor of the fetal zone, which may correspond to its unique function. Reference ranges of Doppler parameters in adrenal artery maybe beneficial for further evaluation of fetal hemodynamics.


Subject(s)
Adrenal Glands/blood supply , Pulsatile Flow/physiology , Umbilical Arteries/diagnostic imaging , Adrenal Glands/diagnostic imaging , Adrenal Glands/embryology , Adult , Female , Humans , Longitudinal Studies , Middle Cerebral Artery/diagnostic imaging , Middle Cerebral Artery/embryology , Pregnancy , Reference Values , Ultrasonography, Doppler, Color , Ultrasonography, Prenatal , Umbilical Arteries/embryology
10.
BMC Pregnancy Childbirth ; 20(1): 570, 2020 Sep 29.
Article in English | MEDLINE | ID: mdl-32993527

ABSTRACT

BACKGROUND: The fetal adrenal gland receives rising awareness as a predictor of spontaneous preterm birth. We hereby provide longitudinal growth assessments of the fetal adrenal gland in a low risk population with an additional focus on trajectories in fetuses born preterm. METHODS: Fetal adrenal gland was assessed via transabdominal ultrasound at gestational weeks (gw) 24-26, 28-30, and 34-36 in a low-risk pregnancy cohort. Longitudinal trajectories of the total gland and the mark (so called fetal zone) as well as ratio of fetal zone width/ total widths (w/W) were analyzed using repeated ANOVA analyses. To compare trajectories of the ratio w/W for preterm and term fetuses respectively, as well as women with and without clinical signs of preterm labor, the propensity score method was applied. RESULTS: Fetal zone width increased over the course of pregnancy (p < 0.0001), while the ratio w/W decreased (p < 0.0001) (n = 327). Comparing the trajectories of the ratio w/W in fetuses born preterm (n = 11) with propensity-score matched term born fetuses (n = 22), a decrease between gw 24-26 and 28-30 was observed in both groups, which continued to decrease for the term born fetuses. However, in preterm born fetuses, the ratio increased above the term born values at gw 34-36. CONCLUSION: Our study provides for the first time longitudinal growth data on the fetal adrenal gland and supports the hypothesis that fetal zone enlargement is associated with preterm birth which could play an important role in risk-prediction.


Subject(s)
Adrenal Glands/anatomy & histology , Adrenal Glands/diagnostic imaging , Fetal Development , Fetus/anatomy & histology , Fetus/diagnostic imaging , Premature Birth/epidemiology , Ultrasonography, Prenatal , Adrenal Glands/embryology , Adult , Female , Gestational Age , Humans , Pregnancy , Risk Assessment
11.
Cancer Cell ; 38(5): 716-733.e6, 2020 11 09.
Article in English | MEDLINE | ID: mdl-32946775

ABSTRACT

Neuroblastoma (NB), which is a subtype of neural-crest-derived malignancy, is the most common extracranial solid tumor occurring in childhood. Despite extensive research, the underlying developmental origin of NB remains unclear. Using single-cell RNA sequencing, we generate transcriptomes of adrenal NB from 160,910 cells of 16 patients and transcriptomes of putative developmental cells of origin of NB from 12,103 cells of early human embryos and fetal adrenal glands at relatively late development stages. We find that most adrenal NB tumor cells transcriptionally mirror noradrenergic chromaffin cells. Malignant states also recapitulate the proliferation/differentiation status of chromaffin cells in the process of normal development. Our findings provide insight into developmental trajectories and cellular states underlying human initiation and progression of NB.


Subject(s)
Adrenal Gland Neoplasms/genetics , Adrenal Glands/embryology , Gene Expression Profiling/methods , Neuroblastoma/genetics , Single-Cell Analysis/methods , Adrenal Glands/chemistry , Cell Differentiation , Cell Proliferation , Chromaffin Cells/chemistry , Chromaffin Cells/cytology , Gene Expression Regulation, Neoplastic , Humans , Phenotype , Sequence Analysis, RNA
12.
Ann Anat ; 231: 151526, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32380196

ABSTRACT

BACKGROUND: Toll-Like Receptors (TLRs) play a critical role in the innate and adaptive immune system. They are the mammalian orthologs of Drosophila melanogaster protein Toll, which has been proved to have an early morphogenetic role in invertebrate embryogenesis that in the adult switches to an immune function. AIM: The aim of this study was to evaluate the expression of TLR4 and TLR7 during dorsal root ganglia (DRG), paravertebral ganglia (PVG), and enteric nervous system (ENS) murine development. METHODS: Mouse embryos from different stages (i.e. E12 to E18) were processed for immunolocalization analysis on formalin-fixed paraffin-embedded sections, and isolated intestine were processed for whole-mount preparations. RESULTS: We observed a differentially regulated expression of TLR4 and TLR7 during embryogenesis and an overall increased expression of both receptors during development. While TLR4 was detectable in neurons of DRG and PVG starting from E14 and only from E18 in the ENS, TLR7 was already expressed in scattered neurons of all the investigated regions at E12. CONCLUSIONS: TLR4 and TRL7 expression temporal patterns suggest a morphogenetic role for these receptors in the development of neural crest derivatives in mammals.


