1
|
Lacroix A, Bourdeau I, Chasseloup F, Kamenický P, Lopez AG, Louiset E, Lefebvre H. Aberrant hormone receptors regulate a wide spectrum of endocrine tumors. Lancet Diabetes Endocrinol 2024; 12:837-855. [PMID: 39326429 DOI: 10.1016/s2213-8587(24)00200-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 06/27/2024] [Accepted: 07/11/2024] [Indexed: 09/28/2024]
Abstract
Aberrant G-protein coupled receptor (GPCR) expression is highly prevalent in cortisol-secreting primary bilateral macronodular adrenal hyperplasia (PBMAH) and unilateral adenomas. The aberrant expression of diverse GPCRs and their ligands play an important role in the over-function of various endocrine tumours. Examples include aberrant expression of MC2R, 5-HT4R, AVPR1A, LHCGR, and GnRHR in primary aldosteronism; GCGR, LHCGR, and 5-HT4R in phaeochromocytomas and paragangliomas; TRHR, GnRHR, GIPR, and GRP101 in pituitary somatotroph tumours; AVPR2, D2DR, and SSTR5 in pituitary corticotroph tumours; GLP1R, GIPR, and somatostatin receptors in medullary thyroid carcinoma; and SSTRs, GLP1R, and GIPR in other neuroendocrine tumours. The genetic mechanisms causing the ectopic expression of GIPR in cortisol-secreting PBMAHs and unilateral adenomas have been identified, but distinct mechanisms are implicated in other endocrine tumours. Development of functional imaging targeting aberrant GPCRs should be useful for identification and for specific therapies of this wide spectrum of tumours. The aim of this review is to show that the regulation of endocrine tumours by aberrant GPCR is not restricted to cortisol-secreting adrenal lesions, but also occurs in tumours of several other organs.
Collapse
Affiliation(s)
- André Lacroix
- Division of Endocrinology, Department of Medicine and Research Center, Centre hospitalier de l' Université de Montréal (CHUM), Montréal, QC, Canada.
| | - Isabelle Bourdeau
- Division of Endocrinology, Department of Medicine and Research Center, Centre hospitalier de l' Université de Montréal (CHUM), Montréal, QC, Canada
| | - Fanny Chasseloup
- Université Paris-Saclay, Inserm, Physiologie et Physiopathologie Endocriniennes, Assistance Publique-Hôpitaux de Paris, Hôpital Bicêtre, Service d'Endocrinologie et des Maladies de la Reproduction, Le Kremlin-Bicêtre, France
| | - Peter Kamenický
- Université Paris-Saclay, Inserm, Physiologie et Physiopathologie Endocriniennes, Assistance Publique-Hôpitaux de Paris, Hôpital Bicêtre, Service d'Endocrinologie et des Maladies de la Reproduction, Le Kremlin-Bicêtre, France
| | - Antoine-Guy Lopez
- Univ Rouen Normandie, Inserm, NorDiC UMR 1239, Rouen, France; Department of Endocrinology, Diabetes and Metabolic Diseases, Rouen, France
| | - Estelle Louiset
- Univ Rouen Normandie, Inserm, NorDiC UMR 1239, Rouen, France; Department of Endocrinology, Diabetes and Metabolic Diseases, Rouen, France
| | - Hervé Lefebvre
- Univ Rouen Normandie, Inserm, NorDiC UMR 1239, Rouen, France; Department of Endocrinology, Diabetes and Metabolic Diseases, Rouen, France
| |
Collapse
|
2
|
Shironomae T, Yamazaki Y, Takeyama J, Sakai K, Sasano H, Suzuki T. Development of aldosterone biosynthesis during fetal and pediatric periods; Histological analysis of CYP11B2-positive cell distribution in the zona glomerulosa of human adrenal. J Steroid Biochem Mol Biol 2024; 244:106593. [PMID: 39094666 DOI: 10.1016/j.jsbmb.2024.106593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 07/23/2024] [Accepted: 07/25/2024] [Indexed: 08/04/2024]
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.
Collapse
Affiliation(s)
- Tsubasa Shironomae
- Department of Anatomic Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan; Department of Urology, Miyagi Children's Hospital, Sendai, Japan
| | - Yuto Yamazaki
- Department of Anatomic Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan.
| | - Junji Takeyama
- Department of Pathology, Miyagi Children's Hospital, Sendai, Japan
| | - Kiyohide Sakai
- Department of Urology, Miyagi Children's Hospital, Sendai, Japan
| | - Hironobu Sasano
- Department of Anatomic Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takashi Suzuki
- Department of Anatomic Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
3
|
Deer LK, Demers CH, Hankin BL, Doom JR, Shields GS, Hoffman MC, Davis EP. Neonatal Hair Cortisol and Birth Outcomes: An Empirical Study and Meta-Analysis. Psychosom Med 2024; 86:720-729. [PMID: 39132972 DOI: 10.1097/psy.0000000000001339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
OBJECTIVE Prenatal stress physiology is often posited as a predictor of birth outcomes, including gestational age at birth and birthweight. However, research has predominantly relied on indicators in the maternal system, with few studies examining hormones of the fetal system. The current study focuses on fetal cortisol in the third trimester, as measured in neonatal hair, as a biological factor that might be associated with birth outcomes (gestational age at birth and birthweight). We report findings from two studies: a longitudinal cohort (Study 1), and a meta-analysis of the existing literature (Study 2). METHODSSTUDY Hair was collected for cortisol analysis from 168 neonates (55.95% female) shortly after birth. Gestational age at birth and birthweight were abstracted from medical records. METHODSSTUDY An exhaustive search of four databases was conducted, yielding 155 total studies for screening. Papers reporting neonatal hair cortisol (collection <2 weeks postpartum) and birth outcomes among human neonates were retained for analysis, including Study 1 results ( k = 9). RESULTSSTUDY Higher neonatal hair cortisol was related to longer gestation ( r = 0.28, p < .001) and higher birthweight, r = 0.16, p = .040. Sex did not moderate either association. RESULTSSTUDY Across the nine studies, higher neonatal hair cortisol predicted both longer gestation ( r = 0.35, p < .001, 95% confidence interval = 0.24-0.45) and higher birthweight ( r = 0.18, p = .001, 95% confidence interval = 0.07-0.28). Neonatal sex did not moderate these associations. CONCLUSIONS Fetal cortisol exposure in the third trimester plays a role in normative maturation of the fetus, and findings reveal that higher cortisol is associated with positive birth outcomes.
Collapse
Affiliation(s)
- LillyBelle K Deer
- From the Department of Psychology (Deer, Doom, Davis), University of Denver, Denver; Department of Psychiatry (Demers, Hoffman), University of Colorado Anschutz Medical Campus, Aurora, Colorado; Department of Psychology (Hankin), University of Illinois Urbana-Champaign, Champaign, Illinois; Department of Psychological Science (Shields), University of Arkansas, Fayetteville, Arkansas; Division of Maternal and Fetal Medicine, Department of Obstetrics and Gynecology (Hoffman), University of Colorado Anschutz Medical Campus, Aurora, Colorado; and Department of Pediatrics (Davis), University of California, Irvine, Irvine, California
| | | | | | | | | | | | | |
Collapse
|
4
|
Pattamathamakul S, Duangkum C, Chaiyarach S, Kongwattanakul K, Saksiriwuttho P, Komwilaisak R, Chantanavilai S, Pongsamakthai M, Sirikarn P. The Impact of Fetal Growth Restriction on Prenatal 2D Ultrasound and Doppler Study of the Fetal Adrenal Gland. J Pregnancy 2024; 2024:9968509. [PMID: 39238897 PMCID: PMC11377111 DOI: 10.1155/2024/9968509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 05/31/2024] [Accepted: 08/12/2024] [Indexed: 09/07/2024] Open
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.
Collapse
Affiliation(s)
- Suphawan Pattamathamakul
- Department of Obstetrics and GynecologyFaculty of MedicineKhon Kaen University, Khon Kaen, Thailand
| | - Chatuporn Duangkum
- Department of Obstetrics and GynecologyFaculty of MedicineKhon Kaen University, Khon Kaen, Thailand
| | - Sukanya Chaiyarach
- Department of Obstetrics and GynecologyFaculty of MedicineKhon Kaen University, Khon Kaen, Thailand
| | - Kiattisak Kongwattanakul
- Department of Obstetrics and GynecologyFaculty of MedicineKhon Kaen University, Khon Kaen, Thailand
| | - Piyamas Saksiriwuttho
- Department of Obstetrics and GynecologyFaculty of MedicineKhon Kaen University, Khon Kaen, Thailand
| | - Ratana Komwilaisak
- Department of Obstetrics and GynecologyFaculty of MedicineKhon Kaen University, Khon Kaen, Thailand
| | | | | | - Prapassara Sirikarn
- Department of Epidemiology and BiostatisticsFaculty of Public HealthKhon Kaen University, Khon Kaen, Thailand
| |
Collapse
|
5
|
Niimi T, Tanaka T, Aoyagi C, Onda Y, Nagamitsu S, Kodama S. Co-culture of vascular endothelial cells enhances corticosterone production in steroid hormone-producing cells generated from adipose-derived mesenchymal stromal cells. Sci Rep 2024; 14:18804. [PMID: 39138321 PMCID: PMC11322653 DOI: 10.1038/s41598-024-69878-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 08/09/2024] [Indexed: 08/15/2024] Open
Abstract
Cell therapy for adrenocortical insufficiency can potentially provide steroid replacement in response to physiological stimuli. Previously, we reported that adipose tissue-derived stromal cells (ADSCs) are transformed into steroid-producing cells by overexpression of nuclear receptor subfamily 5 group A member 1 (NR5A1). The steroidogenic cells are characterized by the production of both adrenal and gonadal steroids. Cytotherapy for adrenocortical insufficiency requires cells with more adrenocortical characteristics. Considering the highly developed vascular network within the adrenal cortex, all adrenocortical cells are adjacent to and interact with vascular endothelial cells (VECs). In this study, NR5A1-induced steroidogenic cells derived from mouse ADSCs (NR5A1-ADSCs) were co-cultured with mouse VECs. Testosterone secretion in NR5A1-ADSCs was not altered; however, corticosterone secretion significantly increased while levels of steroidogenic enzymes significantly increased in the corticosterone synthesis pathway. Co-culture with lymphatic endothelial cells (LECs) or ADSCs, or transwell culture with NR5A1-ADSCs and VECs did not alter corticosterone production. VECs expressed higher levels of collagen and laminin than LECs. Culture in type-IV collagen and laminin-coated dishes increased corticosterone secretion in NR5A1-ADSCs. These results suggest that VECs may characterize ADSC-derived steroidogenic cells into a more corticosterone-producing phenotype, and VECs may be useful for generating adrenal steroidogenic cells from stem cells.
Collapse
Affiliation(s)
- Toshikazu Niimi
- Department of Regenerative Therapy and Transplantation, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-Ku, Fukuoka, 814-0180, Japan
- Department of Pediatrics, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-Ku, Fukuoka, 814-0180, Japan
| | - Tomoko Tanaka
- Department of Regenerative Therapy and Transplantation, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-Ku, Fukuoka, 814-0180, Japan.
| | - Chikao Aoyagi
- Department of Regenerative Therapy and Transplantation, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-Ku, Fukuoka, 814-0180, Japan
| | - Yasuhiro Onda
- Department of Regenerative Therapy and Transplantation, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-Ku, Fukuoka, 814-0180, Japan
- Department of Pediatrics, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-Ku, Fukuoka, 814-0180, Japan
| | - Shinichiro Nagamitsu
- Department of Pediatrics, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-Ku, Fukuoka, 814-0180, Japan
| | - Shohta Kodama
- Department of Regenerative Therapy and Transplantation, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-Ku, Fukuoka, 814-0180, Japan.
| |
Collapse
|
6
|
Aminuddin A, Brown MJ, Azizan EA. Evaluating the role of aldosterone synthesis on adrenal cell fate. Front Endocrinol (Lausanne) 2024; 15:1423027. [PMID: 39170743 PMCID: PMC11335638 DOI: 10.3389/fendo.2024.1423027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 07/23/2024] [Indexed: 08/23/2024] Open
Abstract
Hypertension affects one-third of the adult population worldwide, with primary aldosteronism (PA) accounting for at least 5-10% of these cases. The aldosterone synthase enzyme (CYP11B2) plays a pivotal role in PA manifestation, as increased expression of CYP11B2 leads to excess aldosterone synthesis. Physiological expression of CYP11B2 in humans is normally limited to cells of the adrenal zona glomerulosa under tight homeostatic regulation. In PA, however, there are CYP11B2-positive lesions in the adrenal cortex that autonomously secrete aldosterone, highlighting the dysregulation of adrenal cortex zonation and function as a key aspect of PA pathogenesis. Thus, this review aims to summarize the development of the adrenal glands, the key regulators of adrenal cortex homeostasis, and the dysregulation of this homeostasis. It also discusses the development of CYP11B2 inhibitors for therapeutic use in patients with hypertension, as well as the current knowledge of the effects of CYP11B2 inhibition on adrenal cortex homeostasis and cell fate. Understanding the control of adrenal cell fate may offer valuable insights into both the pathogenesis of PA and the development of alternative treatment approaches for PA.
Collapse
Affiliation(s)
- Amnani Aminuddin
- Department of Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Morris J. Brown
- Endocrine Hypertension, Department of Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
- National Institute for Health Research (NIHR) Barts Biomedical Research Centre, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Elena Aisha Azizan
- Department of Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
- Endocrine Hypertension, Department of Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
- Research Center, Hospital Tunku Ampuan Besar Tuanku Aishah Rohani, Universiti Kebangsaan Malaysia Specialist Children’s Hospital, Kuala Lumpur, Malaysia
| |
Collapse
|
7
|
Deer LK, Hennessey EMP, Doom JR, Gallop RJ, Hoffman MC, Demers CH, Hankin BL, Davis EP. Higher prenatal anxiety predicts lower neonatal hair cortisol. Psychoneuroendocrinology 2024; 165:107044. [PMID: 38657342 PMCID: PMC11139573 DOI: 10.1016/j.psyneuen.2024.107044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 04/04/2024] [Accepted: 04/04/2024] [Indexed: 04/26/2024]
Abstract
BACKGROUND Prenatal glucocorticoids are one of the most widely proposed prenatal programming mechanisms, yet few studies exist that measure fetal cortisol via neonatal hair. Neonatal hair provides a window into the fetal experience and represents cortisol accumulation in the third trimester of pregnancy. In the current study, we test the links between two types of anxiety over the course of gestation (pregnancy-related anxiety and general anxiety) with neonatal hair cortisol. METHOD Pregnant individuals (N = 107) and their neonates (59.8% female) participated in the current study. Prenatal pregnancy-related anxiety and general anxiety were measured using the Pregnancy Related Anxiety Scale (PRAS) and the State-Trait Anxiety Inventory (STAI), in each trimester of pregnancy. Hierarchical linear modeling was used to model the intercept and slope of each type of anxiety over gestation. Neonatal hair samples were collected shortly after birth (Median days = 1.17, IQR = 0.75-2.00). RESULTS Both higher pregnancy-related anxiety and general anxiety at the beginning of pregnancy and a flatter decline of pregnancy-related anxiety over gestation were associated with lower neonatal hair cortisol. After inclusion of gestational age at birth and parity as covariates, pregnancy-related anxiety (intercept: β = -0.614, p =.012; slope: β = -0.681, p =.006), but not general anxiety (intercept: β = -0.389, p =.114; slope: β = -0.302, p =.217) remained a significant predictor. Further, when both general and pregnancy-related anxiety were entered into the same model, only pregnancy-related anxiety (intercept and slope) were significant predictors of neonatal hair cortisol, indicating an association with pregnancy-related anxiety above and beyond general anxiety. CONCLUSION Cortisol plays a central role in maturation of fetal organ systems, and at the end of gestation, higher cortisol has beneficial effects such as promoting fetal lung maturation. Further, lower maternal cortisol is linked to less optimal cognitive development and altered brain development. As maternal higher anxiety in early pregnancy and a flatter decrease over time are both associated with lower neonatal hair cortisol, maternal pregnancy-related anxiety could be a target of future intervention efforts.
