1
|
Dumesic DA, Rasouli MA, Katz JD, Lu GG, Dharanipragada D, Turcu AF, Grogan TR, Flores KE, Magyar CE, Abbott DH, Chazenbalk GD. The Subcutaneous Adipose Microenvironment as a Determinant of Body Fat Development in Polycystic Ovary Syndrome. J Endocr Soc 2024; 8:bvae162. [PMID: 39345868 PMCID: PMC11424691 DOI: 10.1210/jendso/bvae162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Indexed: 10/01/2024] Open
Abstract
Context Adipose steroid metabolism modifies body fat development in polycystic ovary syndrome (PCOS). Objective To determine whether subcutaneous (SC) abdominal adipose aldo-keto reductase 1C3 (AKR1C3; a marker of testosterone generation) is increased in normal-weight women with PCOS vs age- and body mass index (BMI)-matched normoandrogenic ovulatory women (controls) and is related to SC abdominal adipose activator protein-1 (AP-1; a marker of adipocyte differentiation) and/or androgen receptor (AR) protein expression in predicting fat accretion. Design Prospective cohort study. Setting Academic center. Patients Eighteen normal-weight PCOS women; 17 age- and BMI-matched controls. Interventions Circulating hormone/metabolic determinations, intravenous glucose tolerance testing, total body dual-energy x-ray absorptiometry, SC abdominal fat biopsy, immunohistochemistry. Main Outcome Measures Clinical characteristics, hormonal concentrations, body fat distribution, SC adipose AKR1C3, AR, and AP-1 protein expression. Results Women with PCOS had significantly higher serum androgen levels and greater android/gynoid fat mass ratios than controls. SC adipose AKR1C3, AR, and AP-1 protein expressions were comparable between the study groups, but groups differed in correlations. In PCOS women vs controls, SC adipose AKR1C3 protein expression correlated positively with android and gynoid fat masses and negatively with SC adipose AP-1 protein expression. SC adipose AR protein expression correlated negatively with fasting serum free fatty acid and high-density lipoprotein levels. In both study groups, SC adipose AKR1C3 protein expression negatively correlated with serum cortisol levels. Conclusion In normal-weight PCOS women, SC abdominal adipose AKR1C3 protein expression, in combination with intra-adipose AP-1 and AR-dependent events, predicts fat accretion in the presence of physiological cortisol levels.
Collapse
Affiliation(s)
- Daniel A Dumesic
- Department of Obstetrics and Gynecology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Melody A Rasouli
- Department of Obstetrics and Gynecology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jessica D Katz
- Department of Obstetrics and Gynecology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Gwyneth G Lu
- Department of Obstetrics and Gynecology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Devyani Dharanipragada
- Department of Obstetrics and Gynecology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Adina F Turcu
- Division of Metabolism, Endocrinology, Nutrition and Diabetes, University of Michigan, Ann Arbor, MI 48103, USA
| | - Tristan R Grogan
- Department of Medicine Statistics Core, University of California, Los Angeles, Los Angeles, CA 90024, USA
| | - Kimberly E Flores
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Clara E Magyar
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - David H Abbott
- Department of Obstetrics and Gynecology, Wisconsin National Primate Research Center, University of Wisconsin, Madison, Madison, WI 53715, USA
| | - Gregorio D Chazenbalk
- Department of Obstetrics and Gynecology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
2
|
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
|
3
|
Dubinski I, Bechtold-Dalla Pozza S, Bidlingmaier M, Hawley J, Keevil B, Kunz S, Nowotny HF, Reisch N, Schiergens K, Tschaidse L, Schmidt H. Diurnal 11-ketotestosterone and 17-hydroxyprogesterone saliva profiles in paediatric classical congenital adrenal hyperplasia. J Pediatr Endocrinol Metab 2024; 37:419-424. [PMID: 38557593 DOI: 10.1515/jpem-2024-0027] [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: 01/13/2024] [Accepted: 03/15/2024] [Indexed: 04/04/2024]
Abstract
OBJECTIVES The most suitable biochemical markers for therapy adjustment in patients with congenital adrenal hyperplasia are controversial. 11-Oxygenated androgens are a promising new approach. The objective of this study was to investigate the diurnal rhythm of 11-ketotestosterone in children and adolescents in saliva and to correlate it with salivary 17-hydroxyprogesterone. METHODS Fifty-one samples of steroid day-profiles from 17 patients were additionally analysed for 11-ketotestosterone, retrospectively. All patients were treated in our university outpatient clinic for paediatric endocrinology between 2020 and 2022. Steroid day-profiles of 17 patients could be examined. The cohort showed a balanced sex ratio. The median age was 13 years. The measurements for 17-hydroxyprogesterone were carried out during routine care by immunoassay. The measurements of 11-ketotestosterone were performed from frozen saliva samples using an implemented in-house protocol for liquid chromatography-tandem mass spectrometry (LC-MS/MS). The most important outcome were the absolute values for 11-ketotestosterone, their diurnal rhythmicity and the correlation with 17-hydroxyprogesterone. RESULTS Both steroids show a circadian diurnal rhythm. 17-hydroxyprogesterone and 11-ketotestosterone correlate significantly. 11-Ketotestosterone showed a positive correlation with BMI at all times of the day. CONCLUSIONS 11-Ketotestosterone shows circadian rhythmicity in our cohort and correlates with 17-hydroxyprogesterone. These findings serve as an important basis for prospective research into 11-oxygenated androgens as therapeutic markers in paediatrics. However, 11-ketotestosterone appears to be very dependent on BMI.
Collapse
Affiliation(s)
- Ilja Dubinski
- Department of Paediatrics, Division of Paediatric Endocrinology, 74939 Dr. von Hauner Children's Hospital, University Hospital, LMU Munich , Munich, Germany
| | - Susanne Bechtold-Dalla Pozza
- Department of Paediatrics, Division of Paediatric Endocrinology, 74939 Dr. von Hauner Children's Hospital, University Hospital, LMU Munich , Munich, Germany
| | - Martin Bidlingmaier
- Department of Medicine IV, 74939 University Hospital, LMU Munich , Munich, Germany
| | - James Hawley
- Department of Clinical Biochemistry, 5293 Manchester University Foundation NHS Trust, Manchester Academic Health Sciences Centre , Manchester, UK
| | - Brian Keevil
- Department of Clinical Biochemistry, 5293 Manchester University Foundation NHS Trust, Manchester Academic Health Sciences Centre , Manchester, UK
| | - Sonja Kunz
- Department of Medicine IV, 74939 University Hospital, LMU Munich , Munich, Germany
| | | | - Nicole Reisch
- Department of Medicine IV, 74939 University Hospital, LMU Munich , Munich, Germany
| | - Katharina Schiergens
- Department of Paediatrics, Division of Paediatric Endocrinology, 74939 Dr. von Hauner Children's Hospital, University Hospital, LMU Munich , Munich, Germany
| | - Lea Tschaidse
- Department of Medicine IV, 74939 University Hospital, LMU Munich , Munich, Germany
| | - Heinrich Schmidt
- Department of Paediatrics, Division of Paediatric Endocrinology, 74939 Dr. von Hauner Children's Hospital, University Hospital, LMU Munich , Munich, Germany
| |
Collapse
|
4
|
Yazawa T, Imamichi Y, Sato T, Ida T, Umezawa A, Kitano T. Diversity of Androgens; Comparison of Their Significance and Characteristics in Vertebrate Species. Zoolog Sci 2024; 41:77-86. [PMID: 38587520 DOI: 10.2108/zs230064] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Accepted: 10/31/2023] [Indexed: 04/09/2024]
Abstract
Androgen(s) is one of the sex steroids that are involved in many physiological phenomena of vertebrate species. Although androgens were originally identified as male sex hormones, it is well known now that they are also essential in females. As in the case of other steroid hormones, androgen is produced from cholesterol through serial enzymatic reactions. Although testis is a major tissue to produce androgens in all species, androgens are also produced in ovary and adrenal (interrenal tissue). Testosterone is the most common and famous androgen. It represents a major androgen both in males and females of almost vertebrate species. In addition, testosterone is a precursor for producing significant androgens such as11-ketotestosterone, 5α-dihydrotestosterone, 11-ketodihydrotestosterones and 15α-hydroxytestosterone in a species- or sex-dependent manner for their homeostasis. In this article, we will review the significance and characteristics of these androgens, following a description of the history of testosterone discovery and its synthetic pathways.
Collapse
Affiliation(s)
- Takashi Yazawa
- Department of Biochemistry, Asahikawa Medical University, Hokkaido 078-8510, Japan,
| | - Yoshitaka Imamichi
- Faculty of Marine Science and Technology, Fukui Prefectural University, Fukui 917-0003, Japan,
| | - Takahiro Sato
- Division of Molecular Genetics, Institute of Life Sciences, Kurume University, Fukuoka 830-0011, Japan
| | - Takanori Ida
- Center for Animal Disease Control, Frontier Science Research Center, University of Miyazaki, Miyazaki 889-1692, Japan
| | - Akihiro Umezawa
- National Center for Child Health and Development Research Institute, Tokyo 157-8535, Japan
| | - Takeshi Kitano
- Department of Biological Sciences, Graduate School of Science and Technology, Kumamoto University, Kumamoto 860-8555, Japan
| |
Collapse
|
5
|
Wang K, Li Y, Chen Y. Androgen excess: a hallmark of polycystic ovary syndrome. Front Endocrinol (Lausanne) 2023; 14:1273542. [PMID: 38152131 PMCID: PMC10751361 DOI: 10.3389/fendo.2023.1273542] [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: 08/06/2023] [Accepted: 11/22/2023] [Indexed: 12/29/2023] Open
Abstract
Polycystic ovarian syndrome (PCOS) is a metabolic, reproductive, and psychological disorder affecting 6-20% of reproductive women worldwide. However, there is still no cure for PCOS, and current treatments primarily alleviate its symptoms due to a poor understanding of its etiology. Compelling evidence suggests that hyperandrogenism is not just a primary feature of PCOS. Instead, it may be a causative factor for this condition. Thus, figuring out the mechanisms of androgen synthesis, conversion, and metabolism is relatively important. Traditionally, studies of androgen excess have largely focused on classical androgen, but in recent years, adrenal-derived 11-oxygenated androgen has also garnered interest. Herein, this Review aims to investigate the origins of androgen excess, androgen synthesis, how androgen receptor (AR) signaling mediates adverse PCOS traits, and the role of 11-oxygenated androgen in the pathophysiology of PCOS. In addition, it provides therapeutic strategies targeting hyperandrogenism in PCOS.
Collapse
Affiliation(s)
- Kexin Wang
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yanhua Li
- Department of General Practice, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Yu Chen
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
6
|
Sarafoglou K, Merke DP, Reisch N, Claahsen-van der Grinten H, Falhammar H, Auchus RJ. Interpretation of Steroid Biomarkers in 21-Hydroxylase Deficiency and Their Use in Disease Management. J Clin Endocrinol Metab 2023; 108:2154-2175. [PMID: 36950738 PMCID: PMC10438890 DOI: 10.1210/clinem/dgad134] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 03/07/2023] [Indexed: 03/24/2023]
Abstract
The most common form of congenital adrenal hyperplasia is 21-hydroxylase deficiency (21OHD), which in the classic (severe) form occurs in roughly 1:16 000 newborns worldwide. Lifelong treatment consists of replacing cortisol and aldosterone deficiencies, and supraphysiological dosing schedules are typically employed to simultaneously attenuate production of adrenal-derived androgens. Glucocorticoid titration in 21OHD is challenging as it must balance the consequences of androgen excess vs those from chronic high glucocorticoid exposure, which are further complicated by interindividual variability in cortisol kinetics and glucocorticoid sensitivity. Clinical assessment and biochemical parameters are both used to guide therapy, but the specific purpose and goals of each biomarker vary with age and clinical context. Here we review the approach to medication titration for children and adults with classic 21OHD, with an emphasis on how to interpret adrenal biomarker values in guiding this process. In parallel, we illustrate how an understanding of the pathophysiologic and pharmacologic principles can be used to avoid and to correct complications of this disease and consequences of its management using existing treatment options.