Subject(s)
Peripheral Nervous System/embryology , Peripheral Nervous System/metabolism , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 7/metabolism , Adrenal Glands/embryology , Adrenal Glands/growth & development , Adrenal Glands/metabolism , Animals , Female , Fluorescent Antibody Technique , Immunohistochemistry , Male , Mice , Peripheral Nervous System/growth & development
13.
J Clin Ultrasound ; 48(7): 377-387, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32333815

ABSTRACT

PURPOSE: Fetal adrenal gland changes have previously been investigated as novel markers of preterm labor and small for gestational age (SGA) fetuses. We aimed to compare the fetal adrenal gland parameters in SGA and appropriate for gestational age (AGA) fetuses. METHODS: A prospective cohort study was conducted on SGA fetuses with estimated fetal weight (EFW) ≤10th centile and AGA (EFW >10th centile) at 17 to 34 weeks gestation. Fetal adrenal total gland volume (TGV), TGV corrected for EFW (cTGV), fetal zone volume (FZV), FZV corrected for EFW (cFZV), and FZV:TGV ratio were compared and correlated with gestational age and EFW. Receiver operator curves assessed FZV:TGV ratio, cTGV, and cFZV in detecting SGA. RESULTS: Ultrasound examinations from 103 AGA and 50 SGA fetuses showed that (a) SGA fetuses had higher TGV (P = .002), FZV (P = .001), and FZV:TGV (P = .036) compared to AGA fetuses; (b) fetal adrenal TGV, FZV, cFZV, and FZV:TGV increase with advancing gestational age and EFW while cTGV does not; (c) Fetal adrenal changes in cTGV, cFZV, and FZV:TGV have ability to differentiate SGA; (d) FZV:TGV ratio 10 and 25 may be used to identify or exclude SGA in antenatally suspected SGA. CONCLUSIONS: We investigated the concept that SGA fetuses have measurable changes to the adrenal gland. We have shown that fetal TGV, TGV, and FZV:TGV ratio show differences between AGA and SGA with TGV remaining significant after accounting for GA at scan. These findings may be useful as potential biomarkers for diagnosing or excluding SGA.


Subject(s)
Adrenal Glands/diagnostic imaging , Fetal Growth Retardation/diagnosis , Fetus/diagnostic imaging , Infant, Small for Gestational Age , Ultrasonography, Prenatal/methods , Adolescent , Adrenal Glands/embryology , Adult , Female , Fetal Weight , Gestational Age , Humans , Infant, Newborn , Middle Aged , Pregnancy , Pregnancy Trimester, Third , Prospective Studies , Young Adult
14.
Pediatr Radiol ; 50(6): 840-847, 2020 05.
Article in English | MEDLINE | ID: mdl-32060593

ABSTRACT

BACKGROUND: The adrenal gland plays a vital role in fetal growth. Many disease states such as congenital adrenal hyperplasia, hemorrhage and tumors can lead to morphological changes in the gland. Ultrasound measurements of normal adrenal sizes in the fetus reported in the literature have shown a trend of increasing size with gestational age. There is no literature available on standard fetal adrenal sizes or detailed appearance by fetal MRI. OBJECTIVE: The purpose of this study was to provide MR data on the size and signal characteristics of the fetal adrenal gland throughout the second and third trimesters. MATERIALS AND METHODS: In this retrospective review, we selected 185 prenatal MRIs obtained from Jan. 1, 2014, to May 31, 2017, with normal abdominal findings for inclusion. The adrenal glands were identified in coronal, sagittal or axial T2-W planes and coronal T1-W plane when available. We measured the length and thickness of the medial and lateral limbs of the right and left adrenal glands and recorded signal intensity on T1-W and T2-W sequences, gender and gestational age in each case. RESULTS: The gestational age (GA) ranged 18-37 weeks. Visibility of the adrenal glands on T2-W images was high (90.3-97.2%) up to 30 weeks of GA but declined afterward (47.5-62.2% at 31-37 weeks). Visibility on T1-W images increased with GA, ranging from 21.4% visibility at 18-22 weeks and increasing to 40% at 35-37 weeks. Mean lengths of the adrenal gland limbs steadily increased from 8.2 mm at 18-22 weeks to 11.0 mm at 35-37 weeks. In the second trimester, adrenal glands were low in signal intensity on T2-W images and were surrounded by hyperintense perirenal fatty tissue. In the third trimester, the glands became less distinct, with increasing signal and obliteration of perirenal tissue. The glands were moderately hyperintense on T1-W images throughout pregnancy, with increasing visibility as pregnancy progressed. CONCLUSION: Normal sizes and signal intensities for adrenal glands are reported. Visibility of adrenal glands on T2-W images was 90.3-97.2% up to 30 weeks but declined thereafter. Visibility on T1-W images increased in the third trimester. Adrenal gland sizes increased with gestational age.