Collapse
Affiliation(s)
| | | | - Jenalee R Doom
- Department of Psychology, University of Denver, Denver, CO, USA
| | - Robert J Gallop
- Department of Mathematics, West Chester University, West Chester, PA, USA
| | - M Camille Hoffman
- Department of Psychiatry, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Division of Maternal and Fetal Medicine, Department of Obstetrics and Gynecology, University of Colorado Denver School of Medicine, Aurora, CO, USA
| | - Catherine H Demers
- Department of Psychology, University of Denver, Denver, CO, USA; Department of Psychiatry, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Benjamin L Hankin
- Department of Psychology, University of Illinois at Urbana-Champaign, Champaign, IL, USA
| | - Elysia Poggi Davis
- Department of Psychology, University of Denver, Denver, CO, USA; Department of Pediatrics, University of California, Irvine, Irvine, CA, USA
| |
Collapse
|
8
|
Huo Y, Wan Y, Li Y, Lan L, Chen S, Xu S, Xiong C, Xia W. Associations of pentachlorophenol exposure during pregnancy with maternal and infant reproductive hormones based on a birth cohort. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 932:172723. [PMID: 38670355 DOI: 10.1016/j.scitotenv.2024.172723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Revised: 04/18/2024] [Accepted: 04/22/2024] [Indexed: 04/28/2024]
Abstract
Pentachlorophenol (PCP), a typical environmental endocrine disruptor and a new persistent organic pollutant, has been extensively used as a pesticide worldwide. Although its use has been restricted for decades, PCP remains prevalent in both the environment and human bodies. Despite the known endocrine-disrupting and exogenous hormonal effects of PCP, few epidemiological studies examined such impact, especially among sensitive populations and during critical periods. Based on a prospective birth cohort in Wuhan, China, we collected maternal (first trimester; 13.0 ± 1.02 gestational weeks) and infant urine samples (1.16 ± 0.22 months postpartum) from 720 mother-infant pairs. We aimed to examine the association of PCP exposure during early pregnancy with maternal and infant urinary sex steroid hormones, including estrogens (estrone, E1; estradiol, E2; estriol, E3), progestogens (progesterone, P4; pregnenolone, P5; 17α-OH-Progesterone, 17OHP4; 17α-OH-Pregnenolone, 17OHP5), and androgens (testosterone, Testo; dihydrotestosterone, DHT; dehydroepiandrosterone, DHEA; androstenedione, A4). Additionally, gonadotropins [follicle-stimulating hormone (FSH) and luteinizing hormone (LH)] were measured in infant urine. Detection frequencies of all the sex steroid hormones in the maternal urine samples (>99 %) were higher than those in the infants' [most ≥80 %, except for E1 (3.36 %) and E2 (21.4 %)]. Maternal urinary PCP concentration was found to be significantly related with increased maternal sex steroid hormone concentrations; each interquartile increase in PCP concentration was positively related with percent change of the hormones (%Δ) ranging from 26.6 % to 48.5 %. On the other hand, maternal PCP exposure was associated with significantly increased P4 in male infants [%Δ (95 % confidence interval): 10.5 (0.56, 21.4)] but slightly decreased P4 in female infants [-11.9 (-21.8, 0.68)]. In addition, maternal PCP exposure was significantly associated with decreased FSH [%Δ (95 % CI): -9.90 (-17.0, -2.18)] and LH [-8.44 (-16.0, -0.19)] in the female infants, but not in the male infants. Sensitivity analyses, excluding infertility related treatment, pregnancy complications, preterm birth, or low birth weight, showed generally consistent results. Our findings implied that maternal/prenatal PCP exposure might disrupt the homeostasis of maternal and infant reproductive hormones. However, further studies are needed to confirm the findings.
Collapse
Affiliation(s)
- Yitao Huo
- Institute of Maternal and Child Health, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430016, PR China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubation), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, PR China
| | - Yanjian Wan
- Center for Public Health Laboratory Service, Institute of Environmental Health, Wuhan Centers for Disease Control & Prevention, Wuhan, Hubei 430015, PR China
| | - Ying Li
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubation), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, PR China
| | - Liwen Lan
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubation), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, PR China
| | - Silan Chen
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubation), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, PR China
| | - Shunqing Xu
- Institute of Maternal and Child Health, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430016, PR China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubation), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, PR China
| | - Chao Xiong
- Institute of Maternal and Child Health, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430016, PR China.
| | - Wei Xia
- Institute of Maternal and Child Health, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430016, PR China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubation), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, PR China.
| |
Collapse
|
9
|
Rebboah E, Rezaie N, Williams BA, Weimer AK, Shi M, Yang X, Liang HY, Dionne LA, Reese F, Trout D, Jou J, Youngworth I, Reinholdt L, Morabito S, Snyder MP, Wold BJ, Mortazavi A. The ENCODE mouse postnatal developmental time course identifies regulatory programs of cell types and cell states. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.12.598567. [PMID: 38915583 PMCID: PMC11195270 DOI: 10.1101/2024.06.12.598567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Postnatal genomic regulation significantly influences tissue and organ maturation but is under-studied relative to existing genomic catalogs of adult tissues or prenatal development in mouse. The ENCODE4 consortium generated the first comprehensive single-nucleus resource of postnatal regulatory events across a diverse set of mouse tissues. The collection spans seven postnatal time points, mirroring human development from childhood to adulthood, and encompasses five core tissues. We identified 30 cell types, further subdivided into 69 subtypes and cell states across adrenal gland, left cerebral cortex, hippocampus, heart, and gastrocnemius muscle. Our annotations cover both known and novel cell differentiation dynamics ranging from early hippocampal neurogenesis to a new sex-specific adrenal gland population during puberty. We used an ensemble Latent Dirichlet Allocation strategy with a curated vocabulary of 2,701 regulatory genes to identify regulatory "topics," each of which is a gene vector, linked to cell type differentiation, subtype specialization, and transitions between cell states. We find recurrent regulatory topics in tissue-resident macrophages, neural cell types, endothelial cells across multiple tissues, and cycling cells of the adrenal gland and heart. Cell-type-specific topics are enriched in transcription factors and microRNA host genes, while chromatin regulators dominate mitosis topics. Corresponding chromatin accessibility data reveal dynamic and sex-specific regulatory elements, with enriched motifs matching transcription factors in regulatory topics. Together, these analyses identify both tissue-specific and common regulatory programs in postnatal development across multiple tissues through the lens of the factors regulating transcription.
Collapse
Affiliation(s)
- Elisabeth Rebboah
- Developmental and Cell Biology, University of California Irvine, Irvine, USA
- Center for Complex Biological Systems, University of California Irvine, Irvine, USA
| | - Narges Rezaie
- Developmental and Cell Biology, University of California Irvine, Irvine, USA
- Center for Complex Biological Systems, University of California Irvine, Irvine, USA
| | - Brian A. Williams
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, USA
| | - Annika K. Weimer
- Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, USA
| | - Minyi Shi
- Department of Next Generation Sequencing and Microchemistry, Proteomics and Lipidomics, Genentech, San Francisco, USA
| | - Xinqiong Yang
- Department of Genetics, Stanford University School of Medicine, Palo Alto, USA
| | - Heidi Yahan Liang
- Developmental and Cell Biology, University of California Irvine, Irvine, USA
| | | | - Fairlie Reese
- Developmental and Cell Biology, University of California Irvine, Irvine, USA
| | - Diane Trout
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, USA
| | - Jennifer Jou
- Department of Genetics, Stanford University School of Medicine, Palo Alto, USA
| | - Ingrid Youngworth
- Department of Genetics, Stanford University School of Medicine, Palo Alto, USA
| | | | - Samuel Morabito
- Developmental and Cell Biology, University of California Irvine, Irvine, USA
- Center for Complex Biological Systems, University of California Irvine, Irvine, USA
| | - Michael P. Snyder
- Department of Genetics, Stanford University School of Medicine, Palo Alto, USA
| | - Barbara J. Wold
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, USA
| | - Ali Mortazavi
- Developmental and Cell Biology, University of California Irvine, Irvine, USA
- Center for Complex Biological Systems, University of California Irvine, Irvine, USA
| |
Collapse
|
10
|
Duffel MW. Cytosolic sulfotransferases in endocrine disruption. Essays Biochem 2024:EBC20230101. [PMID: 38699885 PMCID: PMC11531609 DOI: 10.1042/ebc20230101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 04/15/2024] [Accepted: 04/17/2024] [Indexed: 05/05/2024]
Abstract
The mammalian cytosolic sulfotransferases (SULTs) catalyze the sulfation of endocrine hormones as well as a broad array of drugs, environmental chemicals, and other xenobiotics. Many endocrine-disrupting chemicals (EDCs) interact with these SULTs as substrates and inhibitors, and thereby alter sulfation reactions responsible for metabolism and regulation of endocrine hormones such as estrogens and thyroid hormones. EDCs or their metabolites may also regulate expression of SULTs through direct interaction with nuclear receptors and other transcription factors. Moreover, some sulfate esters derived from EDCs (EDC-sulfates) may serve as ligands for endocrine hormone receptors. While the sulfation of an EDC can lead to its excretion in the urine or bile, it may also result in retention of the EDC-sulfate through its reversible binding to serum proteins and thereby enable transport to other tissues for intracellular hydrolysis and subsequent endocrine disruption. This mini-review outlines the potential roles of SULTs and sulfation in the effects of EDCs and our evolving understanding of these processes.
Collapse
Affiliation(s)
- Michael W Duffel
- Department of Pharmaceutical Sciences & Experimental Therapeutics, College of Pharmacy, The University of Iowa, Iowa City, IA 52242, U.S.A
| |
Collapse
|
11
|
McLean MA, Weinberg J, Synnes AR, Miller SP, Grunau RE. Relationships between cortisol levels across early childhood and processing speed at age 4.5 years in children born very preterm. Child Neuropsychol 2024:1-19. [PMID: 38406870 DOI: 10.1080/09297049.2024.2314958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 01/11/2024] [Indexed: 02/27/2024]
Abstract
Children born very low gestational age (VLGA, 29-32 weeks gestational age [GA]) display slower processing speed and altered hypothalamic pituitary adrenal (HPA) axis function, with greater effects in those born extremely low gestational age (ELGA; 24-28 weeks GA). We investigated trajectories of HPA axis activity as indexed by cortisol output and patterns across cognitive assessment at ages 1.5, 3 and 4.5 years, comparing children born ELGA and VLGA and associations with 4.5-year processing speed. In a prospective longitudinal cohort study, infants born very preterm (<33 weeks gestation) returned for developmental assessment at ages 1.5, 3, and 4.5 years. At each age, children completed standardized cognitive testing and saliva samples collected before (Pretest), during (During) and after (End) challenging cognitive tasks were assayed for cortisol. For the total group (n = 188), cortisol area under the curve with respect to ground (AUCg) decreased, while cortisol reactivity to challenge (Pre-test to During) increased from 1.5 to 3 years, remaining stable to 4.5 years. This longitudinal pattern was related to higher Processing Speed (WPPSI-IV) scores at 4.5 years. Children born ELGA displayed higher AUCg than VLGA, particularly at age 3, driven by higher Pre-test cortisol levels. Overall, relative to those born VLGA, children born ELGA displayed greater cortisol responsivity to cognitive challenge. A higher setpoint of cortisol levels at age 3-years in children born ELGA may reflect altered HPA axis regulation more broadly and may contribute to difficulties with information processing in this population, critical for academic and social success.
Collapse
Affiliation(s)
- Mia A McLean
- Department of Pediatrics, University of British Columbia, Vancouver, Canada
- BC Children's Hospital Research Institute, Vancouver, Canada
- Department of Psychology and Neuroscience, Auckland University of Technology, Auckland, New Zealand
| | - Joanne Weinberg
- BC Children's Hospital Research Institute, Vancouver, Canada
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada
| | - Anne R Synnes
- Department of Pediatrics, University of British Columbia, Vancouver, Canada
- BC Children's Hospital Research Institute, Vancouver, Canada
| | - Steven P Miller
- Department of Pediatrics, University of British Columbia, Vancouver, Canada
- BC Children's Hospital Research Institute, Vancouver, Canada
| | - Ruth E Grunau
- Department of Pediatrics, University of British Columbia, Vancouver, Canada
- BC Children's Hospital Research Institute, Vancouver, Canada
| |
Collapse
|
12
|
Heckmann M, Runkel AS, Sunny DE, Hartmann MF, Ittermann T, Wudy SA. Steroid Metabolomic Signature in Term and Preterm Infants. Biomolecules 2024; 14:235. [PMID: 38397473 PMCID: PMC10887377 DOI: 10.3390/biom14020235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 02/09/2024] [Accepted: 02/10/2024] [Indexed: 02/25/2024] Open
Abstract
Adrenal function is essential for survival and well-being of preterm babies. In addition to glucocorticoids, it has been hypothesized that C19-steroids (DHEA-metabolites) from the fetal zone of the adrenal gland may play a role as endogenous neuroprotective steroids. In 39 term-born (≥37 weeks gestational age), 42 preterm (30-36 weeks) and 51 early preterm (<30 weeks) infants 38 steroid metabolites were quantified by GC-MS in 24-h urinary samples. In each gestational age group, three distinctive cluster were identified by pattern analysis (k-means clustering). Individual steroidal fingerprints and clinical phenotype were analyzed at the 3rd day of life. Overall, the excretion rates of C21-steroids (glucocorticoid precursors, cortisol, and cortisone metabolites) were low (<99 μg/kg body weight/d) whereas the excretion rates of C19-steroids were up to 10 times higher. There was a shift to higher excretion rates of C19-steroids in both preterm groups compared to term infants but only minor differences in the distribution of C21-steroids. Comparable metabolic patterns were found between gestational age groups: Cluster 1 showed mild elevation of C21- and C19-steroids with the highest incidence of neonatal morbidities in term and severe intraventricular hemorrhage in early preterm infants. In cluster 2 lowest excretion in general was noted but no clinically unique phenotype. Cluster 3 showed highest elevation of C21-steroids and C19-steroids but no clinically unique phenotype. Significant differences in steroid metabolism between clusters are only partly reflected by gestational age and disease severity. In early preterm infants, higher excretion rates of glucocorticoids and their precursors were associated with severe cerebral hemorrhage. High excretion rates of C19-steroids in preterm infants may indicate a biological significance.
Collapse
Affiliation(s)
- Matthias Heckmann
- Department of Neonatology and Pediatric Intensive Care, University Medicine Greifswald, Sauerbruchstraße, 17475 Greifswald, Germany; (A.S.R.); (D.E.S.)
| | - Anna S. Runkel
- Department of Neonatology and Pediatric Intensive Care, University Medicine Greifswald, Sauerbruchstraße, 17475 Greifswald, Germany; (A.S.R.); (D.E.S.)
| | - Donna E. Sunny
- Department of Neonatology and Pediatric Intensive Care, University Medicine Greifswald, Sauerbruchstraße, 17475 Greifswald, Germany; (A.S.R.); (D.E.S.)
| | - Michaela F. Hartmann
- Paediatric Endocrinology & Diabetology, Laboratory for Translational Hormone Analytics, Steroid Research & Mass Spectrometry Unit, Center of Child and Adolescent Medicine, Justus Liebig University, 35392 Giessen, Germany; (M.F.H.); (S.A.W.)
| | - Till Ittermann
- Institute for Community Medicine, University Medicine Greifswald, 17475 Greifswald, Germany;
| | - Stefan A. Wudy
- Paediatric Endocrinology & Diabetology, Laboratory for Translational Hormone Analytics, Steroid Research & Mass Spectrometry Unit, Center of Child and Adolescent Medicine, Justus Liebig University, 35392 Giessen, Germany; (M.F.H.); (S.A.W.)
| |
Collapse
|
13
|
Chen Y, Wang H. The changes in adrenal developmental programming and homeostasis in offspring induced by glucocorticoids exposure during pregnancy. VITAMINS AND HORMONES 2024; 124:463-490. [PMID: 38408809 DOI: 10.1016/bs.vh.2023.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
Clinically, synthetic glucocorticoids are often used to treat maternal and fetal related diseases, such as preterm birth and autoimmune diseases. Although its clinical efficacy is positive, it will expose the fetus to exogenous glucocorticoids. Adverse environments during pregnancy (e.g., exogenous glucocorticoids exposure, malnutrition, infection, hypoxia, and stress) can lead to fetal overexposure to endogenous maternal glucocorticoids. Basal glucocorticoids levels in utero are crucial in determining fetal tissue maturation and its postnatal fate. As the synthesis and secretion organ of glucocorticoids, the adrenal development is crucial for the growth and development of the body. Studies have found that glucocorticoids exposure during pregnancy could cause abnormal fetal adrenal development, which could last after birth or even adulthood. As the key organ of fetal-originated adult disease, the adrenal developmental programming has a profound impact on the health of offspring, which can lead to many chronic diseases in adulthood. However, the aberrant adrenal development in offspring caused by glucocorticoids exposure during pregnancy and its intrauterine programming mechanism have not been systematically clarified. Therefore, this review summarizes recent research progress on the short and long-term hazards of aberrant adrenal development induced by glucocorticoids exposure during pregnancy, which is of great significance for the analysis of aberrant adrenal development and clarify the intrauterine origin mechanism of fetal-originated adult disease.
Collapse
Affiliation(s)
- Yawen Chen
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, P.R. China; Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Hui Wang
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, P.R. China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, P.R. China.
| |
Collapse
|
14
|
Duffel MW, Lehmler HJ. Complex roles for sulfation in the toxicities of polychlorinated biphenyls. Crit Rev Toxicol 2024; 54:92-122. [PMID: 38363552 PMCID: PMC11067068 DOI: 10.1080/10408444.2024.2311270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 01/23/2024] [Indexed: 02/17/2024]
Abstract
Polychlorinated biphenyls (PCBs) are persistent organic toxicants derived from legacy pollution sources and their formation as inadvertent byproducts of some current manufacturing processes. Metabolism of PCBs is often a critical component in their toxicity, and relevant metabolic pathways usually include their initial oxidation to form hydroxylated polychlorinated biphenyls (OH-PCBs). Subsequent sulfation of OH-PCBs was originally thought to be primarily a means of detoxication; however, there is strong evidence that it may also contribute to toxicities associated with PCBs and OH-PCBs. These contributions include either the direct interaction of PCB sulfates with receptors or their serving as a localized precursor for OH-PCBs. The formation of PCB sulfates is catalyzed by cytosolic sulfotransferases, and, when transported into the serum, these metabolites may be retained, taken up by other tissues, and subjected to hydrolysis catalyzed by intracellular sulfatase(s) to regenerate OH-PCBs. Dynamic cycling between PCB sulfates and OH-PCBs may lead to further metabolic activation of the resulting OH-PCBs. Ultimate toxic endpoints of such processes may include endocrine disruption, neurotoxicities, and many others that are associated with exposures to PCBs and OH-PCBs. This review highlights the current understanding of the complex roles that PCB sulfates can have in the toxicities of PCBs and OH-PCBs and research on the varied mechanisms that control these roles.