Collapse
Affiliation(s)
- Kyriakie Sarafoglou
- Department of Pediatrics, Division of Pediatric Endocrinology, University of Minnesota Medical School, Minneapolis, MN 55454, USA
- Department of Experimental and Clinical Pharmacology, University of Minnesota College of Pharmacy, Minneapolis, MN 55455, USA
| | - Deborah P Merke
- Department of Pediatrics, National Institutes of Health Clinical Center, Bethesda, MD 20892, USA
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, USA
| | - Nicole Reisch
- Medizinische Klinik and Poliklinik IV, Klinikum der Universität München, 80336 Munich, Germany
| | - Hedi Claahsen-van der Grinten
- Department of Pediatrics, Amalia Children's Hospital, Radboud University Medical Center, 6500 HB, Nijmegen, The Netherlands
| | - Henrik Falhammar
- Department of Molecular Medicine and Surgery, Karolinska Institutet, SE-17176, Stockholm, Sweden
- Department of Endocrinology, Karolinska University Hospital, SE-17176, Stockholm, Sweden
| | - Richard J Auchus
- Departments of Pharmacology and Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| |
Collapse
|
7
|
Dumesic DA, Turcu AF, Liu H, Grogan TR, Abbott DH, Lu G, Dharanipragada D, Chazenbalk GD. Interplay of Cortisol, Testosterone, and Abdominal Fat Mass in Normal-weight Women With Polycystic Ovary Syndrome. J Endocr Soc 2023; 7:bvad079. [PMID: 37404244 PMCID: PMC10315644 DOI: 10.1210/jendso/bvad079] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Indexed: 07/06/2023] Open
Abstract
Context Ovarian and adrenal steroidogenesis underlie endocrine-metabolic dysfunction in polycystic ovary syndrome (PCOS). Adipocytes express aldo-keto reductase 1C3 and type 1 11β-hydroxysteroid dehydrogenase, which modulate peripheral androgen and cortisol production. Objectives To compare serum adrenal steroids, including 11-oxygenated androgens (11-oxyandrogens), cortisol, and cortisone between normal-weight women with PCOS and body mass index- and age-matched ovulatory women with normal-androgenic profiles (controls), and assess whether adrenal steroids associate with abdominal adipose deposition. Design Prospective, cross-sectional, cohort study. Setting Academic medical center. Patients Twenty normal-weight women with PCOS and 20 body mass index-/age-matched controls. Interventions Blood sampling, IV glucose tolerance testing, and total-body dual-energy x-ray absorptiometry. Main Outcome Measures Clinical characteristics, hormonal concentrations, and body fat distribution. Results Women with PCOS had higher serum total/free testosterone (T) and androstenedione (A4) levels and a greater android/gynoid fat mass than controls (androgens P < .001; android/gynoid fat mass ratio, P = .026). Serum total/free T and A4 levels correlated positively with android/gynoid fat mass ratio in all women combined (P < .025, all values). Serum 11ß-hydroxyA4, 11-ketoA4, 11ß-hydroxyT, 11-ketoT, cortisol, and cortisone levels were comparable between female types and unrelated to body fat distribution. Serum 11-oxyandrogens correlated negatively with % total body fat, but lost significance adjusting for cortisol. Serum cortisol levels, however, correlated inversely with android fat mass (P = .021), with a trend toward reduced serum cortisol to cortisone ratio in women with PCOS vs controls (P = .075), suggesting diminished 11β-hydroxysteroid dehydrogenase activity. Conclusion Reduced cortisol may protect against preferential abdominal fat mass in normal-weight PCOS women with normal serum 11-oxyandrogens.
Collapse
Affiliation(s)
- Daniel A Dumesic
- Correspondence: Daniel A Dumesic, MD, Department Obstetrics and Gynecology, David Geffen School of Medicine at UCLA, 10833 Le Conte Ave, Room 22-178 CHS, Los Angeles, CA 90095, USA.
| | - Adina F Turcu
- Division of Metabolism, Endocrinology, Nutrition and Diabetes, University of Michigan, Ann Arbor, MI 48103, USA
| | - Haiping Liu
- Division of Metabolism, Endocrinology, Nutrition and Diabetes, University of Michigan, Ann Arbor, MI 48103, USA
| | - Tristan R Grogan
- Department of Medicine Statistics Core, University of California, Los Angeles, Los Angeles, CA 90024, USA
| | - David H Abbott
- Department of Obstetrics and Gynecology, Wisconsin National Primate Research Center, University of WI-Madison, Madison, WI 53715, USA
| | - Gwyneth Lu
- Department of Obstetrics and Gynecology, University of California, Los Angeles, 1Los Angeles, CA 90095, USA
| | - Devyani Dharanipragada
- Department of Obstetrics and Gynecology, University of California, Los Angeles, 1Los Angeles, CA 90095, USA
| | - Gregorio D Chazenbalk
- Department of Obstetrics and Gynecology, University of California, Los Angeles, 1Los Angeles, CA 90095, USA
| |
Collapse
|
8
|
Ikegawa K, Hasegawa Y. Adrenal gland involvement in 11-ketotestosterone production analyzed using LC-MS/MS. Front Endocrinol (Lausanne) 2023; 14:1051195. [PMID: 36742384 PMCID: PMC9895773 DOI: 10.3389/fendo.2023.1051195] [Citation(s) in RCA: 2] [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: 09/22/2022] [Accepted: 01/09/2023] [Indexed: 01/22/2023] Open
Abstract
INTRODUCTION 11-ketotestosterone (11KT), which is derived by the bioconversion of testosterone via 11β-hydroxytestosterone (11OHT), is a potent agonist of the human androgen receptor. The adrenal gland is considered an important organ in 11KT production because CYP11B1, which catalyzes testosterone to 11OHT, is expressed in the adrenal glands. The present study aimed to demonstrate adrenal gland involvement in 11KT production in prepubertal children, a topic which has not yet been addressed by any previous studies. METHODS Three, retrospective, observational studies were performed. Study 1 enrolled patients aged 8 months to 7 years with severe Kawasaki disease (KD) who were treated with mPSL pulse. Studies 2 and 3 included patients who had received a corticotropin-releasing hormone (CRH) stimulation test and adrenocorticotropic hormone (ACTH) stimulation test, respectively. Samples were collected before and after treatment or drug administration, and serum 11KT, 11OHT, and other 11-oxygenated androgens were measured by LC-MS/MS. Steroid hormone values before and after medication were analyzed using the Wilcoxon signed rank test. RESULTS Studies 1, 2, and 3 included twenty patients with severe KD, eight patients with a CRH stimulation test, and eight patients with an ACTH stimulation test, respectively. Study 1 demonstrated that the median (IQR) 11KT level was significantly higher before, than after, mPSL pulse (0.39 (0.28-0.47) nmol/L versus 0.064 (0.012-0.075) nmol/L; P < 0.001). Studies 2 and 3 indicated no significant difference in the median 11KT value before and after the CRH or ACTH stimulation test while the 11OHT value was significantly higher after the test. CONCLUSION In conclusion, the mediation of 11KT production by ACTH demonstrated the importance of the adrenal glands in the synthesis of this androgen in prepubertal children.
Collapse
Affiliation(s)
- Kento Ikegawa
- Division of Endocrinology and Metabolism, Tokyo Metropolitan Children’s Medical Center, Tokyo, Japan
- Clinical Research Support Center, Tokyo Metropolitan Children’s Medical Center, Tokyo, Japan
- *Correspondence: Kento Ikegawa,
| | - Yukihiro Hasegawa
- Division of Endocrinology and Metabolism, Tokyo Metropolitan Children’s Medical Center, Tokyo, Japan
- Department of Pediatrics, Keio University of School of Medicine, Tokyo, Japan
| |
Collapse
|
9
|
Snaterse G, Hofland J, Lapauw B. The role of 11-oxygenated androgens in prostate cancer. ENDOCRINE ONCOLOGY (BRISTOL, ENGLAND) 2023; 3:e220072. [PMID: 37434644 PMCID: PMC10305623 DOI: 10.1530/eo-22-0072] [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: 02/09/2023] [Accepted: 03/13/2023] [Indexed: 07/13/2023]
Abstract
11-oxygenated androgens are a class of steroids capable of activating the androgen receptor (AR) at physiologically relevant concentrations. In view of the AR as a key driver of prostate cancer (PC), these steroids are potential drivers of disease and progression. The 11-oxygenated androgens are adrenal-derived, and persist after androgen deprivation therapy (ADT), the mainstay treatment for advanced PC. Consequently, these steroids are of particular interest in the castration-resistant prostate cancer (CRPC) setting. The principal androgen of the pathway, 11-ketotestosterone (11KT), is a potent AR agonist and the predominant circulating active androgen in CRPC patients. Additionally, several precursor steroids are present in the circulation which can be converted into active androgens by steroidogenic enzymes present in PC cells. In vitro evidence suggests that adaptations frequently observed in CRPC favour the intratumoral accumulation of 11-oxygenated androgens in particular. Still, apparent gaps in our understanding of the physiology and role of the 11-oxygenated androgens remain. In particular, in vivo and clinical evidence supporting these in vitro findings is limited. Despite recent advances, a comprehensive assessment of intratumoral concentrations has not yet been performed. The exact contribution of the 11-oxygenated androgens to CRPC progression therefore remains unclear. This review will focus on the current evidence linking the 11-oxygenated androgens to PC, will highlight current gaps in our knowledge, and will provide insight into the potential clinical importance of the 11-oxygenated androgens in the CRPC setting based on the current evidence.
Collapse
Affiliation(s)
- Gido Snaterse
- Department of Endocrinology and Metabolism, Ghent University Hospital, Ghent, Belgium
| | - Johannes Hofland
- Section of Endocrinology, Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Bruno Lapauw
- Department of Endocrinology and Metabolism, Ghent University Hospital, Ghent, Belgium
| |
Collapse
|
10
|
Zeidler R, Biemann R, Ceglarek U, Kratzsch J, Isermann B, Gaudl A. Inclusion of 11-Oxygenated Androgens in a Clinical Routine LC-MS/MS Setup for Steroid Hormone Profiling. Int J Mol Sci 2022; 24:ijms24010539. [PMID: 36613983 PMCID: PMC9820169 DOI: 10.3390/ijms24010539] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/21/2022] [Accepted: 12/23/2022] [Indexed: 12/31/2022] Open
Abstract
11-Oxygenated androgens (11-OAs) are being discussed as potential biomarkers in diagnosis and therapy control of disorders with androgen excess such as congenital adrenal hyperplasia and polycystic ovary syndrome. However, quantification of 11-OAs by liquid chromatography-tandem mass spectrometry (LC-MS/MS) still relies on extensive sample preparation including liquid-liquid extraction, derivatization and partial long runtimes, which is unsuitable for high-throughput analysis under routine laboratory settings. For the first time, an established online-solid-phase extraction-LC-MS/MS (online-SPE-LC-MS/MS) method for the quantitation of seven serum steroids in daily routine use was extended and validated to include 11-ketoandrostenedione, 11-ketotestosterone, 11β-hydroxyandrostenedione and 11β-hydroxytestosterone. Combining a simple protein precipitation step with fast chromatographic separation and ammonium fluoride-modified ionization resulted in a high-throughput method (6.6 min run time) featuring lower limits of quantification well below endogenous ranges (63-320 pmol/L) with recoveries between 85% and 117% (CVs ≤ 15%). Furthermore, the ability of this method to distinguish between adrenal and gonadal androgens was shown by comparing 11-OAs in patients with hyperandrogenemia to healthy controls. Due to the single shot multiplex design of the method, potential clinically relevant ratios of 11-OAs and corresponding androgens were readily available. The fully validated method covering endogenous concentration levels is ready to investigate the diagnostic values of 11-OAs in prospective studies and clinical applications.
