1
|
Ben Simon A, Yackobovitch-Gavan M, Uretzky A, Segev-Becker A, Perl L, Midlij E, Borger O, Brener A, Lebenthal Y. Body composition in prepubertal children with idiopathic premature adrenarche: implications for cardiometabolic health. Pediatr Res 2024:10.1038/s41390-024-03776-2. [PMID: 39695261 DOI: 10.1038/s41390-024-03776-2] [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: 04/11/2024] [Revised: 10/29/2024] [Accepted: 11/20/2024] [Indexed: 12/20/2024]
Abstract
BACKGROUND Premature adrenarche (PA) has been reported as a potential precursor of metabolic disease. We aimed to explore the interaction between body composition and cardiometabolic health of prepubertal children with PA. METHODS This observational study comprised of 87 children with PA (15 boys, 8.0 ± 1.2 years) and 87 healthy sex- and age-matched controls. Body composition was measured by bioelectrical impedance analysis. RESULTS Children with PA had a higher median BMI z-scores (P = 0.001), higher median fat percentage (P = 0.006), and lower muscle-to-fat z-scores (P = 0.050) compared to controls. There were no significant group differences in blood pressure percentiles and lipid profiles. Fat percentage of children with PA was positively correlated and the MFR z-score was negatively correlated with: BMI z-score, systolic blood pressure percentile, and atherogenic dyslipidemia index (P < 0.001 for all). There were significant correlations between parental and offspring MFR z-scores in the control group (father-child: r = 0.528, P = 0.020; mother-child: r = 0.359, P = 0.031), but none in the PA group. CONCLUSIONS Children with PA exhibited an unfavorable body composition in association with their metabolic health even before the onset of puberty. Furthermore, while healthy children displayed heritable body composition traits, children with PA did not, possibly suggesting a relatively greater role for environmental factors in the PA group. IMPACT Prepubertal children with premature adrenarche have a low muscle-to-fat ratio compared to healthy sex- and age- matched controls. The body composition of prepubertal children with premature adrenarche is associated with their systolic blood pressure percentile and atherogenic dyslipidemia index. Children with premature adrenarche and healthy weight exhibited higher adiposity compared to healthy weight controls, and those with overweight/obesity showed higher rates of elevated blood pressure and higher dyslipidemic atherogenic indices compared to overweight/obesity controls. These findings highlight the importance of early identification, intervention, and lifestyle changes for children with premature adrenarche to help prevent cardiometabolic complications.
Collapse
Affiliation(s)
- Asaf Ben Simon
- The Institute of Pediatric Endocrinology, Diabetes and Metabolism, Dana-Dwek Children's Hospital, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel affiliated to the Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | - Michal Yackobovitch-Gavan
- Department of Epidemiology and Preventive Medicine, School of Public Health, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Adi Uretzky
- The Institute of Pediatric Endocrinology, Diabetes and Metabolism, Dana-Dwek Children's Hospital, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel affiliated to the Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Anat Segev-Becker
- The Institute of Pediatric Endocrinology, Diabetes and Metabolism, Dana-Dwek Children's Hospital, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel affiliated to the Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Liat Perl
- The Institute of Pediatric Endocrinology, Diabetes and Metabolism, Dana-Dwek Children's Hospital, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel affiliated to the Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Eyas Midlij
- The Institute of Pediatric Endocrinology, Diabetes and Metabolism, Dana-Dwek Children's Hospital, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel affiliated to the Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ophir Borger
- The Institute of Pediatric Endocrinology, Diabetes and Metabolism, Dana-Dwek Children's Hospital, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel affiliated to the Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- The Nutrition and Dietetics Unit, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Avivit Brener
- The Institute of Pediatric Endocrinology, Diabetes and Metabolism, Dana-Dwek Children's Hospital, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel affiliated to the Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yael Lebenthal
- The Institute of Pediatric Endocrinology, Diabetes and Metabolism, Dana-Dwek Children's Hospital, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel affiliated to the Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
2
|
Saleh FL, Starkman H, Furness A, Pfeifer SM, Kives S. Polycystic Ovary Syndrome in Adolescents. Obstet Gynecol Clin North Am 2024; 51:679-693. [PMID: 39510738 DOI: 10.1016/j.ogc.2024.08.005] [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] [Indexed: 11/15/2024]
Abstract
Polycystic ovary syndrome (PCOS) is a heterogeneous endocrinologic disorder that is characterized by oligomenorrhea or amenorrhea and signs of hyperandrogenism. The cause of PCOS remains unknown, but the syndrome is associated with insulin resistance that in turn leads to hyperandrogenism. An accurate and prompt diagnosis of PCOS is important to understanding an adolescent's risk. In adolescents, both oligoovulation and hyperandrogenism are needed for the diagnosis of PCOS and ultrasound is not recommended. Lifelong health consequences of PCOS are significant and include obesity, diabetes, metabolic syndrome, and anovulatory infertility. The symptoms of PCOS can be disturbing to an adolescent girl.
Collapse
Affiliation(s)
- Farrah L Saleh
- Division of Reproductive Endocrinology and Infertility, Weill Medical College of Cornell University, New York-Presbyterian Hospital, New York, NY, USA
| | | | | | - Samantha M Pfeifer
- Division of Reproductive Endocrinology and Infertility, Weill Medical College of Cornell University, New York-Presbyterian Hospital, New York, NY, USA
| | - Sari Kives
- Division of Pediatric Gynecology, University of Toronto, Hospital for Sick Children, Toronto, Ontario, Canada.
| |
Collapse
|
3
|
Bai J, Wang Z, Yang M, Xiang J, Liu Z. Disrupting CENP-N mediated SEPT9 methylation as a strategy to inhibit aerobic glycolysis and liver metastasis in colorectal cancer. Clin Exp Metastasis 2024; 41:971-988. [PMID: 39424682 DOI: 10.1007/s10585-024-10316-z] [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: 07/12/2024] [Accepted: 09/23/2024] [Indexed: 10/21/2024]
Abstract
Colorectal cancer (CRC) is a prevalent malignancy with a high mortality rate, primarily due to liver metastasis. This study explores the role of centromere protein N (CENP-N) in mediating the methylation of septin 9 (SEPT9) and its subsequent effects on aerobic glycolysis and liver metastasis in CRC. We employed in vitro and in vivo experiments, including single-cell RNA sequencing, methylation-specific PCR (MSP), ChIP assays, and various functional assays to assess the impact of CENP-N and SEPT9 on CRC cell proliferation, migration, invasion, and metabolic reprogramming. Our data reveal that CENP-N directly interacts with SEPT9, enhancing its methylation at specific lysine residues. This modification significantly upregulates key glycolytic enzymes, thereby promoting aerobic glycolysis, CRC cell proliferation, and migration. In vivo studies further demonstrate that the CENP-N/SEPT9 axis facilitates liver metastasis of CRC, as confirmed by fluorescence imaging and histological analysis. This study identifies a novel pathway where CENP-N-mediated methylation of SEPT9 drives metabolic reprogramming and metastasis in CRC. These findings suggest potential therapeutic targets for inhibiting CRC progression and liver metastasis, offering new insights into CRC pathogenesis.
Collapse
Affiliation(s)
- Junge Bai
- Department of Biochemistry and Molecular Biology, Harbin Medical University, 157 Health Road, Nangang District, Harbin, Heilongjiang, 150081, China
| | - Zhexue Wang
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Panjiayuannanli, Chaoyang District, Beijing, 100021, China
| | - Ming Yang
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Panjiayuannanli, Chaoyang District, Beijing, 100021, China
| | - Jun Xiang
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Panjiayuannanli, Chaoyang District, Beijing, 100021, China
| | - Zheng Liu
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Panjiayuannanli, Chaoyang District, Beijing, 100021, China.
| |
Collapse
|
4
|
Huo S, Li B, Du J, Zhang X, Song M, Li Y. Neurotoxic effects of perinatal exposure to Bisphenol F on offspring mice. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 362:124932. [PMID: 39260543 DOI: 10.1016/j.envpol.2024.124932] [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: 05/28/2024] [Revised: 07/26/2024] [Accepted: 09/07/2024] [Indexed: 09/13/2024]
Abstract
Bisphenols constitute a diverse group of endocrine-disrupting chemicals (EDCs) that impact hormone activity. Bisphenol F (BPF) is commonly used as a substitute for Bisphenol A (BPA). The disruption of the immune system by EDCs during embryonic brain development has been suggested as a plausible factor to neurodevelopmental disorders. We investigated the neurotoxic effects of perinatal exposure to BPF on offspring mice. Female mice were exposed to BPF through their drinking water on day 0.5 of pregnancy, and this exposure continued until the offspring mice were weaned, throughout the perinatal period. Our findings revealed that exposure to BPF hindered both growth and neurodevelopment in offspring mice, with a more pronounced effect observed in males. Additionally, transcriptomic analysis was conducted on the brains of male offspring mice exposed to high doses of BPF. In summary, our study indicates that perinatal exposure to BPF results in neurodevelopmental impairments in male offspring mice, linked to oxidative stress, inflammatory responses, and immune dysregulation. These findings underscore that BPF may not be a safe substitute for BPA. Thus, there is a pressing need to reevaluate the current regulation of BPF.
Collapse
Affiliation(s)
- Siming Huo
- Key Laboratory of the Provincial Education, Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Bo Li
- Key Laboratory of the Provincial Education, Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Jiayu Du
- Key Laboratory of the Provincial Education, Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Xuliang Zhang
- Key Laboratory of the Provincial Education, Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Miao Song
- Key Laboratory of the Provincial Education, Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Yanfei Li
- Key Laboratory of the Provincial Education, Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China.
| |
Collapse
|
5
|
Sudman M, Stöger R, Bentley GR, Melamed P. Association of childhood dehydroepiandrosterone sulfate concentration, pubertal development, and DNA methylation at puberty-related genes. Eur J Endocrinol 2024; 191:623-635. [PMID: 39670713 DOI: 10.1093/ejendo/lvae156] [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: 07/10/2024] [Revised: 10/27/2024] [Accepted: 12/11/2024] [Indexed: 12/14/2024]
Abstract
OBJECTIVE High concentrations of dehydroepiandrosterone sulfate (DHEAS) often precede premature puberty and sometimes polycystic ovary syndrome (PCOS). We hypothesized that the underlying mechanisms might involve DNA methylation. As an indicator of the downstream effects of DHEAS, we looked for associations between prepubertal DHEAS concentration, pubertal progression, and DNA methylation at puberty-related genes in blood cells. DESIGN Blood methylome and DHEAS concentration at 7.5 and 8.5 years, respectively, were analyzed in 91 boys and 82 girls. Pubertal development data were collected between 8.1 and 17 years (all from UK birth cohort, Avon Longitudinal Study of Parents and Children [ALSPAC]). METHODS Correlation between DHEAS and pubertal measurements was assessed by Spearman's correlation. DHEAS association with methylation at individual CpGs or regions was evaluated by linear regression, and nearby genes examined by enrichment analysis and intersection with known puberty-related genes. RESULTS Boys and girls with higher childhood DHEAS concentrations had more advanced pubic hair growth throughout puberty; girls also had advanced breast development, earlier menarche, and longer menstrual cycles. DHEAS concentration was associated with methylation at individual CpGs near several puberty-related genes. In boys, 14 genes near CpG islands with DHEAS-associated methylation were detected, and in girls, there were 9 which included LHCGR and SRD5A2; FGFR1 and FTO were detected in both sexes. CONCLUSIONS The association between DHEAS and pubertal development, as reported previously, suggests a physiological connection. Our novel findings showing that DHEAS concentration correlates negatively and linearly with DNA methylation levels at regulatory regions of key puberty-related genes, provide a mechanism for such a functional relationship.
Collapse
Affiliation(s)
- Maya Sudman
- Faculty of Biology, Technion-Israel Institute of Technology, Haifa 32000, Israel
| | - Reinhard Stöger
- School of Biosciences, University of Nottingham, Nottingham LE12 5RD, United Kingdom
| | - Gillian R Bentley
- Department of Anthropology, Durham University, Durham DH1 3LE, United Kingdom
| | - Philippa Melamed
- Faculty of Biology, Technion-Israel Institute of Technology, Haifa 32000, Israel
| |
Collapse
|
6
|
Sessa L, Rotunno G, Sodero G, Pane LC, Rendeli C, Maresca G, Rigante D, Cipolla C. Predictive value of transabdominal pelvic ultrasonography for the diagnosis of central precocious puberty: A single-center observational retrospective study. Clin Pediatr Endocrinol 2024; 33:199-206. [PMID: 39359668 PMCID: PMC11442698 DOI: 10.1297/cpe.2024-0025] [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: 03/04/2024] [Accepted: 06/26/2024] [Indexed: 10/04/2024] Open
Abstract
This single-center, observational, retrospective study aimed to evaluate the diagnostic accuracy of pelvic ultrasonographic parameters for detecting central precocious puberty (CPP) in a cohort of female pediatric patients undergoing gonadotropin stimulation tests. The study population consisted of 47 female patients with a suspicion of CPP. Thirty four out of 47 patients (72.34%) were subsequently diagnosed with CPP based on the current laboratory diagnostic criteria (LH peak > 5 IU/L). The ultrasonography results of 39 out of 47 patients (82.97%) were categorized as pubertal, while 31 out of 34 participants (91.17%) in the CPP group exhibited pubertal ultrasonography features. In 13 out of 47 girls (27.65%), a CPP diagnosis was ruled out; however, among these 13 patients, eight exhibited pubertal ultrasonography features suspicious of CPP. We observed a robust concordance between the GnRH test results indicative of pubertal activation and the presence of pubertal pelvic ultrasonographic features in 31 out of 34 children (91.17%). A significant correlation was found between ovarian volume and basal LH and LH/ FSH ratio, and also for basal LH, LH peak, LH/FSH ratio and peak LH/FSH ratio (p = 0.026, p = 0.011, p = 0.031, p = 0.004, respectively). Pelvic ultrasonography had a sensitivity of 91.17% and a specificity of 38.46% in differentiating CPP from premature thelarche.
Collapse
Affiliation(s)
- Linda Sessa
- Department of Life Sciences and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Giulia Rotunno
- Department of Life Sciences and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Giorgio Sodero
- Department of Life Sciences and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Lucia Celeste Pane
- Department of Life Sciences and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Claudia Rendeli
- Spina Bifida Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Università Cattolica Sacro Cuore, Rome, Italy
| | - Giulia Maresca
- Spina Bifida Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Donato Rigante
- Department of Life Sciences and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Università Cattolica Sacro Cuore, Rome, Italy
| | - Clelia Cipolla
- Department of Life Sciences and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| |
Collapse
|
7
|
Liimatta J, du Toit T, Voegel CD, Jääskeläinen J, Lakka TA, Flück CE. Multiple androgen pathways contribute to the steroid signature of adrenarche. Mol Cell Endocrinol 2024; 592:112293. [PMID: 38838762 DOI: 10.1016/j.mce.2024.112293] [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: 01/26/2024] [Revised: 05/06/2024] [Accepted: 06/01/2024] [Indexed: 06/07/2024]
Abstract
CONTEXT Adrenarche is a normal developmental event in mid-childhood characterized by increasing adrenal androgen secretion. The role of the classic androgen pathway has been well described in adrenarche, but the role of newer active androgens and additional androgen pathways is less clear. OBJECTIVE To study the contribution of novel androgens and related steroid biosynthesis pathways to the development of adrenarche, and to identify additional steroid biomarkers of adrenarche. DESIGN A longitudinal study of children aged 6-8 years at baseline, followed up at ages 8-10 and 14-16 years. A total of 34 children (20 girls) with clinical and/or biochemical signs of adrenarche (cases) and 24 children (11 girls) without these signs (controls) at age 8-10 years were included. Serum steroid profiling was performed by liquid chromatography high-resolution mass spectrometry. MAIN OUTCOME MEASURES Thirty-two steroids compartmentalized in progestagens, gluco- and mineralocorticoid pathways, and four androgen related pathways, including the classic, backdoor, 11-oxy, and 11-oxy backdoor pathways. RESULTS The classic and 11-oxy androgen pathways were more active, and serum concentrations of main androgens in the classic (dehydroepiandrosterone, dehydroepiandrosterone sulfate, androstenedione and androsterone) and 11-oxy (11β-hydroxyandrostenedione, 11β-hydroxytestosterone, 11-ketoandrostenedione, and 11-ketotestosterone) pathways were higher in cases at ages 6-8 and 8-10 years. Pregnenolone concentrations at adrenarchal age (8-10 years) and cortisol concentrations at adolescence (14-16 years) were higher in cases. 11β-hydroxyandrosterone and 11-ketoandrosterone tended to be higher in cases with clinical signs compared to cases who had only biochemical evidence of adrenarche, albeit they were detected at low levels. In biomarker analyses, calculated steroid ratios with cortisol, cortisone, or 11-deoxycortisone as dividers were better classifiers for adrenarche than single steroids. Among these ratios, androstenedione/cortisone was the best. CONCLUSIONS The classic and 11-oxy androgen pathways are active in adrenarche. Children with earlier timing of adrenarche have higher serum cortisol levels at late pubertal age, suggesting that early adrenarche might have long-term effects on adrenal steroidogenesis by increasing the activity of the glucocorticoid pathway. Future studies should employ comprehensive steroid profiling to define novel classifiers and biomarkers for adrenarche and premature adrenarche.