Collapse
Affiliation(s)
- Michael W. Duffel
- Department of Pharmaceutical Sciences & Experimental Therapeutics, College of Pharmacy, The University of Iowa, Iowa City, Iowa, 52242, United States
| | - Hans-Joachim Lehmler
- Department of Occupational and Environmental Health, College of Public Health, The University of Iowa, Iowa City, Iowa, 52242, United States
| |
Collapse
|
15
|
Filetti C, Kane-Grade F, Gunnar M. The Development of Stress Reactivity and Regulation in Children and Adolescents. Curr Neuropharmacol 2024; 22:395-419. [PMID: 37559538 PMCID: PMC10845082 DOI: 10.2174/1570159x21666230808120504] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 02/03/2023] [Accepted: 02/10/2023] [Indexed: 08/11/2023] Open
Abstract
Adversity experienced in early life can have detrimental effects on physical and mental health. One pathway in which these effects occur is through the hypothalamic-pituitary-adrenal (HPA) axis, a key physiological stress-mediating system. In this review, we discuss the theoretical perspectives that guide stress reactivity and regulation research, the anatomy and physiology of the axis, developmental changes in the axis and its regulation, brain systems regulating stress, the role of genetic and epigenetics variation in axis development, sensitive periods in stress system calibration, the social regulation of stress (i.e., social buffering), and emerging research areas in the study of stress physiology and development. Understanding the development of stress reactivity and regulation is crucial for uncovering how early adverse experiences influence mental and physical health.
Collapse
Affiliation(s)
- Clarissa Filetti
- Institute of Child Development, University of Minnesota, Minneapolis, USA
| | - Finola Kane-Grade
- Institute of Child Development, University of Minnesota, Minneapolis, USA
| | - Megan Gunnar
- Institute of Child Development, University of Minnesota, Minneapolis, USA
| |
Collapse
|
16
|
Paul B, Buchholz DR. Minireview: Glucocorticoid-Leptin Crosstalk: Role of Glucocorticoid-Leptin Counterregulation in Metabolic Homeostasis and Normal Development. Integr Comp Biol 2023; 63:1127-1139. [PMID: 37708034 DOI: 10.1093/icb/icad119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 08/16/2023] [Accepted: 08/30/2023] [Indexed: 09/16/2023] Open
Abstract
Glucocorticoids and leptin are two important hormones that regulate metabolic homeostasis by controlling appetite and energy expenditure in adult mammals. Also, glucocorticoids and leptin strongly counterregulate each other, such that chronic stress-induced glucocorticoids upregulate the production of leptin and leptin suppresses glucocorticoid production directly via action on endocrine organs and indirectly via action on food intake. Altered glucocorticoid or leptin levels during development can impair organ development and increase the risk of chronic diseases in adults, but there are limited studies depicting the significance of glucocorticoid-leptin interaction during development and its impact on developmental programming. In mammals, leptin-induced suppression of glucocorticoid production is critical during development, where leptin prevents stress-induced glucocorticoid production by inducing a period of short-hyporesponsiveness when the adrenal glands fail to respond to certain mild to moderate stressors. Conversely, reduced or absent leptin signaling increases glucocorticoid levels beyond what is appropriate for normal organogenesis. The counterregulatory interactions between leptin and glucocorticoids suggest the potential significant involvement of leptin in disorders that occur from stress during development.
Collapse
Affiliation(s)
- Bidisha Paul
- Department of Biological Sciences, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Daniel R Buchholz
- Department of Biological Sciences, University of Cincinnati, Cincinnati, OH 45221, USA
| |
Collapse
|
17
|
Kim JH, Choi Y, Hwang S, Yoon JH, Kim GH, Yoo HW, Choi JH. Clinical Characteristics and Long-Term Outcomes of Adrenal Tumors in Children and Adolescents. Exp Clin Endocrinol Diabetes 2023; 131:515-522. [PMID: 37437600 DOI: 10.1055/a-2127-9292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/14/2023]
Abstract
OBJECTIVE Adrenal tumors are generally rare in children and can be a part of familial cancer syndrome. This research was conducted to examine the clinical outcomes, histopathological results, and genetic etiologies of adrenal tumors in children and adolescents. METHODS Thirty-one children and adolescents with adrenal tumors were included. Data on clinical outcomes and endocrine and radiologic results were retrospectively analyzed. Molecular analysis was conducted in select patients according to their phenotype and family history. RESULTS The median age at diagnosis was 7.9 years (range: 0.8-17.8 years) with 5.1±1.8 cm of maximum tumor diameter. Adrenal adenoma (n=7), carcinoma (n=5), borderline (n=2), isolated micronodular adrenocortical disease (n=2), pheochromocytoma (n=8), paraganglioma (n=3), and ganglioneuroma (n=4) are all pathological diagnoses. The most common presenting symptom was excess production of adrenocortical hormones (n=15), including virilization and Cushing syndrome. Non-functioning adrenocortical tumors were found in a patient with congenital adrenal hyperplasia. Genetic etiologies were identified in TP53 (n=5), VHL (n=4), and PRKACA (n=1). Patients with mutations in TP53 were young (1.5±0.5 years) and had large masses (6.1±2.3 cm). CONCLUSIONS This study describes clinical outcomes and the pathological spectrum of adrenal tumors in children and adolescents. Adrenocortical tumors mostly presented with an excess of the adrenocortical hormone. Patients with genetic defects presented at a young age and large size of tumors, necessitating genetic testing in patients at a young age.
Collapse
Affiliation(s)
- Ja Hye Kim
- Department of Pediatrics, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Yunha Choi
- Department of Pediatrics, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Soojin Hwang
- Department of Pediatrics, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Ji-Hee Yoon
- Department of Pediatrics, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Gu-Hwan Kim
- Medical Genetics Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Han-Wook Yoo
- Department of Pediatrics, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jin-Ho Choi
- Department of Pediatrics, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
18
|
Dugalic S, Todorovic J, Sengul D, Sengul I, Veiga ECDA, Plesinac J, Petronijevic M, Macura M, Kepeci SP, Milinčić M, Pavlovic A, Gojnic M. Highlighting early detection of thyroid pathology and gestational diabetes effects on oxidative stress that provokes preterm delivery in thyroidology: Does that ring a bell? Clinics (Sao Paulo) 2023; 78:100279. [PMID: 37783171 PMCID: PMC10562150 DOI: 10.1016/j.clinsp.2023.100279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 08/05/2023] [Accepted: 08/08/2023] [Indexed: 10/04/2023] Open
Abstract
OBJECTIVES Ad fontes, the status of the thyroid gland, and metabolic disturbance lead to the alteration of oxygenation. In pregnancy, it is particularly crucial to possess all predictive parameters. METHODS This cross-sectional study was conducted at the Clinic of Gynecology and Obstetrics, Clinical Center of Serbia, Belgrade, Serbia, between 2017 and 2021 which study included a total of 99 women who had been admitted for preterm delivery and had undergone thyroid analysis, detected Hashimoto thyroiditis, and Oral Glucose Tolerance Test (OGTT) 40 days after delivery and had pathological Homeostatic Model Assessment for Insulin Response (HOMA IR) indices. In the group of urgent patients with preterm delivery, we looked after not only routine Doppler of the umbilical artery, but we measured specific ratios such as the Cerebroplacental ratio (CP). RESULTS The mean maternal age was 32.23 ± 5.96 years and the mean gestational age was detected as 35.40 ± 2.39 weeks. The delivery was completed vaginally in 77 women (78%) and surgically in 22 (22%). The Mean APGAR score was 8.44 ± 1.18, the mean birth weight was 2666.87 ± 622.17g and the cases undergoing cesarean section had significantly higher values of pulsatility index (1.85 ± 0.27 vs. 1.34 ± 0.31) and CP (1.22 ± 0.26 vs. 0.47 ± 0.17). CONCLUSIONS The introduction of Doppler sonography for blood flow assessment helps to form a complete clinical description of the patient, particularly in conditions where oxidative stress became provocative by the thyroid gland antibodies and gestational diabetes in Thyroidology.
Collapse
Affiliation(s)
- Stefan Dugalic
- Clinic for Obstetrics and Gynecology, University Clinical Centre of Serbia, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Jovana Todorovic
- Institute of Social Medicine, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Demet Sengul
- Department of Pathology, Giresun University Faculty of Medicine, Giresun, Turkey
| | - Ilker Sengul
- Division of Endocrine Surgery, Giresun University Faculty of Medicine, Giresun, Turkey; Department of General Surgery, Giresun University Faculty of Medicine, Giresun, Turkey.
| | - Eduardo Carvalho de Arruda Veiga
- Department of Obstetrics and Gynecology, Universidade de São Paulo (FMRP-USP), Faculdade de Medicina de Ribeirão Preto, Hospital das Clínicas, São Paulo, SP, Brazil
| | - Jovana Plesinac
- Clinic for Obstetrics and Gynecology, University Clinical Centre of Serbia, Belgrade, Serbia
| | | | - Maja Macura
- Clinic for Obstetrics and Gynecology, University Clinical Centre of Serbia, Belgrade, Serbia
| | | | - Miloš Milinčić
- Clinic for Obstetrics and Gynecology, University Clinical Centre of Serbia, Belgrade, Serbia
| | - Andrija Pavlovic
- University of Belgrade, Faculty of Medicine, University Children Clinics, Belgrade, Serbia
| | - Miroslava Gojnic
- Clinic for Obstetrics and Gynecology, University Clinical Centre of Serbia, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
19
|
Belluti S, Imbriano C, Casarini L. Nuclear Estrogen Receptors in Prostate Cancer: From Genes to Function. Cancers (Basel) 2023; 15:4653. [PMID: 37760622 PMCID: PMC10526871 DOI: 10.3390/cancers15184653] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/01/2023] [Accepted: 09/18/2023] [Indexed: 09/29/2023] Open
Abstract
Estrogens are almost ubiquitous steroid hormones that are essential for development, metabolism, and reproduction. They exert both genomic and non-genomic action through two nuclear receptors (ERα and ERβ), which are transcription factors with disregulated functions and/or expression in pathological processes. In the 1990s, the discovery of an additional membrane estrogen G-protein-coupled receptor augmented the complexity of this picture. Increasing evidence elucidating the specific molecular mechanisms of action and opposing effects of ERα and Erβ was reported in the context of prostate cancer treatment, where these issues are increasingly investigated. Although new approaches improved the efficacy of clinical therapies thanks to the development of new molecules targeting specifically estrogen receptors and used in combination with immunotherapy, more efforts are needed to overcome the main drawbacks, and resistance events will be a challenge in the coming years. This review summarizes the state-of-the-art on ERα and ERβ mechanisms of action in prostate cancer and promising future therapies.
Collapse
Affiliation(s)
- Silvia Belluti
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (S.B.); (C.I.)
| | - Carol Imbriano
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (S.B.); (C.I.)
| | - Livio Casarini
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Ospedale di Baggiovara, 41126 Modena, Italy
| |
Collapse
|
20
|
Yoshihara M, Mizutani S, Matsumoto K, Kato Y, Masuo Y, Harumasa A, Iyoshi S, Tano S, Mizutani H, Kotani T, Mizutani E, Shibata K, Kajiyama H. The balance between fetal oxytocin and placental leucine aminopeptidase (P-LAP) controls human uterine contraction around labor onset. Eur J Obstet Gynecol Reprod Biol X 2023; 19:100210. [PMID: 37753515 PMCID: PMC10518509 DOI: 10.1016/j.eurox.2023.100210] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 06/20/2023] [Indexed: 09/28/2023] Open
Abstract
A fetal pituitary hormone, oxytocin which causes uterine contractions, increases throughout gestation, and its increase reaches 10-fold from week 32 afterward. Oxytocin is, on the other hand, degraded by placental leucine aminopeptidase (P-LAP) which exists in both terminal villi and maternal blood. Maternal blood P-LAP increases with advancing gestation under the control of non-genomic effects of progesterone, which is also produced from the placenta. Progesterone is converted to estrogen by CYP17A1 localized in the fetal adrenal gland and placenta at term. The higher oxytocin concentrations in the fetus than in the mother demonstrate not only fetal oxytocin production but also its degradation and/or inhibition of leakage from fetus to mother by P-LAP. Until labor onset, the pregnant uterus is quiescent possibly due to the balance between increasing fetal oxytocin and P-LAP under control of progesterone. A close correlation exists between the feto-placental and maternal units in the placental circulation, although the blood in the two circulations does not necessarily mix. Fetal maturation results in progesterone withdrawal via the CYP17A1 activation accompanied with fetal oxytocin increase. Contribution of fetal oxytocin to labor onset has been acknowledged through the recognition that the effect of fetal oxytocin in the maternal blood is strictly regulated by its degradation by P-LAP under the control of non-genomic effects of progesterone. In all senses, the fetus necessarily takes the initiative in labor onset.
Collapse
Affiliation(s)
- Masato Yoshihara
- Department of Obstetrics & Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | | | - Kunio Matsumoto
- Division of Tumor Dynamics and Regulation, Cancer Institute, Kanazawa University, Kanazawa, Japan
| | - Yukio Kato
- Department of Molecular Pharmacotherapeutics, Facility of Pharmacy, Kanazawa University, Kanazawa, Japan
| | - Yusuke Masuo
- Department of Molecular Pharmacotherapeutics, Facility of Pharmacy, Kanazawa University, Kanazawa, Japan
| | | | - Shohei Iyoshi
- Department of Obstetrics & Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Institute for Advanced Research, Nagoya University, Nagoya, Japan
| | - Sho Tano
- Department of Obstetrics & Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hidesuke Mizutani
- Department of Obstetrics & Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tomomi Kotani
- Department of Obstetrics & Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | | | - Kiyosumi Shibata
- Department of Obstetrics & Gynecology, Bantane Hospital, Fujita Health University, Nagoya, Japan
| | - Hiroaki Kajiyama
- Department of Obstetrics & Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
21
|
Stanczyk FZ, Archer DF. Biosynthesis of estetrol in human pregnancy: Potential pathways. J Steroid Biochem Mol Biol 2023; 232:106359. [PMID: 37390976 DOI: 10.1016/j.jsbmb.2023.106359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/26/2023] [Accepted: 06/26/2023] [Indexed: 07/02/2023]
Abstract
Estetrol (E4) has emerged as a novel and highly promising estrogen for therapeutic use. E4 is a weak natural estrogen produced only in pregnancy. Because of its novelty, there is considerable interest by clinicians in how it is produced in pregnancy. Although the fetal liver plays a key role in its production, the placenta is also involved. A current view is that estradiol (E2) formed in the placenta enters the fetal compartment and is then rapidly sulfated. E2 sulfate then undergoes 15α-/16α-hydroxylation in the fetal liver thereby forming E4 sulfate (phenolic pathway). However, another pathway involving 15α,16α-dihydroxy-DHEAS formed in the fetal liver and converted to E4 in the placenta also plays a significant role (neutral pathway). It is not known which pathway predominates, but both pathways appear to be important in E4 biosynthesis. In this commentary, we summarize the well-established pathways in the formation of estrogens in the nonpregnant and pregnant female. We then review what is known about the biosynthesis of E4 and describe the 2 proposed pathways involving the fetus and placenta.
Collapse
Affiliation(s)
- Frank Z Stanczyk
- Department of Obstetrics and Gynecology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States.
| | - David F Archer
- Department of Obstetrics and Gynecology, Eastern Virginia Medical School, Norfolk, VA, United States
| |
Collapse
|
22
|
Uyan Hendem D, Oluklu D, Menekse Beser D, Yildirim M, Sakcak B, Turgut E, Sahin D. Evaluation of fetal adrenal artery Doppler velocimetry and fetal adrenal gland size in pregnancies after recovery from COVID-19. J Obstet Gynaecol Res 2023; 49:2304-2309. [PMID: 37354108 DOI: 10.1111/jog.15725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 06/14/2023] [Indexed: 06/26/2023]
Abstract
AIM Assessment of the fetal adrenal gland (FAG) size and middle adrenal artery (MAA) Doppler parameters in pregnancy recovered from Coronavirus Disease (COVID-19) and comparison of the values with the healthy control group. METHODS Thirty-eight pregnant women who had recovered from COVID-19 infection and 76 healthy control group between 33 and 35 weeks of gestation were involved in this case-control study. Fetuses were examined for fetal biometry, fetal well-being, adrenal gland dimensions, and Doppler parameters 4-6 weeks after the diagnosis of COVID 19 infection. FAG dimensions were measured in two planes and MAA blood flow velocity was evaluated with pulsed Doppler. Pregnant women with COVID-19 infection were grouped according to the National Institutes of Health for the severity of the disease, and those with mild and moderate infections were examined in the study. RESULTS The total adrenal gland (TAG) height, fetal zone (FZ) length and width, and MAA-Peak Systolic Velocity (MAA-PSV) were significantly higher, and the MAA-Pulsatility Index (MAA-PI) was significantly lower in the COVID-19 group (p < 0.05). The lower in MAA-PI and the higher in MAA-PSV, the width of the FZ, and width of the TAG were found to be significant in the moderate group compared to the mild groups (p < 0.05). CONCLUSION COVID-19 pregnancies might cause early maturation of the FAG and its vasculature depends on the intrauterine stress due to the hyper-inflammation, so fetuses exposed to maternal COVID-19 suggested to have an increase in blood flow to the adrenal gland and fetal adrenal size.