Collapse
|
11
|
Hoseinzadeh M, Molavi N, Norouzi M, Aghaei S, Zeinalian M, Hashemipour M, Tabatabaiefar MA. A Novel Homozygous Pathogenic Variant in CYP11B1 in a Female Iranian Patient with 11B Hydroxylase Deficiency. Lab Med 2022:6885678. [DOI: 10.1093/labmed/lmac141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Abstract
Objective
Congenital adrenal hyperplasia (CAH) addresses a number of autosomal recessive disorders characterized by the enzyme defects in steroid hormones biosynthesis. The second common form of CAH is caused by mutations in the CYP11B1 gene. Here, we reveal a novel mutation in the CYP11B1 gene related to the 11βOHD phenotype.
Methods and Results
Sequence analysis of the CYP11B1 gene in a 19-year-old Iranian woman with the 11βOHD phenotype was performed. In silico analysis and molecular docking were done. A novel missense homozygous variant c.1351C > T (p.L451F) in the CYP11B1 gene was identified in the patient and, according to American College of Medical Genetics and Genomics criteria, was categorized as likely pathogenic. Protein docking showed destructive effects of the variant on the CYP11B1 protein-ligand interactions.
Conclusion
This study broadens the CYP11B1 mutation spectrum and introduces the novel p.L451F likely pathogenic variant leading to destructive effects on protein-ligand interactions. Our results provide reliable information for genetic counseling and molecular diagnostics of CAH.
Collapse
Affiliation(s)
- Marziyeh Hoseinzadeh
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences , Isfahan , Iran
| | - Newsha Molavi
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences , Isfahan , Iran
| | - Mahnaz Norouzi
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences , Isfahan , Iran
| | - Shahrzad Aghaei
- Department of Molecular Medicine, School of Advanced Technologies, Sahrekord University of Medical Sciences , Shahrekord , Iran
| | - Mehrdad Zeinalian
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences , Isfahan , Iran
| | - Mahin Hashemipour
- Metabolic Liver Disease Research Center, Isfahan University of Medical Sciences, Isfahan , Iran
| | - Mohammad Amin Tabatabaiefar
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences , Isfahan , Iran
- Department of Pediatrics, School of Medicine, Isfahan University of Medical Sciences , Isfahan , Iran
- Pediatric Inherited Diseases Research Center, Research Institute for Primordial Prevention of Noncommunicable Disease, Isfahan University of Medical Sciences , Isfahan , Iran
| |
Collapse
|
12
|
Celino-Brady FT, Breves JP, Seale AP. Sex-specific responses to growth hormone and luteinizing hormone in a model teleost, the Mozambique tilapia. Gen Comp Endocrinol 2022; 329:114119. [PMID: 36029822 DOI: 10.1016/j.ygcen.2022.114119] [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: 06/09/2022] [Revised: 08/21/2022] [Accepted: 08/22/2022] [Indexed: 11/20/2022]
Abstract
Across the vertebrate lineage, sexual dimorphism in body size is a common phenomenon that results from trade-offs between growth and reproduction. To address how key hormones that regulate growth and reproduction interact in teleost fishes, we studied Mozambique tilapia (Oreochromis mossambicus) to determine whether the activities of luteinizing hormone (Lh) are modulated by growth hormone (Gh), and conversely, whether targets of Gh are affected by the presence of Lh. In particular, we examined how gonadal morphology and specific gene transcripts responded to ovine GH (oGH) and/or LH (oLH) in hypophysectomized male and female tilapia. Hypophysectomized females exhibited a diminished gonadosomatic index (GSI) concomitant with ovarian follicular atresia. The combination of oGH and oLH restored GSI and ovarian morphology to conditions observed in sham-operated controls. A similar pattern was observed for GSI in males. In control fish, gonadal gh receptor (ghr2) and estrogen receptor β (erβ) expression was higher in females versus males. A combination of oGH and oLH restored erβ and arβ in females. In males, testicular insulin-like growth factor 3 (igf3) expression was reduced following hypophysectomy and subsequently restored to control levels by either oGH or oLH. By contrast, the combination of both hormones was required to recover ovarian igf3 expression in females. In muscle, ghr2 expression was more responsive to oGH in males versus females. In the liver of hypophysectomized males, igf2 expression was diminished by both oGH and oLH; there was no effect of hypophysectomy, oGH, or oLH on igf2 expression in females. Collectively, our results indicate that gene transcripts associated with growth and reproduction exhibit sex-specific responses to oGH and oLH. These responses reflect, at least in part, how hormones mediate trade-offs between growth and reproduction, and thus sexual dimorphism, in teleost fishes.
Collapse
Affiliation(s)
- Fritzie T Celino-Brady
- Department of Human Nutrition, Food and Animal Sciences, University of Hawai'i at Mānoa, 1955 East-West Road, Honolulu, HI 96822, USA
| | - Jason P Breves
- Department of Biology, Skidmore College, 815 N. Broadway, Saratoga Springs, NY 12866, USA
| | - Andre P Seale
- Department of Human Nutrition, Food and Animal Sciences, University of Hawai'i at Mānoa, 1955 East-West Road, Honolulu, HI 96822, USA.
| |
Collapse
|
13
|
Breslow E, Taylor A, Chan CL, Severn C, Pyle L, Torchen L, Sisk R, Legro R, Turcu AF, Auchus RJ, Dunaif A, Kelsey MM, Cree-Green M. 11-Oxygenated Androgen Metabolite Concentrations Are Affected by Pubertal Progression and Obesity. Horm Res Paediatr 2022; 96:412-422. [PMID: 36446347 DOI: 10.1159/000528341] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 10/04/2022] [Indexed: 12/05/2022] Open
Abstract
INTRODUCTION 11-oxygenated C19 steroids (11-oxyandrogens) have been shown to rise during adrenarche and remain higher throughout adulthood than in early childhood. The patterns of circulating 11-oxyandrogens throughout normal puberty have not yet been described. METHODS We conducted a secondary analysis of healthy youth participants, both males and females, enrolled in six prior endocrine studies (N = 249). Participants were classified according to Tanner stage and body mass index (BMI). Concentrations of three adrenal-specific 11-oxygenated androgens, 11β-hydroxyandrostenedione (11OHA4), 11β-hydroxytestosterone (11OHT), and 11-ketotestosterone (11KT), were measured in fasting serum samples. RESULTS 11OHA4 and 11OHT increased modestly between early and late puberty in youth with normal weight (p < 0.05), whereas increases in 11KT did not reach statistical significance (p < 0.06). 11KT levels differed between sexes throughout puberty (p < 0.01), and changes in 11-oxyandrogens were small compared to the marked increases for estradiol in girls or testosterone in boys. The trajectories of 11KT and 11OHA4 changes throughout puberty differed by BMI category (p < 0.05). CONCLUSION Beyond adrenarche, 11-oxyandrogens continue to rise during pubertal development. The differences in 11KT trajectories in males and females are small compared to changes in testosterone for males and estradiol for females during puberty. Obesity appears to influence the trajectories of 11-oxyandrogens during puberty.
Collapse
Affiliation(s)
- Emily Breslow
- Department of Pediatrics, Division of Pediatric Endocrinology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Anya Taylor
- Department of Pediatrics, Division of Pediatric Endocrinology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Christine L Chan
- Department of Pediatrics, Division of Pediatric Endocrinology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Cameron Severn
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Department of Biostatistics and Informatics, Colorado School of Public Health, Aurora, Colorado, USA
| | - Laura Pyle
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Department of Biostatistics and Informatics, Colorado School of Public Health, Aurora, Colorado, USA
| | - Laura Torchen
- Division of Endocrinology, Ann & Robert H Lurie Children's Hospital of Chicago, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Ryan Sisk
- Division of Endocrinology, Metabolism, and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Richard Legro
- Department of Obstetrics and Gynecology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA
| | - Adina F Turcu
- Division of Metabolism, Endocrinology and Diabetes, University of Michigan, Ann Arbor, Michigan, USA
| | - Richard J Auchus
- Division of Metabolism, Endocrinology and Diabetes, University of Michigan, Ann Arbor, Michigan, USA
| | - Andrea Dunaif
- Division of Endocrinology, Diabetes, and Bone Diseases, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Megan Moriarty Kelsey
- Department of Pediatrics, Division of Pediatric Endocrinology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Center for Women's Health Research, Aurora, Colorado, USA
| | - Melanie Cree-Green
- Department of Pediatrics, Division of Pediatric Endocrinology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Center for Women's Health Research, Aurora, Colorado, USA
| |
Collapse
|
14
|
Rosato E, Sciarra F, Anastasiadou E, Lenzi A, Venneri MA. Revisiting the physiological role of androgens in women. Expert Rev Endocrinol Metab 2022; 17:547-561. [PMID: 36352537 DOI: 10.1080/17446651.2022.2144834] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 11/03/2022] [Indexed: 11/09/2022]
Abstract
INTRODUCTION Extensive research underlines the critical functions of androgens in females. Nevertheless, the precise mechanisms of their action are poorly understood. Here, we review the existing literature regarding the physiological role of androgens in women throughout life. AREAS COVERED Several studies show that androgen receptors (ARs) are broadly expressed in numerous female tissues. They are essential for many physiological processes, including reproductive, sexual, cardiovascular, bone, muscle, and brain health. They are also involved in adipose tissue and liver function. Androgen levels change with the menstrual cycle and decrease in the first decades of life, independently of menopause. EXPERT OPINION To date, studies are limited by including small numbers of women, the difficulty of dosing androgens, and their cyclical variations. In particular, whether androgens play any significant role in regulating the establishment of pregnancy is poorly understood. The neural functions of ARs have also been investigated less thoroughly, although it is expressed at high levels in brain structures. Moreover, the mechanism underlying the decline of dehydroepiandrosterone (DHEA) and dehydroepiandrosterone sulfate (DHEAS) with age is unclear. Other factors, including estrogen's effect on adrenal androgen production, reciprocal regulation of ARs, and non-classical effects of androgens, remain to be determined.
Collapse
Affiliation(s)
- Elena Rosato
- Department of Experimental Medicine, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Francesca Sciarra
- Department of Experimental Medicine, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Eleni Anastasiadou
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Andrea Lenzi
- Department of Experimental Medicine, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Mary Anna Venneri
- Department of Experimental Medicine, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| |
Collapse
|
15
|
Alemany M. The Roles of Androgens in Humans: Biology, Metabolic Regulation and Health. Int J Mol Sci 2022; 23:11952. [PMID: 36233256 PMCID: PMC9569951 DOI: 10.3390/ijms231911952] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Revised: 09/26/2022] [Accepted: 09/27/2022] [Indexed: 11/17/2022] Open
Abstract
Androgens are an important and diverse group of steroid hormone molecular species. They play varied functional roles, such as the control of metabolic energy fate and partition, the maintenance of skeletal and body protein and integrity and the development of brain capabilities and behavioral setup (including those factors defining maleness). In addition, androgens are the precursors of estrogens, with which they share an extensive control of the reproductive mechanisms (in both sexes). In this review, the types of androgens, their functions and signaling are tabulated and described, including some less-known functions. The close interrelationship between corticosteroids and androgens is also analyzed, centered in the adrenal cortex, together with the main feedback control systems of the hypothalamic-hypophysis-gonads axis, and its modulation by the metabolic environment, sex, age and health. Testosterone (T) is singled out because of its high synthesis rate and turnover, but also because age-related hypogonadism is a key signal for the biologically planned early obsolescence of men, and the delayed onset of a faster rate of functional losses in women after menopause. The close collaboration of T with estradiol (E2) active in the maintenance of body metabolic systems is also presented Their parallel insufficiency has been directly related to the ravages of senescence and the metabolic syndrome constellation of disorders. The clinical use of T to correct hypoandrogenism helps maintain the functionality of core metabolism, limiting excess fat deposition, sarcopenia and cognoscitive frailty (part of these effects are due to the E2 generated from T). The effectiveness of using lipophilic T esters for T replacement treatments is analyzed in depth, and the main problems derived from their application are discussed.