Collapse
Affiliation(s)
- Jani Liimatta
- Pediatric Endocrinology, Diabetology, and Metabolism, Inselspital, Bern University Hospital, Bern, Switzerland; Department of BioMedical Research (DBMR), University of Bern, Bern, Switzerland; Kuopio Pediatric Research Unit (KuPRU), University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland.
| | - Therina du Toit
- Pediatric Endocrinology, Diabetology, and Metabolism, Inselspital, Bern University Hospital, Bern, Switzerland; Department of BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Clarissa D Voegel
- Department of BioMedical Research (DBMR), University of Bern, Bern, Switzerland; Department of Nephrology and Hypertension, Bern University Hospital, Bern, Switzerland
| | - Jarmo Jääskeläinen
- Kuopio Pediatric Research Unit (KuPRU), University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Timo A Lakka
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio, Finland; Department of Clinical Physiology and Nuclear Medicine, Kuopio University Hospital, Kuopio, Finland; Foundation for Research in Health Exercise and Nutrition, Kuopio Research Institute of Exercise Medicine, Kuopio, Finland
| | - Christa E Flück
- Pediatric Endocrinology, Diabetology, and Metabolism, Inselspital, Bern University Hospital, Bern, Switzerland; Department of BioMedical Research (DBMR), University of Bern, Bern, Switzerland.
| |
Collapse
|
8
|
Lin AE, Scimone ER, Thom RP, Balaguru D, Kinane TB, Moschovis PP, Cohen MS, Tan W, Hague CD, Dannheim K, Levitsky LL, Lilly E, DiGiacomo DV, Masse KM, Kadzielski SM, Zar-Kessler CA, Ginns LC, Neumeyer AM, Colvin MK, Elder JS, Learn CP, Mou H, Weagle KM, Buch KA, Butler WE, Alhadid K, Musolino PL, Sultana S, Bandyopadhyay D, Rapalino O, Peacock ZS, Chou EL, Heidary G, Dorfman AT, Morris SA, Bergin JD, Rayment JH, Schimmenti LA, Lindsay ME. Emergence of the natural history of Myhre syndrome: 47 patients evaluated in the Massachusetts General Hospital Myhre Syndrome Clinic (2016-2023). Am J Med Genet A 2024; 194:e63638. [PMID: 38779990 PMCID: PMC11586855 DOI: 10.1002/ajmg.a.63638] [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: 03/03/2024] [Revised: 04/03/2024] [Accepted: 04/11/2024] [Indexed: 05/25/2024]
Abstract
Myhre syndrome is an increasingly diagnosed ultrarare condition caused by recurrent germline autosomal dominant de novo variants in SMAD4. Detailed multispecialty evaluations performed at the Massachusetts General Hospital (MGH) Myhre Syndrome Clinic (2016-2023) and by collaborating specialists have facilitated deep phenotyping, genotyping and natural history analysis. Of 47 patients (four previously reported), most (81%) patients returned to MGH at least once. For patients followed for at least 5 years, symptom progression was observed in all. 55% were female and 9% were older than 18 years at diagnosis. Pathogenic variants in SMAD4 involved protein residues p.Ile500Val (49%), p.Ile500Thr (11%), p.Ile500Leu (2%), and p.Arg496Cys (38%). Individuals with the SMAD4 variant p.Arg496Cys were less likely to have hearing loss, growth restriction, and aortic hypoplasia than the other variant groups. Those with the p.Ile500Thr variant had moderate/severe aortic hypoplasia in three patients (60%), however, the small number (n = 5) prevented statistical comparison with the other variants. Two deaths reported in this cohort involved complex cardiovascular disease and airway stenosis, respectively. We provide a foundation for ongoing natural history studies and emphasize the need for evidence-based guidelines in anticipation of disease-specific therapies.
Collapse
Affiliation(s)
- Angela E. Lin
- Medical Genetics and Metabolism, Department of Pediatrics, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Eleanor R. Scimone
- Medical Genetics and Metabolism, Department of Pediatrics, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Robyn P. Thom
- Lurie Center for Autism, Mass General for Children, Boston, Massachusetts, USA
- Department of Psychiatry, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Duraisamy Balaguru
- Pediatric Cardiology, Department of Pediatrics, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - T. Bernard Kinane
- Pediatric Pulmonary and Sleep Medicine, Department of Pediatrics, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Peter P. Moschovis
- Pediatric Pulmonary and Sleep Medicine, Department of Pediatrics, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Michael S. Cohen
- Pediatric Otorhinolaryngology, Massachusetts Eye and Ear, Boston, Massachusetts, USA
| | - Weizhen Tan
- Pediatric Nephrology, Department of Pediatrics, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Cole D. Hague
- Department of Psychiatry, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Katelyn Dannheim
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Lynne L. Levitsky
- Pediatric Endocrinology, Department of Pediatrics, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Evelyn Lilly
- Department of Dermatology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Daniel V. DiGiacomo
- Division of Rheumatology, Allergy and Immunology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Kara M. Masse
- Department of Physical Therapy, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Sarah M. Kadzielski
- Lurie Center for Autism, Mass General for Children, Boston, Massachusetts, USA
- Pediatric Gastroenterology, Department of Pediatrics, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Claire A. Zar-Kessler
- Pediatric Gastroenterology, Department of Pediatrics, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Leo C. Ginns
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Ann M. Neumeyer
- Lurie Center for Autism, Mass General for Children, Boston, Massachusetts, USA
- Pediatric Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Mary K. Colvin
- Department of Psychiatry, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Jack S. Elder
- Division of Pediatric Urology, Department of Urology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Christopher P. Learn
- Division of Cardiology, Department of Medicine, Corrigan Minehan Heart Center, Adult Congenital Heart Disease, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Hongmei Mou
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Kathryn M. Weagle
- Department of Child Life, Pediatric Imaging Program, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Karen A. Buch
- Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - William E. Butler
- Department of Neurosurgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Kenda Alhadid
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Patricia L. Musolino
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Sadia Sultana
- Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | | | - Otto Rapalino
- Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Zachary S. Peacock
- Oral and Maxillofacial Surgery, Massachusetts General Hospital and Harvard School of Dental Medicine, Boston, Massachusetts, USA
| | - Elizabeth L. Chou
- Division of Vascular and Endovascular Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Gena Heidary
- Department of Ophthalmology, Boston Children’s Hospital, Boston, Massachusetts, USA
| | - Aaron T. Dorfman
- Division of Cardiology, Department of Pediatrics, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Shaine A. Morris
- Division of Cardiology, Department of Pediatrics, Texas Children’s Hospital and Baylor College of Medicine, Houston, Texas, USA
| | - James D. Bergin
- Division of Cardiology, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Jonathan H. Rayment
- Respiratory Medicine, Department of Pediatrics, British Columbia Children’s Hospital and University of British Columbia, Vancouver, British Columbia, Canada
| | - Lisa A. Schimmenti
- Department of Clinical Genomics, Mayo Clinic, Rochester, Minnesota, USA
- Department of Otorhinolaryngology, Head and Neck Surgery, Mayo Clinic, Rochester, Minnesota, USA
- Department of Ophthalmology, Mayo Clinic, Rochester, Minnesota, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA
| | - Mark E. Lindsay
- Pediatric Cardiology, Department of Pediatrics, Massachusetts General Hospital, Boston, Massachusetts, USA
- Pediatric Cardiology, Department of Pediatrics, Cardiovascular Genetics Program, Cardiology Division, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | | |
Collapse
|
9
|
Jang S, Ryder JR, Kelly AS, Bomberg EM. Association between endogenous sex hormones and adiposity in youth across a weight status spectrum. Pediatr Res 2024:10.1038/s41390-024-03578-6. [PMID: 39294241 DOI: 10.1038/s41390-024-03578-6] [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: 09/19/2023] [Revised: 07/29/2024] [Accepted: 09/05/2024] [Indexed: 09/20/2024]
Abstract
BACKGROUND While endogenous sex hormones (e.g., testosterone, estradiol) are important factors regulating adipose tissue distribution, studies evaluating such relationships in youth across a wide weight status spectrum are limited. METHODS We performed a cross-sectional analysis of 8-21-year-old youth. Multiple linear regression models were used to evaluate associations between sex hormones and adiposity measures (android/gynoid ratio (A/G), total fat mass (FM), visceral adipose tissue (VAT), waist circumference (WC)) and total lean mass (LM), adjusting for pubertal stage and race/ethnicity, and stratified by sex and weight status. RESULTS Among 342 youth, the mean age was 13.0 ± 2.8 years old (52.6% female; 38.9% normal weight [NW]; 27.8% overweight/obesity [OW/OB]; 33.3% severe obesity [SO]). Testosterone was positively associated with LM among males with NW (1462 g, 95% CI: 255-2668 g) and OW/OB (3792 g, 95% CI: 1244-6340 g), with A/G and WC among males with NW (0.01, 95% CI: 0-0.2 and 10 mm, 95% CI: 4-16 mm, respectively), and negatively associated with WC among males with SO (-43 mm, -81 to -5 mm). Estradiol was positively associated with A/G, FM, and WC among males with SO, and VAT in females with NW. CONCLUSION Our findings showed that sex hormones were associated with adipose tissue deposition in youth across the weight spectrum. IMPACT STATEMENT Sex hormones (e.g., testosterone, estradiol) are associated with various adiposity measures among male and female children and adolescents across a weight status spectrum. We evaluated associations between sex hormones and various adiposity measures among 8-21-year-olds across a weight status spectrum (normal weight, overweight/class 1 obesity, class 2-3 obesity). We found that estradiol was positively associated with total fat mass, android/gynoid ratio, and waist circumference in males with class 2-3 obesity, and testosterone was positively associated with lean mass in males with normal weight and overweight/class 1 obesity. Sex hormones may influence, or may be influenced by, adiposity in youth.
Collapse
Affiliation(s)
- Subin Jang
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, USA.
- Center for Pediatric Obesity Medicine, Department of Pediatrics, University of Minnesota Medical School, Minneapolis, MN, USA.
| | - Justin R Ryder
- Department of Surgery, Lurie Children's Hospital, Chicago, IL, USA
| | - Aaron S Kelly
- Center for Pediatric Obesity Medicine, Department of Pediatrics, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Eric M Bomberg
- Center for Pediatric Obesity Medicine, Department of Pediatrics, University of Minnesota Medical School, Minneapolis, MN, USA
- Division of Endocrinology, Department of Pediatrics, University of Minnesota Medical School, Minneapolis, MN, USA
| |
Collapse
|
10
|
Cuda S. Special considerations for the child with obesity: An Obesity Medicine Association (OMA) clinical practice statement (CPS) 2024. OBESITY PILLARS 2024; 11:100113. [PMID: 38953014 PMCID: PMC11216014 DOI: 10.1016/j.obpill.2024.100113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 05/21/2024] [Accepted: 05/22/2024] [Indexed: 07/03/2024]
Abstract
Background This Obesity Medicine Association (OMA) Clinical Practice Statement (CPS) details assessment and management of the child with overweight or obesity. The term "child" is defined as the child between 2 and 12 years of age. Because children are in a continual state of development during this age range, we will specify when our discussion applies to subsets within this age range. For the purposes of this CPS, we will use the following definitions: overweight in the child is a body mass index (BMI) ≥ 85th and <95th percentile, obesity in the child is a BMI ≥95th percentile, and severe obesity is a BMI ≥120% of the 95th percentile. Methods The information and clinical guidance in this OMA Clinical Practice Statement are based on scientific evidence, supported by medical literature, and derived from the clinical perspectives of the authors. Results This OMA Clinical Practice Statement provides an overview of prevalence of disease in this population, reviews precocious puberty in the child with obesity, discusses the current and evolving landscape of the use of anti-obesity medications in children in this age range, discusses the child with obesity and special health care needs, and reviews hypothalamic obesity in the child. Conclusions This OMA Clinical Practice Statement on the child with obesity is an evidence based review of the literature and an overview of current recommendations. This CPS is intended to provide a roadmap to the improvement of the health of children with obesity, especially those with metabolic, physiological, psychological complications and/or special healthcare needs. This CPS addresses treatment recommendations and is designed to help the clinician with clinical decision making.
Collapse
Affiliation(s)
- Suzanne Cuda
- Alamo City Healthy Kids and Families, 1919 Oakwell Farms Parkway, Ste 145, San Antonio, TX, 78218, USA
| |
Collapse
|
11
|
Hashemipour M, Saleh R. The spectrum of clinical, hormonal findings in children with congenital adrenal hyperplasia in Isfahan province; a 20-year review. Horm Mol Biol Clin Investig 2024; 0:hmbci-2022-0116. [PMID: 39138818 DOI: 10.1515/hmbci-2022-0116] [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: 12/31/2022] [Accepted: 07/09/2024] [Indexed: 08/15/2024]
Abstract
OBJECTIVES Congenital adrenal hyperplasia is an autosomal recessive disorder caused by complete or partial defects in one of the several steroidogenic enzymes involved in synthesizing of cortisol from cholesterol in the adrenal gland. Prompt and proper treatment of the disease would reduce symptoms and the level of androgens in patients. The present study aimed to evaluate the demographic characteristics and clinical findings of these patients. METHODS This retrospective investigation was conducted in 146 patients with congenital adrenal hyperplasia participated. Their clinical and paraclinical findings were accurately recorded in the file and extracted from the records. RESULTS Among all 146 patients, 119(81.5 %) was 21-OH Deficiency type;11-OH Deficiency type was 13(8.9 %), 10(6.8 %) was 3β-HSD type, StAR was 2(1.4 %) and 17 alpha(α)-hydroxylase Deficiency was 2(1.4 %). The mean age of disease onset in these patients was 2.45 ± 1.16 years. Macropenis was the most frequent clinical finding in 39 cases of 64 boys (60.9 %), and Clitoromgaly was the most clinical presentation in 40 cases of 82 girls (48.7 %). The levels of testosterone, dehydroepiandrosterone sulfate, and 17-OHP significantly decreased in the last visit compared to the initial diagnosis. CONCLUSIONS Based on the clinical findings in every infant or child with ambiguous genitalia, macropenis, clitoromegaly, hirsutism, and premature pubarche, we should consider congenital adrenal hyperplasia. Prompt and proper treatment and disease control would reduce symptoms and the level of androgens in patients.
Collapse
Affiliation(s)
- Mahin Hashemipour
- Metabolic Liver Disease Research Center, 48455 Isfahan University of Medical Sciences , Isfahan, Iran
- Isfahan Endocrine & Metabolism Research Center, 48455 Isfahan University of Medical Sciences , Isfahan, Iran
| | - Rana Saleh
- Isfahan Endocrine & Metabolism Research Center, 48455 Isfahan University of Medical Sciences , Isfahan, Iran
- Child Growth and Development Research Center, 48455 Isfahan University of Medical Sciences , Isfahan, Iran
| |
Collapse
|
12
|
Zhang ZZ, Peckins MK, Beal SJ, Schnabel DJ, Shenk CE, Dorn LD. The Impact of Time Since Menarche for Depressive and Anxiety Symptom Severity in Adolescence and Young Adulthood. J Adolesc Health 2024; 75:281-287. [PMID: 38739057 DOI: 10.1016/j.jadohealth.2024.03.010] [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: 09/01/2023] [Revised: 02/15/2024] [Accepted: 03/19/2024] [Indexed: 05/14/2024]
Abstract
PURPOSE The study mapped depressive and anxiety symptom trajectories throughout adolescence and early adulthood, arrayed by time since menarche, a novel indicator of pubertal change and examined the effect of age of menarche and pubertal timing, more frequently used variables, on depressive and anxiety symptom severity trajectories. METHODS Secondary analysis of a cross-sequential prospective longitudinal investigation included a community sample of 262 US, adolescent females. Participants were enrolled in age cohorts of 11, 13, 15, and 17 years. Four annual waves of data were collected. Self-report of age at menarche was categorized into pubertal timing categories. A novel measure "time since menarche" (chronological age at each wave minus age at menarche), was measured along with depressive and anxiety symptom severity. Two-piece growth curve modeling with landmark registration examined depressive and anxiety symptom severity trajectories according to time since menarche. RESULTS There was no change (p > .05) in depression and anxiety symptom severity before menarche; however, in the years leading away from menarche, depression and anxiety symptom severity decreased (p < .05). Age at menarche was not associated with change in depressive and anxiety symptom severity (p > .05) and there were no moderating effects of pubertal timing. DISCUSSION Depressive and anxiety symptoms decrease in the years leading away from menarche, suggesting puberty-related psychopathology may be transitory in some individuals. Time since menarche may be a clinically relevant indicator of psychological functioning in pubescent adolescent females. Future studies should examine this variable in larger samples, including more adolescents in the earlier stages of puberty.
