1
|
Yang Z, Ping YQ, Wang MW, Zhang C, Zhou SH, Xi YT, Zhu KK, Ding W, Zhang QY, Song ZC, Zhao RJ, He ZL, Wang MX, Qi L, Ullmann C, Ricken A, Schöneberg T, Gan ZJ, Yu X, Xiao P, Yi F, Liebscher I, Sun JP. Identification, structure, and agonist design of an androgen membrane receptor. Cell 2025:S0092-8674(25)00035-2. [PMID: 39884271 DOI: 10.1016/j.cell.2025.01.006] [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: 12/14/2023] [Revised: 10/29/2024] [Accepted: 01/03/2025] [Indexed: 02/01/2025]
Abstract
Androgens, such as 5α-dihydrotestosterone (5α-DHT), regulate numerous functions by binding to nuclear androgen receptors (ARs) and potential unknown membrane receptors. Here, we report that the androgen 5α-DHT activates membrane receptor GPR133 in muscle cells, thereby increasing intracellular cyclic AMP (cAMP) levels and enhancing muscle strength. Further cryoelectron microscopy (cryo-EM) structural analysis of GPR133-Gs in complex with 5α-DHT or its derivative methenolone (MET) reveals the structural basis for androgen recognition. Notably, the presence of the "Φ(F/L)2.64-F3.40-W6.53" and the "F7.42××N/D7.46" motifs, which recognize the hydrophobic steroid core and polar groups, respectively, are common in adhesion GPCRs (aGPCRs), suggesting that many aGPCRs may recognize different steroid hormones. Finally, we exploited in silico screening methods to identify a small molecule, AP503, which activates GPR133 and separates the beneficial muscle-strengthening effects from side effects mediated by AR. Thus, GPR133 represents an androgen membrane receptor that contributes to normal androgen physiology and has important therapeutic potentials.
Collapse
Affiliation(s)
- Zhao Yang
- Key Laboratory Experimental Teratology of the Ministry of Education, New Cornerstone Science Laboratory, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, and Advanced Medical Research Institute, NHC Key Laboratory of Otorhinolaryngology, Qilu hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Yu-Qi Ping
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Ming-Wei Wang
- Key Laboratory Experimental Teratology of the Ministry of Education, New Cornerstone Science Laboratory, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, and Advanced Medical Research Institute, NHC Key Laboratory of Otorhinolaryngology, Qilu hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, China; Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Chao Zhang
- Key Laboratory Experimental Teratology of the Ministry of Education, New Cornerstone Science Laboratory, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, and Advanced Medical Research Institute, NHC Key Laboratory of Otorhinolaryngology, Qilu hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Shu-Hua Zhou
- Key Laboratory Experimental Teratology of the Ministry of Education, New Cornerstone Science Laboratory, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, and Advanced Medical Research Institute, NHC Key Laboratory of Otorhinolaryngology, Qilu hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, China; Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Yue-Tong Xi
- Key Laboratory Experimental Teratology of the Ministry of Education, New Cornerstone Science Laboratory, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, and Advanced Medical Research Institute, NHC Key Laboratory of Otorhinolaryngology, Qilu hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Kong-Kai Zhu
- Key Laboratory Experimental Teratology of the Ministry of Education, New Cornerstone Science Laboratory, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, and Advanced Medical Research Institute, NHC Key Laboratory of Otorhinolaryngology, Qilu hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Wei Ding
- Laboratory of Soft Matter Physics, Institute of Physics, Chinese Academy of Sciences, Beijing, China
| | - Qi-Yue Zhang
- Key Laboratory Experimental Teratology of the Ministry of Education, New Cornerstone Science Laboratory, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, and Advanced Medical Research Institute, NHC Key Laboratory of Otorhinolaryngology, Qilu hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Zhi-Chen Song
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Ru-Jia Zhao
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Zi-Lu He
- Key Laboratory Experimental Teratology of the Ministry of Education, New Cornerstone Science Laboratory, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, and Advanced Medical Research Institute, NHC Key Laboratory of Otorhinolaryngology, Qilu hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Meng-Xin Wang
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Lei Qi
- Biomedical Research Center for Structural Analysis, Shandong University, Jinan 250012, Shandong, China
| | - Christian Ullmann
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, 04103 Leipzig, Germany
| | - Albert Ricken
- Institute of Anatomy, Medical Faculty, Leipzig University, 04103 Leipzig, Germany
| | - Torsten Schöneberg
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, 04103 Leipzig, Germany
| | - Zhen-Ji Gan
- Medical School of Nanjing University, Nanjing University, Nanjing 210061, China
| | - Xiao Yu
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China.