Collapse
Affiliation(s)
- Marià Alemany
- Facultat de Biologia, Universitat de Barcelona, Av. Diagonal, 635, 08028 Barcelona, Catalonia, Spain;
- Institut de Biomedicina, Universitat de Barcelona, 08028 Barcelona, Catalonia, Spain
| |
Collapse
|
16
|
Advances in the Current Understanding of the Mechanisms Governing the Acquisition of Castration-Resistant Prostate Cancer. Cancers (Basel) 2022; 14:cancers14153744. [PMID: 35954408 PMCID: PMC9367587 DOI: 10.3390/cancers14153744] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 07/29/2022] [Indexed: 11/17/2022] Open
Abstract
Despite aggressive treatment and androgen-deprivation therapy, most prostate cancer patients ultimately develop castration-resistant prostate cancer (CRPC), which is associated with high mortality rates. However, the mechanisms governing the development of CRPC are poorly understood, and androgen receptor (AR) signaling has been shown to be important in CRPC through AR gene mutations, gene overexpression, co-regulatory factors, AR shear variants, and androgen resynthesis. A growing number of non-AR pathways have also been shown to influence the CRPC progression, including the Wnt and Hh pathways. Moreover, non-coding RNAs have been identified as important regulators of the CRPC pathogenesis. The present review provides an overview of the relevant literature pertaining to the mechanisms governing the molecular acquisition of castration resistance in prostate cancer, providing a foundation for future, targeted therapeutic efforts.
Collapse
|
17
|
Buchanan K, Greenup E, Hurst ACE, Sunil B, Ashraf AP. Case report: 11-ketotestosterone may potentiate advanced bone age as seen in some cases of Wiedemann-Steiner Syndrome. Front Endocrinol (Lausanne) 2022; 13:1004114. [PMID: 36263329 PMCID: PMC9574220 DOI: 10.3389/fendo.2022.1004114] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
CONTEXT Wiedemann-Steiner Syndrome (WSS) is a genetic disorder associated with an array of clinical phenotypes, including advanced bone age and short stature. 11-ketotestosterone (11KT) is a member of the group known as 11-oxygenated C19 androgens that are implicated in premature adrenarche. CASE DESCRIPTION Case 1: The patient is a 3 year and 11-month-old female diagnosed with WSS due to deletion of KMT2A detected on CGH microarray. At two years and 11 months, imaging revealed an advanced bone age. We obtained an 11KT level on this patient. 11KT in case 1 was elevated at 26.3 ng/dL, while the normal reference range is 7.3-10.9 ng/dL and the reference interval for premature adrenarche is 12.3-22.9 ng/dL, The repeat 11KT at follow up (chronological age 4 years and 6 months) was still elevated at 33.8 ng/dL Case 2: A second child with WSS and a 5kb intragenic KMT2A deletion was evaluated at 11 months of age; his 11KT was 4.5 ng/dL. CONCLUSIONS The elevated 11KT may indicate maturational changes related to increasing adrenal gland androgenic activation and may explain the advanced bone age seen in some patients with WSS. To our knowledge, this is the first case report that describes 11KT as a bioactive androgen potentially causing bone age advancement in WSS. Lack of elevation of 11KT in the second child who is an infant suggests increasing androgenic precursors and metabolites related to premature adrenarche may need to be longitudinally followed.
Collapse
Affiliation(s)
- Katherine Buchanan
- University of Alabama at Birmingham Marnix E. Heersink School of Medicine, Birmingham, AL, United States
- *Correspondence: Katherine Buchanan,
| | - Erin Greenup
- Division of Pediatric Endocrinology, Department of Pediatrics, Orlando Health Arnold Palmer Hospital for Children, Orlando, FL, United States
| | - Anna C. E. Hurst
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Bhuvana Sunil
- Division of Pediatric Endocrinology and Diabetes, Mary Bridge Children’s Hospital, Tacoma, WA, United States
| | - Ambika P. Ashraf
- Divison of Pediatric Endocrinology and Diabetes, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
18
|
Lee BY, Jo JB, Choi D, Lee SH, Cheon YP. A Chronic-Low-Dose Exposing of DEHP with OECD TG 443 Altered the
Histological Characteristics and Steroidogeic Gene Expression of Adrenal Gland
in Female Mice. Dev Reprod 2021; 25:257-268. [PMID: 35141451 PMCID: PMC8807134 DOI: 10.12717/dr.2021.25.4.257] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 11/13/2021] [Accepted: 12/04/2021] [Indexed: 11/17/2022]
Affiliation(s)
- Bo Young Lee
- Division of Developmental Biology and
Physiology, Center for Development and Program Research, Department of
Biotechnology, Institute of Basic Sciences, Sungshin
University, Seoul 02844, Korea
| | - Jeong Bin Jo
- Division of Developmental Biology and
Physiology, Center for Development and Program Research, Department of
Biotechnology, Institute of Basic Sciences, Sungshin
University, Seoul 02844, Korea
| | - Donchan Choi
- Dept. of Life Science, College of
Environmental Sciences, Yong-In University, Yongin
17092, Korea
| | - Sung-Ho Lee
- Dept. of Biotechnology, Sangmyung
University, Seoul 03016, Korea
| | - Yong-Pil Cheon
- Division of Developmental Biology and
Physiology, Center for Development and Program Research, Department of
Biotechnology, Institute of Basic Sciences, Sungshin
University, Seoul 02844, Korea
- Corresponding author Yong-Pil Cheon,
Division of Developmental Biology and Physiology, Department of Biotechnology,
Institute of Basic Sciences, Sungshin University, Seoul 02844, Korea. Tel:
+82-2-920-7639, Fax: +82-2-920-2736,
E-mail:
| |
Collapse
|
19
|
Abstract
Adrenarche is the maturational increase in adrenal androgen production that normally begins in early childhood. It results from changes in the secretory response to adrenocorticotropin (ACTH) that are best indexed by dehydroepiandrosterone sulfate (DHEAS) rise. These changes are related to the development of the zona reticularis (ZR) and its unique gene/enzyme expression pattern of low 3ß-hydroxysteroid dehydrogenase type 2 with high cytochrome b5A, sulfotransferase 2A1, and 17ß-hydroxysteroid dehydrogenase type 5. Recently 11-ketotestosterone was identified as an important bioactive adrenarchal androgen. Birth weight, body growth, obesity, and prolactin are related to ZR development. Adrenarchal androgens normally contribute to the onset of sexual pubic hair (pubarche) and sebaceous and apocrine gland development. Premature adrenarche causes ≥90% of premature pubarche (PP). Its cause is unknown. Affected children have a significantly increased growth rate with proportionate bone age advancement that typically does not compromise growth potential. Serum DHEAS and testosterone levels increase to levels normal for early female puberty. It is associated with mildly increased risks for obesity, insulin resistance, and possibly mood disorder and polycystic ovary syndrome. Between 5% and 10% of PP is due to virilizing disorders, which are usually characterized by more rapid advancement of pubarche and compromise of adult height potential than premature adrenarche. Most cases are due to nonclassic congenital adrenal hyperplasia. Algorithms are presented for the differential diagnosis of PP. This review highlights recent advances in molecular genetic and developmental biologic understanding of ZR development and insights into adrenarche emanating from mass spectrometric steroid assays.
Collapse
Affiliation(s)
- Robert L Rosenfield
- University of Chicago Pritzker School of Medicine, Section of Adult and Pediatric Endocrinology, Metabolism, and Diabetes, Chicago, IL, USA.,Department of Pediatrics, University of California, San Francisco, CA, USA
| |
Collapse
|
20
|
Han B, Zhu H, Yao H, Ren J, O'Day P, Wang H, Zhu W, Cheng T, Auchus RJ, Qiao J. Differences of adrenal-derived androgens in 5α-reductase deficiency versus androgen insensitivity syndrome. Clin Transl Sci 2021; 15:658-666. [PMID: 34755921 PMCID: PMC8932821 DOI: 10.1111/cts.13184] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 09/22/2021] [Accepted: 09/26/2021] [Indexed: 11/27/2022] Open
Abstract
Steroid 5α‐reductase type 2 deficiency (5α‐RD2) and androgen insensitivity syndrome (AIS) are difficult to distinguish clinically and biochemically, and adrenal‐derived androgens have not been investigated in these conditions using modern methods. The objective of the study was to compare Chinese patients with 5α‐RD2, AIS, and healthy men. Sixteen patients with 5α‐RD2, 10 patients with AIS, and 39 healthy men were included. Serum androgen profiles were compared in these subjects using liquid chromatography/tandem mass spectrometry (LC‐MS/MS). Based on clinical features and laboratory tests, 5α‐RD2 and AIS were diagnosed and confirmed by genotyping. Dihydrotestosterone (DHT) and testosterone (T) were both significantly lower in patients with 5α‐RD2 than AIS (p < 0.0001). The T/DHT ratio was higher in 5α‐RD2 (4.5–88.6) than AIS (13.4–26.7) or healthy men (7.6–40.5). Using LC‐MS/MS, a cutoff T/DHT value of 27.3 correctly diagnosed 5α‐RD2 versus AIS with sensitivity 93.8% and specificity 100%. Among the adrenal‐derived 11‐oxygenated androgens, 11β‐hydroxyandrostenedione (11OHA4) and 11‐ketoandrostenedione (11KA4) were also lower in patients with 5α‐RD2 than those of patients with AIS. In contrast, 11β‐hydroxytestosterone (11OHT) was higher in 5α‐RD2 than AIS. Furthermore, a 11OHT/11OHA4 cutoff value of 0.048 could also distinguish 5α‐RD2 from AIS. Thus, both elevated T/DHT values above 27.3 and the unexpected 11‐oxygenated androgen profile, with a 11OHT/11OHA4 ratio greater than 0.048, distinguished 5α‐RD2 from AIS. These data suggest that the metabolism of both gonadal and adrenal‐derived androgens is altered in 5α‐RD2.
Collapse
Affiliation(s)
- Bing Han
- Department of Endocrinology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hui Zhu
- Department of Endocrinology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haijun Yao
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jianwei Ren
- Division of Metabolism, Endocrinology and Diabetes, Department of Internal Medicine, Department of Pharmacology, the Program for Disorders of Sexual Development, University of Michigan, Ann Arbor, Michigan, USA
| | - Patrick O'Day
- Division of Metabolism, Endocrinology and Diabetes, Department of Internal Medicine, Department of Pharmacology, the Program for Disorders of Sexual Development, University of Michigan, Ann Arbor, Michigan, USA
| | - Hao Wang
- Department of Endocrinology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenjiao Zhu
- Department of Endocrinology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tong Cheng
- Department of Endocrinology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Richard J Auchus
- Division of Metabolism, Endocrinology and Diabetes, Department of Internal Medicine, Department of Pharmacology, the Program for Disorders of Sexual Development, University of Michigan, Ann Arbor, Michigan, USA
| | - Jie Qiao
- Department of Endocrinology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
21
|
Stárka L, Dušková M. Remarks on the Hormonal Background of the Male Equivalent of Polycystic Ovary Syndrome. Prague Med Rep 2021; 122:73-79. [PMID: 34137683 DOI: 10.14712/23362936.2021.8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
The hypothesis that the most common female endocrine disease, the polycystic ovarian syndrome (PCOS), has a male equivalent, has recently become more widely accepted. The male form of PCOS is marked by alterations in the secretion of gonadotropins, increased insulin resistance, and changes of the levels of several steroid hormones, with clinical manifestations including premature androgenic alopecia (AGA). Because these symptoms are not always found in men with genetic predispositions, knowledge of the male equivalent of PCOS needs to be supplemented by measurements of adrenal 11-oxygenated C19 steroids, particularly 11-keto-, and 11β-hydroxy-derivatives of testosterone and dihydrotestosterone, by focusing on the newly-realized role of skin as an endocrine organ, and by confirming any age-related factors in glucose metabolism disorders in such predisposed men.