Collapse
Affiliation(s)
- Zhenyu Z Zhang
- Department of Psychology, The Pennsylvania State University, University Park, Pennsylvania
| | | | - Sarah J Beal
- Behavioral Medicine and Clinical Psychology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - David J Schnabel
- Ross and Carol Nese College of Nursing, The Pennsylvania State University, University Park, Pennsylvania
| | - Chad E Shenk
- Department of Human Development and Family Studies, The Pennsylvania State University, University Park, Pennsylvania; The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Lorah D Dorn
- Ross and Carol Nese College of Nursing, The Pennsylvania State University, University Park, Pennsylvania.
| |
Collapse
|
13
|
Wagner F, Zeidler R, Ceglarek U, Kiess W, Kratzsch J, Gaudl A, Biemann R, Vogel M. Obesity Is Associated with Increased 11-Oxyandrogen Serum Concentrations during Puberty. Horm Res Paediatr 2024:1-10. [PMID: 39038449 DOI: 10.1159/000540433] [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: 03/20/2024] [Accepted: 07/16/2024] [Indexed: 07/24/2024] Open
Abstract
INTRODUCTION While the influence of various factors on classical androgen synthesis in children and adolescents and its impact on puberty has been widely investigated, there appear to be gaps and contradictory findings regarding the association of overweight and obesity with the synthesis of adrenal-derived 11-oxygenated androgen (11-OA) serum levels. With this study, we aimed to examine how overweight and obesity affect 11-OA serum levels during puberty in a large cohort of children and adolescents. METHODS Our cohort comprised 1,054 healthy children aged 6-19 years providing serum samples at a total of 1,734 visits. Liquid chromatography-tandem mass spectrometry was used to quantify 11-ketotestosterone (11-KT), 11-ketoandrostendione (11-KA4), 11-β-hydroxytestosterone (11-OHT), 11-β-hydroxyandrostendione (11-OHA4), testosterone, androstenedione, and DHEAS. In addition, we assessed BMI-SDSs, skinfold thicknesses, and Tanner stages. The significance level α was set to α = 0.05. RESULTS Increases in 11-KT, 11-KA4, 11-OHT, and 11-OHA4 levels were observed in boys and girls during puberty. 11-KT (β = 0.2, p < 0.001), 11-KA4 (β = 0.16, p < 0.001), and 11-OHA4 (β = 0.12, p = 0.003) were positively correlated with BMI in boys aged 13 years and under. 11-KT (β = 0.1, p = 0.047) was positively correlated with BMI in girls aged 11 years and under. 11-OHT was positively correlated with BMI independent of age (boys 13 years and under: β = 0.17, p < 0.001; over 13 years: β = 0.14, p = 0.001; girls 11 years and under: β = 0.17, p < 0.001; over 11 years: β = 0.18, p < 0.001). CONCLUSION We found increasing 11-OA serum levels throughout all Tanner stages. 11-OAs were observed to be associated with BMI and skinfold thickness, suggesting that overweight and obesity may be associated with pubertal alterations in 11-OA serum levels.
Collapse
Affiliation(s)
- Friederike Wagner
- Hospital for Children and Adolescents, Center for Pediatric Research, Leipzig University Medical Center, Leipzig, Germany
- LIFE Child, LIFE Leipzig Research Center for Civilization Diseases, Leipzig University, Leipzig, Germany
| | - Robert Zeidler
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany
| | - Uta Ceglarek
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany
| | - Wieland Kiess
- Hospital for Children and Adolescents, Center for Pediatric Research, Leipzig University Medical Center, Leipzig, Germany
- LIFE Child, LIFE Leipzig Research Center for Civilization Diseases, Leipzig University, Leipzig, Germany
| | - Jürgen Kratzsch
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany
| | - Alexander Gaudl
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany
| | - Ronald Biemann
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany
| | - Mandy Vogel
- Hospital for Children and Adolescents, Center for Pediatric Research, Leipzig University Medical Center, Leipzig, Germany
- LIFE Child, LIFE Leipzig Research Center for Civilization Diseases, Leipzig University, Leipzig, Germany
| |
Collapse
|
14
|
Rosenfield RL. The Search for the Causes of Common Hyperandrogenism, 1965 to Circa 2015. Endocr Rev 2024; 45:553-592. [PMID: 38457123 DOI: 10.1210/endrev/bnae007] [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: 08/27/2023] [Revised: 12/23/2023] [Accepted: 03/06/2024] [Indexed: 03/09/2024]
Abstract
From 1965 to 2015, immense strides were made into understanding the mechanisms underlying the common androgen excess disorders, premature adrenarche and polycystic ovary syndrome (PCOS). The author reviews the critical discoveries of this era from his perspective investigating these disorders, commencing with his early discoveries of the unique pattern of plasma androgens in premature adrenarche and the elevation of an index of the plasma free testosterone concentration in most hirsute women. The molecular genetic basis, though not the developmental biologic basis, for adrenarche is now known and 11-oxytestosterones shown to be major bioactive adrenal androgens. The evolution of the lines of research into the pathogenesis of PCOS is historically traced: research milestones are cited in the areas of neuroendocrinology, insulin resistance, hyperinsulinism, type 2 diabetes mellitus, folliculogenesis, androgen secretion, obesity, phenotyping, prenatal androgenization, epigenetics, and complex genetics. Large-scale genome-wide association studies led to the 2014 discovery of an unsuspected steroidogenic regulator DENND1A (differentially expressed in normal and neoplastic development). The splice variant DENND1A.V2 is constitutively overexpressed in PCOS theca cells in long-term culture and accounts for their PCOS-like phenotype. The genetics are complex, however: DENND1A intronic variant copy number is related to phenotype severity, and recent data indicate that rare variants in a DENND1A regulatory network and other genes are related to PCOS. Obesity exacerbates PCOS manifestations via insulin resistance and proinflammatory cytokine excess; excess adipose tissue also forms testosterone. Polycystic ovaries in 40 percent of apparently normal women lie on the PCOS functional spectrum. Much remains to be learned.
Collapse
Affiliation(s)
- Robert L Rosenfield
- Department of Pediatrics and Medicine, The University of Chicago, Chicago, IL 94109, USA
- Department of Pediatrics, The University of California, San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
15
|
Rohayem J, Alexander EC, Heger S, Nordenström A, Howard SR. Mini-Puberty, Physiological and Disordered: Consequences, and Potential for Therapeutic Replacement. Endocr Rev 2024; 45:460-492. [PMID: 38436980 PMCID: PMC11244267 DOI: 10.1210/endrev/bnae003] [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: 07/21/2023] [Indexed: 03/05/2024]
Abstract
There are 3 physiological waves of central hypothalamic-pituitary-gonadal (HPG) axis activity over the lifetime. The first occurs during fetal life, the second-termed "mini-puberty"-in the first months after birth, and the third at puberty. After adolescence, the axis remains active all through adulthood. Congenital hypogonadotropic hypogonadism (CHH) is a rare genetic disorder characterized by a deficiency in hypothalamic gonadotropin-releasing hormone (GnRH) secretion or action. In cases of severe CHH, all 3 waves of GnRH pulsatility are absent. The absence of fetal HPG axis activation manifests in around 50% of male newborns with micropenis and/or undescended testes (cryptorchidism). In these boys, the lack of the mini-puberty phase accentuates testicular immaturity. This is characterized by a low number of Sertoli cells, which are important for future reproductive capacity. Thus, absent mini-puberty will have detrimental effects on later fertility in these males. The diagnosis of CHH is often missed in infants, and even if recognized, there is no consensus on optimal therapeutic management. Here we review physiological mini-puberty and consequences of central HPG axis disorders; provide a diagnostic approach to allow for early identification of these conditions; and review current treatment options for replacement of mini-puberty in male infants with CHH. There is evidence from small case series that replacement with gonadotropins to mimic "mini-puberty" in males could have beneficial outcomes not only regarding testis descent, but also normalization of testis and penile sizes. Moreover, such therapeutic replacement regimens in disordered mini-puberty could address both reproductive and nonreproductive implications.
Collapse
Affiliation(s)
- Julia Rohayem
- Department of Pediatric Endocrinology and Diabetology, Children's Hospital of Eastern Switzerland, 9006 St. Gallen, Switzerland
- University of Muenster, 48149 Muenster, Germany
| | - Emma C Alexander
- Centre for Endocrinology, William Harvey Research Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | - Sabine Heger
- Department of Pediatric Endocrinology, Children's Hospital Auf der Bult, 30173 Hannover, Germany
| | - Anna Nordenström
- Pediatric Endocrinology, Karolinska Institutet, Astrid Lindgren Children's Hospital, Karolinska University Hospital, 17176 Stockholm, Sweden
| | - Sasha R Howard
- Centre for Endocrinology, William Harvey Research Institute, Queen Mary University of London, London EC1M 6BQ, UK
- Department of Paediatric Endocrinology, Royal London Children's Hospital, Barts Health NHS Trust, London E1 1FR, UK
| |
Collapse
|
16
|
Tennilä J, Muukkonen L, Utriainen P, Voutilainen R, Jääskeläinen J, Liimatta J. Cognitive performance in young adult women with a history of premature adrenarche. Pediatr Res 2024:10.1038/s41390-024-03380-4. [PMID: 38969813 DOI: 10.1038/s41390-024-03380-4] [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: 12/12/2023] [Revised: 04/23/2024] [Accepted: 06/19/2024] [Indexed: 07/07/2024]
Abstract
BACKGROUND Girls with premature adrenarche (PA) mature earlier than peers and have been found to have greater symptom accounts reflecting anxiety compared to peers. It is not known, however, whether PA effects cognitive development. This longitudinal case-control cohort study aimed: (1) To investigate whether a history of PA leads to measurable changes in adulthood cognitive performance, and (2) to assess whether findings characteristic of PA girls predict adulthood cognitive performance. METHODS Twenty-seven girls with PA and 27 age-matched control girls were examined and followed from mid-childhood (mean age 7.2 years) until early adult age (18.5 years). Wechsler Adult Intelligence Scale, Fourth Edition scores were used as main outcome measure. RESULTS Allostatic load (AL) scores, which compile multisystem variables to reflect the overall wear and tear of the body from increased and prolonged stress, were higher in the PA group in both prepuberty and adulthood, but there were no differences in WAIS-IV results between the groups (full-scale IQ 92.7 vs. 97.5, p 0.376; no differences in separate indexes). Childhood androgen levels, glucose metabolism biomarkers, and AL scores failed to predict adulthood cognitive performance outcomes. CONCLUSION The study suggests that PA does not predispose to adverse adulthood outcomes of cognitive development. IMPACT The study suggests that a history of premature adrenarche (PA) does not affect cognitive performance in adult age. Childhood androgen levels and biomarkers of glucose metabolism failed to predict adulthood cognitive outcomes in this study. Allostatic load scores were elevated in the PA group both in childhood and adulthood but did not predict adulthood cognitive outcomes.
Collapse
Affiliation(s)
- Jussi Tennilä
- Kuopio Pediatric Research Unit, University of Eastern Finland and Kuopio University Hospital, 70211, Kuopio, Finland.
| | | | - Pauliina Utriainen
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, 00014, Helsinki, Finland
- Pediatric Research Center, Children's Hospital, Helsinki University Hospital, 00029, Helsinki, Finland
| | - Raimo Voutilainen
- Kuopio Pediatric Research Unit, University of Eastern Finland and Kuopio University Hospital, 70211, Kuopio, Finland
| | - Jarmo Jääskeläinen
- Kuopio Pediatric Research Unit, University of Eastern Finland and Kuopio University Hospital, 70211, Kuopio, Finland
| | - Jani Liimatta
- Kuopio Pediatric Research Unit, University of Eastern Finland and Kuopio University Hospital, 70211, Kuopio, Finland
- Department of BioMedical Research, University of Bern, 3012, Bern, Switzerland
| |
Collapse
|
17
|
Gu M, Wang Y, Yu Y. Ovarian fibrosis: molecular mechanisms and potential therapeutic targets. J Ovarian Res 2024; 17:139. [PMID: 38970048 PMCID: PMC11225137 DOI: 10.1186/s13048-024-01448-7] [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: 05/11/2024] [Accepted: 06/03/2024] [Indexed: 07/07/2024] Open
Abstract
Ovarian fibrosis, characterized by the excessive proliferation of ovarian fibroblasts and the accumulation of extracellular matrix (ECM), serves as one of the primary causes of ovarian dysfunction. Despite the critical role of ovarian fibrosis in maintaining the normal physiological function of the mammalian ovaries, research on this condition has been greatly underestimated, which leads to a lack of clinical treatment options for ovarian dysfunction caused by fibrosis. This review synthesizes recent research on the molecular mechanisms of ovarian fibrosis, encompassing TGF-β, extracellular matrix, inflammation, and other profibrotic factors contributing to abnormal ovarian fibrosis. Additionally, we summarize current treatment approaches for ovarian dysfunction targeting ovarian fibrosis, including antifibrotic drugs, stem cell transplantation, and exosomal therapies. The purpose of this review is to summarize the research progress on ovarian fibrosis and to propose potential therapeutic strategies targeting ovarian fibrosis for the treatment of ovarian dysfunction.
Collapse
Affiliation(s)
- Mengqing Gu
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China
- Key Laboratory of Assisted Reproduction (Peking University), Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Ministry of Education, Beijing, 100191, China
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, 100191, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China
| | - Yibo Wang
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China.
- Key Laboratory of Assisted Reproduction (Peking University), Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Ministry of Education, Beijing, 100191, China.
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, 100191, China.
- Institute of Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China.
| | - Yang Yu
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China.
- Key Laboratory of Assisted Reproduction (Peking University), Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Ministry of Education, Beijing, 100191, China.
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, 100191, China.
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China.
| |
Collapse
|
18
|
Brown ER, Gettler LT, Rosenbaum S. Effects of social environments on male primate HPG and HPA axis developmental programming. Dev Psychobiol 2024; 66:e22491. [PMID: 38698633 DOI: 10.1002/dev.22491] [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: 09/01/2023] [Revised: 03/05/2024] [Accepted: 04/07/2024] [Indexed: 05/05/2024]
Abstract
Developmental plasticity is particularly important for humans and other primates because of our extended period of growth and maturation, during which our phenotypes adaptively respond to environmental cues. The hypothalamus-pituitary-gonadal (HPG) and hypothalamus-pituitary-adrenal (HPA) axes are likely to be principal targets of developmental "programming" given their roles in coordinating fitness-relevant aspects of the phenotype, including sexual development, adult reproductive and social strategies, and internal responses to the external environment. In social animals, including humans, the social environment is believed to be an important source of cues to which these axes may adaptively respond. The effects of early social environments on the HPA axis have been widely studied in humans, and to some extent, in other primates, but there are still major gaps in knowledge specifically relating to males. There has also been relatively little research examining the role that social environments play in developmental programming of the HPG axis or the HPA/HPG interface, and what does exist disproportionately focuses on females. These topics are likely understudied in males in part due to the difficulty of identifying developmental milestones in males relative to females and the general quiescence of the HPG axis prior to maturation. However, there are clear indicators that early life social environments matter for both sexes. In this review, we examine what is known about the impact of social environments on HPG and HPA axis programming during male development in humans and nonhuman primates, including the role that epigenetic mechanisms may play in this programming. We conclude by highlighting important next steps in this research area.
Collapse
Affiliation(s)
- Ella R Brown
- Department of Anthropology, University of Michigan, Ann Arbor, Michigan, USA
| | - Lee T Gettler
- Department of Anthropology, University of Notre Dame, Notre Dame, Indiana, USA
- Eck Institute for Global Health, University of Notre Dame, Notre Dame, Indiana, USA
| | - Stacy Rosenbaum
- Department of Anthropology, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
19
|
Pötzl B, Kürzinger L, Kendl S, Stopper H, Kurlbaum M, Fassnacht M, Dischinger U. Disruptive effects of plasticizers bisphenol A, F, and S on steroidogenesis of adrenocortical cells. Front Endocrinol (Lausanne) 2024; 15:1387133. [PMID: 38966215 PMCID: PMC11222671 DOI: 10.3389/fendo.2024.1387133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 06/03/2024] [Indexed: 07/06/2024] Open
Abstract
Introduction Endocrine disrupting chemicals (EDCs) are known to interfere with endocrine homeostasis. Their impact on the adrenal cortex and steroidogenesis has not yet been sufficiently elucidated. This applies in particular to the ubiquitously available bisphenols A (BPA), F (BPF), and S (BPS). Methods NCI-H295R adrenocortical cells were exposed to different concentrations (1nM-1mM) of BPA, BPF, BPS, and an equimolar mixture of them (BPmix). After 72 hours, 15 endogenous steroids were measured using LC-MS/MS. Ratios of substrate and product of CYP-regulated steps were calculated to identify most influenced steps of steroidogenesis. mRNA expression of steroidogenic enzymes was determined by real-time PCR. Results Cell viability remained unaffected at bisphenol concentrations lower than 250 µM. All tested bisphenols and their combination led to extensive alterations in the quantified steroid levels. The most profound fold changes (FC) in steroid concentrations after exposure to BPA (>10µM) were seen for androstenedione, e.g. a 0.37±0.11-fold decrease at 25µM (p≤0.0001) compared to vehicle-treated controls. For BPF, levels of 17-hydroxyprogesterone were significantly increased by 25µM (FC 2.57±0.49, p≤0.001) and 50µM (FC 2.65±0.61, p≤0.0001). BPS treatment led to a dose-dependent decrease of 11-deoxycorticosterone at >1µM (e.g. FC 0.24±0.14, p≤0.0001 at 10µM). However, when combining all three bisphenols, additive effects were detected: e.g. 11-deoxycortisosterone was decreased at doses >10µM (FC 0.27±0.04, p≤0.0001, at 25µM), whereas 21-deoxycortisol was increased by 2.92±0.20 (p≤0.01) at 10µM, and by 3.21±0.45 (p≤0.001) at 50µM. While every measured androgen (DHEA, DHEAS, androstenedione, testosterone, DHT) was lowered in all experiments, estradiol levels were significantly increased by BPA, BPF, BPS, and BPmix (e.g. FC 3.60±0.54, p≤0.0001 at 100µM BPF). Calculated substrate-product ratios indicated an inhibition of CYP17A1-, and CYP21A2 mediated conversions, whereas CYP11B1 and CYP19A1 showed higher activity in the presence of bisphenols. Based on these findings, most relevant mRNA expression of CYP genes were analysed. mRNA levels of StAR, CYP11B1, and CYP17A1 were significantly increased by BPF, BPS, and BPmix. Discussion In cell culture, bisphenols interfere with steroidogenesis at non-cytotoxic levels, leading to compound-specific patterns of significantly altered hormone levels. These results justify and call for additional in-vivo studies to evaluate effects of EDCs on adrenal gland functionality.