Collapse
Affiliation(s)
- Derya Uyan Hendem
- Department of Obstetrics and Gynecology, Turkish Ministry of Health Ankara City Hospital, Ankara, Turkey
- Division of Perinatology, Turkish Ministry of Health Ankara City Hospital, Ankara, Turkey
| | - Deniz Oluklu
- Department of Obstetrics and Gynecology, Turkish Ministry of Health Ankara City Hospital, Ankara, Turkey
- Division of Perinatology, Turkish Ministry of Health Ankara City Hospital, Ankara, Turkey
| | - Dilek Menekse Beser
- Department of Obstetrics and Gynecology, Turkish Ministry of Health Ankara City Hospital, Ankara, Turkey
- Division of Perinatology, Turkish Ministry of Health Ankara City Hospital, Ankara, Turkey
| | - Muradiye Yildirim
- Department of Obstetrics and Gynecology, Turkish Ministry of Health Ankara City Hospital, Ankara, Turkey
- Division of Perinatology, Turkish Ministry of Health Ankara City Hospital, Ankara, Turkey
| | - Bedri Sakcak
- Department of Obstetrics and Gynecology, Turkish Ministry of Health Ankara City Hospital, Ankara, Turkey
- Division of Perinatology, Turkish Ministry of Health Ankara City Hospital, Ankara, Turkey
| | - Ezgi Turgut
- Department of Obstetrics and Gynecology, Turkish Ministry of Health Ankara City Hospital, Ankara, Turkey
- Division of Perinatology, Turkish Ministry of Health Ankara City Hospital, Ankara, Turkey
| | - Dilek Sahin
- Department of Obstetrics and Gynecology, Turkish Ministry of Health Ankara City Hospital, University of Health Sciences, Ankara, Turkey
- Division of Perinatology, Turkish Ministry of Health Ankara City Hospital, University of Health Sciences, Ankara, Turkey
| |
Collapse
|
23
|
del Valle I, Young MD, Kildisiute G, Ogunbiyi OK, Buonocore F, Simcock IC, Khabirova E, Crespo B, Moreno N, Brooks T, Niola P, Swarbrick K, Suntharalingham JP, McGlacken-Byrne SM, Arthurs OJ, Behjati S, Achermann JC. An integrated single-cell analysis of human adrenal cortex development. JCI Insight 2023; 8:e168177. [PMID: 37440461 PMCID: PMC10443814 DOI: 10.1172/jci.insight.168177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 05/31/2023] [Indexed: 07/15/2023] Open
Abstract
The adrenal glands synthesize and release essential steroid hormones such as cortisol and aldosterone, but many aspects of human adrenal gland development are not well understood. Here, we combined single-cell and bulk RNA sequencing, spatial transcriptomics, IHC, and micro-focus computed tomography to investigate key aspects of adrenal development in the first 20 weeks of gestation. We demonstrate rapid adrenal growth and vascularization, with more cell division in the outer definitive zone (DZ). Steroidogenic pathways favored androgen synthesis in the central fetal zone, but DZ capacity to synthesize cortisol and aldosterone developed with time. Core transcriptional regulators were identified, with localized expression of HOPX (also known as Hop homeobox/homeobox-only protein) in the DZ. Potential ligand-receptor interactions between mesenchyme and adrenal cortex were seen (e.g., RSPO3/LGR4). Growth-promoting imprinted genes were enriched in the developing cortex (e.g., IGF2, PEG3). These findings reveal aspects of human adrenal development and have clinical implications for understanding primary adrenal insufficiency and related postnatal adrenal disorders, such as adrenal tumor development, steroid disorders, and neonatal stress.
Collapse
Affiliation(s)
- Ignacio del Valle
- Genetics and Genomic Medicine Research and Teaching Department, University College London (UCL) Great Ormond Street Institute of Child Health, UCL, London, United Kingdom
| | - Matthew D. Young
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, United Kingdom
| | - Gerda Kildisiute
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, United Kingdom
| | - Olumide K. Ogunbiyi
- Department of Histopathology, Great Ormond Street Hospital for Children National Health Service (NHS) Foundation Trust, London, United Kingdom
- Developmental Biology and Cancer Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, UCL, London, United Kingdom
| | - Federica Buonocore
- Genetics and Genomic Medicine Research and Teaching Department, University College London (UCL) Great Ormond Street Institute of Child Health, UCL, London, United Kingdom
| | - Ian C. Simcock
- Department of Clinical Radiology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
- National Institute of Health Research (NIHR) Great Ormond Street Biomedical Research Centre, London, United Kingdom
- Population, Policy and Practice Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, UCL, London, United Kingdom
| | - Eleonora Khabirova
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, United Kingdom
| | - Berta Crespo
- Developmental Biology and Cancer Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, UCL, London, United Kingdom
| | - Nadjeda Moreno
- Developmental Biology and Cancer Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, UCL, London, United Kingdom
| | - Tony Brooks
- UCL Genomics, Zayed Centre for Research, UCL Great Ormond Street Institute of Child Health, UCL, London, United Kingdom
| | - Paola Niola
- UCL Genomics, Zayed Centre for Research, UCL Great Ormond Street Institute of Child Health, UCL, London, United Kingdom
| | - Katherine Swarbrick
- Department of Histopathology, Great Ormond Street Hospital for Children National Health Service (NHS) Foundation Trust, London, United Kingdom
- Developmental Biology and Cancer Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, UCL, London, United Kingdom
| | - Jenifer P. Suntharalingham
- Genetics and Genomic Medicine Research and Teaching Department, University College London (UCL) Great Ormond Street Institute of Child Health, UCL, London, United Kingdom
| | - Sinead M. McGlacken-Byrne
- Genetics and Genomic Medicine Research and Teaching Department, University College London (UCL) Great Ormond Street Institute of Child Health, UCL, London, United Kingdom
| | - Owen J. Arthurs
- Department of Clinical Radiology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
- National Institute of Health Research (NIHR) Great Ormond Street Biomedical Research Centre, London, United Kingdom
- Population, Policy and Practice Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, UCL, London, United Kingdom
| | - Sam Behjati
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, United Kingdom
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
- Department of Paediatrics, University of Cambridge, Cambridge, United Kingdom
| | - John C. Achermann
- Genetics and Genomic Medicine Research and Teaching Department, University College London (UCL) Great Ormond Street Institute of Child Health, UCL, London, United Kingdom
| |
Collapse
|
24
|
Kang Y, Laprocina K, Zheng HS, Huang CCJ. Current insight into the transient X-zone in the adrenal gland cortex. VITAMINS AND HORMONES 2023; 124:297-339. [PMID: 38408801 PMCID: PMC11023618 DOI: 10.1016/bs.vh.2023.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
Mouse models have been widely used in the study of adrenal gland development and diseases. The X-zone is a unique structure of the mouse adrenal gland and lineage-tracing studies show that the X-zone is a remnant of the fetal adrenal cortex. Although the X-zone is considered analogous to the fetal zone in the human adrenal cortex, the functional significance of the X-zone has remained comparatively more obscure. The X-zone forms during the early postnatal stages of adrenal development and regresses later in a remarkable sexually dimorphic fashion. The formation and regression of the X-zone can be different in mice with different genetic backgrounds. Mouse models with gene mutations, hormone/chemical treatments, and/or gonadectomy can also display an aberrant development of the X-zone or alternatively a dysregulated X-zone regression. These models have shed light on the molecular mechanisms regulating the development and regression of these unique adrenocortical cells. This review paper briefly describes the development of the adrenal gland including the formation and regression processes of the X-zone. It also summarizes and lists mouse models that demonstrate different X-zone phenotypes.
Collapse
Affiliation(s)
- Yuan Kang
- Department of Anatomy, Physiology & Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
| | - Karly Laprocina
- Department of Anatomy, Physiology & Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
| | - Huifei Sophia Zheng
- Department of Anatomy, Physiology & Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
| | - Chen-Che Jeff Huang
- Department of Anatomy, Physiology & Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States.
| |
Collapse
|
25
|
Melau C, Gayete Mor B, Lundgaard Riis M, Nielsen JE, Dreisler E, Aaboe K, Tutein Brenøe P, Langhoff Thuesen L, Juul Hare K, Mitchell RT, Frederiksen H, Juul A, Jørgensen A. Dexamethasone affects human fetal adrenal steroidogenesis and subsequent ACTH response in an ex vivo culture model. Front Endocrinol (Lausanne) 2023; 14:1114211. [PMID: 37484942 PMCID: PMC10358843 DOI: 10.3389/fendo.2023.1114211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Accepted: 06/13/2023] [Indexed: 07/25/2023] Open
Abstract
Introduction Administration of dexamethasone (DEX) has been used experimentally to suppress androgenization of external genitalia in 46,XX fetuses with congenital adrenal hyperplasia. Despite this, the prenatal biological mechanism-of-action of DEX on fetal development is not known. This study aimed to examine direct effects of DEX on human fetal adrenal (HFA) steroidogenic activity including possible effects on the subsequent response to ACTH-stimulation. Methods Human fetal adrenal (HFA) tissue from 30 fetuses (1st trimester) were cultured ex vivo with A) DEX (10 µm) for 14 days, or B) DEX (10 µm) for 10 days followed by ACTH (1 nM) for 4 days. DEX-mediated effects on HFA morphology, viability, and apoptosis (immunohistochemistry), gene expression (quantitative PCR), and steroid hormone secretion (LC-MS/MS) were investigated. Results DEX-treatment caused decreased androstenedione (p<0.05) and increased cortisol (p<0.01) secretion suggesting that direct effects on the adrenal gland may contribute to the negative feedback on the hypothalamic-pituitary-adrenal axis in vivo. An altered response to ACTH stimulation in HFA pre-treated with DEX included increased androgen (p<0.05) and reduced cortisol production (p<0.05), supporting clinical observations of a temporary decreased ACTH-response following prenatal DEX-treatment. Additionally, the secretion of corticosterone was decreased (p<0.0001) following ACTH-stimulation in the initially DEX-treated HFAs. Discussion The observed effects suggest that prenatal DEX-treatment can cause direct effects on HFA steroidogenesis and in the subsequent response to ACTH-stimulation. This may indicate a requirement for careful monitoring of adrenal function in prenatally DEX-treated neonates, with particular focus on their mineralocorticoid levels.
Collapse
Affiliation(s)
- Cecilie Melau
- Department of Growth and Reproduction, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Berta Gayete Mor
- Department of Growth and Reproduction, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Malene Lundgaard Riis
- Department of Growth and Reproduction, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - John E. Nielsen
- Department of Growth and Reproduction, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Eva Dreisler
- Department of Gynaecology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Kasper Aaboe
- Department of Gynaecology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Pia Tutein Brenøe
- Department of Obstetrics and Gynaecology, Copenhagen University Hospital, Herlev and Gentofte Hospital, Herlev, Denmark
| | - Lea Langhoff Thuesen
- Department of Obstetrics and Gynaecology, Copenhagen University Hospital, Hvidovre and Amager Hospital, Hvidovre, Denmark
| | - Kristine Juul Hare
- Department of Obstetrics and Gynaecology, Copenhagen University Hospital, Hvidovre and Amager Hospital, Hvidovre, Denmark
| | - Rod T. Mitchell
- Medical Research Council (MRC) Centre for Reproductive Health, The Queen’s Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Hanne Frederiksen
- Department of Growth and Reproduction, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Anders Juul
- Department of Growth and Reproduction, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Anne Jørgensen
- Department of Growth and Reproduction, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
26
|
Yarsilikal Guleroglu F, Balkan Ozmen A, Turan Bakirci I, Ekmez M, Cetin A. Relationship among anogenital distance, adrenal gland volume, and penile length and width at 22-36 weeks of pregnancy. J Perinat Med 2023; 51:356-362. [PMID: 35985035 DOI: 10.1515/jpm-2022-0239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Accepted: 08/03/2022] [Indexed: 11/15/2022]
Abstract
OBJECTIVES The subject of current work was to determine the relationship of fetal ultrasonographic biomarkers, including anogenital distance (AGD), adrenal gland volume, and penile length and width in mothers with male fetuses at 22-36 weeks of gestation for the assessment of the effect of fetal adrenal gland producing androgens on the male anogenital structures that are exposed to androgen effects as anogenital region and penis. METHODS This study is a prospective cross-sectional study conducted in our hospital's outpatient perinatal care unit. One hundred and seventy pregnant women with a male fetus aged 22-36 weeks of gestation were included in the study. The fetal adrenal gland length, width, and depth for the calculation of adrenal volume, AGD, and penile length and width were measured for each participant. The Pearson coefficients were calculated to assess the correlation among these parameters. RESULTS The adrenal gland volume had a meaningful, positive moderate relationship with both the AGD (r=0.60) and penile length and width (r=0.57 and r=0.59, respectively; p<0.001). The AGD had a positive, strong correlation with the penile length and width (r=0.74 and r=0.76, respectively; p<0.001). CONCLUSIONS The fetal adrenal gland as one of the androgen sources of the fetus is an influencer of the development of the anogenital and penile region. The findings of the current study support that the adrenal gland considerably affects the masculinization of male fetuses, since there were remarkable correlations among the AGD, adrenal gland volume, and penile length and width.
Collapse
Affiliation(s)
- Filiz Yarsilikal Guleroglu
- Department of Obstetrics and Gynecology, Istanbul Haseki Training and Research Hospital affiliated with the University of Health Sciences, Istanbul, Turkey
| | - Aliye Balkan Ozmen
- Department of Obstetrics and Gynecology, Bursa City Hospital, Bursa, Turkey
| | - Isil Turan Bakirci
- Department of Obstetrics and Gynecology, Basaksehir Cam ve Sakura City Hospital, Istanbul, Turkey
| | - Murat Ekmez
- Department of Obstetrics and Gynecology, Istanbul Haseki Training and Research Hospital affiliated with the University of Health Sciences, Istanbul, Turkey
| | - Ali Cetin
- Department of Obstetrics and Gynecology, Istanbul Haseki Training and Research Hospital affiliated with the University of Health Sciences, Istanbul, Turkey
| |
Collapse
|
27
|
Pawlicki P, Koziorowska A, Koziorowski M, Pawlicka B, Duliban M, Wieczorek J, Płachno BJ, Pardyak L, Korzekwa AJ, Kotula-Balak M. Senescence and autophagy relation with the expressional status of non-canonical estrogen receptors in testes and adrenals of roe deer (Capreolus capreolus) during the pre-rut period. Theriogenology 2023; 198:141-152. [PMID: 36586352 DOI: 10.1016/j.theriogenology.2022.12.023] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 12/14/2022] [Accepted: 12/14/2022] [Indexed: 12/24/2022]
Abstract
The roe deer bucks represent a spontaneous model to study the synchronized testicular involution and recrudescence cycles. However, cellular processes and hormonal control of steroidogenic glands are scarcely known. For the present study testes and adrenal glands obtained from roe deer during the pre-rut season were used. We aimed to determine (i) senescence and autophagy involvement in testis atrophy (immunohistochemical analysis for tumor suppressor protein encoded by the cyclin-dependent kinase inhibitor 2A; p16 and microtubule-associated protein 1A/1B-light chain 3; LC3, respectively), (ii) the size of the adrenal cortex and medulla (morphometric analysis), (iii) G-protein coupled estrogen receptor (GPER) and estrogen-related receptors (ERRs; type α, β, and Y) distribution and expression (qRT-PCR and immunohistochemical analyses) and (iv) serum testosterone and estradiol levels (immunoassay ELISA). This study revealed pre-rut characteristics of testis structure with the presence of both senescence and autophagy-positive cells and higher involvement of senescence, especially in spermatogenic cells (P < 0.05). In the adrenal cortex, groups of cells exhibiting shrinkage were observed. The presence of ERRs in cells of the seminiferous epithelium and interstitial Leydig cells and GPER presence distinctly in Leydig cells was revealed. In adrenals, these receptors were localized in groups of normal-looking cells and those with shrinkage. Morphometric analysis showed differences in cortex width which was smaller (P < 0.05) than that of the medulla. A weak immunohistochemical signal was observed for ERRβ when compared to ERRα and ERRγ. The mRNA expression level of ERRα and ERRγ was lower (P < 0.001 and P < 0.05, respectively) while ERRβ was higher (P < 0.001) in adrenals when compared to testes. mRNA GPER expression was similar in both glands. In the pre-rut season, the testosterone level was 4.89 ng/ml while the estradiol level was 0.234 ng/ml. We postulate that: (i) senescence and autophagy may be involved in both reinitiation of testis function and/or induction of abnormal processes, (ii) hormonal modulation of testis inactivity may affect adrenal cortex causing cell shrinkage, (iii) ERRs and GPER localization in spermatogenic cells and interstitial cells, as well as cortex cells, may maintain and control the morpho-functional status of both glands, and (iv) androgens and estrogens (via ERRs and GPER) drive cellular processes in the testis and adrenal pre-rut physiology.
Collapse
Affiliation(s)
- Piotr Pawlicki
- Center of Experimental and Innovative Medicine, University of Agriculture in Krakow, Redzina 1c, 30-248, Krakow, Poland
| | - Anna Koziorowska
- College of Natural Sciences, Institute of Material Engineering, University of Rzeszow, Pigonia 1, 35-310, Rzeszow, Poland; College of Natural Sciences, Institute of Biology and Biotechnology, University of Rzeszów, Pigonia 1, 35-310, Rzeszów, Poland
| | - Marek Koziorowski
- College of Natural Sciences, Institute of Material Engineering, University of Rzeszow, Pigonia 1, 35-310, Rzeszow, Poland; Department of Animal Physiology and Reproduction, Faculty of Biotechnology, University of Rzeszow, Pigonia 1, 35-310, Rzeszów, Poland
| | - Bernadetta Pawlicka
- Department of Genetics and Evolutionism, Institute of Zoology and Biomedical Research, Gronostajowa 9, 30-387, Jagiellonian University in Krakow, Krakow, Poland
| | - Michal Duliban
- Department of Endocrinology, Institute of Zoology and Biomedical Research, Gronostajowa 9, 30-387, Jagiellonian University in Krakow, Krakow, Poland
| | - Jarosław Wieczorek
- Department of Clinical Diagnostics and Internal Animal Diseases, University Centre of Veterinary Medicine JU-UA, University of Agriculture in Krakow, Mickiewicza 24/28, 30-059, Krakow, Poland
| | - Bartosz J Płachno
- Department of Plant Cytology and Embryology, Institute of Botany, Jagiellonian University in Krakow, Gronostajowa 9, 30-387, Krakow, Poland
| | - Laura Pardyak
- Center of Experimental and Innovative Medicine, University of Agriculture in Krakow, Redzina 1c, 30-248, Krakow, Poland
| | - Anna J Korzekwa
- Department of Biodiversity Protection, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Tuwima 10, 10-748, Olsztyn, Poland
| | - Malgorzata Kotula-Balak
- Department of Animal Anatomy and Preclinical Sciences, University Centre of Veterinary Medicine JU-UA, University of Agriculture in Kraków, 30-059, Krakow, Poland.
| |
Collapse
|
28
|
Abstract
The adrenal cortex undergoes multiple structural and functional rearrangements to satisfy the systemic needs for steroids during fetal life, postnatal development, and adulthood. A fully functional adrenal cortex relies on the proper subdivision in regions or 'zones' with distinct but interconnected functions, which evolve from the early embryonic stages to adulthood, and rely on a fine-tuned gene network. In particular, the steroidogenic activity of the fetal adrenal is instrumental in maintaining normal fetal development and growth. Here, we review and discuss the most recent advances in our understanding of embryonic and fetal adrenal development, including the known causes for adrenal dys-/agenesis, and the steroidogenic pathways that link the fetal adrenal with the hormone system of the mother through the fetal-placental unit. Finally, we discuss what we think are the major open questions in the field, including, among others, the impact of osteocalcin, thyroid hormone, and other hormone systems on adrenal development and function, and the reliability of rodents as models of adrenal pathophysiology.