Collapse
|
22
|
Yazawa T, Sato T, Nemoto T, Nagata S, Imamichi Y, Kitano T, Sekiguchi T, Uwada J, Islam MS, Mikami D, Nakajima I, Takahashi S, Khan MRI, Suzuki N, Umezawa A, Ida T. 11-Ketotestosterone is a major androgen produced in porcine adrenal glands and testes. J Steroid Biochem Mol Biol 2021; 210:105847. [PMID: 33609691 DOI: 10.1016/j.jsbmb.2021.105847] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 02/09/2021] [Accepted: 02/10/2021] [Indexed: 12/12/2022]
Abstract
Porcine steroid hormone profiles have some unique characteristics. We previously studied human and murine steroidogenesis using steroidogenic cells-derived from mesenchymal stem cells (MSCs). To investigate porcine steroidogenesis, we induced steroidogenic cells from porcine subcutaneous preadipocytes (PSPA cells), which originate from MSCs. Using cAMP, adenovirus-mediated introduction of steroidogenic factor-1 (SF-1)/adrenal 4-binding protein (Ad4BP) induced the differentiation of PSPA cells into sex steroid-producing cells. Introducing SF-1/Ad4BP also induced the aldo-keto reductase 1C1 (AKR1C1) gene. Porcine AKR1C1 had 17β-hydroxysteroid dehydrogenase activity, which converts androstenedione and 11-ketoandrostenedione into testosterone (T) and 11-ketotestosteorne (11KT). Furthermore, differentiated cells expressed hydroxysteroid 11β-dehydrogenase 2 (HSD11B2) and produced 11KT. HSD11B2 was expressed in testicular Leydig cells and the adrenal cortex. 11KT was present in the plasma of both immature male and female pigs, with slightly higher levels in the male pigs. T levels were much higher in the male pigs. It is noteworthy that in the female pigs, the 11KT levels were >10-fold higher than the T levels. However, castration altered the 11KT and T plasma profiles in the male pigs to near those of the females. 11KT induced endothelial nitric oxide synthase (eNOS) in porcine vascular endothelial cells. These results indicate that 11KT is produced in porcine adrenal glands and testes, and may regulate cardiovascular functions through eNOS expression.
Collapse
Affiliation(s)
- Takashi Yazawa
- Department of Biochemistry, Asahikawa Medical University, Hokkaido 078-8510, Japan.
| | - Takahiro Sato
- Division of Molecular Genetics, Institute of Life Sciences, Kurume University, Fukuoka 830-0011, Japan
| | - Takahiro Nemoto
- Department of Physiology, Nippon Medical School, Tokyo 113-8602, Japan
| | - Sayaka Nagata
- Circulatory and Body Fluid Regulation, Faculty of Medicine, University of Miyazaki, Miyazaki 889-1692, Japan
| | - Yoshitaka Imamichi
- Department of Pharmacology, Asahikawa Medical University, Hokkaido 078-8510, Japan
| | - Takeshi Kitano
- Department of Biological Sciences, Graduate School of Science and Technology, Kumamoto University, Kumamoto 860-8555, Japan
| | - Toshio Sekiguchi
- Noto Marine Laboratory, Division of Marine Environmental Studies, Institute of Nature and Environmental Technology, Kanazawa University, Ishikawa 927-0553, Japan
| | - Junsuke Uwada
- Department of Biochemistry, Asahikawa Medical University, Hokkaido 078-8510, Japan
| | | | - Daisuke Mikami
- Department of Nephrology, University of Fukui, Fukui 910-1193, Japan
| | - Ikuyo Nakajima
- Institute of Livestock and Grassland Science, NARO, Tsukuba, Ibaraki 305-0901, Japan
| | - Satoru Takahashi
- Department of Pediatrics, Asahikawa Medical University, Hokkaido 078-8510, Japan
| | - Md Rafiqul Islam Khan
- Department of Biochemistry, Asahikawa Medical University, Hokkaido 078-8510, Japan; Department of Pharmacy, University of Rajshahi, Rajshahi, Bangladesh
| | - Nobuo Suzuki
- Noto Marine Laboratory, Division of Marine Environmental Studies, Institute of Nature and Environmental Technology, Kanazawa University, Ishikawa 927-0553, Japan
| | - Akihiro Umezawa
- Department of Reproduction, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan
| | - Takanori Ida
- Center for Animal Disease Control, University of Miyazaki, Miyazaki 889-1692, Japan
| |
Collapse
|
23
|
Dumontet T, Martinez A. Adrenal androgens, adrenarche, and zona reticularis: A human affair? Mol Cell Endocrinol 2021; 528:111239. [PMID: 33676986 DOI: 10.1016/j.mce.2021.111239] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/11/2021] [Accepted: 03/01/2021] [Indexed: 12/11/2022]
Abstract
In humans, reticularis cells of the adrenal cortex fuel the production of androgen steroids, constituting the driver of numerous morphological changes during childhood. These steps are considered a precocious stage of sexual maturation and are grouped under the term "adrenarche". This review describes the molecular and enzymatic characteristics of the zona reticularis, along with the possible signals and mechanisms that control its emergence and the associated clinical features. We investigate the differences between species and discuss new studies such as genetic lineage tracing and transcriptomic analysis, highlighting the rodent inner cortex's cellular and molecular heterogeneity. The recent development and characterization of mouse models deficient for Prkar1a presenting with adrenocortical reticularis-like features prompt us to review our vision of the mouse adrenal gland maturation. We expect these new insights will help increase our understanding of the adrenarche process and the pathologies associated with its deregulation.
Collapse
Affiliation(s)
- Typhanie Dumontet
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, USA; Training Program in Organogenesis, Center for Cell Plasticity and Organ Design, University of Michigan, Ann Arbor, MI, USA.
| | - Antoine Martinez
- Génétique, Reproduction et Développement (GReD), Centre National de La Recherche Scientifique CNRS, Institut National de La Santé & de La Recherche Médicale (INSERM), Université Clermont-Auvergne (UCA), France.
| |
Collapse
|
24
|
Ly LK, Doden HL, Ridlon JM. Gut feelings about bacterial steroid-17,20-desmolase. Mol Cell Endocrinol 2021; 525:111174. [PMID: 33503463 PMCID: PMC8886824 DOI: 10.1016/j.mce.2021.111174] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 01/17/2021] [Accepted: 01/20/2021] [Indexed: 12/12/2022]
Abstract
Advances in technology are only beginning to reveal the complex interactions between hosts and their resident microbiota that have co-evolved over centuries. In this review, we present compelling evidence that implicates the host-associated microbiome in the generation of 11β-hydroxyandrostenedione, leading to the formation of potent 11-oxy-androgens. Microbial steroid-17,20-desmolase cleaves the side-chain of glucocorticoids (GC), including cortisol (and its derivatives of cortisone, 5α-dihydrocortisol, and also (allo)- 3α, 5α-tetrahydrocortisol, but not 3α-5β-tetrahydrocortisol) and drugs (prednisone and dexamethasone). In addition to side-chain cleavage, we discuss the gut microbiome's robust potential to transform a myriad of steroids, mirroring much of the host's metabolism. We also explore the overlooked role of intestinal steroidogenesis and efflux pumps as a potential route for GC transport into the gut. Lastly, we propose several health implications from microbial steroid-17,20-desmolase function, including aberrant mineralocorticoid, GC, and androgen receptor signaling in colonocytes, immune cells, and prostate cells, which may exacerbate disease states.
Collapse
Affiliation(s)
- Lindsey K Ly
- Microbiome Metabolic Engineering Theme, Carl R. Woese Institute for Genomic Biology, Urbana, IL, 61801, USA; Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA; Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Heidi L Doden
- Microbiome Metabolic Engineering Theme, Carl R. Woese Institute for Genomic Biology, Urbana, IL, 61801, USA; Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Jason M Ridlon
- Microbiome Metabolic Engineering Theme, Carl R. Woese Institute for Genomic Biology, Urbana, IL, 61801, USA; Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA; Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA; Cancer Center of Illinois, University of Illinois at Urbana-Champaign, Urbana, IL, USA; Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA.
| |
Collapse
|
25
|
Reinehr T, Kulle A, Barth A, Ackermann J, Holl RW, Holterhus PM. Transition from gynaecomastia to lipomastia in pubertal boys. Clin Endocrinol (Oxf) 2021; 94:583-589. [PMID: 33351202 DOI: 10.1111/cen.14403] [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: 09/30/2020] [Revised: 12/14/2020] [Accepted: 12/16/2020] [Indexed: 11/30/2022]
Abstract
OBJECTIVE Gynaecomastia is frequent in pubertal boys and is regarded as a self-limiting abnormality. However, longitudinal studies proving this hypothesis are scarce. DESIGN Longitudinal follow-up study (median 2.4, range 1.0-4.8 years). METHODS The regression of breast diameter was analysed in 31 pubertal boys aged 11.7-16.1 (median 13.2) years with gynaecomastia. Furthermore, weight changes (as BMI-SDS) and pubertal stage, oestradiol [E2], oestriol, oestrone, androstenedione, testosterone [T], dihydrotestosterone, gonadotropins, IGF-1, and IGFBP-3 serum concentrations determined at first clinical presentation were related to breast diameter regression determined by palpation and disappearance of breast glandular tissue in ultrasound in follow-up to identify possible predictors of breast regression. RESULTS During the observation period, the breast diameter decreased (in median -1 (interquartile range [IQR] -5 to +1) cm). At follow-up, 6% of boys had no breast enlargement any more, and 65% developed lipomastia. Gynaecomastia was still present in 29%. None of the analysed hormones was related significantly to breast diameter regression or disappearance of breast glandular tissue. In multiple linear regression analyses adjusted for observational period, as well as age and BMI-SDS at first presentation, changes in BMI-SDS (β-coefficient 6.0 ± 2.3, p = .015) but not the E2/T ratio or any other hormone determined at baseline was related to changes in breast diameter. CONCLUSIONS Breast diameter regression seems not to be predictable by a hormone profile in pubertal boys with gynaecomastia. In pubertal boys presenting with gynaecomastia, conversion to lipomastia of smaller volume is common. The reduction of weight status was the best predictor of breast diameter regression.
Collapse
Affiliation(s)
- Thomas Reinehr
- Department of Pediatric Endocrinology, Diabetes and Nutrition Medicine, Vestische Hospital for Children and Adolescents Datteln, University of Witten/Herdecke, Datteln, Germany
| | - Alexandra Kulle
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics, University Hospital of Schleswig - Holstein, Campus Kiel / Christian - Albrechts University of Kiel, Kiel, Germany
| | - Andre Barth
- Department of Pediatric Endocrinology, Diabetes and Nutrition Medicine, Vestische Hospital for Children and Adolescents Datteln, University of Witten/Herdecke, Datteln, Germany
| | - Jonas Ackermann
- Department of Pediatric Endocrinology, Diabetes and Nutrition Medicine, Vestische Hospital for Children and Adolescents Datteln, University of Witten/Herdecke, Datteln, Germany
| | - Reinhard W Holl
- Institute of Epidemiology and Medical Biometry, German Center for Diabetes Research, University of Ulm, Ulm, Germany
| | - Paul-Martin Holterhus
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics, University Hospital of Schleswig - Holstein, Campus Kiel / Christian - Albrechts University of Kiel, Kiel, Germany
| |
Collapse
|
26
|
Glass SM, Reddish MJ, Child SA, Wilkey CJ, Stec DF, Guengerich FP. Characterization of human adrenal cytochrome P450 11B2 products of progesterone and androstenedione oxidation. J Steroid Biochem Mol Biol 2021; 208:105787. [PMID: 33189850 PMCID: PMC7954869 DOI: 10.1016/j.jsbmb.2020.105787] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 10/19/2020] [Accepted: 10/26/2020] [Indexed: 11/17/2022]
Abstract
Cytochrome P450 (P450) 11B1 and 11B2 both catalyze the 11β-hydroxylation of 11-deoxycorticosterone and the subsequent 18-hydroxylation of the product. P450 11B2, but not P450 11B1, catalyzes a further C-18 oxidation to yield aldosterone. 11-Oxygenated androgens are of interest, and 11-hydroxy progesterone has been reported to be a precursor of these. Oxidation of progesterone by purified recombinant P450 11B2 yielded a mono-hydroxy derivative as the major product, and co-chromatography with commercial standards and 2-D NMR spectroscopy indicated 11β-hydroxylation. 18-Hydroxyprogesterone and a dihydroxyprogesterone were also formed. Similarly, oxidation of androstenedione by P450 11B2 yielded 11β-hydroxyandrostenedione, 18-hydroxyandrostenedione, and a dihydroxyandrostenedione. The steady-state kinetic parameters for androstenedione and progesterone 11β-hydroxylation were similar to those reported for the classic substrate 11-deoxycorticosterone. The source of 11α-hydroxyprogesterone in humans remains unresolved.