Collapse
Affiliation(s)
- Benedikt Pötzl
- Division of Endocrinology and Diabetology, Department of Internal Medicine I, University Hospital of Würzburg, Würzburg, Germany
| | - Lydia Kürzinger
- Division of Endocrinology and Diabetology, Department of Internal Medicine I, University Hospital of Würzburg, Würzburg, Germany
| | - Sabine Kendl
- Central Laboratory, Core Unit Clinical Mass Spectrometry, University Hospital, University of Würzburg, Würzburg, Germany
| | - Helga Stopper
- Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany
| | - Max Kurlbaum
- Division of Endocrinology and Diabetology, Department of Internal Medicine I, University Hospital of Würzburg, Würzburg, Germany
- Central Laboratory, Core Unit Clinical Mass Spectrometry, University Hospital, University of Würzburg, Würzburg, Germany
| | - Martin Fassnacht
- Division of Endocrinology and Diabetology, Department of Internal Medicine I, University Hospital of Würzburg, Würzburg, Germany
- Central Laboratory, Core Unit Clinical Mass Spectrometry, University Hospital, University of Würzburg, Würzburg, Germany
| | - Ulrich Dischinger
- Division of Endocrinology and Diabetology, Department of Internal Medicine I, University Hospital of Würzburg, Würzburg, Germany
| |
Collapse
|
20
|
Kaplowitz PB. Premature Pubarche: A Pragmatic Approach. Endocrinol Metab Clin North Am 2024; 53:203-209. [PMID: 38677863 DOI: 10.1016/j.ecl.2024.02.001] [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] [Indexed: 04/29/2024]
Abstract
Premature pubarche (PP) is a common and usually benign variant of normal puberty most often seen in 5-year-old to 9-year-old children. Some providers routinely order laboratory testing and a bone age to try to rule out other diagnoses including nonclassic congenital adrenal hyperplasia and gonadal or adrenal tumors. I review the natural history of PP and studies which suggest that without clinical features such as rapid growth and progression or genital enlargement, it is unlikely that a treatable condition will be found. Therefore it is recommended that patients with PP not undergo testing unless there are red flags at the time of the initial visit.
Collapse
Affiliation(s)
- Paul B Kaplowitz
- Division of Endocrinology, Children's National Hospital, Washington, DC, USA.
| |
Collapse
|
21
|
Bangalore Krishna K, Silverman LA. Diagnosis of Central Precocious Puberty. Endocrinol Metab Clin North Am 2024; 53:217-227. [PMID: 38677865 DOI: 10.1016/j.ecl.2024.02.002] [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] [Indexed: 04/29/2024]
Abstract
A thorough history and physical examination including Tanner staging and growth assessments can guide differential diagnosis and aid in the evaluation of precocious puberty. Basal luteinizing hormone levels measured using a highly sensitive assay can be helpful in diagnosing central precocious puberty (CPP). Brain MRI is indicated with males diagnosed with CPP and females under the age of 6 with CPP. As more information becomes available regarding the genetic etiologies of CPP, genetic testing may preclude the need for imaging studies and other hormonal testing, especially in familial cases.
Collapse
Affiliation(s)
- Kanthi Bangalore Krishna
- Division of Pediatric Endocrinology and Diabetes, UPMC Childrens Hospital of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA 15224, USA.
| | - Lawrence A Silverman
- Division of Pediatric Endocrinology, Goryeb Children's Hospital, Atlantic Health System, 100 Madison Avenue, Morristown, NJ 07960, USA
| |
Collapse
|
22
|
Jiang JH, Wang YF, Zheng J, Lei YM, Chen ZY, Guo Y, Guo YJ, Guo BQ, Lv YF, Wang HH, Xie JJ, Liu YX, Jin TW, Li BQ, Zhu XS, Jiang YH, Mo ZN. Human-like adrenal features in Chinese tree shrews revealed by multi-omics analysis of adrenal cell populations and steroid synthesis. Zool Res 2024; 45:617-632. [PMID: 38766745 PMCID: PMC11188597 DOI: 10.24272/j.issn.2095-8137.2023.280] [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: 11/07/2023] [Accepted: 12/25/2023] [Indexed: 05/22/2024] Open
Abstract
The Chinese tree shrew ( Tupaia belangeri chinensis) has emerged as a promising model for investigating adrenal steroid synthesis, but it is unclear whether the same cells produce steroid hormones and whether their production is regulated in the same way as in humans. Here, we comprehensively mapped the cell types and pathways of steroid metabolism in the adrenal gland of Chinese tree shrews using single-cell RNA sequencing, spatial transcriptome analysis, mass spectrometry, and immunohistochemistry. We compared the transcriptomes of various adrenal cell types across tree shrews, humans, macaques, and mice. Results showed that tree shrew adrenal glands expressed many of the same key enzymes for steroid synthesis as humans, including CYP11B2, CYP11B1, CYB5A, and CHGA. Biochemical analysis confirmed the production of aldosterone, cortisol, and dehydroepiandrosterone but not dehydroepiandrosterone sulfate in the tree shrew adrenal glands. Furthermore, genes in adrenal cell types in tree shrews were correlated with genetic risk factors for polycystic ovary syndrome, primary aldosteronism, hypertension, and related disorders in humans based on genome-wide association studies. Overall, this study suggests that the adrenal glands of Chinese tree shrews may consist of closely related cell populations with functional similarity to those of the human adrenal gland. Our comprehensive results (publicly available at http://gxmujyzmolab.cn:16245/scAGMap/) should facilitate the advancement of this animal model for the investigation of adrenal gland disorders.
Collapse
Affiliation(s)
- Jing-Hang Jiang
- Center for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi 530021, China
- Department of Urology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
- Reproductive Medicine Center, Jingmen People's Hospital, JingChu University of Technology Affiliated Central Hospital, Jingmen, Hubei 448000, China
| | - Yi-Fu Wang
- Center for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi 530021, China
- Department of Urology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Jie Zheng
- Center for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi 530021, China
- Department of Urology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Yi-Ming Lei
- School of Computer Science and Engineering, Yulin Normal University, Yulin, Guangxi 537000, China
| | - Zhong-Yuan Chen
- Center for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi 530021, China
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Yi Guo
- Department of Urology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Ya-Jie Guo
- Center for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Bing-Qian Guo
- Center for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Yu-Fang Lv
- Center for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Hong-Hong Wang
- Center for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Juan-Juan Xie
- Center for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Yi-Xuan Liu
- Center for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Ting-Wei Jin
- Center for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Bi-Qi Li
- Department of Pathology, Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530007, China
| | - Xiao-Shu Zhu
- School of Computer Science and Engineering, Yulin Normal University, Yulin, Guangxi 537000, China. E-mail:
| | - Yong-Hua Jiang
- Center for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi 530021, China
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning, Guangxi 530021, China
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530007, China. E-mail:
| | - Zeng-Nan Mo
- Center for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi 530021, China
- Department of Urology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China. E-mail:
| |
Collapse
|
23
|
Augsburger P, Liimatta J, Flück CE. Update on Adrenarche-Still a Mystery. J Clin Endocrinol Metab 2024; 109:1403-1422. [PMID: 38181424 DOI: 10.1210/clinem/dgae008] [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: 09/11/2023] [Revised: 12/29/2023] [Accepted: 01/04/2024] [Indexed: 01/07/2024]
Abstract
CONTEXT Adrenarche marks the timepoint of human adrenal development when the cortex starts secreting androgens in increasing amounts, in healthy children at age 8-9 years, with premature adrenarche (PA) earlier. Because the molecular regulation and significance of adrenarche are unknown, this prepubertal event is characterized descriptively, and PA is a diagnosis by exclusion with unclear long-term consequences. EVIDENCE ACQUISITION We searched the literature of the past 5 years, including original articles, reviews, and meta-analyses from PubMed, ScienceDirect, Web of Science, Embase, and Scopus, using search terms adrenarche, pubarche, DHEAS, steroidogenesis, adrenal, and zona reticularis. EVIDENCE SYNTHESIS Numerous studies addressed different topics of adrenarche and PA. Although basic studies on human adrenal development, zonation, and zona reticularis function enhanced our knowledge, the exact mechanism leading to adrenarche remains unsolved. Many regulators seem involved. A promising marker of adrenarche (11-ketotestosterone) was found in the 11-oxy androgen pathway. By current definition, the prevalence of PA can be as high as 9% to 23% in girls and 2% to 10% in boys, but only a subset of these children might face related adverse health outcomes. CONCLUSION New criteria for defining adrenarche and PA are needed to identify children at risk for later disease and to spare children with a normal variation. Further research is therefore required to understand adrenarche. Prospective, long-term studies should characterize prenatal or early postnatal developmental pathways that modulate trajectories of birth size, early postnatal growth, childhood overweight/obesity, adrenarche and puberty onset, and lead to abnormal sexual maturation, fertility, and other adverse outcomes.
Collapse
Affiliation(s)
- Philipp Augsburger
- Pediatric Endocrinology, Diabetology, and Metabolism, Inselspital, Bern University Hospital, 3010 Bern, Switzerland
- Department of BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland
| | - Jani Liimatta
- Pediatric Endocrinology, Diabetology, and Metabolism, Inselspital, Bern University Hospital, 3010 Bern, Switzerland
- Department of BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland
- Kuopio Pediatric Research Unit (KuPRU), University of Eastern Finland and Kuopio University Hospital, 70029 Kuopio, Finland
| | - Christa E Flück
- Pediatric Endocrinology, Diabetology, and Metabolism, Inselspital, Bern University Hospital, 3010 Bern, Switzerland
- Department of BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland
| |
Collapse
|
24
|
Helvacıoğlu D, Güran T. Bone Phenotype is Always Present But Androgen Excess is Less Frequently Seen in PAPSS2 Deficiency. J Clin Res Pediatr Endocrinol 2024; 16:4-10. [PMID: 38084048 PMCID: PMC10938522 DOI: 10.4274/jcrpe.galenos.2023.2023-12-10] [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: 12/10/2023] [Accepted: 12/11/2023] [Indexed: 03/12/2024] Open
Abstract
3’-Phosphoadenosine 5’-phosphosulfate synthase 2 (PAPSS2) deficiency is a rare disorder due to biallelic pathogenic variants in the PAPSS2 gene. This disorder was first described in 1998 by Ahmad et al. and Faiyaz ul Haque et al. To date, 79 patients with PAPSS2 deficiency have been reported. The main reported features of these patients are related to bone abnormalities and clinical/biochemical androgen excess. Disproportionate short stature and symptoms associated with spondylar skeletal dysplasia are the most common clinical features that require clinical attention. Androgen excess has been described much less commonly. This review summarizes the currently published clinical, molecular, and biochemical features of patients with PAPSS2 deficiency.
Collapse
Affiliation(s)
- Didem Helvacıoğlu
- Marmara University Faculty of Medicine, Department of Pediatric Endocrinology and Diabetes, İstanbul, Turkey
| | - Tülay Güran
- Marmara University Faculty of Medicine, Department of Pediatric Endocrinology and Diabetes, İstanbul, Turkey
| |
Collapse
|
25
|
Liu D, Liu WV, Zhang L, Qin Y, Li Y, Ding G, Zhou Y, Xie Y, Chen P, Zhu W. Diagnostic value of adenohypophyseal MRI features in female children with precocious puberty. Clin Radiol 2024; 79:179-188. [PMID: 38114375 DOI: 10.1016/j.crad.2023.11.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 11/01/2023] [Accepted: 11/15/2023] [Indexed: 12/21/2023]
Abstract
AIM To evaluate the diagnostic value of adenohypophyseal magnetic resonance imaging (MRI) features for precocious puberty (PP) in female children and also to establish a non-invasive diagnostic approach in clinics. MATERIALS AND METHODS A total of 126 female children (37, 57, and 32 female children clinically diagnosed with central PP [CPP], incomplete PP [IPP], and controls, respectively) were enrolled in this study. Data were collected and analysed using analysis of variance. Pearson correlation and stepwise multivariate linear regression analysis were used to examine the association and build prediction models. Receiver operating characteristic (ROC) analysis was used to evaluate the diagnostic efficacy. RESULTS The values of adenohypophysis volume (aPV), adenohypophysis height (aPH), and signal-intensity ratio (SIR), height, weight, and seven laboratory testing characteristics were correlated closely with the activation status of the hypothalamic-pituitary-gonad axis in the different groups (all p<0.05). Model 1 including aPV, weight, and aPH and Model 2 including SIR, aPV, and height were built to obtain predicted luteinising hormone (LH; R2 = 0.271) and LH/follicle stimulating hormone (FSH; R2 = 0.311). ROC analysis showed the predicted LH, predicted LH/FSH, and aPV were the top 3 best predictors in distinguishing CPP from controls (AUC = 0.969, 0.949, and 0.938) while predicted LH/FSH was the best predictor in distinguishing CPP from IPP and controls (AUC = 0.829 and 0.828). CONCLUSION The adenohypophysis volume itself and the prediction models including main adenohypophyseal MRI features increased diagnostic efficiency for PP and offered a non-invasive and credible diagnostic method.
Collapse
Affiliation(s)
- D Liu
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China.
| | - W V Liu
- MR Research, GE Healthcare, Beijing 100176, China
| | - L Zhang
- Department of Hematology and Tumor, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430016, Hubei, China
| | - Y Qin
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Y Li
- Department of Endocrinology and Metabolism, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430016, Hubei, China
| | - G Ding
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Y Zhou
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Y Xie
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - P Chen
- Department of Pediatric Pediatric Department, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - W Zhu
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| |
Collapse
|
26
|
Wang Q, Wang X, Liu B, Ma S, Zhang F, Sun S, Jing Y, Fan Y, Ding Y, Xiong M, Li J, Zhai Q, Zheng Y, Liu C, Xu G, Yang J, Wang S, Ye J, Izpisua Belmonte JC, Qu J, Liu GH, Zhang W. Aging induces region-specific dysregulation of hormone synthesis in the primate adrenal gland. NATURE AGING 2024; 4:396-413. [PMID: 38503993 DOI: 10.1038/s43587-024-00588-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 02/05/2024] [Indexed: 03/21/2024]
Abstract
Adrenal glands, vital for steroid secretion and the regulation of metabolism, stress responses and immune activation, experience age-related decline, impacting systemic health. However, the regulatory mechanisms underlying adrenal aging remain largely uninvestigated. Here we established a single-nucleus transcriptomic atlas of both young and aged primate suprarenal glands, identifying lipid metabolism and steroidogenic pathways as core processes impacted by aging. We found dysregulation in centripetal adrenocortical differentiation in aged adrenal tissues and cells in the zona reticularis region, responsible for producing dehydroepiandrosterone sulfate (DHEA-S), were highly susceptible to aging, reflected by senescence, exhaustion and disturbed hormone production. Remarkably, LDLR was downregulated in all cell types of the outer cortex, and its targeted inactivation in human adrenal cells compromised cholesterol uptake and secretion of dehydroepiandrosterone sulfate, as observed in aged primate adrenal glands. Our study provides crucial insights into endocrine physiology, holding therapeutic promise for addressing aging-related adrenal insufficiency and delaying systemic aging.