Subject(s)
Adrenal Glands/diagnostic imaging , Adrenal Glands/embryology , Magnetic Resonance Imaging/methods , Female , Gestational Age , Humans , Pregnancy , Reference Values , Retrospective Studies
15.
Endocrinol Metab (Seoul) ; 35(4): 765-773, 2020 12.
Article in English | MEDLINE | ID: mdl-33397037

ABSTRACT

The adrenal gland plays a pivotal role in an organism's health span by controlling the endocrine system. Decades of research on the adrenal gland have provided multiscale insights into the development and maintenance of this essential organ. A particularly interesting finding is that founder stem/progenitor cells participate in adrenocortical development and enable the adult adrenal cortex to regenerate itself in response to hormonal stress and injury. Since major advances have been made in understanding the dynamics of the developmental process and the remarkable regenerative capacity of the adrenal gland, understanding the mechanisms underlying adrenal development, maintenance, and regeneration will be of interest to basic and clinical researchers. Here, we introduce the developmental processes of the adrenal gland and discuss current knowledge regarding stem/progenitor cells that regulate adrenal cortex remodeling and regeneration. This review will provide insights into the fascinating ongoing research on the development and regeneration of the adrenal cortex.


Subject(s)
Adrenal Cortex/embryology , Adrenal Glands/embryology , Embryonic Development/physiology , Stem Cells/metabolism , Adrenal Cortex/growth & development , Adrenal Cortex/metabolism , Adrenal Glands/growth & development , Adrenal Glands/metabolism , Adult , Animals , Gene Expression Regulation, Developmental , Humans , Regeneration , Signal Transduction
16.
Proc Natl Acad Sci U S A ; 116(44): 22294-22299, 2019 10 29.
Article in English | MEDLINE | ID: mdl-31611378

ABSTRACT

Androgen biosynthesis in the human fetus proceeds through the adrenal sex steroid precursor dehydroepiandrosterone, which is converted to testosterone in the gonads, followed by further activation to 5α-dihydrotestosterone in genital skin, thereby facilitating male external genital differentiation. Congenital adrenal hyperplasia due to P450 oxidoreductase deficiency results in disrupted dehydroepiandrosterone biosynthesis, explaining undervirilization in affected boys. However, many affected girls are born virilized, despite low circulating androgens. We hypothesized that this is due to a prenatally active, alternative androgen biosynthesis pathway from 17α-hydroxyprogesterone to 5α-dihydrotestosterone, which bypasses dehydroepiandrosterone and testosterone, with increased activity in congenital adrenal hyperplasia variants associated with 17α-hydroxyprogesterone accumulation. Here we employ explant cultures of human fetal organs (adrenals, gonads, genital skin) from the major period of sexual differentiation and show that alternative pathway androgen biosynthesis is active in the fetus, as assessed by liquid chromatography-tandem mass spectrometry. We found androgen receptor expression in male and female genital skin using immunohistochemistry and demonstrated that both 5α-dihydrotestosterone and adrenal explant culture supernatant induce nuclear translocation of the androgen receptor in female genital skin primary cultures. Analyzing urinary steroid excretion by gas chromatography-mass spectrometry, we show that neonates with P450 oxidoreductase deficiency produce androgens through the alternative androgen pathway during the first weeks of life. We provide quantitative in vitro evidence that the corresponding P450 oxidoreductase mutations predominantly support alternative pathway androgen biosynthesis. These results indicate a key role of alternative pathway androgen biosynthesis in the prenatal virilization of girls affected by congenital adrenal hyperplasia due to P450 oxidoreductase deficiency.


Subject(s)
17-alpha-Hydroxyprogesterone/metabolism , Androgens/biosynthesis , Antley-Bixler Syndrome Phenotype/genetics , Fetus/metabolism , Receptors, Androgen/genetics , Virilism/metabolism , Adrenal Glands/embryology , Adrenal Glands/metabolism , Androgens/genetics , Cells, Cultured , Female , Fetus/embryology , Genitalia/embryology , Genitalia/metabolism , Gonads/embryology , Gonads/metabolism , Humans , Male , Receptors, Androgen/metabolism , Sex Differentiation , Virilism/genetics
17.
PLoS One ; 14(9): e0221719, 2019.
Article in English | MEDLINE | ID: mdl-31483805