Collapse
Affiliation(s)
- Sarah M Glass
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, 37232-0146, United States
| | - Michael J Reddish
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, 37232-0146, United States; Department of Chemistry and Fermentation Sciences, Appalachian State University, Boone, NC, 28608, United States
| | - Stella A Child
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, 37232-0146, United States
| | - Clayton J Wilkey
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, 37232-0146, United States
| | - Donald F Stec
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN, 37122, United States
| | - F Peter Guengerich
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, 37232-0146, United States.
| |
Collapse
|
27
|
Determination of Intraprostatic and Intratesticular Androgens. Int J Mol Sci 2021; 22:ijms22010466. [PMID: 33466491 PMCID: PMC7796479 DOI: 10.3390/ijms22010466] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 12/25/2020] [Accepted: 12/31/2020] [Indexed: 12/19/2022] Open
Abstract
Androgens represent the main hormones responsible for maintaining hormonal balance and function in the prostate and testis. As they are involved in prostate and testicular carcinogenesis, more detailed information of their active concentration at the site of action is required. Since the introduction of the term intracrinology as the local formation of active steroid hormones from inactive precursors of the adrenal gland, mainly dehydroepiandrosterone (DHEA) and DHEA-S, it is evident that blood circulating levels of sex steroid hormones need not reflect their actual concentrations in the tissue. Here, we review and critically evaluate available methods for the analysis of human intraprostatic and intratesticular steroid concentrations. Since analytical approaches have much in common in both tissues, we discuss them together. Preanalytical steps, including various techniques for separation of the analytes, are compared, followed by the end-point measurement. Advantages and disadvantages of chromatography-mass spectrometry (LC-MS, GC-MS), immunoanalytical methods (IA), and hybrid (LC-IA) are discussed. Finally, the clinical information value of the determined steroid hormones is evaluated concerning differentiating between patients with cancer or benign hyperplasia and between patients with different degrees of infertility. Adrenal-derived 11-oxygenated androgens are mentioned as perspective prognostic markers for these purposes.
Collapse
|
28
|
Rosenfield RL, Cooke DW, Radovick S. Puberty in the Female and Its Disorders. SPERLING PEDIATRIC ENDOCRINOLOGY 2021:528-626. [DOI: 10.1016/b978-0-323-62520-3.00016-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
29
|
Torchen LC, Sisk R, Legro RS, Turcu AF, Auchus RJ, Dunaif A. 11-Oxygenated C19 Steroids Do Not Distinguish the Hyperandrogenic Phenotype of PCOS Daughters from Girls with Obesity. J Clin Endocrinol Metab 2020; 105:5892800. [PMID: 32797203 PMCID: PMC7500474 DOI: 10.1210/clinem/dgaa532] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 08/07/2020] [Indexed: 12/13/2022]
Abstract
CONTEXT Hyperandrogenemia (HA) is a consistent reproductive phenotype in women with polycystic ovary syndrome (PCOS) and their relatives. Increased testosterone levels are present in premenarchal daughters of affected women (PCOS-d). Obese girls (OB-g) without a family history of PCOS also have peripubertal HA. The sources and significance of HA in these groups remains unknown. OBJECTIVE 11-oxygenated 19-carbon (C19) steroids are adrenally derived androgens that are elevated in hyperandrogenic disorders, including PCOS. We performed this study to test the hypothesis that peripheral serum 11-oxygenated steroids would differ in PCOS-d compared with OB-g suggesting distinct etiologies of HA in affected girls. DESIGN, SETTING, AND PARTICIPANTS We compared peripheral serum 11-oxygenated steroid levels in 21 PCOS-d, 29 OB-g, and 17 lean control girls (LC) of comparable age at an academic medical center. RESULTS Body mass index (BMI) differed by design (P < 0.001). 11β-hydroxyandrostenedione, 11-ketoandrostenedione, and 11β-hydroxytestosterone levels did not differ between the groups. Compared with LC, PCOS-d and OB-g had similar elevations in 11-ketotestosterone (11KT) (analysis of variance [ANOVA] P = 0.03; PCOS-d vs LC, P = 0.04; OB-g vs LC, P = 0.05; PCOS-d vs OB-g, P = 0.97). In multivariate regression, 11KT levels were associated with DHEAS (P = 0.008), but not with BMI z score, breast Tanner stage, testosterone, anti-Müllerian hormone or sex hormone-binding globulin levels. CONCLUSIONS Circulating 11KT levels were similarly elevated in peripubertal PCOS-d and OB-g, suggesting an adrenal component of HA in both groups. We found that 11-oxygenated 19-carbon steroid profiles did not identify subtypes of HA girls.
Collapse
Affiliation(s)
- Laura C Torchen
- Division of Endocrinology, Ann & Robert H Lurie Children’s Hospital of Chicago, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Correspondence and Reprint Requests: Laura Torchen, MD, Ann & Robert H Lurie Children’s Hospital of Chicago, 225 E Chicago Ave, Box 54, Chicago, IL, 60611, USA. E-mail:
| | - Ryan Sisk
- Division of Endocrinology, Metabolism, and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Richard S Legro
- Department of Obstetrics and Gynecology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Adina F Turcu
- Division of Metabolism, Endocrinology and Diabetes, University of Michigan, Ann Arbor, Michigan
| | - Richard J Auchus
- Division of Metabolism, Endocrinology and Diabetes, University of Michigan, Ann Arbor, Michigan
| | - Andrea Dunaif
- Division of Endocrinology, Diabetes, and Bone Diseases, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
30
|
Is Idiopathic Hirsutism Truly Idiopathic? J Obstet Gynaecol India 2020; 70:366-370. [PMID: 33041554 DOI: 10.1007/s13224-020-01324-6] [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: 12/16/2019] [Accepted: 05/07/2020] [Indexed: 10/24/2022] Open
Abstract
Objective To determine whether other androgens [androstenedione (A4), 17-hydroxy progesterone (17OHP) and dehydroepiandrosterone (DHEA)] were elevated in women with classically defined idiopathic hirsutism (IH)/patient-important hirsutism (PIH). Study Design Retrospective analysis. Setting Outpatient endocrine department of a tertiary care hospital. Patients In total, 30 consecutive women with IH/PIH were included. IH/PIH was defined as presentation with hirsutism with normal menstrual cycles (25-35 days), normal total (< 45 ng/dL) and free T (fT) (< 0.6 ng/dL) and normal ovaries sonologically (transabdominal ultrasonogram ovarian volume < 10 cm3) without any other signs of virilization. Clinical and biochemical details were collected and analyzed. Androgens were measured by LC-MS/MS. A4 ≥ 2.5 ng/mL, DHEA ≥ 15 (age < 18) or ≥ 11.8 (age ≥ 18) ng/mL, DHEAS ≥ 2847 ng/mL or 17OHP ≥ 2 ng/mL were considered high. Results With the mean age of 22 years and mean BMI of 25 kg/m2, 12/30 (40%) had IH and remaining PIH. DHEA alone was elevated in 60% and A4 alone in 33%. Overall, 23/30 (73%) had any one elevated androgen with normal total and free testosterone. There was no correlation with modified Ferriman-Gallwey score, and there was no significant difference in androgens between IH and PIH. Conclusion A high proportion of women with classically defined IH/PIH have elevated DHEA and/or A4. Though on pharmacotherapy basis, there would be no change in management, the role of hyperandrogenemia detected by sensitive assays on metabolic functions and cardiovascular risk has to be studied.
Collapse
|
31
|
Stárka L, Dušková M, Vítků J. 11-Keto-testosterone and other androgens of adrenal origin. Physiol Res 2020; 69:S187-S192. [PMID: 33094617 DOI: 10.33549/physiolres.934516] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
The adrenal glands produce significant amounts of steroid hormones and their metabolites, with various levels of androgenic activities. Until recently, the androgenic potency of these adrenal-derived compounds were not well known, but some recent studies have shown that the production of 11-oxo- and 11beta-hydroxy-derived testosterone and dihydrotestosterone evidently have high androgenic activity. This fact has clinical importance, for instance, in various types of congenital adrenal hyperplasia with androgenization or polycystic ovarian syndrome, and laboratory determinations of these substances could help to better evaluate the total androgen pressure in patients with these disorders. Another area of concern is the treatment of prostate cancer with androgen deprivation, which loses effectiveness after a certain time. The concurrent blocking of the secretion of adrenal C(19)-steroids, whether using corticoids or adrenostatics, could increase the effectiveness of androgen-deprivation therapy.
Collapse
Affiliation(s)
- L Stárka
- Institute of Endocrinology, Prague, Czech Republic.
| | | | | |
Collapse
|
32
|
Barnard L, Nikolaou N, Louw C, Schiffer L, Gibson H, Gilligan LC, Gangitano E, Snoep J, Arlt W, Tomlinson JW, Storbeck KH. The A-ring reduction of 11-ketotestosterone is efficiently catalysed by AKR1D1 and SRD5A2 but not SRD5A1. J Steroid Biochem Mol Biol 2020; 202:105724. [PMID: 32629108 DOI: 10.1016/j.jsbmb.2020.105724] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 06/25/2020] [Accepted: 06/30/2020] [Indexed: 11/23/2022]
Abstract
Testosterone and its 5α-reduced form, 5α-dihydrotestosterone, were previously thought to represent the only active androgens in humans. However, recent studies have shown that the potent androgen, 11-ketotestosterone, derived from the adrenal androgen precursor, 11β-hydroxyandrostenedione, may in fact serve as the primary androgen in healthy women. Yet, despite recent renewed interest in these steroids, their downstream metabolism has remained undetermined. We therefore set out to investigate the metabolism of 11-ketotestosterone by characterising the 5α- or 5β-reduction commitment step. We show that inactivation of 11-ketotestosterone is predominantly driven by AKR1D1, which efficiently catalyses the 5β-reduction of 11-ketotestosterone, committing it to a metabolic pathway that terminates in 11-ketoetiocholanolone. We demonstrate that 5α-reduction of 11-ketotestosterone is catalysed by SRD5A2, but not SRD5A1, and terminates in 11-ketoandrosterone, but is only responsible for a minority of 11-ketotestosterone inactivation. However, as 11-ketoetiocholanolone is also generated by the metabolism of the glucocorticoid cortisone, 11-ketoandrosterone should be considered a more specific urinary marker of 11-ketotestosterone production.