Collapse
Affiliation(s)
- Qiaoran Wang
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xuebao Wang
- University of Chinese Academy of Sciences, Beijing, China
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Beibei Liu
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China
| | - Shuai Ma
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Feng Zhang
- Division of Endocrinology, Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou, China
- The Joint Innovation Center for Engineering in Medicine, Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou, China
| | - Shuhui Sun
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Yaobin Jing
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- International Center for Aging and Cancer, Hainan Medical University, Haikou, China
| | - Yanling Fan
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China
| | - Yingjie Ding
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Muzhao Xiong
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jiaming Li
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Qiaocheng Zhai
- Division of Endocrinology, Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou, China
- The Joint Innovation Center for Engineering in Medicine, Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou, China
| | - Yandong Zheng
- University of Chinese Academy of Sciences, Beijing, China
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Chengyu Liu
- University of Chinese Academy of Sciences, Beijing, China
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Gang Xu
- Liver Transplant Center, Organ Transplant Center, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Liver Transplantation, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital of Sichuan University, Chengdu, China
| | - Jiayin Yang
- Liver Transplant Center, Organ Transplant Center, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Liver Transplantation, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital of Sichuan University, Chengdu, China
| | - Si Wang
- Aging Translational Medicine Center, International Center for Aging and Cancer, Xuanwu Hospital, Capital Medical University, Beijing, China
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing, China
- The Fifth People's Hospital of Chongqing, Chongqing, China
- Aging Biomarker Consortium, Beijing, China
| | - Jinlin Ye
- The Joint Innovation Center for Engineering in Medicine, Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou, China
| | | | - Jing Qu
- University of Chinese Academy of Sciences, Beijing, China.
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.
- Aging Biomarker Consortium, Beijing, China.
| | - Guang-Hui Liu
- University of Chinese Academy of Sciences, Beijing, China.
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
- International Center for Aging and Cancer, Hainan Medical University, Haikou, China.
- Aging Translational Medicine Center, International Center for Aging and Cancer, Xuanwu Hospital, Capital Medical University, Beijing, China.
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing, China.
- Aging Biomarker Consortium, Beijing, China.
| | - Weiqi Zhang
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China.
- University of Chinese Academy of Sciences, Beijing, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.
- Aging Biomarker Consortium, Beijing, China.
| |
Collapse
|
27
|
Li H, Liu Y, Meng F, Chen J, Han X. Adrenarche-accompanied rise of adrenal sex steroid precursors prevents NAFLD in Young Female rats by converting into active androgens and inactivating hepatic Srebf1 signaling. BMC Genomics 2024; 25:190. [PMID: 38369486 PMCID: PMC10875776 DOI: 10.1186/s12864-024-10107-6] [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: 08/18/2023] [Accepted: 02/09/2024] [Indexed: 02/20/2024] Open
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD) has rapidly become the most common cause of chronic liver disease in children and adolescents, but its etiology remains largely unknown. Adrenarche is a critical phase for hormonal changes, and any disturbance during this period has been linked to metabolic disorders, including obesity and dyslipidemia. However, whether there is a causal linkage between adrenarche disturbance and the increasing prevalence of NAFLD in children remains unclear. RESULTS Using the young female rat as a model, we found that the liver undergoes a transient slowdown period of growth along with the rise of adrenal-derived sex steroid precursors during adrenarche. Specifically blocking androgen actions across adrenarche phase using androgen receptor antagonist flutamide largely increased liver weight by 47.97% and caused marked fat deposition in liver, thus leading to severe NAFLD in young female rats. Conversely, further administrating nonaromatic dihydrotestosterone (DHT) into young female rats across adrenarche phase could effectively reduce liver fat deposition. But, administration of the aromatase inhibitor, formestane across adrenarche had minimal effects on hepatic de novo fatty acid synthesis and liver fat deposition, suggesting adrenal-derived sex steroid precursors exert their anti-NAFLD effects in young females by converting into active androgens rather than into active estrogens. Mechanistically, transcriptomic profiling and integrated data analysis revealed that active androgens converted from the adrenal sex steroid precursors prevent NAFLD in young females primarily by inactivating hepatic sterol regulatory element-binding transcription factor 1 (Srebf1) signaling. CONCLUSIONS We firstly evidenced that adrenarche-accompanied rise of sex steroid precursors plays a predominant role in preventing the incidence of NAFLD in young females by converting into active androgens and inactivating hepatic Srebf1 signaling. Our novel finding provides new insights into the etiology of NAFLD and is crucial in developing effective prevention and management strategies for NAFLD in children.
Collapse
Affiliation(s)
- Haoqing Li
- College of Life Science, Sichuan Agricultural University, Ya'an, 625014, China
| | - Yingyu Liu
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Fengyan Meng
- College of Life Science, Sichuan Agricultural University, Ya'an, 625014, China
| | - Junan Chen
- College of Life Science, Sichuan Agricultural University, Ya'an, 625014, China
| | - Xingfa Han
- College of Life Science, Sichuan Agricultural University, Ya'an, 625014, China.
| |
Collapse
|
28
|
Liimatta J, Jääskeläinen J, Mäntyselkä A, Häkkinen MR, Auriola S, Voutilainen R, Flück CE, Lakka TA. Accelerated Early Childhood Growth Is Associated With the Development of Earlier Adrenarche and Puberty. J Endocr Soc 2024; 8:bvae026. [PMID: 38425434 PMCID: PMC10904224 DOI: 10.1210/jendso/bvae026] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Indexed: 03/02/2024] Open
Abstract
Context Small birth size and increased postnatal growth have been associated with earlier timing of adrenarche and puberty, but it is not well known whether these factors alone or together lead to earlier maturation. Objective This work aimed to search for different growth trajectories using a clustering approach to analyze the effects of birth size and postnatal growth on adrenarchal and pubertal development. Methods Altogether 351 children (48% girls) were examined prospectively at ages 6 to 9 and 9 to 11 years. Birth and early-growth data were collected retrospectively. Main outcome measures included clinical signs of adrenarche and puberty, and serum androgen concentrations (dehydroepiandrosterone, dehydroepiandrosterone sulfate, androstenedione, testosterone). Results We detected 4 clusters with different birth sizes and postnatal growth trajectories: 1) children with average birth size and increased postnatal growth (AI), 2) children with small birth size and increased postnatal growth (SI), 3) children with average birth size and postnatal growth (AA), and 4) children with small birth size and average postnatal growth (SA). Thelarche at age 9 to 11 was most common and serum androgens at ages 6 to 9 and 9 to 11 years were highest in girls belonging to the AI and SI groups. Similar patterns in the onset of puberty and in androgen levels were not seen in the SA group. Conclusion Increased early growth and weight gain predict higher serum androgen concentrations and earlier onset of puberty in girls. Adrenarche and puberty do not appear to be shifted earlier in children with small birth size who do not have catch-up growth.
Collapse
Affiliation(s)
- Jani Liimatta
- Kuopio Pediatric Research Unit (KuPRU), University of Eastern Finland, 70029 Kuopio, Finland
- Department of BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland
- Pediatric Endocrinology, Diabetology, and Metabolism, Inselspital, Bern University Hospital, 3010 Bern, Switzerland
| | - Jarmo Jääskeläinen
- Kuopio Pediatric Research Unit (KuPRU), University of Eastern Finland, 70029 Kuopio, Finland
- Department of Pediatrics, Kuopio University Hospital, 70029 Kuopio, Finland
| | - Aino Mäntyselkä
- Kuopio Pediatric Research Unit (KuPRU), University of Eastern Finland, 70029 Kuopio, Finland
- Department of Pediatrics, Kuopio University Hospital, 70029 Kuopio, Finland
| | - Merja R Häkkinen
- School of Pharmacy, University of Eastern Finland, 70211 Kuopio, Finland
- Department of Health Security, Finnish Institute for Health and Welfare (THL), 70701 Kuopio, Finland
| | - Seppo Auriola
- School of Pharmacy, University of Eastern Finland, 70211 Kuopio, Finland
| | - Raimo Voutilainen
- Kuopio Pediatric Research Unit (KuPRU), University of Eastern Finland, 70029 Kuopio, Finland
- Department of Pediatrics, Kuopio University Hospital, 70029 Kuopio, Finland
| | - Christa E Flück
- Department of BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland
- Pediatric Endocrinology, Diabetology, and Metabolism, Inselspital, Bern University Hospital, 3010 Bern, Switzerland
| | - Timo A Lakka
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, 70211 Kuopio, Finland
- Department of Clinical Physiology and Nuclear Medicine, Kuopio University Hospital, 70210 Kuopio, Finland
- Foundation for Research in Health Exercise and Nutrition, Kuopio Research Institute of Exercise Medicine, 70211 Kuopio, Finland
| |
Collapse
|
29
|
Reder SR, Fritzen I, Brockmann MA, Hardt J, Elsner K, Petrowski K, Bjelopavlovic M. Comparing a common clavicle maturation-based age estimation method to ordinary regression analyses with quadratic and sex-specific interaction terms in adolescents. Sci Rep 2024; 14:2754. [PMID: 38307929 PMCID: PMC10837444 DOI: 10.1038/s41598-024-52980-x] [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: 09/06/2023] [Accepted: 01/25/2024] [Indexed: 02/04/2024] Open
Abstract
Established methods of age estimation are based on correlating defined maturation stages of bony structures with tables representing the observed range of biological ages in the majority of cases. In this retrospective monocentric study in southwestern Germany, common age estimation methodology was assessed in n = 198 subjects at the age of 25 or younger by analyzing the influence of age, quadratic age, biological sex and age-sex interaction on the ossification stages of the medial epiphysis fugue. Three readers (ICC ≥ 0.81 for left/right side) evaluated routine care computed tomography images of the clavicle with a slice thickness of 1 mm. By using least square regression analyses, to determine the real biological age a quadratic function was determined corrected for the age estimated by established methods and sex (R2 = 0.6 each side), reducing the mean absolute error and root mean squared error in the age estimation of women (2.57 and 3.19) and men (2.57 and 3.47) to 1.54 and 1.82 for women, and 1.54 and 2.25 for men. In women, the medial clavicle epiphysis seem to fuse faster, which was particularly observable from approximately 18 years of age. Before that age, the estimation method was relatively close to the ideal correlation between assessed and real age. To conclude, the presented new method enables more precise age estimation in individuals and facilitates the determination and quantification of additional variables, quantifying their influence on the maturation of the medial clavicle epiphysis based on the established ossification stages.
Collapse
Affiliation(s)
- Sebastian R Reder
- Department of Neuroradiology, University Medical Center, Johannes Gutenberg-University of Mainz, 55131, Mainz, Germany.
| | - Isabel Fritzen
- Department of Prosthetic Dentistry, University Medical Center of the Johannes Gutenberg-University Mainz, Augustusplatz 2, 55131, Mainz, Germany
| | - Marc A Brockmann
- Department of Neuroradiology, University Medical Center, Johannes Gutenberg-University of Mainz, 55131, Mainz, Germany
| | - Jochen Hardt
- Department of Medical Psychology and Medical Sociology, University Medical Center of the Johannes Gutenberg-University Mainz, Duesbergweg 6, 55131, Mainz, Germany
| | - Katrin Elsner
- Institute of Legal Medicine, University Medical Center of the Johannes Gutenberg-University of Mainz, Am Pulverturm 9, 55131, Mainz, Germany
| | - Katja Petrowski
- Department of Medical Psychology and Medical Sociology, University Medical Center of the Johannes Gutenberg-University Mainz, Duesbergweg 6, 55131, Mainz, Germany
| | - Monika Bjelopavlovic
- Department of Prosthetic Dentistry, University Medical Center of the Johannes Gutenberg-University Mainz, Augustusplatz 2, 55131, Mainz, Germany
| |
Collapse
|
30
|
Tamer G, Arets HGM, van der Ent CK, van Santen HM, van der Kamp HJ. BMI increase during early childhood in boys with cystic fibrosis and early adrenarche. Pediatr Pulmonol 2024. [PMID: 38270329 DOI: 10.1002/ppul.26861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 11/21/2023] [Accepted: 01/02/2024] [Indexed: 01/26/2024]
Abstract
BACKGROUND Increase in body mass index (BMI) in early childhood (1-6 years) was found to be a contributing factor for impaired final height in boys with Cystic Fibrosis (CF). Early adrenarche (before age 9 years in boys) may contribute to an impaired final height by triggering an early acceleration of bone age resulting in a compromised growth spurt during puberty. We aimed to analyze the timing of adrenarche in boys with CF and to associate BMI increase in early childhood to timing of adrenarche. METHODS Boys with CF, aged 8-9 years, visiting the CF expertize center Utrecht were included. Since 2018, anthropomorphic, pubertal and endocrine data were collected. Early adrenarche in boys was defined as a dehydroepiandrosterone sulfate (DHEAS) ≥ 1 µmol/L before the age of 9 years. RESULTS Thirteen boys (mean age 8.55 ± 0.27 years) were enrolled. The median (IQR) DHEAS-level was 1.3 µmol/L (0.71-2.40). Eight boys (61.5%) had an early rise in DHEAS-levels ≥ 1 µmol/L. Mean increase in BMI Z-score between 1 and 6 years of age (ΔBMI1-6 ) was -0.07 ± 0.86. A significant correlation was found between ΔBMI1-6 and DHEAS-levels at the age of 8-9 years (r = 0.624, p = 0.040). In five boys with early rise in DHEAS, accelerated bone age was found (average 1.55 ± 0.96 years). CONCLUSION In this small cohort, 61.5% of boys with CF between 8 and 9 years had an early rise of DHEAS, which was correlated to ΔBMI1 -6 between 1 and 6 years. Early adrenarche may be caused by ΔBMI1 -6 .
Collapse
Affiliation(s)
- Gizem Tamer
- Department of Pediatric Endocrinology, Wilhelmina Children's Hospital, University Medical Center, Utrecht, the Netherlands
- Department of Pediatric Pulmonology, Wilhelmina Children's Hospital, University Medical Center, Utrecht, the Netherlands
| | - Hubertus Gerardus Maria Arets
- Department of Pediatric Pulmonology, Wilhelmina Children's Hospital, University Medical Center, Utrecht, the Netherlands
| | - Cornelis Kors van der Ent
- Department of Pediatric Pulmonology, Wilhelmina Children's Hospital, University Medical Center, Utrecht, the Netherlands
| | - Hanneke Margo van Santen
- Department of Pediatric Endocrinology, Wilhelmina Children's Hospital, University Medical Center, Utrecht, the Netherlands
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Hetty Jacoba van der Kamp
- Department of Pediatric Endocrinology, Wilhelmina Children's Hospital, University Medical Center, Utrecht, the Netherlands
| |
Collapse
|
31
|
Heijboer AC, Hannema SE. Androgen Excess and Deficiency: Analytical and Diagnostic Approaches. Clin Chem 2023; 69:1361-1373. [PMID: 37794651 DOI: 10.1093/clinchem/hvad146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 08/18/2023] [Indexed: 10/06/2023]
Abstract
BACKGROUND Androgens are synthesized from cholesterol through sequential conversions by enzymes in the adrenal glands and gonads. Serum levels of androgens change during the different phases of life and regulate important developmental and maturational processes. Androgen excess or deficiency can therefore present at various ages in various ways. CONTENT The diagnostic approach for atypical genitalia, premature pubarche, delayed pubertal onset or progression, and hirsutism or virilization, including measurement of androgens (testosterone, androstenedione, 17-OHprogesterone, dehydroepiandrosterone, and dihydrotestosterone) is discussed in the current review. Androgens can be measured in serum, saliva, urine, or dried blood spots. Techniques to measure androgens, including immunoassays and LC-MS, have their own advantages and pitfalls. In addition, pre- and postanalytical issues are important when measuring androgens. SUMMARY During clinical interpretation of androgen measurements, it is important to take preanalytical circumstances, such as time of blood withdrawal, into account. As immunoassays have major drawbacks, especially in samples from women and neonates, concentrations measured using these assays should be interpreted with care. Reference intervals can only be used in relation to the measurement technique and the standardization of the assay. In the near future, new androgens will probably be added to the current repertoire to further improve the diagnosis and follow-up of androgen excess or deficiency.
Collapse
Affiliation(s)
- Annemieke C Heijboer
- Endocrine Laboratory, Department of Laboratory Medicine, Amsterdam UMC location University of Amsterdam, Amsterdam, the Netherlands
- Amsterdam Gastroenterology, Endocrinology & Metabolism, Amsterdam, the Netherlands
- Endocrine Laboratory, Department of Laboratory Medicine, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Amsterdam Reproduction & Development Research Institute, Amsterdam, The Netherlands
| | - Sabine E Hannema
- Amsterdam Gastroenterology, Endocrinology & Metabolism, Amsterdam, the Netherlands
- Amsterdam Reproduction & Development Research Institute, Amsterdam, The Netherlands
- Department of Pediatric Endocrinology, Amsterdam UMC location Vrije Universiteit, Emma Children's Hospital, Amsterdam, the Netherlands
| |
Collapse
|
32
|
Liimatta J, Flück CE, Mäntyselkä A, Häkkinen MR, Auriola S, Voutilainen R, Jääskeläinen J, Lakka TA. Effects of 2-Year Physical Activity and Dietary Intervention on Adrenarchal and Pubertal Development: The PANIC Study. J Clin Endocrinol Metab 2023; 108:e1603-e1613. [PMID: 37329220 PMCID: PMC10655527 DOI: 10.1210/clinem/dgad367] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 06/10/2023] [Accepted: 06/15/2023] [Indexed: 06/18/2023]
Abstract
CONTEXT Childhood overweight has been linked to earlier development of adrenarche and puberty, but it remains unknown if lifestyle interventions influence sexual maturation in general populations. OBJECTIVE To investigate if a 2-year lifestyle intervention influences circulating androgen concentrations and sexual maturation in a general population of children. METHODS We conducted a 2-year physical activity and dietary intervention study in which 421 prepubertal and mostly normal-weight 6- to 9-year-old children were allocated either to a lifestyle intervention group (119 girls, 132 boys) or a control group (84 girls, 86 boys). The main outcome measures were serum dehydroepiandrosterone (DHEA), dehydroepiandrosterone sulfate (DHEAS), androstenedione (A4), and testosterone concentrations, and clinical adrenarchal and pubertal signs. RESULTS The intervention and control groups had no differences in body size and composition, clinical signs of androgen action, and serum androgens at baseline. The intervention attenuated the increase of DHEA (P = .032), DHEAS (P = .001), A4 (P = .003), and testosterone (P = .007) and delayed pubarche (P = .038) in boys but it only attenuated the increase of DHEA (P = .013) and DHEAS (P = .003) in girls. These effects of lifestyle intervention on androgens and the development of pubarche were independent of changes in body size and composition, but the effects of intervention on androgens were partly explained by changes in fasting serum insulin. CONCLUSION A combined physical activity and dietary intervention attenuates the increase of serum androgen concentrations and sexual maturation in a general population of prepubertal and mostly normal-weight children, independently of changes in body size and composition.