Collapse
Affiliation(s)
- Emanuele Pignatti
- Department of Pediatrics, Division of Endocrinology, Diabetology and Metabolism, University Hospital Inselspital, University of Bern, 3010, Bern, Switzerland.
- Department for BioMedical Research, University Hospital Inselspital, University of Bern, 3010, Bern, Switzerland.
| | - Therina du Toit
- Department for BioMedical Research, University Hospital Inselspital, University of Bern, 3010, Bern, Switzerland.
| | - Christa E Flück
- Department of Pediatrics, Division of Endocrinology, Diabetology and Metabolism, University Hospital Inselspital, University of Bern, 3010, Bern, Switzerland
- Department for BioMedical Research, University Hospital Inselspital, University of Bern, 3010, Bern, Switzerland
| |
Collapse
|
29
|
Flück CE, Kuiri-Hänninen T, Silvennoinen S, Sankilampi U, Groessl M. The Androgen Metabolome of Preterm Infants Reflects Fetal Adrenal Gland Involution. J Clin Endocrinol Metab 2022; 107:3111-3119. [PMID: 35994776 DOI: 10.1210/clinem/dgac482] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Indexed: 11/19/2022]
Abstract
CONTEXT The human adrenal cortex changes with fetal-neonatal transition from the fetal to the adult organ, accompanied by changes in the steroid metabolome. OBJECTIVE As it is unclear how the observed developmental changes differ between preterm and full-term neonates, we investigated whether the involution of the fetal adrenals is following a fixed time course related to postmenstrual age or whether it is triggered by birth. Furthermore, the fetal and postnatal androgen metabolome of preterm infants was characterized in comparison to term babies. METHODS This was a prospective, longitudinal, 2-center study collecting spot urines of preterm and term infants during the first 12 to 18 months of life. Steroid metabolites were measured from spot urines by gas chromatography-mass spectrometry. Data relating were modeled according to established pre- and postnatal pathways. RESULTS Fetal adrenal involution occurs around term-equivalent age in preterm infants and is not triggered by premature birth. Testosterone levels are higher in preterm infants at birth and decline slower until term compared to full-term babies. Dihydrotestosterone levels and the activity of the classic androgen biosynthesis pathway are lower in premature infants as is 5α-reductase activity. No difference was found in the activity of the alternate backdoor pathway for androgen synthesis. CONCLUSION Human adrenal involution follows a strict timing that is not affected by premature birth. By contrast, prematurity is associated with an altered androgen metabolome after birth. Whether this reflects altered androgen biosynthesis in utero remains to be investigated.
Collapse
Affiliation(s)
- Christa E Flück
- Department of Pediatrics, Division of Endocrinology, Diabetology and Metabolism, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
- Department of BioMedical Research, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
| | - Tanja Kuiri-Hänninen
- Department of Pediatrics, Kuopio University Hospital and University of Eastern Finland, 70029 Kuopio, Finland
| | - Sanna Silvennoinen
- Department of Pediatrics, Kuopio University Hospital and University of Eastern Finland, 70029 Kuopio, Finland
| | - Ulla Sankilampi
- Department of Pediatrics, Kuopio University Hospital and University of Eastern Finland, 70029 Kuopio, Finland
| | - Michael Groessl
- Department of BioMedical Research, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
- Department of Nephrology and Hypertension, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
| |
Collapse
|
30
|
Sakata Y, Cheng K, Mayama M, Seita Y, Detlefsen AJ, Mesaros CA, Penning TM, Shishikura K, Yang W, Auchus RJ, Strauss JF, Sasaki K. Reconstitution of human adrenocortical specification and steroidogenesis using induced pluripotent stem cells. Dev Cell 2022; 57:2566-2583.e8. [PMID: 36413950 PMCID: PMC9927873 DOI: 10.1016/j.devcel.2022.10.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 09/18/2022] [Accepted: 10/25/2022] [Indexed: 11/23/2022]
Abstract
The mechanisms leading to adrenal cortex development and steroid synthesis in humans remain poorly understood due to the paucity of model systems. Herein, we recapitulate human fetal adrenal cortex specification processes through stepwise induction of human-induced pluripotent stem cells through posterior intermediate mesoderm-like and adrenocortical progenitor-like states to ultimately generate fetal zone adrenal-cortex-like cells (FZLCs), as evidenced by histomorphological, ultrastructural, and transcriptome features and adrenocorticotropic hormone (ACTH)-independent Δ5 steroid biosynthesis. Furthermore, FZLC generation is promoted by SHH and inhibited by NOTCH, ACTIVIN, and WNT signaling, and steroid synthesis is amplified by ACTH/PKA signaling and blocked by inhibitors of Δ5 steroid synthesis enzymes. Finally, NR5A1 promotes FZLC survival and steroidogenesis. Together, these findings provide a framework for understanding and reconstituting human adrenocortical development in vitro, paving the way for cell-based therapies of adrenal insufficiency.
Collapse
Affiliation(s)
- Yuka Sakata
- Department of Biomedical Sciences, University of Pennsylvania, School of Veterinary Medicine, Philadelphia, PA 19104, USA.,Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.,These authors contributed equally to this work
| | - Keren Cheng
- Department of Biomedical Sciences, University of Pennsylvania, School of Veterinary Medicine, Philadelphia, PA 19104, USA.,Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.,These authors contributed equally to this work
| | - Michinori Mayama
- Department of Biomedical Sciences, University of Pennsylvania, School of Veterinary Medicine, Philadelphia, PA 19104, USA.,Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.,These authors contributed equally to this work
| | - Yasunari Seita
- Department of Biomedical Sciences, University of Pennsylvania, School of Veterinary Medicine, Philadelphia, PA 19104, USA.,Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.,Bell Research Center for Reproductive Health and Cancer, Nagoya 460-0003, Japan
| | - Andrea J. Detlefsen
- Department of Biochemistry and Biophysics, Perelman School of Medicine University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Clementina A. Mesaros
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.,Center of Excellence in Environmental Toxicology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Trevor M. Penning
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.,Center of Excellence in Environmental Toxicology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kyosuke Shishikura
- Department of Chemistry, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Wenli Yang
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.,Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.,Penn Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Richard J. Auchus
- Division of Metabolism, Endocrinology, and Diabetes, Departments of Internal Medicine and Pharmacology, University of Michigan, Ann Arbor, MI 48105, USA
| | - Jerome F. Strauss
- Department of Obstetrics and Gynecology, University of Pennsylvania, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kotaro Sasaki
- Department of Biomedical Sciences, University of Pennsylvania, School of Veterinary Medicine, Philadelphia, PA 19104, USA; Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Center of Excellence in Environmental Toxicology, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Pathology and Laboratory Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA.
| |
Collapse
|
31
|
Pourquet A, Teoli J, Bouty A, Renault L, Roucher F, Mallet D, Rigaud C, Dijoud F, Mouriquand P, Mure PY, Sanlaville D, Ecochard R, Plotton I. Steroid profiling in the amniotic fluid: reference range for 12 steroids and interest in 21-hydroxylase deficiency. J Clin Endocrinol Metab 2022; 108:e129-e138. [PMID: 36402139 DOI: 10.1210/clinem/dgac656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 10/25/2022] [Accepted: 11/10/2022] [Indexed: 11/21/2022]
Abstract
INTRODUCTION Determination of steroid levels in the amniotic fluid gives some insight on foetal adrenal and gonadal functions. Our objectives were to establish reference ranges of 12 steroid levels throughout pregnancy and to compare them with steroid levels from pregnancies with foetuses presenting 21-hydroxylase deficiency (21OHD). MATERIALS AND METHODS Liquid chromatography coupled with tandem mass spectrometry (LC-MS/MS) was applied to 145 "control" amniotic fluid samples from gynaecology activity (12 + 6 to 32 + 4 Gestational Weeks, GW). The following steroids were analysed according to gestational age and compared to 23 amniotic fluid samples from foetuses with classic 21-hydroxylase deficiency confirmed by molecular studies: delta-4-androstenedione (D4), dehydroepiandrosterone (DHEA), 17-hydroxyprogesterone (17OHP), 11-deoxycortisol (11OH), 21-deoxycortisol (21OH), corticosterone, deoxycorticosterone (DOC), testosterone, pregnenolone, 17-hydroxypregnenolone (17Pregn), cortisol and cortisone. Chromosomal sex was determined by karyotype and gestational age by biometric measurements. RESULTS Analysis of "control" samples showed a statistically significant difference for D4 and testosterone levels according to foetal sex. Cortisol, corticosterone, and DOC had lower concentrations before 20 GW than after 20 GW, whereas 17Pregn and pregnenolone had higher concentrations before 20 GW. This allowed us to establish age- and sex-dependant reference values. We observed higher 21OH, 17Pregn, D4 and testosterone levels in females 21OHD than female controls. The ratios 17OHP/17Pregn, D4/DHEA and 11OH/17OHP appeared discriminant for the diagnosis of 21OHD. CONCLUSION Our study provides information on foetal steroidogenesis and suggests reference values for 12 steroids during pregnancy. This allows a prenatal diagnosis of 21-hydroxylase deficiency within 24 hours and might be useful in the diagnosis of other variations of sex development (VSD).
Collapse
Affiliation(s)
- Anne Pourquet
- Department of Clinical Biochemistry, University Hospital of Lyon, Lyon, France
- Department of Pediatric Surgery, University Hospital of Lyon, Lyon, France
- Claude Bernard Lyon 1 University
| | - Jordan Teoli
- Department of Clinical Biochemistry, University Hospital of Lyon, Lyon, France
- Claude Bernard Lyon 1 University
| | - Aurore Bouty
- Department of Pediatric Surgery, University Hospital of Lyon, Lyon, France
- Claude Bernard Lyon 1 University
| | - Lucie Renault
- Department of Clinical Biochemistry, University Hospital of Lyon, Lyon, France
- Claude Bernard Lyon 1 University
| | - Florence Roucher
- Department of Clinical Biochemistry, University Hospital of Lyon, Lyon, France
- Claude Bernard Lyon 1 University
| | - Delphine Mallet
- Department of Clinical Biochemistry, University Hospital of Lyon, Lyon, France
- Claude Bernard Lyon 1 University
| | - Chantal Rigaud
- Department of Clinical Biochemistry, University Hospital of Lyon, Lyon, France
| | - Frédérique Dijoud
- Department of Pathology, University Hospital of Lyon, Lyon, France
- Claude Bernard Lyon 1 University
| | - Pierre Mouriquand
- Department of Pediatric Surgery, University Hospital of Lyon, Lyon, France
- Claude Bernard Lyon 1 University
| | - Pierre-Yves Mure
- Department of Pediatric Surgery, University Hospital of Lyon, Lyon, France
- Claude Bernard Lyon 1 University
| | - Damien Sanlaville
- Department of Medical Genetics, University Hospital of Lyon, Lyon, France
- Claude Bernard Lyon 1 University
| | - René Ecochard
- Department of Biostatistics, University Hospital of Lyon, Lyon, France
- Claude Bernard Lyon 1 University
| | - Ingrid Plotton
- Department of Clinical Biochemistry, University Hospital of Lyon, Lyon, France
- Claude Bernard Lyon 1 University
| |
Collapse
|
32
|
Ssewanyana D, Knight JA, Matthews SG, Wong J, Khani NA, Lye J, Murphy KE, Foshay K, Okeke J, Lye SJ, Hung RJ. Maternal prenatal psychological distress and vitamin intake with children's neurocognitive development. Pediatr Res 2022; 92:1450-1457. [PMID: 35288638 DOI: 10.1038/s41390-022-02003-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 01/06/2022] [Accepted: 02/06/2022] [Indexed: 11/09/2022]
Abstract
BACKGROUND Maternal prenatal psychological distress (PPD) is increasingly linked to sub-optimal child neurodevelopment. Daily intake of prenatal vitamin during pre-conception and early pregnancy may ameliorate the effects of PPD on cognition in the offspring. METHODS PPD was assessed in early (12-16 weeks) and late (28-32 weeks) gestation in the Ontario Birth Study. Prenatal vitamin supplement intake information was collected in early gestation. Child cognition at 4 years was assessed using the NIH Toolbox. Poisson regression was used to investigate associations between PPD and/or prenatal vitamin intake and child cognition. RESULTS Four hundred and eighteen mother-child dyads were assessed. Moderate-severe PPD experienced during early gestation was associated with reduced cognition (adjusted incidence rate ratio (IRRadj) = 3.71, 95% confidence interval (CI): 1.57-8.77, P = 0.003). Daily intake of prenatal vitamins was not associated with cognition (IRRadj = 1.34, 95% CI: 0.73-2.46, P = 0.34). Upon stratification, the experience of mild-severe PPD with daily intake of prenatal vitamins was associated with higher incident rates of suboptimal cognition compared to children of women with daily prenatal vitamin intake without any episode of PPD (IRRadj = 2.88, 95% CI: 1.1-7.4). CONCLUSIONS Moderate-severe PPD in early pregnancy is associated with poor cognition in children and daily intake of prenatal vitamin did not ameliorate this association. IMPACT Our findings expand on existing literature by highlighting that exposure to prenatal psychological distress (PPD), in moderate-to-severe form, in the early stages of pregnancy, can have detrimental effects on the offspring's cognitive development at 4 years. Overall, prenatal vitamin intake did not ameliorate the effects of PPD. Early screening and treatment of prenatal maternal mental illness is crucial.
Collapse
Affiliation(s)
- Derrick Ssewanyana
- Alliance for Human Development, Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, ON, Canada.,Prosserman Centre for Population Health Research, Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, ON, Canada
| | - Julia A Knight
- Prosserman Centre for Population Health Research, Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, ON, Canada.,Division of Epidemiology, Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| | - Stephen G Matthews
- Alliance for Human Development, Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, ON, Canada.,Department of Physiology, University of Toronto, Toronto, ON, Canada.,Department of Medicine, University of Toronto, Toronto, ON, Canada.,Department of Obstetrics and Gynaecology, University of Toronto, Toronto, ON, Canada.,Department of Obstetrics and Gynaecology, Sinai Health System, Toronto, ON, Canada
| | - Jody Wong
- Prosserman Centre for Population Health Research, Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, ON, Canada
| | - Nadya Adel Khani
- Prosserman Centre for Population Health Research, Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, ON, Canada
| | - Jennifer Lye
- Prosserman Centre for Population Health Research, Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, ON, Canada
| | - Kellie E Murphy
- Department of Obstetrics and Gynaecology, University of Toronto, Toronto, ON, Canada.,Department of Obstetrics and Gynaecology, Sinai Health System, Toronto, ON, Canada
| | - Kim Foshay
- Department of Obstetrics and Gynaecology, Sinai Health System, Toronto, ON, Canada
| | - Justin Okeke
- Department of Obstetrics and Gynaecology, Sinai Health System, Toronto, ON, Canada
| | - Stephen J Lye
- Alliance for Human Development, Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, ON, Canada.,Department of Physiology, University of Toronto, Toronto, ON, Canada.,Department of Medicine, University of Toronto, Toronto, ON, Canada.,Department of Obstetrics and Gynaecology, University of Toronto, Toronto, ON, Canada.,Department of Obstetrics and Gynaecology, Sinai Health System, Toronto, ON, Canada
| | - Rayjean J Hung
- Prosserman Centre for Population Health Research, Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, ON, Canada. .,Division of Epidemiology, Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
33
|
Nilsson AK, Hellgren G, Sjöbom U, Landin A, Ryberg H, Wackernagel D, Ley D, Hansen Pupp I, Poutanen M, Ohlsson C, Hellstrom A. Preterm infant circulating sex steroid levels are not altered by transfusion with adult male plasma: a retrospective multicentre cohort study. Arch Dis Child Fetal Neonatal Ed 2022; 107:577-582. [PMID: 35232892 PMCID: PMC9606499 DOI: 10.1136/archdischild-2021-323433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 02/12/2022] [Indexed: 12/02/2022]
Abstract
OBJECTIVE To determine if plasma transfusions with male donor plasma to very preterm infants affect circulatory levels of sex steroids. DESIGN AND PATIENTS Retrospective multicentre cohort study in 19 infants born at gestational age <29 weeks requiring plasma transfusion during their first week of life. SETTING Three neonatal intensive care units in Sweden. MAIN OUTCOME MEASURES Concentrations of sex steroids and sex hormone-binding globulin (SHBG) in donor plasma and infant plasma measured before and after a plasma transfusion and at 6, 12, 24 and 72 hours. RESULTS The concentrations of progesterone, dehydroepiandrosterone and androstenedione were significantly lower in donor plasma than in infant plasma before the transfusion (median (Q1-Q3) 37.0 (37.0-37.0), 1918 (1325-2408) and 424 (303-534) vs 901 (599-1774), 4119 (2801-14 645) and 842 (443-1684) pg/mL), while oestrone and oestradiol were higher in donor plasma (17.4 (10.4-20.1) and 16.0 (11.7-17.2) vs 3.1 (1.1-10.2) and 0.25 (0.25-0.25) pg/mL). Median testosterone and dihydrotestosterone (DHT) levels were 116-fold and 21-fold higher in donor plasma than pre-transfusion levels in female infants, whereas the corresponding difference was not present in male infants. Plasma sex steroid levels were unchanged after completed transfusion compared with pre-transfusion levels, irrespective of the gender of the receiving infant. The SHBG concentration was significantly higher in donor than in recipient plasma (22.8 (17.1-33.5) vs 10.2 (9.1-12.3) nmol/L) before transfusion but did not change in the infants after the transfusion. CONCLUSIONS A single transfusion of adult male plasma to preterm infants had no impact on circulating sex steroid levels.