Collapse
Affiliation(s)
- Lise Barnard
- Department of Biochemistry, Stellenbosch University, Stellenbosch, 7600, South Africa
| | - Nikolaos Nikolaou
- Oxford Centre for Diabetes, Endocrinology and Metabolism, NIHR Oxford Biomedical Research Centre, University of Oxford, Churchill Hospital, Oxford, OX3 7LE, UK
| | - Carla Louw
- Department of Biochemistry, Stellenbosch University, Stellenbosch, 7600, South Africa
| | - Lina Schiffer
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, B15 2TT, UK
| | - Hylton Gibson
- Department of Biochemistry, Stellenbosch University, Stellenbosch, 7600, South Africa
| | - Lorna C Gilligan
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, B15 2TT, UK
| | - Elena Gangitano
- Oxford Centre for Diabetes, Endocrinology and Metabolism, NIHR Oxford Biomedical Research Centre, University of Oxford, Churchill Hospital, Oxford, OX3 7LE, UK; Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Jacky Snoep
- Department of Biochemistry, Stellenbosch University, Stellenbosch, 7600, South Africa; Molecular Cell Physiology, VU, Amsterdam, the Netherlands
| | - Wiebke Arlt
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, B15 2TT, UK; NIHR Birmingham Biomedical Research Centre, University of Birmingham and University Hospitals Birmingham NHS Foundation Trust, Birmingham, B15 3GW, UK
| | - Jeremy W Tomlinson
- Oxford Centre for Diabetes, Endocrinology and Metabolism, NIHR Oxford Biomedical Research Centre, University of Oxford, Churchill Hospital, Oxford, OX3 7LE, UK
| | - Karl-Heinz Storbeck
- Department of Biochemistry, Stellenbosch University, Stellenbosch, 7600, South Africa; Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, B15 2TT, UK.
| |
Collapse
|
33
|
Endo S, Morikawa Y, Kudo Y, Suenami K, Matsunaga T, Ikari A, Hara A. Human dehydrogenase/reductase SDR family member 11 (DHRS11) and aldo-keto reductase 1C isoforms in comparison: Substrate and reaction specificity in the reduction of 11-keto-C 19-steroids. J Steroid Biochem Mol Biol 2020; 199:105586. [PMID: 31926269 DOI: 10.1016/j.jsbmb.2020.105586] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 01/07/2020] [Accepted: 01/07/2020] [Indexed: 10/25/2022]
Abstract
Recent studies have shown that an adrenal steroid 11β-hydroxy-4-androstene-3,17-dione serves as the precursor to androgens, 11-ketotestosterone and 11-ketodihydrotestosterone (11KDHT). The biosynthetic pathways include the reduction of 3- and 17-keto groups of the androgen precursors 11-keto-C19-steroids, which has been reported to be mediated by three human enzymes; aldo-keto reductase (AKR)1C2, AKR1C3 and 17β-hydroxysteroid dehydrogenase (HSD) type-3. To explore the contribution of the enzymes in the reductive metabolism, we kinetically compared the substrate specificity for 11-keto-C19-steroids among purified recombinant preparations of four AKRs (1C1, 1C2,1C3 and 1C4) and DHRS11, which shows 17β-HSD activity. Although AKR1C1 did not reduce the 11-keto-C19-steroids, AKR1C3 and DHRS11 reduced 17-keto groups of 11-keto-4-androstene-3,17-dione, 11-keto-5α-androstane-3,17-dione (11K-Adione) and 11-ketoandrosterone with Km values of 5-28 μM. The 3-keto groups of 11KDHT and 11K-Adione were reduced by AKR1C4 (Km 1 μM) more efficiently than by AKR1C2 (Km 5 and 8 μM, respectively). GC/MS analysis of the products showed that DHRS11 acts as 17β-HSD, and that AKR1C2 and AKR1C4 are predominantly 3α-HSDs, but formed a minor 3β-metabolite from 11KDHT. Since DHRS11 was thus newly identified as 11-keto-C19-steroid reductase, we also investigated its substrate-binding mode by molecular docking and site-directed mutagenesis of Thr163 and Val200, and found the following structural features: 1). There is a space that accommodates the 11-keto group of the 11-keto-C19-steroids in the substrate-binding site. 2) Val200 is a critical determinant for exhibiting the strict 17β-HSD activity of the enzyme, because the Val200Leu mutation resulted in both significant impairment of the 17β-HSD activity and emergence of 3β-HSD activity towards 5α-androstanes including 11KDHT.
Collapse
Affiliation(s)
- Satoshi Endo
- Laboratory of Biochemistry, Gifu Pharmaceutical University, Gifu, 501-1196, Japan.
| | - Yoshifumi Morikawa
- Forensic Science Laboratory, Gifu Prefectural Police Headquarters, Gifu, 500-8501, Japan
| | - Yudai Kudo
- Laboratory of Biochemistry, Gifu Pharmaceutical University, Gifu, 501-1196, Japan
| | - Koichi Suenami
- Forensic Science Laboratory, Gifu Prefectural Police Headquarters, Gifu, 500-8501, Japan
| | - Toshiyuki Matsunaga
- Laboratory of Biochemistry, Gifu Pharmaceutical University, Gifu, 501-1196, Japan
| | - Akira Ikari
- Laboratory of Biochemistry, Gifu Pharmaceutical University, Gifu, 501-1196, Japan
| | - Akira Hara
- Faculty of Engineering, Gifu University, Gifu, 501-1193, Japan
| |
Collapse
|
34
|
Abstract
Although the fundamental symptoms of polycystic ovary syndrome (PCOS) relate most directly to ovarian dysfunction, central neuroendocrine systems play a prominent role in its pathophysiology. Gonadotropin-releasing hormone (GnRH) pulse generator resistance to negative feedback contributes to rapid GnRH pulse secretion, which promotes gonadotropin abnormalities that foster ovarian hyperandrogenemia and ovulatory dysfunction. The causes of GnRH neuron dysfunction, however, have remained enigmatic. In this review, we highlight a number of recent preclinical and clinical studies pertinent to the neuroendocrine abnormalities of PCOS, including those that have provided important insights into the relevance of animal models with PCOS-like features, the potential roles of kisspeptin and γ-aminobutyric acid (GABA)-ergic neurons, and the potential role of anti-Müllerian hormone.
Collapse
|
35
|
Reinehr T, Kulle A, Barth A, Ackermann J, Lass N, Holterhus PM. Sex Hormone Profile in Pubertal Boys With Gynecomastia and Pseudogynecomastia. J Clin Endocrinol Metab 2020; 105:5717688. [PMID: 31996898 DOI: 10.1210/clinem/dgaa044] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 01/27/2020] [Indexed: 12/19/2022]
Abstract
CONTENT Gynecomastia (defined by proliferation of glandular elements) and pseudogynecomastia (defined by adipose tissue) are frequent in pubertal boys. An association with sex hormones and the growth hormone axis has been discussed. OBJECTIVE The objective of this work is to compare sex hormones, insulin-like growth factor 1 (IGF-1), and insulin-like growth factor binding protein 3 (IGFBP-3) between boys with gynecomastia and pseudogynecomastia (separation by ultrasound). DESIGN An observational study was performed. SETTING The setting of this study was an outpatient clinic. PARTICIPANTS A total of 124 pubertal boys (mean age 14 ± 2 years) with breast enlargement and 84 healthy boys (mean age 14 ± 2 years) without breast enlargement participated in this study. INTERVENTIONS No interventions were performed. MAIN OUTCOME MEASURES Measurements were taken for sex hormones (progesterone, estradiol [E2], estriol, estrone, androstendione, testosterone [T], dihydrotestosterone) measured by liquid chromatography-tandem mass spectrometry, as well as gonadotropins, prolactin, IGF-1, and IGFBP-3. RESULTS Eighty-six boys suffered from gynecomastia and 38 from pseudogynecomastia. In boys with gynecomastia, the E2/T ratio (median 22, interquartile range [IQR] 8-75) was significantly (P < .05) higher compared to boys with pseudogynecomastia (median 12, IQR 5-21) or healthy controls without breast enlargement (median 18, IQR 6-44) even after adjustment for testes volume. T concentrations were significantly (P < .05) lower in boys with gynecomastia (median 1.8, IQR 0.7-4.2 nM/L) compared to boys with pseudogynecomastia (median 4.3, IQR 1.4-6.9 nM/L) or healthy controls without breast enlargement (median 3.1, IQR 0.6-7.6 nM/L). Boys with gynecomastia did not differ from boys with pseudogynecomastia according to other sex hormones, prolactin, IGF-1, or IGFBP-3 concentrations. CONCLUSIONS True gynecomastia is characterized by a relative T deficiency to E2 concentrations in contrast to pseudogynecomastia.
Collapse
Affiliation(s)
- Thomas Reinehr
- Department of Pediatric Endocrinology, Diabetes and Nutrition Medicine, Vestische Hospital for Children and Adolescents Datteln, University of Witten/Herdecke, Datteln, Germany
| | - Alexandra Kulle
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics, University Hospital of Schleswig-Holstein, Campus Kiel/Christian-Albrechts University of Kiel, Kiel, Germany
| | - Andre Barth
- Department of Pediatric Endocrinology, Diabetes and Nutrition Medicine, Vestische Hospital for Children and Adolescents Datteln, University of Witten/Herdecke, Datteln, Germany
| | - Jonas Ackermann
- Department of Pediatric Endocrinology, Diabetes and Nutrition Medicine, Vestische Hospital for Children and Adolescents Datteln, University of Witten/Herdecke, Datteln, Germany
| | - Nina Lass
- Department of Pediatric Endocrinology, Diabetes and Nutrition Medicine, Vestische Hospital for Children and Adolescents Datteln, University of Witten/Herdecke, Datteln, Germany
| | - Paul-Martin Holterhus
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics, University Hospital of Schleswig-Holstein, Campus Kiel/Christian-Albrechts University of Kiel, Kiel, Germany
| |
Collapse
|
36
|
Barnard M, Mostaghel EA, Auchus RJ, Storbeck KH. The role of adrenal derived androgens in castration resistant prostate cancer. J Steroid Biochem Mol Biol 2020; 197:105506. [PMID: 31672619 PMCID: PMC7883395 DOI: 10.1016/j.jsbmb.2019.105506] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 10/17/2019] [Accepted: 10/22/2019] [Indexed: 01/02/2023]
Abstract
Castration resistant prostate cancer (CRPC) remains androgen dependant despite castrate levels of circulating testosterone following androgen deprivation therapy, the first line of treatment for advanced metstatic prostate cancer. CRPC is characterized by alterations in the expression levels of steroidgenic enzymes that enable the tumour to derive potent androgens from circulating adrenal androgen precursors. Intratumoral androgen biosynthesis leads to the localized production of both canonical androgens such as 5α-dihydrotestosterone (DHT) as well as less well characterized 11-oxygenated androgens, which until recently have been overlooked in the context of CRPC. In this review we discuss the contribution of both canonical and 11-oxygenated androgen precursors to the intratumoral androgen pool in CRPC. We present evidence that CRPC remains androgen dependent and discuss the alterations in steroidogenic enzyme expression and how these affect the various pathways to intratumoral androgen biosynthesis. Finally we summarize the current treatment strategies for targeting adrenal derived androgen biosynthesis.