Collapse
Affiliation(s)
- Jani Liimatta
- Kuopio Pediatric Research Unit (KuPRU), University of Eastern Finland, 70029 Kuopio, Finland
- Department of BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland
- Pediatric Endocrinology, Diabetology, and Metabolism, Inselspital, Bern University Hospital, 3010 Bern, Switzerland
| | - Christa E Flück
- Department of BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland
- Pediatric Endocrinology, Diabetology, and Metabolism, Inselspital, Bern University Hospital, 3010 Bern, Switzerland
| | - Aino Mäntyselkä
- Kuopio Pediatric Research Unit (KuPRU), University of Eastern Finland, 70029 Kuopio, Finland
- Department of Pediatrics, Kuopio University Hospital, 70029 Kuopio, Finland
| | - Merja R Häkkinen
- School of Pharmacy, University of Eastern Finland, 70211 Kuopio, Finland
- Department of Health Security, Finnish Institute for Health and Welfare (THL), 70701 Kuopio, Finland
| | - Seppo Auriola
- School of Pharmacy, University of Eastern Finland, 70211 Kuopio, Finland
| | - Raimo Voutilainen
- Kuopio Pediatric Research Unit (KuPRU), University of Eastern Finland, 70029 Kuopio, Finland
- Department of Pediatrics, Kuopio University Hospital, 70029 Kuopio, Finland
| | - Jarmo Jääskeläinen
- Kuopio Pediatric Research Unit (KuPRU), University of Eastern Finland, 70029 Kuopio, Finland
- Department of Pediatrics, Kuopio University Hospital, 70029 Kuopio, Finland
| | - Timo A Lakka
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, 70211 Kuopio, Finland
- Department of Clinical Physiology and Nuclear Medicine, Kuopio University Hospital, 70210 Kuopio, Finland
- Foundation for Research in Health Exercise and Nutrition, Kuopio Research Institute of Exercise Medicine, 70211 Kuopio, Finland
| |
Collapse
|
33
|
Shi LL, Hang JG, Lou J, Dong JJ, Feng H, Wang Z, Shen B, Nakayama SF, Kido T, Ma C, Sun XL, Jung CR. Multiple exposures to heavy metals and changes in steroid hormones production in 4-year-old children. JOURNAL OF EXPOSURE SCIENCE & ENVIRONMENTAL EPIDEMIOLOGY 2023; 33:865-873. [PMID: 36973528 DOI: 10.1038/s41370-023-00539-9] [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: 08/31/2022] [Revised: 03/07/2023] [Accepted: 03/14/2023] [Indexed: 06/18/2023]
Abstract
BACKGROUND Prenatal exposure to multiple heavy metals can interfere with early neurodevelopment, lead to changes in sex hormone concentrations in children, and affect female reproductive health. To date, the influence of prenatal exposure to heavy metals on the endocrine system of children in Chinese electronic waste (e-waste) recycling areas has not been elucidated. METHODS Four weeks after delivery, 10 mL of human milk was collected for analysis of three heavy metals (lead (Pb), cadmium (Cd), and mercury (Hg)) via inductively coupled plasma mass spectrometry (ICP-MS). Four serum steroid hormones, including progesterone, testosterone, androstenedione (A-dione), and dehydroepiandrosterone (DHEA), were analyzed in 4-year-old children (25 boys and 17 girls). A multiple linear regression (MLR) model was implemented to investigate the association between each individual metal and serum steroid hormone. The exposure-response relationships were explored by generalized additive models (GAMs). Additionally, a Bayesian kernel machine regression (BKMR) model was used to assess the effects of multiple heavy metal exposures on each steroid hormone. RESULTS The MLR results show a significant positive association between a natural log unit increase in Hg and DHEA levels after adjusting for confounders (β = 65.50, 95% confidence interval (CI) = 4.37, 126.62). According to the GAM, the univariate exposure-response relationship of Hg on DHEA was almost linear. However, this association was attenuated based on the multiple metal MLR and BKMR results after accounting for multiple heavy metal exposures. SIGNIFICANCE Prenatal Hg exposure may affect sex hormones in children by affecting DHEA levels. IMPACT STATEMENT Prenatal maternal exposure to Hg may have long-term effects on the next generation. Hence, regulatory measures to reduce Hg exposure and long-term monitoring of children's health in e-waste areas are needed.
Collapse
Affiliation(s)
- Li Li Shi
- School of Medicine, and Huzhou Key Laboratory for Precise Prevention and Control of Major Chronic Diseases, Huzhou University, Huzhou, China
- School of Medicine, Jiaxing University, Jiaxing, China
| | - Jin Guo Hang
- Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, China
| | - Jianlin Lou
- School of Medicine, and Huzhou Key Laboratory for Precise Prevention and Control of Major Chronic Diseases, Huzhou University, Huzhou, China
| | | | - Hao Feng
- School of Medicine, Jiaxing University, Jiaxing, China
| | - Zheng Wang
- School of Medicine, Jiaxing University, Jiaxing, China
| | - Bin Shen
- School of Medicine, Jiaxing University, Jiaxing, China
| | - Shoji F Nakayama
- Japan Environment and Children's Study Programme Office, National Institute for Environmental Studies, Tsukuba, Japan
| | - Teruhiko Kido
- Faculty of Health Sciences, Institute of Medical, Pharmaceutical, and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Chaochen Ma
- Cancer Control Center, Osaka International Cancer Institute, Osaka, Japan
| | - Xian Liang Sun
- School of Medicine, and Huzhou Key Laboratory for Precise Prevention and Control of Major Chronic Diseases, Huzhou University, Huzhou, China.
- School of Medicine, Jiaxing University, Jiaxing, China.
- Faculty of Health Sciences, Institute of Medical, Pharmaceutical, and Health Sciences, Kanazawa University, Kanazawa, Japan.
| | - Chau-Ren Jung
- Japan Environment and Children's Study Programme Office, National Institute for Environmental Studies, Tsukuba, Japan.
- Department of Public Health, College of Public Health, China Medical University, Taichung, Taiwan.
| |
Collapse
|
34
|
Schneider G, Ruggiero C, Renault L, Doghman-Bouguerra M, Durand N, Hingrai G, Dijoud F, Plotton I, Lalli E. ACTH and prolactin synergistically and selectively regulate CYP17 expression and adrenal androgen production in human foetal adrenal organ cultures. Eur J Endocrinol 2023; 189:327-335. [PMID: 37638769 DOI: 10.1093/ejendo/lvad118] [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: 04/23/2023] [Revised: 07/12/2023] [Accepted: 08/10/2023] [Indexed: 08/29/2023]
Abstract
OBJECTIVE The essential role of ACTH on the growth and function of the human foetal adrenal (HFA) has long been recognized. In addition, many studies have suggested a role of the pituitary hormone prolactin (PRL) in the regulation of the HFA, but the effects of this hormone on steroidogenesis and gene expression are still unknown. Our objective was to investigate the effect of ACTH and PRL on the steroidogenic capacities of the HFA. DESIGN In vitro/ex vivo experimental study. METHODS We used a hanging drop in vitro organ culture system. First trimester HFA samples were cultured for 14 days in basal conditions or treated with ACTH, PRL, or a combination of the 2 (3 to 11 replicates depending on the experiment). Steroids were measured by liquid chromatography/tandem mass spectrometry or immunoassay, gene expression by RT-qPCR, and protein expression by immunoblot. RESULTS ACTH significantly increased corticosterone, cortisol, and cortisone production, both by itself and when used together with PRL. PRL stimulation by itself had no effect. Combined stimulation with ACTH + PRL synergistically and selectively increased adrenal androgen (DHEAS and Δ4-androstenedione) production and CYP17A1 expression in the HFA, while treatment with each single hormone had no significant effect on those steroids. CONCLUSIONS These results have important implications for our understanding of the hormonal cues regulating adrenal steroidogenesis in the HFA during the first trimester in physiological and pathological conditions and warrant further studies to characterize the molecular mechanisms of converging ACTH and PRL signalling to regulate CYP17A1 expression.
Collapse
Affiliation(s)
- Grégoire Schneider
- Department of Pediatric Surgery, University Hospital of Lyon, 69002 Lyon, France
- Claude Bernard Lyon 1 University, 69100 Villeurbanne, France
| | - Carmen Ruggiero
- Institut de Pharmacologie Moléculaire et Cellulaire CNRS UMR 7275, 06560 Valbonne, France
- Université Côte d'Azur, 06560 Valbonne, France
| | - Lucie Renault
- Claude Bernard Lyon 1 University, 69100 Villeurbanne, France
- Reproductive Medicine and Biology, University Hospital of Lyon, 69002 Lyon, France
| | - Mabrouka Doghman-Bouguerra
- Institut de Pharmacologie Moléculaire et Cellulaire CNRS UMR 7275, 06560 Valbonne, France
- Université Côte d'Azur, 06560 Valbonne, France
| | - Nelly Durand
- Institut de Pharmacologie Moléculaire et Cellulaire CNRS UMR 7275, 06560 Valbonne, France
- Université Côte d'Azur, 06560 Valbonne, France
| | - Guillaume Hingrai
- Orthogenics Department, University Hospital of Lyon, 69002 Lyon, France
| | - Frédérique Dijoud
- Claude Bernard Lyon 1 University, 69100 Villeurbanne, France
- Inserm U1208, 69675 Bron, France
- Department of Pathology, University Hospital of Lyon, 69002 Lyon, France
| | - Ingrid Plotton
- Claude Bernard Lyon 1 University, 69100 Villeurbanne, France
- Reproductive Medicine and Biology, University Hospital of Lyon, 69002 Lyon, France
- Inserm U1208, 69675 Bron, France
- Department of Clinical Biochemistry, University Hospital of Lyon, 69002 Lyon, France
| | - Enzo Lalli
- Institut de Pharmacologie Moléculaire et Cellulaire CNRS UMR 7275, 06560 Valbonne, France
- Université Côte d'Azur, 06560 Valbonne, France
- Inserm, 06560 Valbonne, France
| |
Collapse
|
35
|
Jee YH, Jumani S, Mericq V. The Association of Accelerated Early Growth, Timing of Puberty, and Metabolic Consequences in Children. J Clin Endocrinol Metab 2023; 108:e663-e670. [PMID: 37029976 PMCID: PMC10686698 DOI: 10.1210/clinem/dgad202] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 03/29/2023] [Accepted: 04/04/2023] [Indexed: 04/09/2023]
Abstract
Accelerated early growth and early timing of puberty or pubertal variant have been noticed as risk factors for metabolic syndrome, more frequently observed in children born small for gestational age (SGA) or children with premature adrenarche (PA). Children with SGA, especially if they make an accelerated catch-up growth in early life, carry a higher risk for long-term metabolic consequences, such as type 2 diabetes, insulin resistance, and cardiovascular diseases. Furthermore, multiple studies support that these children, either born SGA or with a history of PA, may have earlier pubertal timing, which is also associated with various metabolic risks. This review aims to summarize the recent studies investigating the association between early infantile growth, the timing of puberty, and metabolic risks to expand our knowledge and gain more insight into the underlying pathophysiology.
Collapse
Affiliation(s)
- Youn Hee Jee
- Section on Growth, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD 20892, USA
- Division of Endocrinology and Center for Genetic Medicine Research, Children's National Hospital, Washington, DC 20010, USA
- Department of Pediatrics, George Washington University School of Medicine and Health Sciences, Washington, DC 20012, USA
| | - Sanjay Jumani
- Section on Growth, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD 20892, USA
| | - Veronica Mericq
- Institute of Maternal and Child Research, School of Medicine, University of Chile, Santiago 13101, Chile
| |
Collapse
|
36
|
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
|
37
|
Vanhorebeek I, Coppens G, Güiza F, Derese I, Wouters PJ, Joosten KF, Verbruggen SC, Van den Berghe G. Abnormal DNA methylation within genes of the steroidogenesis pathway two years after paediatric critical illness and association with stunted growth in height further in time. Clin Epigenetics 2023; 15:116. [PMID: 37468957 PMCID: PMC10354984 DOI: 10.1186/s13148-023-01530-9] [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: 05/24/2023] [Accepted: 07/04/2023] [Indexed: 07/21/2023] Open
Abstract
BACKGROUND Former critically ill children show an epigenetic age deceleration 2 years after paediatric intensive care unit (PICU) admission as compared with normally developing healthy children, with stunted growth in height 2 years further in time as physical correlate. This was particularly pronounced in children who were 6 years or older at the time of critical illness. As this age roughly corresponds to the onset of adrenarche and further pubertal development, a relation with altered activation of endocrine pathways is plausible. We hypothesised that children who have been admitted to the PICU, sex- and age-dependently show long-term abnormal DNA methylation within genes involved in steroid hormone synthesis or steroid sulphation/desulphation, possibly aggravated by in-PICU glucocorticoid treatment, which may contribute to stunted growth in height further in time after critical illness. RESULTS In this preplanned secondary analysis of the multicentre PEPaNIC-RCT and its follow-up, we compared the methylation status of genes involved in the biosynthesis of steroid hormones (aldosterone, cortisol and sex hormones) and steroid sulphation/desulphation in buccal mucosa DNA (Infinium HumanMethylation EPIC BeadChip) from former PICU patients at 2-year follow-up (n = 818) and healthy children with comparable sex and age (n = 392). Adjusting for technical variation and baseline risk factors and corrected for multiple testing (false discovery rate < 0.05), former PICU patients showed abnormal DNA methylation of 23 CpG sites (within CYP11A1, POR, CYB5A, HSD17B1, HSD17B2, HSD17B3, HSD17B6, HSD17B10, HSD17B12, CYP19A1, CYP21A2, and CYP11B2) and 4 DNA regions (within HSD17B2, HSD17B8, and HSD17B10) that were mostly hypomethylated. These abnormalities were partially sex- (1 CpG site) or age-dependent (7 CpG sites) and affected by glucocorticoid treatment (3 CpG sites). Finally, multivariable linear models identified robust associations of abnormal methylation of steroidogenic genes with shorter height further in time, at 4-year follow-up. CONCLUSIONS Children who have been critically ill show abnormal methylation within steroidogenic genes 2 years after PICU admission, which explained part of the stunted growth in height at 4-year follow-up. The abnormalities in DNA methylation may point to a long-term disturbance in the balance between active sex steroids and mineralocorticoids/glucocorticoids after paediatric critical illness, which requires further investigation.
Collapse
Affiliation(s)
- Ilse Vanhorebeek
- Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, Louvain, Herestraat 49, B-3000, Leuven, Belgium
| | - Grégoire Coppens
- Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, Louvain, Herestraat 49, B-3000, Leuven, Belgium
| | - Fabian Güiza
- Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, Louvain, Herestraat 49, B-3000, Leuven, Belgium
| | - Inge Derese
- Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, Louvain, Herestraat 49, B-3000, Leuven, Belgium
| | - Pieter J Wouters
- Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, Louvain, Herestraat 49, B-3000, Leuven, Belgium
| | - Koen F Joosten
- Division of Paediatric ICU, Department of Neonatal and Paediatric ICU, Erasmus Medical Centre, Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Sascha C Verbruggen
- Division of Paediatric ICU, Department of Neonatal and Paediatric ICU, Erasmus Medical Centre, Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Greet Van den Berghe
- Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, Louvain, Herestraat 49, B-3000, Leuven, Belgium.
| |
Collapse
|
38
|
Ma Data Analysis C, Xu H, Zhang X, Feng Data Analysis G, Shi L, Su Y, Yang L, Zhao R, Qiao J. Association of classic and 11-oxygenated androgens with polycystic ovaries and menstrual cycle prolongation in infertile women with PCOS. Clin Chim Acta 2023:117440. [PMID: 37311505 DOI: 10.1016/j.cca.2023.117440] [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: 02/17/2023] [Revised: 06/06/2023] [Accepted: 06/07/2023] [Indexed: 06/15/2023]
Abstract
BACKGROUND AND AIMS The etiology of polycystic ovary syndrome (PCOS) is unclear. This study aimed to evaluate the role of classic and 11-oxygenated (11oxyC19) androgens in two typical signs of PCOS, polycystic ovary morphology (PCOM) and menstrual cycle prolongation. MATERIALS AND METHODS A total of 462 infertile women with diagnosed PCOS and/or commonly accompanied metabolic disorders were recruited. Classic and 11oxyC19 androgens were determined with a sensitive high-performance liquid chromatography-differential mobility spectrometry tandem mass spectrometry apparatus. Least absolute shrinkage and selection operator logistic regression with fivefold cross-validation was applied to construct prediction models. RESULTS For PCOM, the most significant contributing androgen was testosterone (T), with the weight of 51.6%. The AUC of the prediction model was 0.824 in validation set. For menstrual cycle prolongation, androstenedione (A4) was the most significant contributing androgen with weights of 77.5%. The AUC the prediction model was less than 0.75. When including other variables, the most significant variable turned to be AMH both in PCOM and in menstrual cycle prolongation. CONCLUSION Androgens had more contribution in PCOM than in menstrual cycle prolongation. The classic androgen T or A4 contributed more than 11oxyC19 androgens. However, their contributions were diminished when other factors were considered, especially AMH.