ABSTRACT

Biochemical changes in utero may alter normal fetal development, resulting in disease later in life, a phenomenon known as fetal programming. Recent epidemiological studies link fetal programming to negative health outcomes, such as low birth weight and hypertension in adulthood. Here, we used a WKY rat model and studied the molecular changes triggered by prenatal glucocorticoid (GC) exposure on the development of hypertension, and on the regulation of phenylethanolamine N-methyl transferase (PNMT), the enzyme responsible for biosynthesis of epinephrine, and a candidate gene linked to hypertension. Clinically, high doses of the synthetic GC dexamethasone (DEX) are used to treat infant respiratory distress syndrome. Elevated maternal GCs have been correlated with fetal programming of hypertension. The aim of this study was to determine if lower doses of DEX would not lead to detrimental fetal programming effects such as hypertension. Our data suggests that prenatal stress programs for increased expression of PNMT and altered regulation of PNMT in males and females. Importantly, we identified that DEX mediated programming was more apparent in the male rats, and the lower dose 10µg/kg/day of DEX did not lead to changes in blood pressure (BP) in female rats suggesting that this dose is below the threshold for programming of hypertension. Furthermore, sex-specific differences were observed in regards to programming mechanisms that may account for hypertension in males.


Subject(s)
Adrenal Glands/enzymology , Dexamethasone/adverse effects , Fetal Development/drug effects , Glucocorticoids/adverse effects , Hypertension/chemically induced , Phenylethanolamine N-Methyltransferase/metabolism , Sex Characteristics , Adrenal Glands/embryology , Animals , Corticosterone/blood , Dose-Response Relationship, Drug , Epinephrine/blood , Female , Hypertension/metabolism , Male , Pregnancy , Rats , Rats, Inbred WKY , Stress, Physiological/drug effects , Stress, Physiological/genetics , Transcription, Genetic/drug effects
18.
J Perinat Med ; 47(9): 941-946, 2019 Nov 26.
Article in English | MEDLINE | ID: mdl-31562804

ABSTRACT

Background The aim of this study was to compare the adrenal gland size of fetuses of women with gestational diabetes mellitus (GDM) with that of healthy control fetuses. Methods This prospective cross-sectional study included measurements of the adrenal gland size of 62 GDM fetuses (GDM group) and 370 normal controls (control group) between the 19th and 41st week of gestation. A standardized transversal plane was used to measure the total width and the medulla width. The cortex width and an adrenal gland ratio (total width/medulla width) were calculated from these data. Adrenal gland size measurements were adjusted to the week of gestation and compared between the two groups in a multivariable linear regression analysis. A variance decomposition metric was used to compare the relative importance of predictors of the different adrenal gland size measurements. Results For all the investigated parameters of the adrenal gland size, increased values were found in the case of GDM (P < 0.05), while adjusting for the week of gestation. GDM seems to have a greater impact on the size of the cortex than on the size of the medulla. Conclusion The fetal adrenal gland is enlarged in pregnancy complicated by GDM. The width of the cortex seems to be particularly affected.


Subject(s)
Adrenal Glands/embryology , Diabetes, Gestational/physiopathology , Fetal Development , Adrenal Glands/diagnostic imaging , Adrenal Glands/pathology , Case-Control Studies , Cross-Sectional Studies , Diabetes, Gestational/diagnostic imaging , Female , Gestational Age , Humans , Linear Models , Multivariate Analysis , Pregnancy , Prospective Studies , Ultrasonography, Prenatal
19.
J Endocrinol ; 241(1): R51-R63, 2019 04.
Article in English | MEDLINE | ID: mdl-30817316

ABSTRACT

The X-zone is a transient cortical region enriched in eosinophilic cells located in the cortical-medullary boundary of the mouse adrenal gland. Similar to the X-zone, the fetal zone in human adrenals is also a transient cortical compartment, comprising the majority of the human fetal adrenal gland. During adrenal development, fetal cortical cells are gradually replaced by newly formed adult cortical cells that develop into outer definitive zones. In mice, the regression of this fetal cell population is sexually dimorphic. Many mouse models with mutations associated with endocrine factors have been reported with X-zone phenotypes. Increasing findings indicate that the cell fate of this aged cell population of the adrenal cortex can be manipulated by many hormonal and nonhormonal factors. This review summarizes the current knowledge of this transient adrenocortical zone with an emphasis on genes and signaling pathways that affect X-zone cells.


Subject(s)
Adrenal Cortex/metabolism , Adrenal Glands/metabolism , Adrenal Medulla/metabolism , Adrenocorticotropic Hormone/metabolism , Adrenal Cortex/cytology , Adrenal Cortex/embryology , Adrenal Glands/cytology , Adrenal Glands/embryology , Adrenal Medulla/cytology , Adrenal Medulla/embryology , Animals , Apoptosis/genetics , Fetal Development/genetics , Gene Expression Regulation, Developmental , Humans , Mice
SELECTION OF CITATIONS
SEARCH DETAIL