Collapse
Affiliation(s)
- Monique Barnard
- Department of Biochemistry, Stellenbosch University, Stellenbosch, South Africa
| | - Elahe A Mostaghel
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA; Department of Medicine, University of Washington, Seattle, WA, USA; Geriatric Research, Education and Clinical Center, VA Puget Sound Health Care System, Seattle, WA, USA
| | - Richard J Auchus
- Division of Metabolism, Endocrinology and Diabetes, University of Michigan, Ann Arbor, MI, USA; Department of Pharmacology, University of Michigan, Ann Arbor, MI, USA
| | - Karl-Heinz Storbeck
- Department of Biochemistry, Stellenbosch University, Stellenbosch, South Africa.
| |
Collapse
|
37
|
Bacila I, Adaway J, Hawley J, Mahdi S, Krone R, Patel L, Alvi S, Randell T, Gevers E, Dattani M, Cheetham T, Kyriakou A, Schiffer L, Ryan F, Crowne E, Davies JH, Ahmed SF, Keevil B, Krone N. Measurement of Salivary Adrenal-Specific Androgens as Biomarkers of Therapy Control in 21-Hydroxylase Deficiency. J Clin Endocrinol Metab 2019; 104:6417-6429. [PMID: 31361321 DOI: 10.1210/jc.2019-00031] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Accepted: 07/22/2019] [Indexed: 02/12/2023]
Abstract
BACKGROUND Monitoring of hormonal control represents a key part of the management of congenital adrenal hyperplasia (CAH). Monitoring strategies remain suboptimal because they rely on frequent blood tests and are not specific for adrenal-derived hormones. Recent evidence suggests the crucial role of adrenal-specific 11-oxygenated-C19 androgens in the pathogenesis of CAH. OBJECTIVE To establish a correlation between plasma and salivary adrenal-specific androgens in CAH as a noninvasive monitoring strategy. DESIGN This prospective cross-sectional study recruited patients between 2015 and 2018. SETTING Multicenter study including 13 tertiary centers in the United Kingdom. PARTICIPANTS Seventy-eight children with CAH and 62 matched healthy controls. METHODS Using liquid chromatography-tandem mass spectrometry, plasma and salivary concentrations of five steroids were measured: 17-hydroxyprogesterone (17OHP), androstenedione (A4), testosterone (T), 11-hydroxyandrostenedione (11OHA4), and 11-ketotestosterone (11KT). The correlation between plasma and salivary steroids was analyzed to assess their use in clinical practice. RESULTS Strong correlations between plasma and salivary steroid concentrations in patients with CAH were detected: 17OHP (rs = 0.871; P < 0.001), A4 (rs = 0.931; P < 0.001), T (rs = 0.867; P < 0.001), 11OH4A (rs = 0.876; P < 0.001), and 11KT (rs = 0.944; P < 0.001). These results were consistent for patient subgroups based on sex and age. Analysis of patient subgroups based on 17OHP concentrations established clear correlations between plasma and salivary concentrations of the adrenal-specific androgen 11KT. CONCLUSIONS The current study identified tight correlations between plasma and saliva for the adrenal-derived 11-oxygenated C19 androgen 11KT, as well as 17OHP and A4, which are widely used for monitoring treatment in CAH. This combination of steroid hormones will serve as an improved noninvasive salivary test for disease monitoring in patients with CAH.
Collapse
Affiliation(s)
- Irina Bacila
- Academic Unit of Child Health, Department of Oncology and Metabolism, University of Sheffield, Sheffield, United Kingdom
| | - Jo Adaway
- Department of Biochemistry, Manchester University NHS Foundation Trust, Manchester, United Kingdom
| | - James Hawley
- Department of Biochemistry, Manchester University NHS Foundation Trust, Manchester, United Kingdom
| | - Sundus Mahdi
- Academic Unit of Child Health, Department of Oncology and Metabolism, University of Sheffield, Sheffield, United Kingdom
| | - Ruth Krone
- Birmingham Women's & Children's Hospital, Birmingham, United Kingdom
| | - Leena Patel
- University of Manchester, Manchester, United Kingdom
| | - Sabah Alvi
- Leeds General Infirmary, Leeds, United Kingdom
| | | | - Evelien Gevers
- Queen Mary University London and Barts Health NHS Trust, The Royal London Hospital, London, United Kingdom
| | - Mehul Dattani
- Great Ormond Street Hospital, London, United Kingdom
| | - Timothy Cheetham
- Great North Children's Hospital, University of Newcastle, Newcastle, United Kingdom
| | - Andreas Kyriakou
- Developmental Endocrinology Research Group, University of Glasgow, Glasgow, United Kingdom
| | - Lina Schiffer
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom
| | - Fiona Ryan
- Oxford Children's Hospital, Oxford, United Kingdom
| | - Elizabeth Crowne
- Bristol Royal Hospital for Children, University Hospitals Bristol Foundation Trust, Bristol, United Kingdom
| | - Justin H Davies
- University Hospital Southampton, Southampton, United Kingdom
| | - Syed Faisal Ahmed
- Developmental Endocrinology Research Group, University of Glasgow, Glasgow, United Kingdom
| | - Brian Keevil
- Department of Clinical Biochemistry, Wythenshawe Hospital, Manchester, United Kingdom
| | - Nils Krone
- Academic Unit of Child Health, Department of Oncology and Metabolism, University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
38
|
Schiffer L, Barnard L, Baranowski ES, Gilligan LC, Taylor AE, Arlt W, Shackleton CHL, Storbeck KH. Human steroid biosynthesis, metabolism and excretion are differentially reflected by serum and urine steroid metabolomes: A comprehensive review. J Steroid Biochem Mol Biol 2019; 194:105439. [PMID: 31362062 PMCID: PMC6857441 DOI: 10.1016/j.jsbmb.2019.105439] [Citation(s) in RCA: 232] [Impact Index Per Article: 38.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 07/24/2019] [Accepted: 07/25/2019] [Indexed: 02/07/2023]
Abstract
Advances in technology have allowed for the sensitive, specific, and simultaneous quantitative profiling of steroid precursors, bioactive steroids and inactive metabolites, facilitating comprehensive characterization of the serum and urine steroid metabolomes. The quantification of steroid panels is therefore gaining favor over quantification of single marker metabolites in the clinical and research laboratories. However, although the biochemical pathways for the biosynthesis and metabolism of steroid hormones are now well defined, a gulf still exists between this knowledge and its application to the measured steroid profiles. In this review, we present an overview of steroid hormone biosynthesis and metabolism by the liver and peripheral tissues, specifically highlighting the pathways linking and differentiating the serum and urine steroid metabolomes. A brief overview of the methodology used in steroid profiling is also provided.
Collapse
Affiliation(s)
- Lina Schiffer
- Institute of Metabolism and Systems Research (IMSR), University of Birmingham, Birmingham, UK
| | - Lise Barnard
- Department of Biochemistry, Stellenbosch University, Stellenbosch, South Africa
| | - Elizabeth S Baranowski
- Institute of Metabolism and Systems Research (IMSR), University of Birmingham, Birmingham, UK; Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, UK; Department of Paediatric Endocrinology and Diabetes, Birmingham Women's and Children's Hospital NHS Foundation Trust, Birmingham, UK
| | - Lorna C Gilligan
- Institute of Metabolism and Systems Research (IMSR), University of Birmingham, Birmingham, UK
| | - Angela E Taylor
- Institute of Metabolism and Systems Research (IMSR), University of Birmingham, Birmingham, UK
| | - Wiebke Arlt
- Institute of Metabolism and Systems Research (IMSR), University of Birmingham, Birmingham, UK; Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, UK; NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust & University of Birmingham, Birmingham, UK
| | - Cedric H L Shackleton
- Institute of Metabolism and Systems Research (IMSR), University of Birmingham, Birmingham, UK; UCSF Benioff Children's Hospital Oakland Research Institute, Oakland, CA, USA
| | - Karl-Heinz Storbeck
- Institute of Metabolism and Systems Research (IMSR), University of Birmingham, Birmingham, UK; Department of Biochemistry, Stellenbosch University, Stellenbosch, South Africa.
| |
Collapse
|
39
|
Abstract
PURPOSE OF REVIEW Polycystic ovary syndrome (PCOS) is often difficult to diagnose in adolescents. Recent recommendations and concepts regarding the diagnosis and treatment of PCOS in the adolescent girl are considered. RECENT FINDINGS The diagnosis of PCOS in adolescents should be primarily based on clinical and biochemical signs of hyperandrogenism and presentation with irregular menses. Because of the similarity of normal pubertal development and features of PCOS, the diagnosis should be deferred until at least 2 years following menarche. For girls who do not fulfill the diagnostic criteria, the focus should be on treatment of symptoms. SUMMARY PCOS is a complex, multifaceted disorder, and should be diagnosed and treated in adolescents after taking into consideration the patient's full diagnostic picture, metabolic risks, and individual concerns, to both avoid overdiagnosis but yet be able to provide early and meaningful interventions.
Collapse
|
40
|
Engels M, Span PN, van Herwaarden AE, Sweep FCGJ, Stikkelbroeck NMML, Claahsen-van der Grinten HL. Testicular Adrenal Rest Tumors: Current Insights on Prevalence, Characteristics, Origin, and Treatment. Endocr Rev 2019; 40:973-987. [PMID: 30882882 DOI: 10.1210/er.2018-00258] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 01/23/2019] [Indexed: 11/19/2022]
Abstract
This review provides the reader with current insights on testicular adrenal rest tumors (TARTs), a complication in male patients with congenital adrenal hyperplasia (CAH). In recent studies, an overall TART prevalence of 40% (range, 14% to 89%) in classic patients with CAH is found. Reported differences are mainly caused by the method of detection and the selected patient population. Biochemically, histologically, and molecularly, TARTs exhibit particular adrenal characteristics and were therefore thought to originate from aberrant adrenal cells. More recently, TARTs have been found to also exhibit testicular characteristics. This has led to the hypothesis of pluripotent cells as the origin of TARTs. High concentrations of ACTH could cause hyperplasia of these pluripotent cells, as TARTs appear to be associated with poor hormonal control with concomitant elevated ACTH. Unfortunately, as yet there are no methods to prevent the development of TARTs, nor are there guidelines to treat patients with TARTs. Intensified glucocorticoid treatment could improve fertility status in some cases, although studies report contradicting results. TARTs can also lead to irreversible testicular damage, and therefore semen cryopreservation could be offered to patients with TARTs. Further research should focus on the etiology and pharmacological treatment to prevent TART development or to treat TARTs and improve the fertility status of patients with TARTs.
Collapse
Affiliation(s)
- Manon Engels
- Department of Pediatrics, Amalia Children's Hospital, Radboud University Medical Center, Nijmegen, Netherlands.,Department of Laboratory Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Paul N Span
- Department of Radiation Oncology, Radiotherapy and OncoImmunology Laboratory, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Antonius E van Herwaarden
- Department of Laboratory Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Fred C G J Sweep
- Department of Laboratory Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | | | | |
Collapse
|
41
|
Rege J, Turcu AF, Else T, Auchus RJ, Rainey WE. Steroid biomarkers in human adrenal disease. J Steroid Biochem Mol Biol 2019; 190:273-280. [PMID: 30707926 PMCID: PMC6707065 DOI: 10.1016/j.jsbmb.2019.01.018] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 01/24/2019] [Accepted: 01/27/2019] [Indexed: 01/24/2023]
Abstract
Adrenal steroidogenesis is a robust process, involving a series of enzymatic reactions that facilitate conversion of cholesterol into biologically active steroid hormones under the stimulation of angiotensin II, adrenocorticotropic hormone and other regulators. The biosynthesis of mineralocorticoids, glucocorticoids, and adrenal-derived androgens occur in separate adrenocortical zones as a result of the segregated expression of steroidogenic enzymes and cofactors. This mini review provides the principles of adrenal steroidogenesis, including the classic and under-appreciated 11-oxygenated androgen pathways. Several adrenal diseases result from dysregulated adrenal steroid synthesis. Herein, we review growing evidence that adrenal diseases exhibit characteristic modifications from normal adrenal steroid pathways that provide opportunities for the discovery of biomarker steroids that would improve diagnosis and monitoring of adrenal disorders.
Collapse
Affiliation(s)
- Juilee Rege
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, United States
| | - Adina F Turcu
- Division of Metabolism, Endocrinology, and Diabetes, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, United States
| | - Tobias Else
- Division of Metabolism, Endocrinology, and Diabetes, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, United States
| | - Richard J Auchus
- Division of Metabolism, Endocrinology, and Diabetes, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, United States; Department of Pharmacology, University of Michigan, Ann Arbor, MI 48109, United States
| | - William E Rainey
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, United States; Division of Metabolism, Endocrinology, and Diabetes, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, United States.
| |
Collapse
|