Collapse
Affiliation(s)
- Congcong Ma Data Analysis
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China; National Clinical Research Center for Obstetrics and Gynecology, Beijing 100191, China; Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing 100191, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing 100191, China
| | - Huiyu Xu
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China; National Clinical Research Center for Obstetrics and Gynecology, Beijing 100191, China; Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing 100191, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing 100191, China
| | - Xianhua Zhang
- Department of Pharmacy, Peking University Third Hospital, Beijing 100191, China; Therapeutic Drug Monitoring and Clinical Toxicology Center of Peking University, Beijing 100191, China
| | - Guoshuang Feng Data Analysis
- Big Data Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Li Shi
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China; National Clinical Research Center for Obstetrics and Gynecology, Beijing 100191, China; Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing 100191, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing 100191, China
| | - Yuan Su
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China; National Clinical Research Center for Obstetrics and Gynecology, Beijing 100191, China; Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing 100191, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing 100191, China
| | - Li Yang
- Department of Pharmacy, Peking University Third Hospital, Beijing 100191, China; Therapeutic Drug Monitoring and Clinical Toxicology Center of Peking University, Beijing 100191, China
| | - Rongsheng Zhao
- Department of Pharmacy, Peking University Third Hospital, Beijing 100191, China; Therapeutic Drug Monitoring and Clinical Toxicology Center of Peking University, Beijing 100191, China.
| | - Jie Qiao
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China; National Clinical Research Center for Obstetrics and Gynecology, Beijing 100191, China; Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing 100191, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing 100191, China; Beijing Advanced Innovation Center for Genomics, Beijing 100871, People's Republic of China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, People's Republic of China.
| |
Collapse
|
39
|
Santos-Silva R, Fontoura M, Severo M, Santos AC. Dehydroepiandrosterone sulfate levels at 7 years old and cardio-metabolic factors at 10 and 13 years old - the generation XXI birth cohort. J Pediatr Endocrinol Metab 2023:jpem-2022-0593. [PMID: 37141272 DOI: 10.1515/jpem-2022-0593] [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: 11/23/2022] [Accepted: 04/17/2023] [Indexed: 05/05/2023]
Abstract
OBJECTIVES Premature adrenarche is often linked to a cluster of endocrine-metabolic risk factors. Our objective was to explore the association of dehydroepiandrosterone sulfate (DHEAS) levels at age 7 with cardio-metabolic traits at ages 10 and 13, independently of adiposity and pubertal stage. METHODS Longitudinal study of 603 individuals (301 girls/302 boys) from the Generation XXI birth cohort. DHEAS at age 7 was measured by immunoassay. Anthropometrics, pubertal staging, blood pressure, and metabolic outcomes were evaluated at ages 7, 10, and 13. Pearson correlations between DHEAS and cardio-metabolic traits (insulin, HOMA-IR, triglycerides, LDL-cholesterol, high-sensitivity C-reactive protein, and systolic and diastolic blood pressure) were computed. Path analysis was used to estimate the effect of DHEAS at age 7 on cardiometabolic traits at ages 10 and 13, adjusted for body mass index (BMI) z-score and Tanner stage. RESULTS DHEAS at age 7 correlated positively with insulin and HOMA-IR at ages 7 and 10 in both sexes, and at age 13 in girls, but not in boys. In girls, DHEAS levels at age 7 directly influenced HOMA-IR at age 13, controlling for BMI and Tanner stage. In boys, DHEAS at age 7 did not influence HOMA-IR at ages 10 and 13. DHEAS at age 7 did not influence the other cardio-metabolic outcomes analyzed. CONCLUSIONS DHEAS levels in mid-childhood have a positive longitudinal association with on insulin-resistance that persists, in girls, but not in boys, at least until age 13. No association was found regarding dyslipidemia, hypertension, or low-grade inflammation.
Collapse
Affiliation(s)
- Rita Santos-Silva
- EPIUnit, Instituto de Saúde Pública, Universidade do Porto, Porto, Portugal
- Unidade de Endocrinologia Pediátrica, Serviço de Pediatria, Centro Hospitalar Universitário de S. João, Porto, Portugal
- Departamento de Ginecologia-Obstetrícia e Pediatria, Faculdade de Medicina, Universidade do Porto, Porto, Portugal
- ITR - Laboratório para a Investigação Integrativa e Translacional em Saúde Populacional, Porto, Portugal
| | - Manuel Fontoura
- Unidade de Endocrinologia Pediátrica, Serviço de Pediatria, Centro Hospitalar Universitário de S. João, Porto, Portugal
- Departamento de Ginecologia-Obstetrícia e Pediatria, Faculdade de Medicina, Universidade do Porto, Porto, Portugal
| | - Milton Severo
- EPIUnit, Instituto de Saúde Pública, Universidade do Porto, Porto, Portugal
- ITR - Laboratório para a Investigação Integrativa e Translacional em Saúde Populacional, Porto, Portugal
- Departamento de Ciências da Saúde Pública e Forenses e Educação Médica, Faculdade de Medicina, Universidade do Porto, Porto, Portugal
| | - Ana Cristina Santos
- EPIUnit, Instituto de Saúde Pública, Universidade do Porto, Porto, Portugal
- ITR - Laboratório para a Investigação Integrativa e Translacional em Saúde Populacional, Porto, Portugal
- Departamento de Ciências da Saúde Pública e Forenses e Educação Médica, Faculdade de Medicina, Universidade do Porto, Porto, Portugal
| |
Collapse
|
40
|
Takeda Y, Demura M, Kometani M, Karashima S, Yoneda T, Takeda Y. Molecular and Epigenetic Control of Aldosterone Synthase, CYP11B2 and 11-Hydroxylase, CYP11B1. Int J Mol Sci 2023; 24:ijms24065782. [PMID: 36982850 PMCID: PMC10054571 DOI: 10.3390/ijms24065782] [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: 12/23/2022] [Revised: 03/15/2023] [Accepted: 03/15/2023] [Indexed: 03/30/2023] Open
Abstract
Aldosterone and cortisol serve important roles in the pathogenesis of cardiovascular diseases and metabolic disorders. Epigenetics is a mechanism to control enzyme expression by genes without changing the gene sequence. Steroid hormone synthase gene expression is regulated by transcription factors specific to each gene, and methylation has been reported to be involved in steroid hormone production and disease. Angiotensin II or potassium regulates the aldosterone synthase gene, CYP11B2. The adrenocorticotropic hormone controls the 11b-hydroxylase, CYP11B1. DNA methylation negatively controls the CYP11B2 and CYP11B1 expression and dynamically changes the expression responsive to continuous stimulation of the promoter gene. Hypomethylation status of the CYP11B2 promoter region is seen in aldosterone-producing adenomas. Methylation of recognition sites of transcription factors, including cyclic AMP responsive element binding protein 1 or nerve growth factor-induced clone B, diminish their DNA-binding activity. A methyl-CpG-binding protein 2 cooperates directly with the methylated CpG dinucleotides of CYP11B2. A low-salt diet, treatment with angiotensin II, and potassium increase the CYP11B2 mRNA levels and induce DNA hypomethylation in the adrenal gland. A close association between a low DNA methylation ratio and an increased CYP11B1 expression is seen in Cushing's adenoma and aldosterone-producing adenoma with autonomous cortisol secretion. Epigenetic control of CYP11B2 or CYP11B1 plays an important role in autonomic aldosterone or cortisol synthesis.
Collapse
Affiliation(s)
- Yoshimichi Takeda
- Endocrinology and Metabolism, Kanazawa University Hospital, Kanazawa 920-8641, Japan
- Department of Hygiene, Graduate School of Medical Science, Kanazawa University, Kanazawa 920-1192, Japan
| | - Masashi Demura
- Department of Hygiene, Graduate School of Medical Science, Kanazawa University, Kanazawa 920-1192, Japan
| | - Mitsuhiro Kometani
- Endocrinology and Metabolism, Kanazawa University Hospital, Kanazawa 920-8641, Japan
| | - Shigehiro Karashima
- Institute of Liberal Arts and Science, Kanazawa University, Kanazawa 920-1192, Japan
| | - Takashi Yoneda
- Institute of Liberal Arts and Science, Kanazawa University, Kanazawa 920-1192, Japan
| | - Yoshiyu Takeda
- Endocrinology and Metabolism, Kanazawa University Hospital, Kanazawa 920-8641, Japan
- Endocrine and Diabetes Center, Asanogawa General Hospital, Kanazawa 920-0811, Japan
| |
Collapse
|
41
|
Premature Pubarche: Time to Revise the Diagnostic Approach? J Clin Med 2023; 12:jcm12062187. [PMID: 36983190 PMCID: PMC10054674 DOI: 10.3390/jcm12062187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 03/08/2023] [Accepted: 03/09/2023] [Indexed: 03/18/2023] Open
Abstract
Premature pubarche (PP) could represent the first manifestation of non-classic congenital adrenal hyperplasia caused by 21 hydroxylase deficiency (NC21OHD) (10–30% of cases). In the last 20 years, the necessity of performing an ACTH test to diagnose NC21OHD in all cases with PP has been questioned, with conflicting results. This study aims to retrospectively evaluate the predictive value of the basal androgens, 17-OHP levels, and auxological features in suggesting the presence of NC21OHD and, thus, the need for a standard ACTH test to confirm the diagnosis. In all, 111 consecutive patients (87 females) with PP and advanced bone age underwent an ACTH test. Of these, 6/111 cases (1 male) were diagnosed with NC21OHD. The mean baseline 17 hydroxyprogesterone (17-OHP), dehydroepiandrosterone (DHEA), dehydroepiandrosterone sulfate (DHEA-S), delta 4 androstenedione (Δ4A), and testosterone serum levels were higher in NC21OHD patients than in the others (p < 0.05). We found three predictive features for NC21OHD: basal 17 OHP of >200 ng/mL, bone age advance of >2 years, and DHEA-S levels of >228 ng/mL with sensitivity and specificity of 83.3% and 97.1%, 83.3% and 65.7%, and 83.3% and 96.2%, respectively. Our data confirm that the prevalence of NC21OHD is low among patients with PP. Serum 17-OHP of >200 ng/mL could be helpful to decide, in most cases, which patients should undergo the ACTH test. Bone age advance represented an inadequately specific predictive marker of NC21OHD.
Collapse
|
42
|
Long-term impact of paediatric critical illness on the difference between epigenetic and chronological age in relation to physical growth. Clin Epigenetics 2023; 15:8. [PMID: 36639798 PMCID: PMC9840263 DOI: 10.1186/s13148-023-01424-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 01/07/2023] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Altered DNA-methylation affects biological ageing in adults and developmental processes in children. DNA-methylation is altered by environmental factors, trauma and illnesses. We hypothesised that paediatric critical illness, and the nutritional management in the paediatric intensive care unit (PICU), affects DNA-methylation changes that underly the developmental processes of childhood ageing. RESULTS We studied the impact of critical illness, and of the early use of parenteral nutrition (early-PN) versus late-PN, on "epigenetic age-deviation" in buccal mucosa of 818 former PICU-patients (406 early-PN, 412 late-PN) who participated in the 2-year follow-up of the multicentre PEPaNIC-RCT (ClinicalTrials.gov-NCT01536275), as compared with 392 matched healthy children, and assessed whether this relates to their impaired growth. The epigenetic age-deviation (difference between PedBE clock-estimated epigenetic age and chronological age) was calculated. Using bootstrapped multivariable linear regression models, we assessed the impact hereon of critical illness, and of early-PN versus late-PN. As compared with healthy children, epigenetic age of patients assessed 2 years after PICU-admission deviated negatively from chronological age (p < 0.05 in 51% of bootstrapped replicates), similarly in early-PN and late-PN groups. Next, we identified vulnerable subgroups for epigenetic age-deviation using interaction analysis. We revealed that DNA-methylation age-deceleration in former PICU-patients was dependent on age at time of illness (p < 0.05 for 83% of bootstrapped replicates), with vulnerability starting from 6 years onwards. Finally, we assessed whether vulnerability to epigenetic age-deviation could be related to impaired growth from PICU-admission to follow-up at 2 and 4 years. Multivariable repeated measures ANOVA showed that former PICU-patients, as compared with healthy children, grew less in height (p = 0.0002) and transiently gained weight (p = 0.0003) over the 4-year time course. Growth in height was more stunted in former PICU-patients aged ≥ 6-years at time of critical illness (p = 0.002) than in the younger patients. CONCLUSIONS As compared with healthy children, former PICU-patients, in particular those aged ≥ 6-years at time of illness, revealed epigenetic age-deceleration, with a physical correlate revealing stunted growth in height. Whether this vulnerability around the age of 6 years for epigenetic age-deceleration and stunted growth years later relates to altered endocrine pathways activated at the time of adrenarche requires further investigation.
Collapse
|
43
|
Graves LE, Torpy DJ, Coates PT, Alexander IE, Bornstein SR, Clarke B. Future directions for adrenal insufficiency: cellular transplantation and genetic therapies. J Clin Endocrinol Metab 2023; 108:1273-1289. [PMID: 36611246 DOI: 10.1210/clinem/dgac751] [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: 07/19/2022] [Revised: 12/21/2022] [Accepted: 12/23/2022] [Indexed: 01/09/2023]
Abstract
Primary adrenal insufficiency occurs in 1 in 5-7000 adults. Leading aetiologies are autoimmune adrenalitis in adults and congenital adrenal hyperplasia (CAH) in children. Oral replacement of cortisol is lifesaving, but poor quality of life, repeated adrenal crises and dosing uncertainty related to lack of a validated biomarker for glucocorticoid sufficiency, persists. Adrenocortical cell therapy and gene therapy may obviate many of the shortcomings of adrenal hormone replacement. Physiological cortisol secretion regulated by pituitary adrenocorticotropin, could be achieved through allogeneic adrenocortical cell transplantation, production of adrenal-like steroidogenic cells from either stem cells or lineage conversion of differentiated cells, or for CAH, gene therapy to replace or repair a defective gene. The adrenal cortex is a high turnover organ and thus failure to incorporate progenitor cells within a transplant will ultimately result in graft exhaustion. Identification of adrenocortical progenitor cells is equally important in gene therapy where new genetic material must be specifically integrated into the genome of progenitors to ensure a durable effect. Delivery of gene editing machinery and a donor template, allowing targeted correction of the 21-hydroxylase gene, has the potential to achieve this. This review describes advances in adrenal cell transplants and gene therapy that may allow physiological cortisol production for children and adults with primary adrenal insufficiency.