Collapse
Affiliation(s)
- Anders K Nilsson
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, University of Gothenburg Sahlgrenska Academy, Gothenburg, Sweden
| | - Gunnel Hellgren
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, University of Gothenburg Sahlgrenska Academy, Gothenburg, Sweden,Institute of Biomedicine, University of Gothenburg Sahlgrenska Academy, Gothenburg, Sweden
| | - Ulrika Sjöbom
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, University of Gothenburg Sahlgrenska Academy, Gothenburg, Sweden,Institute of Health and Care Sciences, University of Gothenburg Sahlgrenska Academy, Gothenburg, Sweden
| | - Andreas Landin
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, University of Gothenburg Sahlgrenska Academy, Gothenburg, Sweden
| | - Henrik Ryberg
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, University of Gothenburg Sahlgrenska Academy, Gothenburg, Sweden,Department of Clinical Chemistry, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Dirk Wackernagel
- Department of Neonatology, Karolinska University Hospital, Stockholm, Sweden,Department of Clinical Science CLINTEC, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - David Ley
- Department of Pediatrics, Institute of Clinical Sciences Lund, Lund University Hospital, Lund, Sweden
| | - Ingrid Hansen Pupp
- Department of Pediatrics, Institute of Clinical Sciences Lund, Lund University Hospital, Lund, Sweden
| | - Matti Poutanen
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology and Turku Center for Disease Modeling, University of Turku, Turku, Finland,Centre for Bone and Arthritis Research, Sahlgrenska Osteoporosis Centre, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, University of Gothenburg Sahlgrenska Academy, Gothenburg, Sweden
| | - Claes Ohlsson
- Centre for Bone and Arthritis Research, Sahlgrenska Osteoporosis Centre, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, University of Gothenburg Sahlgrenska Academy, Gothenburg, Sweden,Department of Drug Treatment, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Ann Hellstrom
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, University of Gothenburg Sahlgrenska Academy, Gothenburg, Sweden
| |
Collapse
|
34
|
Junhasavasdikul S, Panburana P, Bumrungphuet S, Dulyaphat W. The Correlation Between Three-Dimensional Ultrasound Measurement of Fetal Adrenal Gland and Maternal Serum Fructosamine Level in Gestational Diabetes Mellitus: Prospective Cohort Study. Int J Womens Health 2022; 14:1465-1476. [PMID: 36277447 PMCID: PMC9586166 DOI: 10.2147/ijwh.s373087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 09/26/2022] [Indexed: 12/03/2022] Open
Abstract
OBJECTIVE Gestational diabetes mellitus (GDM) has an impact on fetal adrenal gland size and volume, which are greater in the GDM population. This study used 2D and 3D ultrasound scanning to determine the correlation of fetal adrenal gland size and volume with fructosamine levels, HbA1c levels, estimated fetal weight (EFW), and neonatal birth weight in GDM patients. METHODS This study included eighty singleton pregnant women diagnosed with GDM between 24-28 weeks of gestation. During weeks 32-34 of gestation, the length, width, and depth of the fetal zone and total adrenal gland were measured using transabdominal 2D ultrasound. Virtual organ computer-aided analysis (VOCAL) software was used to evaluate fetal zone and total adrenal gland volume in 3D ultrasound. All the participants were followed until delivery. Pearson's correlation analysis was conducted to examine the correlation between fetal adrenal gland ultrasound measurements and the factors of interest. RESULTS The study consisted of sixty-six (82.5%) pregnant women with diet-controlled GDM (GDMA1) and fourteen (17.5%) pregnant women with insulin-managed GDM (GDMA2). There was no difference in fetal adrenal gland measurements between the diet-controlled (GDMA1) and the insulin-managed (GDMA2) groups. All the participants had achieved optimal glucose levels at the time of ultrasound acquisition. The total adrenal gland length and fetal zone volume had statistically significant positive correlations with EFW (r = 0.69, p = 0.02 and r = 0.84, p = 0.01, respectively). After adjusting for EFW, only the fetal zone volume was significantly correlated with fructosamine levels (adjusted-OR = 2.4, 95% CI: 1.5, 3.9, p = 0.01) and HbA1c levels (adjusted-OR = 2.5, 95% CI: 1.6, 4.3, p = 0.01). CONCLUSION The fetal zone volume is correlated with EFW, fructosamine levels, and HbA1c levels. This non-invasive technique may be beneficial as an indirect marker for glycemic monitoring in GDM.
Collapse
Affiliation(s)
- Saowapak Junhasavasdikul
- Department of Obstetrics and Gynecology, Division of Maternal Fetal Medicine, Faculty of Medicine, Mahidol University, Ramathibodi Hospital, Bangkok, Thailand
| | - Panyu Panburana
- Department of Obstetrics and Gynecology, Division of Maternal Fetal Medicine, Faculty of Medicine, Mahidol University, Ramathibodi Hospital, Bangkok, Thailand
| | - Sommart Bumrungphuet
- Department of Obstetrics and Gynecology, Division of Maternal Fetal Medicine, Faculty of Medicine, Mahidol University, Ramathibodi Hospital, Bangkok, Thailand
| | - Wirada Dulyaphat
- Department of Obstetrics and Gynecology, Division of Maternal Fetal Medicine, Faculty of Medicine, Mahidol University, Ramathibodi Hospital, Bangkok, Thailand,Correspondence: Wirada Dulyaphat, Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, 10400, Thailand, Tel +66-873017028, Fax +66 02-2011416, Email
| |
Collapse
|
35
|
Elghareeb MM, Elshopakey GE, Elkhooly TA, Salama B, Samy A, Bazer FW, Elmetwally MA, Almutairi MH, Aleya L, Abdel-Daim MM, Rezk S. Estradiol and zinc-doped nano hydroxyapatite as therapeutic agents in the prevention of osteoporosis; oxidative stress status, inflammation, bone turnover, bone mineral density, and histological alterations in ovariectomized rats. Front Physiol 2022; 13:989487. [PMID: 36200054 PMCID: PMC9527315 DOI: 10.3389/fphys.2022.989487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 08/22/2022] [Indexed: 11/28/2022] Open
Abstract
Osteoporosis (OP) is a serious health problem, and the most popular therapeutic strategy for OP is hormone replacement (estrogen); however, it increases the risk of reproductive cancers. Hydroxyapatite (HA) nanoparticles have a similar chemical structure to the bone mineral component and can be used as a new remedy for OP. This study was designed to investigate the osteoporosis-protective potential of nano zinc hydroxyapatite (ZnHA-NPs) and/or estradiol (E2) combined therapy. A total of 35 adult female rats were assigned into five groups (n = 7): 1) control group; 2) ovariectomized group (OVX); 3) OVX received oral estradiol replacement therapy (OVX/E2); 4) OVX received ZnHA replacement therapy (OVX/ZnHA); and 5) OVX received both estradiol and ZnHA-NPs combined therapy (OVX/E2+ZnHA). After 3 months of treatment, serum bone markers and estrogen level, oxidative/antioxidant, and inflammatory cytokines were determined. Additionally, femoral expression of estrogen receptors alpha and beta (ESR1; ESR2), receptor activator of nuclear factor-kappa B (RANKL) ligand, osteoprotegerin (OPG), bone mineral density (BMD), histological alterations, and immunohistochemical expression of vascular endothelial growth factor (VEGF) and proliferating cell nuclear antigen (PCNA) were assessed. ALP, PINP, Ca, and P concentrations improved significantly (p < 0.05) in all treatment groups, especially in the OVX/E + ZnHA group. MDA and NO were higher in OVX rats, while SOD activity and GSH were lower (p < 0.05). E2 alone or with ZnHA-NPs restored the estimated antioxidant molecules and cytokines toward normal levels in OVX rats (p < 0.05). On the other hand, E2 and ZnHA increased OPG and OC expression in femurs while decreasing ESR1, ESR2, and NF-kB expression (p < 0.05). The combination treatment was superior in the restoration of normal femoral histoarchitecture and both cortical and trabecular BMD (p < 0.05). Overall, the combined therapy of OVX/E2+ZnHA was more effective than the individual treatments in attenuating excessive bone turnover and preventing osteoporosis.
Collapse
Affiliation(s)
- Mona M. Elghareeb
- Department of Physiology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Gehad E. Elshopakey
- Department of Clinical Pathology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Tarek A. Elkhooly
- Nanomedicine Research Unit, Faculty of Medicine, Delta University for Science and Technology, Belqas, Egypt
- Refractories, Ceramics, and Building Materials Department, National Research Centre, Giza, Egypt
- Department of Physics, Faculty of Science, New Mansoura University, New Mansoura City, Egypt
| | - Basma Salama
- Department of Biochemistry and Chemistry of Nutrition, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Alaa Samy
- Department of Surgery, Anesthesiology, and Radiology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Fuller W Bazer
- Department of Animal Science, Texas A&M University, College Station, TX, United States
| | - Mohammed A Elmetwally
- Department of Theriogenology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Mikhlid H. Almutairi
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Lotfi Aleya
- UMR CNRS 6249, Chrono-Environnement Laboratory, Bourgogne, Franche-Comté University, Besançon, France
| | - Mohamed M. Abdel-Daim
- Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, Egypt
| | - Shaymaa Rezk
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| |
Collapse
|
36
|
Stoye DQ, Boardman JP, Osmond C, Sullivan G, Lamb G, Black GS, Homer NZM, Nelson N, Theodorsson E, Mörelius E, Reynolds RM. Saliva cortisol diurnal variation and stress responses in term and preterm infants. Arch Dis Child Fetal Neonatal Ed 2022; 107:558-564. [PMID: 35256524 PMCID: PMC9411886 DOI: 10.1136/archdischild-2021-321593] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Accepted: 02/11/2022] [Indexed: 01/15/2023]
Abstract
OBJECTIVE To determine if preterm birth is associated with adaptation of the hypothalamic-pituitary-adrenal (HPA) axis and whether HPA axis programming relates to the degree of prematurity (defined as extremely preterm birth at <28 weeks or very preterm birth at 28-32 weeks gestation). DESIGN This study reports findings from a prospective birth cohort. Saliva cortisol concentrations were measured prevaccination and postvaccination, and in the morning and evening, at 4 months chronological age. SETTING Infants born at a single Scottish hospital. PARTICIPANTS 45 term-born, 42 very preterm and 16 extremely preterm infants. OUTCOMES Cortisol stress response to vaccination (postvaccination minus prevaccination cortisol concentrations), diurnal slope (log-transformed morning minus log-transformed evening cortisol values) and mean log-transformed daily cortisol. RESULTS Compared with infants born at term, infants born extremely preterm had a blunted cortisol response to vaccination (5.8 nmol/L vs 13.1 nmol/L, difference in means: -7.3 nmol/L, 95% CI -14.0 to -0.6) and a flattened diurnal slope (difference in geometric means: -72.9%, 95% CI -87.1 to -42.8). In contrast, the cortisol response to vaccination (difference in means -2.7 nmol/L, 95% CI -7.4 to 2.0) and diurnal slope at 4 months (difference in geometric means: -33.6%, 95% CI -62.0 to 16.0) did not differ significantly in infants born very preterm compared with infants born at term. CONCLUSIONS Infants born extremely preterm have blunted cortisol reactivity and a flattened diurnal slope. These patterns of HPA axis regulation are commonly seen after childhood adversity and could contribute to later metabolic and neurodevelopmental phenotypes observed in this population.
Collapse
Affiliation(s)
- David Q Stoye
- MRC Centre for Reproductive Health, The University of Edinburgh, Edinburgh, UK
| | - James P Boardman
- MRC Centre for Reproductive Health, The University of Edinburgh, Edinburgh, UK
- Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, UK
| | - Clive Osmond
- MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton, UK
| | - Gemma Sullivan
- MRC Centre for Reproductive Health, The University of Edinburgh, Edinburgh, UK
| | - Gillian Lamb
- MRC Centre for Reproductive Health, The University of Edinburgh, Edinburgh, UK
| | - Gill S Black
- MRC Centre for Reproductive Health, The University of Edinburgh, Edinburgh, UK
| | - Natalie Z M Homer
- Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh, UK
| | - Nina Nelson
- Department of Clinical and Experimental Medicine, Linköping University, Linkoping, Sweden
- Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Stockholm, Sweden
| | - Elvar Theodorsson
- Division of Clinical Chemistry, Department of Biomedical and Clinical Sciences, Faculty of Medicine and Health Sciences, Linköping University, Linkoping, Sweden
| | - Evalotte Mörelius
- School of Nursing and Midwifery, Edith Cowan University, Joondalup, Western Australia, Australia
| | - Rebecca M Reynolds
- MRC Centre for Reproductive Health, The University of Edinburgh, Edinburgh, UK
- Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh, UK
| |
Collapse
|
37
|
Tamburello M, Altieri B, Sbiera I, Sigala S, Berruti A, Fassnacht M, Sbiera S. FGF/FGFR signaling in adrenocortical development and tumorigenesis: novel potential therapeutic targets in adrenocortical carcinoma. Endocrine 2022; 77:411-418. [PMID: 35583844 PMCID: PMC9385797 DOI: 10.1007/s12020-022-03074-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 05/08/2022] [Indexed: 12/14/2022]
Abstract
FGF/FGFR signaling regulates embryogenesis, angiogenesis, tissue homeostasis and wound repair by modulating proliferation, differentiation, survival, migration and metabolism of target cells. Understandably, compelling evidence for deregulated FGF signaling in the development and progression of different types of tumors continue to emerge and FGFR inhibitors arise as potential targeted therapeutic agents, particularly in tumors harboring aberrant FGFR signaling. There is first evidence of a dual role of the FGF/FGFR system in both organogenesis and tumorigenesis, of which this review aims to provide an overview. FGF-1 and FGF-2 are expressed in the adrenal cortex and are the most powerful mitogens for adrenocortical cells. Physiologically, they are involved in development and maintenance of the adrenal gland and bind to a family of four tyrosine kinase receptors, among which FGFR1 and FGFR4 are the most strongly expressed in the adrenal cortex. The repeatedly proven overexpression of these two FGFRs also in adrenocortical cancer is thus likely a sign of their participation in proliferation and vascularization, though the exact downstream mechanisms are not yet elucidated. Thus, FGFRs potentially offer novel therapeutic targets also for adrenocortical carcinoma, a type of cancer resistant to conventional antimitotic agents.
Collapse
Affiliation(s)
- Mariangela Tamburello
- Division of Endocrinology, Department of Internal Medicine I, University Hospital, University of Würzburg, Würzburg, Germany
- Section of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Barbara Altieri
- Division of Endocrinology, Department of Internal Medicine I, University Hospital, University of Würzburg, Würzburg, Germany
| | - Iuliu Sbiera
- Division of Endocrinology, Department of Internal Medicine I, University Hospital, University of Würzburg, Würzburg, Germany
| | - Sandra Sigala
- Section of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Alfredo Berruti
- Oncology Unit, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia and ASST Spedali Civili di Brescia, Brescia, Italy
| | - Martin Fassnacht
- Division of Endocrinology, Department of Internal Medicine I, University Hospital, University of Würzburg, Würzburg, Germany
- Comprehenssive Cancer Center Mainfranken, University of Würzburg, Würzburg, Germany
| | - Silviu Sbiera
- Division of Endocrinology, Department of Internal Medicine I, University Hospital, University of Würzburg, Würzburg, Germany.
| |
Collapse
|
38
|
Murphy HR, Gu Y, Wu Q, Brunner J, Panisch LS, Best M, Arnold MS, Duberstein ZT, Putzig J, Carnahan J, Groth SW, Barrett ES, Qiu X, O'Connor TG. Prenatal diurnal cortisol: Normative patterns and associations with affective symptoms and stress. Psychoneuroendocrinology 2022; 143:105856. [PMID: 35797838 DOI: 10.1016/j.psyneuen.2022.105856] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 06/28/2022] [Accepted: 06/28/2022] [Indexed: 11/30/2022]
Abstract
The hypothalamic-pituitary-adrenal (HPA) axis in pregnancy has attracted considerable research attention, in part, because it may be a mechanism by which diverse prenatal exposures alter perinatal and child health outcomes. Symptoms of affective disturbance and stress are among the most-studied prenatal factors associated with HPA axis alterations, but there remains uncertainty about the nature of the association because of the limitations to, and variability in, data collection and analytic approaches. The current study capitalized on a prospective, longitudinal pregnancy cohort that examined salivary diurnal cortisol, collected at 5 time points across the day, at each trimester in a diverse sample of women. Detailed data on affective symptoms and major life events were collected at each trimester, as were data on health behaviors, medication, and socio-demographics. Results indicated modest stability of individual differences in diurnal cortisol across pregnancy, which was evident for diurnal slope (ICC = .20) and measures of total output (area under the curve, ICC = .25); substantial gestation-related increases in total cortisol output across pregnancy was also observed (p < .001). Adjusting for health behaviors, medication, and socio-demographic covariates, elevated levels of depressive symptoms and major life events were significantly (p < .05) associated with a higher morning awakening value and flatter diurnal slope, which was evident across all trimesters. In addition to the normative gestation-related changes in cortisol production, our results demonstrate selective but robust associations between psychological symptoms, stressors, and the HPA axis across gestation, and suggest both methodological and mechanistic strategies for future study.
Collapse
Affiliation(s)
- Hannah R Murphy
- Wynne Center for Family Research, University of Rochester, 601 Elmwood Ave., Rochester, NY 14642, USA; Obstetrics and Gynecology, University of Rochester, 601 Elmwood Ave., Rochester, NY 14642, USA
| | - Yu Gu
- Biostatistics and Computational Biology, University of Rochester, 265 Crittenden Blvd., Box 630, Rochester, NY 14642, USA
| | - Qiuyi Wu
- Biostatistics and Computational Biology, University of Rochester, 265 Crittenden Blvd., Box 630, Rochester, NY 14642, USA
| | - Jessica Brunner
- Obstetrics and Gynecology, University of Rochester, 601 Elmwood Ave., Rochester, NY 14642, USA
| | - Lisa S Panisch
- Wayne State University School of Social Work, 5447 Woodward Ave., Detroit, MI 48202, USA
| | - Meghan Best
- Obstetrics and Gynecology, University of Rochester, 601 Elmwood Ave., Rochester, NY 14642, USA
| | - Molly S Arnold
- Wynne Center for Family Research, University of Rochester, 601 Elmwood Ave., Rochester, NY 14642, USA; Psychology, University of Rochester, Meliora Hall, P.O. Box 270266, Rochester, NY 14627, USA
| | - Zoe T Duberstein
- Wynne Center for Family Research, University of Rochester, 601 Elmwood Ave., Rochester, NY 14642, USA; Psychology, University of Rochester, Meliora Hall, P.O. Box 270266, Rochester, NY 14627, USA
| | - Jenelle Putzig
- Pediatrics Infectious Diseases, University of Rochester, 601 Elmwood Ave., Rochester, NY 14642, USA
| | - Jennifer Carnahan
- Pediatrics Infectious Diseases, University of Rochester, 601 Elmwood Ave., Rochester, NY 14642, USA
| | - Susan W Groth
- School of Nursing, University of Rochester, 255 Crittenden Blvd., Rochester, NY, USA
| | - Emily S Barrett
- Biostatistics and Epidemiology, Rutgers School of Public Health, 683 Hoes Lane West, Piscataway, NJ 08854, USA; Environmental and Occupational Health Sciences Institute, Rutgers University, 170 Frelinghuysen Rd., Piscataway, NJ 08854, USA
| | - Xing Qiu
- Biostatistics and Computational Biology, University of Rochester, 265 Crittenden Blvd., Box 630, Rochester, NY 14642, USA
| | - Thomas G O'Connor
- Wynne Center for Family Research, University of Rochester, 601 Elmwood Ave., Rochester, NY 14642, USA; Obstetrics and Gynecology, University of Rochester, 601 Elmwood Ave., Rochester, NY 14642, USA; Psychology, University of Rochester, Meliora Hall, P.O. Box 270266, Rochester, NY 14627, USA; Neuroscience, University of Rochester, 601 Elmwood Avenue, Box 603, KMRB G.9602, Rochester, NY 14642, USA; Psychiatry, University of Rochester, 300 Crittenden Blvd., Rochester, NY 14642, USA.
| |
Collapse
|
39
|
Androgens Tend to Be Higher, but What about Altered Progesterone Metabolites in Boys and Girls with Autism? Life (Basel) 2022; 12:life12071004. [PMID: 35888093 PMCID: PMC9324026 DOI: 10.3390/life12071004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 06/21/2022] [Accepted: 07/04/2022] [Indexed: 11/17/2022] Open
Abstract
Background: Evidence exists that steroid hormones are altered in individuals with autism, especially androgens. Despite lower prevalence in girls than boys, evidence of potential alterations in progesterone metabolites is sparse, so the aim of this study was to elucidate different progesterone metabolites in affected children with autism versus healthy controls. Material and Methods: Circadian urine samples from 48 boys and 16 girls with autism spectrum disorders and a matched case−control group were analysed for progesterone metabolites by gas chromatography−mass spectrometry and normalised for creatinine excretion. Results: In boys with autism, the majority of progesterone metabolites were reduced, such as progesterone, 6a-OH-3a5b-TH-progesterone, or 20a-DH-progesterone (p < 0.01 for all). In girls with autism, a similar pattern of reduction in progesterone metabolites was detected; however, potentially due to the relatively small sample, this pattern was only detectable on the level of a trend. Discussion: As stated, androgen levels are higher in boys and girls with autism, but evidence for progesterone metabolites is much sparser. The pattern of a decrease in progesterone metabolites suggests the existence of an altered routing of steroid metabolites, probably in combination with a dysregulation of the HPAG axis. As, recently, increased CYP17A1 activity has been suggested, the stronger routing towards androgens is further implied in line with our findings of lower progesterone concentrations in boys and girls with autism than healthy controls.
Collapse
|
40
|
Hwang JK, Kang HN, Ahn JH, Lee HJ, Park HK, Kim CR. Effects of Ponderal Index on Neonatal Mortality and Morbidities in Extremely Premature Infants. J Korean Med Sci 2022; 37:e198. [PMID: 35726149 PMCID: PMC9247722 DOI: 10.3346/jkms.2022.37.e198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 05/17/2022] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND To evaluate how intrauterine stress affects extremely premature infants in terms of intrauterine growth restriction. We hypothesized that extremely premature infants with mildly-low ponderal index (MPI) would have better neonatal outcomes. METHODS We selected 2,721 subjects of 23 to 28 weeks of gestation between 2013 and 2015 from Korean Neonatal Network database. They were divided into 4 groups based on ponderal index (PI) percentile; PI ≤ 3rd as severely-low PI (SPI, n = 82), 3rd < PI ≤ 10th as MPI (n = 190), 10th < PI ≤ 90th as adequate PI (API, n = 2,179), and PI > 90th as high PI (HPI, n = 270). RESULTS The mortality in MPI and API groups was comparable (16.3% vs. 16.9%). It was significantly lower than that in the SPI and HPI groups (30.5% and 24.9%, respectively; P = 0.001). The MPI and API groups had better neonatal morbidities compared with the SPI and/or HPI groups, while the MPI group (8.2%) showed a lower incidence of severe intraventricular hemorrhage (IVH) than the other groups (SPI, 21.3%; API, 15.0%; HPI, 19.7%, respectively; P = 0.004). The MPI group had a trend of a bottom in neonatal mortality and morbidities in extremely premature infants. CONCLUSION The MPI and API groups had lower mortality, massive pulmonary hemorrhage, severe bronchopulmonary dysplasia or death, pulmonary hypertension and neonatal seizure rates than the SPI and/or HPI groups, while the MPI group showed a lower incidence of severe IVH than the other groups. We speculate that the lower incidence of neonatal morbidities and mortality in the MPI group indicating mild intrauterine stress might accelerate fetal maturation resulting in better outcomes in extremely premature infants.
Collapse
Affiliation(s)
- Jae Kyoon Hwang
- Department of Pediatrics, Hanyang University Guri Hospital, Guri, Korea
| | - Ha-Na Kang
- Department of Pediatrics, Cheongju St. Mary's Hospital, Cheongju, Korea
| | - Ja-Hye Ahn
- Department of Pediatrics, Hanyang University Seoul Hospital, Seoul, Korea
| | - Hyun Ju Lee
- Department of Pediatrics, Hanyang University Seoul Hospital, Seoul, Korea
- Department of Pediatrics, College of Medicine, Hanyang University, Seoul, Korea
| | - Hyun-Kyung Park
- Department of Pediatrics, Hanyang University Seoul Hospital, Seoul, Korea
- Department of Pediatrics, College of Medicine, Hanyang University, Seoul, Korea
| | - Chang-Ryul Kim
- Department of Pediatrics, Hanyang University Guri Hospital, Guri, Korea
- Department of Pediatrics, College of Medicine, Hanyang University, Seoul, Korea.
| |
Collapse
|
41
|
Cheng K, Seita Y, Moriwaki T, Noshiro K, Sakata Y, Hwang YS, Torigoe T, Saitou M, Tsuchiya H, Iwatani C, Hosaka M, Ohkouchi T, Watari H, Umazume T, Sasaki K. The developmental origin and the specification of the adrenal cortex in humans and cynomolgus monkeys. SCIENCE ADVANCES 2022; 8:eabn8485. [PMID: 35442744 PMCID: PMC9020778 DOI: 10.1126/sciadv.abn8485] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Development of the adrenal cortex, a vital endocrine organ, originates in the adrenogonadal primordium, a common progenitor for both the adrenocortical and gonadal lineages in rodents. In contrast, we find that in humans and cynomolgus monkeys, the adrenocortical lineage originates in a temporally and spatially distinct fashion from the gonadal lineage, arising earlier and more anteriorly within the coelomic epithelium. The adrenal primordium arises from adrenogenic coelomic epithelium via an epithelial-to-mesenchymal transition, which then progresses into the steroidogenic fetal zone via both direct and indirect routes. Notably, we find that adrenocortical and gonadal lineages exhibit distinct HOX codes, suggesting distinct anterior-posterior regionalization. Together, our assessment of the early divergence of these lineages provides a molecular framework for understanding human adrenal and gonadal disorders.
Collapse
Affiliation(s)
- Keren Cheng
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yasunari Seita
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Bell Research Center for Reproductive Health and Cancer, Nagoya 460-0003, Japan
| | - Taku Moriwaki
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kiwamu Noshiro
- Department of Obstetrics and Gynecology, Hokkaido University Graduate School of Medicine, Sapporo 060-8638, Japan
| | - Yuka Sakata
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Young Sun Hwang
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Toshihiko Torigoe
- Department of Pathology, Sapporo Medical University Graduate School of Medicine, Sapporo 060-8556, Japan
| | - Mitinori Saitou
- Department of Anatomy and Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
- Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Kyoto 606-8501, Japan
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
| | - Hideaki Tsuchiya
- Research Center for Animal Life Science, Shiga University of Medical Science, Otsu 520-2192, Japan
| | - Chizuru Iwatani
- Research Center for Animal Life Science, Shiga University of Medical Science, Otsu 520-2192, Japan
| | - Masayoshi Hosaka
- Fukuzumi Obstetrics and Gynecology Hospital, Sapporo 062-0043, Japan
| | | | - Hidemichi Watari
- Department of Obstetrics and Gynecology, Hokkaido University Graduate School of Medicine, Sapporo 060-8638, Japan
| | - Takeshi Umazume
- Department of Obstetrics and Gynecology, Hokkaido University Graduate School of Medicine, Sapporo 060-8638, Japan
| | - Kotaro Sasaki
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Corresponding author.
| |
Collapse
|
42
|
Ji B, Lei J, Xu T, Zhao M, Cai H, Qiu J, Gao Q. Effects of prenatal hypoxia on placental glucocorticoid barrier: mechanistic insight from experiments in rats. Reprod Toxicol 2022; 110:78-84. [DOI: 10.1016/j.reprotox.2022.03.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 03/25/2022] [Accepted: 03/29/2022] [Indexed: 11/25/2022]
|
43
|
Yi J, Yum SY, Kim D, Han S, Ha J, Kim J, Jung D, Jang G, Lee W, Moon J. Differences in hormone levels around parturition in Hanwoo cattle (Bos taurus coreanae) following artificial insemination and embryo transfer. Vet Med Sci 2022; 8:1258-1263. [PMID: 35193169 PMCID: PMC9122465 DOI: 10.1002/vms3.777] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Background With unique genetic traits, Hanwoo cattle (Bos taurus coreanae) are well‐adapted to the Korean environment. However, their perinatal mortality rate is 2%–3%, which imposes an economic burden. Objective Due to insufficient data on hormonal changes around parturition, the timing of parturition is often predicted subjectively; few studies have examined hormones in Hanwoo cattle. We measured the changes in various hormones around parturition, to seek an objective predictor of parturition time. Methods In 14 female Hanwoo cattle, we measured progesterone, prolactin and cortisol concentrations daily in jugular vein blood samples, beginning 6 days before parturition until 7 days after parturition. Conception was induced in five animals using artificial insemination. Nine animals received embryo transfer. Results During parturition, the concentration of progesterone decreased significantly in the embryo transfer group (n = 9) and in the total population (n = 14); it did not change significantly in the artificial insemination group (n = 5). Prolactin concentration increased on the day of parturition but did not differ significantly among the groups. Cortisol remained constant throughout the study course. Conclusion We concluded that parturition time can be predicted in Hanwoo cattle using progesterone concentration. This knowledge can reduce perinatal mortality, which would help to improve farm income and animal welfare.
Collapse
Affiliation(s)
- Junkoo Yi
- Livestock Research Institute, Yeongju-si, Gyeongsangbuk-do, Republic of Korea
| | - Soo-Young Yum
- Lartbio Co., Ltd., Gangnam-gu, Seoul, Republic of Korea
| | - Daehyun Kim
- Livestock Research Institute, Yeongju-si, Gyeongsangbuk-do, Republic of Korea
| | - Sera Han
- Lartbio Co., Ltd., Gangnam-gu, Seoul, Republic of Korea
| | - Jaejung Ha
- Livestock Research Institute, Yeongju-si, Gyeongsangbuk-do, Republic of Korea
| | - Jisu Kim
- Lartbio Co., Ltd., Gangnam-gu, Seoul, Republic of Korea
| | - Daejin Jung
- Livestock Research Institute, Yeongju-si, Gyeongsangbuk-do, Republic of Korea
| | - Goo Jang
- Lartbio Co., Ltd., Gangnam-gu, Seoul, Republic of Korea.,Laboratory of Theriogenology and Biotechnology, Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Seoul National University, Gwanak-gu, Seoul, Republic of Korea
| | - Wonyou Lee
- Lartbio Co., Ltd., Gangnam-gu, Seoul, Republic of Korea
| | - Joonho Moon
- Lartbio Co., Ltd., Gangnam-gu, Seoul, Republic of Korea
| |
Collapse
|
44
|
Wang Y, Guo B, Guo Y, Qi N, Lv Y, Ye Y, Huang Y, Long X, Chen H, Su C, Zhang L, Zhang Q, Li M, Liao J, Yan Y, Mao X, Zeng Y, Jiang J, Chen Z, Guo Y, Gao S, Cheng J, Jiang Y, Mo Z. A spatiotemporal steroidogenic regulatory network in human fetal adrenal glands and gonads. Front Endocrinol (Lausanne) 2022; 13:1036517. [PMID: 36465633 PMCID: PMC9713933 DOI: 10.3389/fendo.2022.1036517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Accepted: 10/28/2022] [Indexed: 11/18/2022] Open
Abstract
Human fetal adrenal glands produce substantial amounts of dehydroepiandrosterone (DHEA), which is one of the most important precursors of sex hormones. However, the underlying biological mechanism remains largely unknown. Herein, we sequenced human fetal adrenal glands and gonads from 7 to 14 gestational weeks (GW) via 10× Genomics single-cell transcriptome techniques, reconstructed their location information by spatial transcriptomics. Relative to gonads, adrenal glands begin to synthesize steroids early. The coordination among steroidogenic cells and multiple non-steroidogenic cells promotes adrenal cortex construction and steroid synthesis. Notably, during the window of sexual differentiation (8-12 GW), key enzyme gene expression shifts to accelerate DHEA synthesis in males and cortisol synthesis in females. Our research highlights the robustness of the action of fetal adrenal glands on gonads to modify the process of sexual differentiation.
Collapse
Affiliation(s)
- Yifu Wang
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Medical University, Guangxi, China
- Institute of Urology and Nephrology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Bingqian Guo
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Medical University, Guangxi, China
- Guangxi Collaborative Innovation Center for Biomedicine (Guangxi-Association of Southeast Asian Nations (ASEAN) Collaborative Innovation Center for Major Disease Prevention and Treatment), Guangxi Medical University, Nanning, Guangxi, China
| | - Yajie Guo
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Medical University, Guangxi, China
- Guangxi Collaborative Innovation Center for Biomedicine (Guangxi-Association of Southeast Asian Nations (ASEAN) Collaborative Innovation Center for Major Disease Prevention and Treatment), Guangxi Medical University, Nanning, Guangxi, China
| | - Nana Qi
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Medical University, Guangxi, China
- Guangxi Collaborative Innovation Center for Biomedicine (Guangxi-Association of Southeast Asian Nations (ASEAN) Collaborative Innovation Center for Major Disease Prevention and Treatment), Guangxi Medical University, Nanning, Guangxi, China
| | - Yufang Lv
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Medical University, Guangxi, China
- Institute of Urology and Nephrology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Yu Ye
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Medical University, Guangxi, China
- Institute of Urology and Nephrology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Department of Emergency, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Department of Urology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Yan Huang
- Department of Obstetrics, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Xinyang Long
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Medical University, Guangxi, China
- School of Public Health of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China
| | - Hongfei Chen
- Department of Obstetrics, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Cheng Su
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Medical University, Guangxi, China
- Institute of Urology and Nephrology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Liying Zhang
- Department of Gynecology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Qingyun Zhang
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Medical University, Guangxi, China
- Institute of Urology and Nephrology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Department of Urology, The Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Minxi Li
- Department of Gynecology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Jinling Liao
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Medical University, Guangxi, China
- Guangxi Collaborative Innovation Center for Biomedicine (Guangxi-Association of Southeast Asian Nations (ASEAN) Collaborative Innovation Center for Major Disease Prevention and Treatment), Guangxi Medical University, Nanning, Guangxi, China
| | - Yunkun Yan
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Medical University, Guangxi, China
- Institute of Urology and Nephrology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Xingning Mao
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Medical University, Guangxi, China
- Guangxi Collaborative Innovation Center for Biomedicine (Guangxi-Association of Southeast Asian Nations (ASEAN) Collaborative Innovation Center for Major Disease Prevention and Treatment), Guangxi Medical University, Nanning, Guangxi, China
| | - Yanyu Zeng
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Medical University, Guangxi, China
- Guangxi Collaborative Innovation Center for Biomedicine (Guangxi-Association of Southeast Asian Nations (ASEAN) Collaborative Innovation Center for Major Disease Prevention and Treatment), Guangxi Medical University, Nanning, Guangxi, China
| | - Jinghang Jiang
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Medical University, Guangxi, China
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Zhongyuan Chen
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Medical University, Guangxi, China
- Guangxi Collaborative Innovation Center for Biomedicine (Guangxi-Association of Southeast Asian Nations (ASEAN) Collaborative Innovation Center for Major Disease Prevention and Treatment), Guangxi Medical University, Nanning, Guangxi, China
| | - Yi Guo
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Medical University, Guangxi, China
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Shuai Gao
- College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Jiwen Cheng
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Medical University, Guangxi, China
- Institute of Urology and Nephrology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Yonghua Jiang
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Medical University, Guangxi, China
- Guangxi Collaborative Innovation Center for Biomedicine (Guangxi-Association of Southeast Asian Nations (ASEAN) Collaborative Innovation Center for Major Disease Prevention and Treatment), Guangxi Medical University, Nanning, Guangxi, China
- Department of Obstetrics, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Life Sciences Institute, Guangxi Medical University, Nanning, Guangxi, China
- *Correspondence: Zengnan Mo, ; Yonghua Jiang,
| | - Zengnan Mo
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Medical University, Guangxi, China
- Institute of Urology and Nephrology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- *Correspondence: Zengnan Mo, ; Yonghua Jiang,
| |
Collapse
|
45
|
Abdellatif AB, Fernandes-Rosa FL, Boulkroun S, Zennaro MC. Vascular and hormonal interactions in the adrenal gland. Front Endocrinol (Lausanne) 2022; 13:995228. [PMID: 36506065 PMCID: PMC9731668 DOI: 10.3389/fendo.2022.995228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 11/07/2022] [Indexed: 11/25/2022] Open
Abstract
Primary aldosteronism is the most common form of secondary arterial hypertension, due to excessive aldosterone production from the adrenal gland. Although somatic mutations have been identified in aldosterone producing adenoma, the exact mechanisms leading to increased cell proliferation and nodule formation remain to be established. One hypothesis is that changes in vascular supply to the adrenal cortex, due to phenomena of atherosclerosis or high blood pressure, may influence the morphology of the adrenal cortex, resulting in a compensatory growth and nodule formation in response to local hypoxia. In this review, we will summarize our knowledge on the mechanisms regulating adrenal cortex development and function, describe adrenal vascularization in normal and pathological conditions and address the mechanisms allowing the cross-talk between the hormonal and vascular components to allow the extreme tissue plasticity of the adrenal cortex in response to endogenous and exogenous stimuli. We will then address recent evidence suggesting a role for alterations in the vascular compartment that could eventually be involved in nodule formation and the development of primary aldosteronism.