Collapse
Affiliation(s)
- Lara E Graves
- Institute of Endocrinology and Diabetes, The Children's Hospital at Westmead, Sydney, NSW, Australia
- Gene Therapy Research Unit, Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney and Sydney Children's Hospitals Network, Westmead, NSW, Australia
- Discipline of Child and Adolescent Health, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Westmead, Australia
| | - David J Torpy
- Endocrine and Metabolic Unit, Royal Adelaide Hospital, Adelaide, SA, Australia
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
| | - P Toby Coates
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
- Central Northern Adelaide Renal and Transplantation Service, Royal Adelaide Hospital, Adelaide, SA, Australia
| | - Ian E Alexander
- Gene Therapy Research Unit, Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney and Sydney Children's Hospitals Network, Westmead, NSW, Australia
- Discipline of Child and Adolescent Health, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Westmead, Australia
| | - Stefan R Bornstein
- University Clinic Carl Gustav Carus, Fetscherstrasse 74, 01307 Dresden, Germany
| | - Brigette Clarke
- Endocrine and Metabolic Unit, Royal Adelaide Hospital, Adelaide, SA, Australia
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
44
|
Bezen D, Tütüncüler Kökenli F, Dilek E, Ağ Seleci D, Erbaş H. Evaluation of Glucose Metabolism and Cardiovascular Risk Factors in Prepubertal Girls with Premature Pubarche. J Clin Res Pediatr Endocrinol 2022; 14:385-392. [PMID: 35633646 PMCID: PMC9724051 DOI: 10.4274/jcrpe.galenos.2022.2022-1-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE Premature pubarche (PP) is a risk factor for metabolic syndrome (MS). The aim was to evaluate if glucose-insulin metabolism, cardiovascular risk factors, familial cardiovascular risk factors (FCVRF) created a risk for insulin resistance (IR) and if PP was a risk factor alone for MS in normal weight prepubertal girls with PP. METHODS Thirty-five prepubertal, non-obese girls with PP with normal birth weight and 35 age-matched control girls were evaluated for FCVRF, anthropometric measurements, blood pressure, lipid profile, fasting blood glucose-insulin, hemoglobin A1c (HbA1c), sex hormone binding globulin (SHBG), leptin, adiponectin, tumor necrosis factor-alpha (TNF-α), androgen levels, and bone age. Oral glucose tolerance test was performed in PP participants. Homeostasis model of assessment of IR (HOMA-IR), fasting glucose/insulin ratio, atherogenic index (AI), and free androgen index (FAI) were calculated. PP participants were further stratified by FCVRF. RESULTS HbA1c, lipid profile, testosterone, leptin, adiponectin, TNF-α, HOMA-IR, glucose/insulin ratio, AI, and fasting glucose-insulin levels were similar. In the PP group FAI was significantly higher (p=0.001), whereas SHBG was significantly lower (p=0.010) than the control group. Leptin levels of FCVRF+ and FCVRF- subgroups were 15.2±9.1 and 9.7±7.2 ng/mL, respectively and the difference was significant (p=0.016). CONCLUSION As PP does not appear to be a risk factor alone for impaired glucose metabolism and IR in prepubertal non-obese girls with normal birth weight, it is our opinion that it is unnecessary to examine in detail such cases before puberty. Low SHBG levels in the PP group and high leptin levels in FCVRF+ subgroup might suggest that these may be predictive for MS in the future.
Collapse
Affiliation(s)
- Diğdem Bezen
- University of Health Sciences Turkey, İstanbul Prof. Dr. Cemil Taşçıoğlu City Hospital, Clinic of Pediatrics, Division of Pediatric Endocrinology, İstanbul, Turkey,* Address for Correspondence: University of Health Sciences Turkey, İstanbul Prof. Dr. Cemil Taşçıoğlu City Hospital, Clinic of Pediatrics, Division of Pediatric Endocrinology, İstanbul, Turkey Phone: +90 532 628 37 71 E-mail:
| | - Filiz Tütüncüler Kökenli
- Trakya University Faculty of Medicine, Department of Pediatrics, Clinic of Pediatric Endocrinology, Edirne, Turkey
| | - Emine Dilek
- Trakya University Faculty of Medicine, Department of Pediatrics, Clinic of Pediatric Endocrinology, Edirne, Turkey
| | - Didem Ağ Seleci
- Trakya University Faculty of Medicine, Department of Biochemistry, Edirne, Turkey
| | - Hakan Erbaş
- Trakya University Faculty of Medicine, Department of Biochemistry, Edirne, Turkey
| |
Collapse
|
45
|
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: 1.0] [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
|
46
|
Li W, Liu C, Yang Q, Zhou Y, Liu M, Shan H. Oxidative stress and antioxidant imbalance in ovulation disorder in patients with polycystic ovary syndrome. Front Nutr 2022; 9:1018674. [PMID: 36386912 PMCID: PMC9650267 DOI: 10.3389/fnut.2022.1018674] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Accepted: 10/07/2022] [Indexed: 07/30/2023] Open
Abstract
Polycystic ovary syndrome (PCOS) is a common reproductive endocrine disease that is characterized by oligo-ovulation or anovulation, hyperandrogenism, and polycystic ovaries observed using ultrasound with high clinical heterogeneity. At present, the etiology of PCOS is not clear but is thought to be related to genetic, metabolic, endocrine and environmental factors. Hyperandrogenism interacts with insulin resistance and overweight/obesity, forming a vicious cycle of mutual promotion and participating in the occurrence and progression of PCOS. Oxidative stress (OS) refers to the imbalance between the oxidation system and antioxidation system in the human body, which is associated with the occurrence and development of various diseases. Recent studies have shown that OS may be closely related to ovulation disorders in PCOS, and antioxidants can improve the oxidative stress state of PCOS. However, previous studies did not examine the effect of the interaction between OS and hyperandrogenism, insulin resistance or overweight/obesity on ovulation disorders in PCOS. This article reviews the interaction between OS and hyperandrogenism, insulin resistance and overweight/obesity; the effects of OS, hyperandrogenism, insulin resistance and overweight/obesity on ovulation disorders in PCOS; and the application of antioxidants in PCOS.
Collapse
Affiliation(s)
- Wenqian Li
- Department of Reproductive Medicine, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Chang Liu
- Department of Reproductive Medicine, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Qingmei Yang
- Department of Reproductive Medicine, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Ying Zhou
- Department of Reproductive Medicine, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Min Liu
- Department of Reproductive Medicine, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Hongying Shan
- Department of Reproductive Medicine, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
- Department of Obstetrics and Gynecology, Reproductive Medical Center, Peking University Third Hospital, Beijing, China
| |
Collapse
|
47
|
Lalli E, Figueiredo BC. Prolactin as an adrenocorticotropic hormone: Prolactin signalling is a conserved key regulator of sexually dimorphic adrenal gland function in health and disease. Bioessays 2022; 44:e2200109. [PMID: 36000778 DOI: 10.1002/bies.202200109] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 07/26/2022] [Accepted: 08/11/2022] [Indexed: 11/08/2022]
Abstract
A large number of previous reports described an effect of the pituitary hormone prolactin (PRL) on steroid hormone production by the adrenal cortex. However, those studies remained anecdotal and were never converted into a conceptual and mechanistic framework, let alone being translated into clinical care. In the light of our recently published landmark study where we described PRL signalling as a pivotal regulator of the sexually dimorphic adrenal phenotype in mouse and of adrenal androgen production in humans, we present here the overarching hypothesis that PRL signalling increases the activity of Steroidogenic Factor-1 (SF-1/NR5A1), a transcription factor that has an essential role in adrenal gland development and function, to regulate adrenal cortex growth and hormonal production in physiological and pathological conditions. PRL can then be considered as a bona fide adrenocorticotropic hormone synergizing with ACTH in the endocrine control of adrenal cortex function.
Collapse
Affiliation(s)
- Enzo Lalli
- EXPOGEN-CANCER CNRS International Research Project, 660 route des Lucioles, Sophia Antipolis, Valbonne, 06560, France.,Inserm, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France.,Université Côte d'Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France.,Pelé Pequeno Principe Research Institute, Curitiba, PR, Brazil
| | - Bonald C Figueiredo
- EXPOGEN-CANCER CNRS International Research Project, 660 route des Lucioles, Sophia Antipolis, Valbonne, 06560, France.,Pelé Pequeno Principe Research Institute, Curitiba, PR, Brazil
| |
Collapse
|
48
|
Ye L, Zhao Z, Ren H, Wang W, Zhou W, Zheng S, Han R, Zhang J, Li H, Wan Z, Tang C, Sun S, Wang W, Ning G. A Multiclassifier System to Identify and Subtype Congenital Adrenal Hyperplasia Based on Circulating Steroid Hormones. J Clin Endocrinol Metab 2022; 107:e3304-e3312. [PMID: 35512387 PMCID: PMC9282246 DOI: 10.1210/clinem/dgac271] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Indexed: 12/04/2022]
Abstract
CONTEXT Measurement of plasma steroids is necessary for diagnosis of congenital adrenal hyperplasia (CAH). We sought to establish an efficient strategy for detection and subtyping of CAH with a machine-learning algorithm. METHODS Clinical phenotype and genetic testing were used to provide CAH diagnosis and subtype. We profiled 13 major steroid hormones by liquid chromatography-tandem mass spectrometry. A multiclassifier system was established to distinguish 11β-hydroxylase deficiency (11βOHD), 17α-hydroxylase/17,20-lyase deficiency (17OHD), and 21α-hydroxylase deficiency (21OHD) in a discovery cohort (n = 226). It was then validated in an independent cohort (n = 111) and finally applied in a perspective cohort of 256 patients. The diagnostic performance on the basis of area under receiver operating characteristic curves (AUCs) was evaluated. RESULTS A cascade logistic regression model, we named the "Steroidogenesis Score", was able to discriminate the 3 most common CAH subtypes: 11βOHD, 17OHD, and 21OHD. In the perspective application cohort, the steroidogenesis score had a high diagnostic accuracy for all 3 subtypes, 11βOHD (AUC, 0.994; 95% CI, 0.983-1.000), 17OHD (AUC, 0.993; 95% CI, 0.985-1.000), and 21OHD (AUC, 0.979; 95% CI, 0.964-0.994). For nonclassic 21OHD patients, the tool presented with significantly higher sensitivity compared with measurement of basal 17α-hydroxyprogesterone (17OHP) (0.973 vs 0.840, P = 0.005) and was not inferior to measurement of basal vs stimulated 17OHP (0.973 vs 0.947, P = 0.681). CONCLUSIONS The steroidogenesis score was biochemically interpretable and showed high accuracy in identifying CAH patients, especially for nonclassic 21OHD patients, thus offering a standardized approach to diagnose and subtype CAH.
Collapse
Affiliation(s)
| | | | | | | | | | - Sichang Zheng
- Shanghai Clinical Center for Endocrine and Metabolic Diseases, Department of Endocrinology, Ruijin Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Rulai Han
- Shanghai Key Laboratory for Endocrine Tumors, Shanghai Institute of Endocrine and Metabolic Diseases, Shanghai, China
| | - Jie Zhang
- Shanghai Clinical Center for Endocrine and Metabolic Diseases, Department of Endocrinology, Ruijin Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Haorong Li
- Shanghai Clinical Center for Endocrine and Metabolic Diseases, Department of Endocrinology, Ruijin Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Zhihan Wan
- Shanghai Clinical Center for Endocrine and Metabolic Diseases, Department of Endocrinology, Ruijin Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Chao Tang
- Center for quantitative biology, Peking University, Beijing, China
| | - Shouyue Sun
- Correspondence: Shouyue Sun, MD, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Department of Endocrinology, Ruijin Hospital, Shanghai Jiaotong University, School of Medicine, 197 Ruijin 2nd Rd, Shanghai 200025, P.R. China.
| | - Weiqing Wang
- Weiqing Wang, MD, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Department of Endocrinology, Ruijin Hospital, Shanghai Jiaotong University, School of Medicine, 197 Ruijin 2nd Rd, Shanghai 200025, P.R. China.
| | - Guang Ning
- Shanghai Clinical Center for Endocrine and Metabolic Diseases, Department of Endocrinology, Ruijin Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, China
- Shanghai Key Laboratory for Endocrine Tumors, Shanghai Institute of Endocrine and Metabolic Diseases, Shanghai, China
| |
Collapse
|
49
|
Pignatti E, Altinkilic EM, Bräutigam K, Grössl M, Perren A, Zavolan M, Flück CE. Cholesterol Deprivation Drives DHEA Biosynthesis in Human Adrenals. Endocrinology 2022; 163:6588149. [PMID: 35583599 DOI: 10.1210/endocr/bqac076] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Indexed: 11/19/2022]
Abstract
Adrenarche is an early event in sexual maturation in prepubertal children and corresponds to the postnatal development of the adrenocortical zona reticularis (zR). However, the molecular mechanisms that govern the onset and maturation of zR remain unknown. Using tissue laser microdissection combined with transcript quantification and immunodetection, we showed that the human zR receives low levels of cholesterol in comparison with other adrenal layers. To model this metabolic condition, we challenged adrenal cells in vitro using cholesterol deprivation. This resulted in reprogramming the steroidogenic pathway toward inactivation of 3-beta-hydroxysteroid dehydrogenase type 2 (HSD3B2), increased CYB5A expression, and increased biosynthesis of dehydroepiandrosterone (DHEA), 3 key features of zR maturation during adrenarche. Finally, we found that cholesterol deprivation leads to decreased transcriptional activity of POU3F2, which normally stimulates the expression of HSD3B2 by directly binding to its promoter. These findings demonstrate that cholesterol deprivation can account, at least in part, for the acquisition of a zR-like androgenic program in humans.
Collapse
Affiliation(s)
- Emanuele Pignatti
- Division of Pediatric Endocrinology, Diabetology and Metabolism, Department of Pediatrics, University Hospital Inselspital, University of Bern, 3010 Bern, Switzerland
- Department for BioMedical Research, University Hospital Inselspital, University of Bern, 3010 Bern, Switzerland
| | - Emre Murat Altinkilic
- Division of Pediatric Endocrinology, Diabetology and Metabolism, Department of Pediatrics, University Hospital Inselspital, University of Bern, 3010 Bern, Switzerland
- Department for BioMedical Research, University Hospital Inselspital, University of Bern , 3010 Bern, Switzerland
| | | | - Michael Grössl
- Department for BioMedical Research, University Hospital Inselspital, University of Bern , 3010 Bern, Switzerland
- Department of Nephrology and Hypertension, Inselspital, Bern University Hospital, 3010, Bern, Switzerland
| | - Aurel Perren
- Institute of Pathology, University of Bern , 3008 Bern, Switzerland
| | | | - Christa E Flück
- Division of Pediatric Endocrinology, Diabetology and Metabolism, Department of Pediatrics, University Hospital Inselspital, University of Bern, 3010 Bern, Switzerland
- Department for BioMedical Research, University Hospital Inselspital, University of Bern, 3010 Bern, Switzerland
| |
Collapse
|
50
|
Madsen A, Almås B, Bruserud IS, Oehme NHB, Nielsen CS, Roelants M, Hundhausen T, Ljubicic ML, Bjerknes R, Mellgren G, Sagen JV, Juliusson PB, Viste K. Reference Curves for Pediatric Endocrinology: Leveraging Biomarker Z-Scores for Clinical Classifications. J Clin Endocrinol Metab 2022; 107:2004-2015. [PMID: 35299255 PMCID: PMC9202734 DOI: 10.1210/clinem/dgac155] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Indexed: 12/13/2022]
Abstract
CONTEXT Hormone reference intervals in pediatric endocrinology are traditionally partitioned by age and lack the framework for benchmarking individual blood test results as normalized z-scores and plotting sequential measurements onto a chart. Reference curve modeling is applicable to endocrine variables and represents a standardized method to account for variation with gender and age. OBJECTIVE We aimed to establish gender-specific biomarker reference curves for clinical use and benchmark associations between hormones, pubertal phenotype, and body mass index (BMI). METHODS Using cross-sectional population sample data from 2139 healthy Norwegian children and adolescents, we analyzed the pubertal status, ultrasound measures of glandular breast tissue (girls) and testicular volume (boys), BMI, and laboratory measurements of 17 clinical biomarkers modeled using the established "LMS" growth chart algorithm in R. RESULTS Reference curves for puberty hormones and pertinent biomarkers were modeled to adjust for age and gender. Z-score equivalents of biomarker levels and anthropometric measurements were compiled in a comprehensive beta coefficient matrix for each gender. Excerpted from this analysis and independently of age, BMI was positively associated with female glandular breast volume (β = 0.5, P < 0.001) and leptin (β = 0.6, P < 0.001), and inversely correlated with serum levels of sex hormone-binding globulin (SHBG) (β = -0.4, P < 0.001). Biomarker z-score profiles differed significantly between cohort subgroups stratified by puberty phenotype and BMI weight class. CONCLUSION Biomarker reference curves and corresponding z-scores provide an intuitive framework for clinical implementation in pediatric endocrinology and facilitate the application of machine learning classification and covariate precision medicine for pediatric patients.
Collapse
Affiliation(s)
- Andre Madsen
- Correspondence: André Madsen, PhD, Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, N-5021 Bergen, Norway.
| | - Bjørg Almås
- Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
| | - Ingvild S Bruserud
- Faculty of Health, VID Specialized University, Bergen, Norway
- Department of Pediatrics, Haukeland University Hospital, Bergen, Norway
| | | | - Christopher Sivert Nielsen
- Department of Chronic Diseases and Ageing, Norwegian Institute of Public Health, Oslo, Norway
- Department of Pain Management and Research, Oslo University Hospital, Oslo, Norway
| | - Mathieu Roelants
- Environment and Health, Department of Public Health and Primary Care, KU Leuven, University of Leuven, Leuven, Belgium
| | - Thomas Hundhausen
- Department of Medical Biochemistry, Southern Norway Hospital Trust, Kristiansand, Norway
- Department of Natural Sciences, University of Agder, Kristiansand, Norway
| | - Marie Lindhardt Ljubicic
- Department of Growth and Reproduction, Rigshospitalet, University of Copenhagen, and International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Copenhagen, Denmark
| | - Robert Bjerknes
- Department of Pediatrics, Haukeland University Hospital, Bergen, Norway
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Gunnar Mellgren
- Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Mohn Nutrition Research Laboratory, University of Bergen, Bergen, Norway
| | - Jørn V Sagen
- Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | | | | |
Collapse
|