Collapse
Affiliation(s)
| | | | - Sheerazed Boulkroun
- Université Paris Cité, PARCC, INSERM, Paris, France
- *Correspondence: Maria-Christina Zennaro, ; Sheerazed Boulkroun,
| | - Maria-Christina Zennaro
- Université Paris Cité, PARCC, INSERM, Paris, France
- Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Service de Génétique, Paris, France
- *Correspondence: Maria-Christina Zennaro, ; Sheerazed Boulkroun,
| |
Collapse
|
46
|
Pitsava G, Stratakis CA. Adrenal hyperplasias in childhood: An update. Front Endocrinol (Lausanne) 2022; 13:937793. [PMID: 35992119 PMCID: PMC9382287 DOI: 10.3389/fendo.2022.937793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 07/08/2022] [Indexed: 11/13/2022] Open
Abstract
Pediatric adrenocortical hyperplasias are rare; they usually present with Cushing syndrome (CS); of them, isolated micronodular adrenal disease and its variant, primary pigmented adrenocortical disease are the most commonly encountered. Most cases are due to defects in the cyclic AMP/protein kinase A (cAMP/PKA) pathway, although a few cases remain without an identified genetic defect. Another cause of adrenal hyperplasia in childhood is congenital adrenal hyperplasia, a group of autosomal recessive disorders that affect steroidogenic enzymes in the adrenal cortex. Clinical presentation varies and depends on the extent of the underlying enzymatic defect. The most common form is due to 21-hydroxylase deficiency; it accounts for more than 90% of the cases. In this article, we discuss the genetic etiology of adrenal hyperplasias in childhood.
Collapse
Affiliation(s)
- Georgia Pitsava
- Division of Intramural Research, Division of Population Health Research, Eunice Kennedy Shriver National Institutes of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
- Section on Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
- *Correspondence: Georgia Pitsava,
| | - Constantine A. Stratakis
- Section on Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
- Human Genetics and Precision Medicine, Institute of Molecular Biology and Biotechnology of the Foundation for Research and Technology Hellas (IMBB-FORTH), Heraklion, Greece
- ELPEN Research Institute, ELPEN, Athens, Greece
| |
Collapse
|
47
|
Holmstrom LE, Jnah AJ. Relative Adrenal Insufficiency: Crisis Averted? Neonatal Netw 2021; 40:369-376. [PMID: 34845087 DOI: 10.1891/11-t-703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/19/2020] [Indexed: 11/25/2022]
Abstract
Relative adrenal insufficiency (AI) is a disease process commonly associated with preterm birth and critical illness. Further, the incidence of AI is inversely proportional to gestational age. The incidence of AI is likely underreported; however, it is reported to occur in 150-280/1,000,000 live births worldwide. Functional development of the adrenal gland does not occur until after 30 weeks of gestation; however, advances made in neonatal care increase the survivability of infants born well before this period. Among infants with AI, the adrenal gland is transiently incapable of secreting physiologic levels of cortisol in response to stressors. Common and nonspecific signs include hypotension, poor perfusion, and dysregulation of fluid, electrolytes, and euglycemia. Recognition, diagnosis, and steroid therapy is critical, as inappropriately managed AI can lead to an adrenal crisis, shock, and death. Understanding the presentation and common risk factors for developing relative AI is crucial for quick diagnosis and timely management to prevent morbidity and mortality in this vulnerable population.
Collapse
|
48
|
Long C, Tordiffe A, Sauther M, Cuozzo F, Millette J, Ganswindt A, Scheun J. Seasonal drivers of faecal glucocorticoid metabolite concentrations in an African strepsirrhine primate, the thick-tailed greater galago ( Otolemur crassicaudatus). CONSERVATION PHYSIOLOGY 2021; 9:coab081. [PMID: 34707874 PMCID: PMC8543700 DOI: 10.1093/conphys/coab081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 05/14/2021] [Accepted: 09/30/2021] [Indexed: 06/13/2023]
Abstract
As global non-human primate populations show dramatic declines due to climate change, land transformation and other anthropogenic stressors, it has become imperative to study physiological responses to environmental change in order to understand primate adaptability and enhance species conservation strategies. We examined the effects of seasonality on faecal glucocorticoid metabolite (fGCM) concentrations of free-ranging male and female thick-tailed greater galagos (Otolemur crassicaudatus) in an Afromontane habitat. To do so, we established an enzyme immunoassay (EIA) for monitoring fGCM concentrations in the species using a biological validation. Following this, faecal samples were collected each month over the course of a year from free-ranging males and females situated in the Soutpansberg Mountains, Limpopo, South Africa. Multivariate analyses revealed lactation period was a driver of fGCM levels, whereas sex and food availability mostly influenced seasonal fGCM concentrations in the total population. Thus far, the results of this study show that drivers of fGCM levels, an indication of increased adrenocortical activity, in O. crassicaudatus are numerous and complex within the natural environment. The species may be adapted to such conditions and an extreme change to any one component may result in elevated fGCM levels. This increases our understanding of strepsirrhine primate physiology and offers initial insights into species adaptability to a challenging environment.
Collapse
Affiliation(s)
- Channen Long
- Department of Paraclinical Sciences, Faculty of Veterinary Science, University of Pretoria, Onderstepoort, 0110, South Africa
- National Zoological Garden, South African National Biodiversity Institute, Pretoria, 0001, South Africa
| | - Adrian Tordiffe
- Department of Paraclinical Sciences, Faculty of Veterinary Science, University of Pretoria, Onderstepoort, 0110, South Africa
| | - Michelle Sauther
- Department of Anthropology, University of Colorado, Boulder, CO 80309, USA
| | - Frank Cuozzo
- Lajuma Research Centre, Louis Trichardt (Makhado), 0920, South Africa
- Department of Zoology and Entomology, Mammal Research Institute, University of Pretoria, Pretoria, 0028, South Africa
| | - James Millette
- Department of Anthropology, University of Colorado, Boulder, CO 80309, USA
| | - Andre Ganswindt
- National Zoological Garden, South African National Biodiversity Institute, Pretoria, 0001, South Africa
- Department of Zoology and Entomology, Mammal Research Institute, University of Pretoria, Pretoria, 0028, South Africa
| | - Juan Scheun
- National Zoological Garden, South African National Biodiversity Institute, Pretoria, 0001, South Africa
- Department of Zoology and Entomology, Mammal Research Institute, University of Pretoria, Pretoria, 0028, South Africa
- Department of Life and Consumer Sciences, University of South Africa, Johannesburg, 1710, South Africa
| |
Collapse
|
49
|
Riedmeier M, Decarolis B, Haubitz I, Müller S, Uttinger K, Börner K, Reibetanz J, Wiegering A, Härtel C, Schlegel PG, Fassnacht M, Wiegering V. Adrenocortical Carcinoma in Childhood: A Systematic Review. Cancers (Basel) 2021; 13:5266. [PMID: 34771430 PMCID: PMC8582500 DOI: 10.3390/cancers13215266] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 10/13/2021] [Accepted: 10/14/2021] [Indexed: 01/01/2023] Open
Abstract
Adrenocortical tumors are rare in children. This systematic review summarizes the published evidence on pediatric adrenocortical carcinoma (ACC) to provide a basis for a better understanding of the disease, investigate new molecular biomarkers and therapeutic targets, and define which patients may benefit from a more aggressive therapeutic approach. We included 137 studies with 3680 ACC patients (~65% female) in our analysis. We found no randomized controlled trials, so this review mainly reflects retrospective data. Due to a specific mutation in the TP53 gene in ~80% of Brazilian patients, that cohort was analyzed separately from series from other countries. Hormone analysis was described in 2569 of the 2874 patients (89%). Most patients were diagnosed with localized disease, whereas 23% had metastasis at primary diagnosis. Only 72% of the patients achieved complete resection. In 334 children (23%), recurrent disease was reported: 81%-local recurrence, 19% (n = 65)-distant metastases at relapse. Patients < 4 years old had a different distribution of tumor stages and hormone activity and better overall survival (p < 0.001). Although therapeutic approaches are typically multimodal, no consensus is available on effective standard treatments for advanced ACC. Thus, knowledge regarding pediatric ACC is still scarce and international prospective studies are needed to implement standardized clinical stratifications and risk-adapted therapeutic strategies.
Collapse
Affiliation(s)
- Maria Riedmeier
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University Children’s Hospital, University of Wuerzburg, Josef-Schneiderstr. 2, 97080 Wuerzburg, Germany; (M.R.); (I.H.); (C.H.); (P.-G.S.)
| | - Boris Decarolis
- Department of Pediatric Oncology and Hematology, Medical Faculty, University Children’s Hospital of Cologne, 50937 Cologne, Germany;
| | - Imme Haubitz
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University Children’s Hospital, University of Wuerzburg, Josef-Schneiderstr. 2, 97080 Wuerzburg, Germany; (M.R.); (I.H.); (C.H.); (P.-G.S.)
| | - Sophie Müller
- Department of General, Visceral, Transplantation, Vascular and Pediatric Surgery, University Hospital, University of Wuerzburg, Oberduerrbacherstr. 6, 97080 Wuerzburg, Germany; (S.M.); (K.U.); (K.B.); (J.R.); (A.W.)
| | - Konstantin Uttinger
- Department of General, Visceral, Transplantation, Vascular and Pediatric Surgery, University Hospital, University of Wuerzburg, Oberduerrbacherstr. 6, 97080 Wuerzburg, Germany; (S.M.); (K.U.); (K.B.); (J.R.); (A.W.)
| | - Kevin Börner
- Department of General, Visceral, Transplantation, Vascular and Pediatric Surgery, University Hospital, University of Wuerzburg, Oberduerrbacherstr. 6, 97080 Wuerzburg, Germany; (S.M.); (K.U.); (K.B.); (J.R.); (A.W.)
| | - Joachim Reibetanz
- Department of General, Visceral, Transplantation, Vascular and Pediatric Surgery, University Hospital, University of Wuerzburg, Oberduerrbacherstr. 6, 97080 Wuerzburg, Germany; (S.M.); (K.U.); (K.B.); (J.R.); (A.W.)
| | - Armin Wiegering
- Department of General, Visceral, Transplantation, Vascular and Pediatric Surgery, University Hospital, University of Wuerzburg, Oberduerrbacherstr. 6, 97080 Wuerzburg, Germany; (S.M.); (K.U.); (K.B.); (J.R.); (A.W.)
- Department of Biochemistry and Molecular Biology, University of Wuerzburg, Am Hubland, 97074 Wuerzburg, Germany
- Comprehensive Cancer Centre Mainfranken, University of Wuerzburg Medical Centre, Josef-Schneiderstr. 2, 97080 Wuerzburg, Germany;
| | - Christoph Härtel
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University Children’s Hospital, University of Wuerzburg, Josef-Schneiderstr. 2, 97080 Wuerzburg, Germany; (M.R.); (I.H.); (C.H.); (P.-G.S.)
- Comprehensive Cancer Centre Mainfranken, University of Wuerzburg Medical Centre, Josef-Schneiderstr. 2, 97080 Wuerzburg, Germany;
| | - Paul-Gerhardt Schlegel
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University Children’s Hospital, University of Wuerzburg, Josef-Schneiderstr. 2, 97080 Wuerzburg, Germany; (M.R.); (I.H.); (C.H.); (P.-G.S.)
- Comprehensive Cancer Centre Mainfranken, University of Wuerzburg Medical Centre, Josef-Schneiderstr. 2, 97080 Wuerzburg, Germany;
| | - Martin Fassnacht
- Comprehensive Cancer Centre Mainfranken, University of Wuerzburg Medical Centre, Josef-Schneiderstr. 2, 97080 Wuerzburg, Germany;
- Department of Medicine, Division of Endocrinology and Diabetes, University Hospital, University of Wuerzburg, Oberduerrbacherstr. 6, 97080 Wuerzburg, Germany
| | - Verena Wiegering
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University Children’s Hospital, University of Wuerzburg, Josef-Schneiderstr. 2, 97080 Wuerzburg, Germany; (M.R.); (I.H.); (C.H.); (P.-G.S.)
- Comprehensive Cancer Centre Mainfranken, University of Wuerzburg Medical Centre, Josef-Schneiderstr. 2, 97080 Wuerzburg, Germany;
| |
Collapse
|
50
|
Viengchareun S, Pussard E, Castanet M, Sachs LM, Vu TA, Boileau P, Lombès M, Martinerie L. The invention of aldosterone, how the past resurfaces in pediatric endocrinology. Mol Cell Endocrinol 2021; 535:111375. [PMID: 34197901 DOI: 10.1016/j.mce.2021.111375] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 06/22/2021] [Accepted: 06/24/2021] [Indexed: 12/23/2022]
Abstract
Sodium and water homeostasis are drastically modified at birth, in mammals, by the transition from aquatic life to terrestrial life. Accumulating evidence during the past ten years underscores the central role for the mineralocorticoid signaling pathway, in the fine regulation of this equilibrium, at this critical period of development. Interestingly, regarding evolution, while the mineralocorticoid receptor is expressed in fish, the appearance of its related ligand, aldosterone, coincides with terrestrial life, as it is first detected in lungfish and amphibian. Thus, aldosterone is likely one of the main hormones regulating the transition from an aquatic environment to an air environment. This review will focus on the different actors of the mineralocorticoid signaling pathway from aldosterone secretion in the adrenal gland, to mineralocorticoid receptor expression in the kidney, summarizing their regulation and roles throughout fetal and neonatal development, in the light of evolution.
Collapse
Affiliation(s)
- Say Viengchareun
- Université Paris-Saclay, Inserm, Physiologie et Physiopathologie Endocriniennes, 94276, Le Kremlin-Bicêtre, France
| | - Eric Pussard
- Université Paris-Saclay, Inserm, Physiologie et Physiopathologie Endocriniennes, 94276, Le Kremlin-Bicêtre, France; Service de Génétique Moléculaire, Pharmacogénétique et Hormonologie, Hôpital de Bicêtre, Assistance Publique-Hôpitaux de Paris, 94275, Le Kremlin Bicêtre, France
| | - Mireille Castanet
- Normandie Univ, UNIROUEN, Inserm U1239, CHU Rouen, Department of Pediatrics, F-76000, Rouen, France
| | - Laurent M Sachs
- UMR 7221 Molecular Physiology and Adaption, Department Adaptation of Life, Centre National de La Recherche Scientifique, Muséum National d'Histoire Naturelle, Paris, France
| | - Thi An Vu
- Université Paris-Saclay, Inserm, Physiologie et Physiopathologie Endocriniennes, 94276, Le Kremlin-Bicêtre, France
| | - Pascal Boileau
- Université Paris-Saclay, Inserm, Physiologie et Physiopathologie Endocriniennes, 94276, Le Kremlin-Bicêtre, France; Department of Neonatal Pediatrics, Centre Hospitalier Intercommunal de Poissy-Saint-Germain, 10, Rue du Champ Gaillard 78300 Poissy France; Université Paris-Saclay, UVSQ, 78180, Montigny-Le-Bretonneux, France
| | - Marc Lombès
- Université Paris-Saclay, Inserm, Physiologie et Physiopathologie Endocriniennes, 94276, Le Kremlin-Bicêtre, France
| | - Laetitia Martinerie
- Université Paris-Saclay, Inserm, Physiologie et Physiopathologie Endocriniennes, 94276, Le Kremlin-Bicêtre, France; Université de Paris, 75019, Paris, France; Pediatric Endocrinology Department, AP-HP, Hôpital Universitaire Robert-Debre, 75019, Paris, France.
| |
Collapse
|