| | - Peng Xiao
- Key Laboratory Experimental Teratology of the Ministry of Education, New Cornerstone Science Laboratory, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, and Advanced Medical Research Institute, NHC Key Laboratory of Otorhinolaryngology, Qilu hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, China; Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, China.
| | - Fan Yi
- Key Laboratory of Infection and Immunity of Shandong Province, Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan 250012, China.
| | - Ines Liebscher
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, 04103 Leipzig, Germany.
| | - Jin-Peng Sun
- Key Laboratory Experimental Teratology of the Ministry of Education, New Cornerstone Science Laboratory, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, and Advanced Medical Research Institute, NHC Key Laboratory of Otorhinolaryngology, Qilu hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, China; Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University, Beijing 100191, China.
| |
Collapse
|
2
|
Su Z, Shi C, Fan Y, Gao Y, Wang L, Ren X, Shen Y, Wang M. Engineering a two-enzyme system in Mycolicibacterium neoaurum for efficient biotransformation of phytosterols to dihydrotestosterone. Int J Biol Macromol 2025; 287:138443. [PMID: 39645107 DOI: 10.1016/j.ijbiomac.2024.138443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 11/30/2024] [Accepted: 12/04/2024] [Indexed: 12/09/2024]
Abstract
Dihydrotestosterone (DHT) is a valuable steroid drug with widespread clinical applications, but traditional chemical synthesis is environmentally harmful and requires complex reaction conditions. This study introduces a one-step microbial transformation method for the production of DHT from inexpensive phytosterols (PS) using engineered Mycolicibacterium neoaurum (MNR). The heterologous expression of 5α-reductase (5α-R) and 17β-hydroxysteroid dehydrogenase (17β-HSD) in MNR enabled the efficient conversion of PS to DHT. To further enhance 5α-R activity, semi-rational mutagenesis was employed, which significantly increasing DHT production. Molecular dynamics simulations provided insights into the underlying mechanisms driving this enhancement. Additionally, the incorporation of a PntAB-based NADPH regeneration system further improved DHT yield. Process optimization resulted in a maximum DHT concentration of 1.123 g·L-1, representing the first gram-scale microbial production of DHT. Compared to traditional chemical synthesis, this biotransformation method offers milder reaction conditions, reduced environmental impact, and eliminates the need for toxic catalysts. This work demonstrates a sustainable and efficient microbial method for DHT production, with significant industrial potential for the greener manufacturing of steroid hormone drugs.
Collapse
Affiliation(s)
- Zhenhua Su
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin Key Laboratory of Industrial Microbiology, College of Biotechnology, Tianjin University of Science & Technology, Tianjin 300457, China
| | - Chang Shi
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin Key Laboratory of Industrial Microbiology, College of Biotechnology, Tianjin University of Science & Technology, Tianjin 300457, China
| | - Yunshuang Fan
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin Key Laboratory of Industrial Microbiology, College of Biotechnology, Tianjin University of Science & Technology, Tianjin 300457, China
| | - Yuanyuan Gao
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin Key Laboratory of Industrial Microbiology, College of Biotechnology, Tianjin University of Science & Technology, Tianjin 300457, China
| | - Lu Wang
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin Key Laboratory of Industrial Microbiology, College of Biotechnology, Tianjin University of Science & Technology, Tianjin 300457, China
| | - Xiaoxian Ren
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin Key Laboratory of Industrial Microbiology, College of Biotechnology, Tianjin University of Science & Technology, Tianjin 300457, China
| | - Yanbing Shen
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin Key Laboratory of Industrial Microbiology, College of Biotechnology, Tianjin University of Science & Technology, Tianjin 300457, China.
| | - Min Wang
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin Key Laboratory of Industrial Microbiology, College of Biotechnology, Tianjin University of Science & Technology, Tianjin 300457, China.
| |
Collapse
|
3
|
Angeloni E, Germelli L, Costa B, Martini C, Da Pozzo E. Neurosteroids and Translocator Protein (TSPO) in neuroinflammation. Neurochem Int 2025; 182:105916. [PMID: 39681140 DOI: 10.1016/j.neuint.2024.105916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 11/28/2024] [Accepted: 12/12/2024] [Indexed: 12/18/2024]
Abstract
Neurosteroids have a crucial role in physiological intrinsic regulations of the Central Nervous System functions. They are derived from peripheral steroidogenic sources and from the de novo neurosteroidogenic capacity of brain cells. Significant alterations of neurosteroid levels have been frequently observed in neuroinflammation and neurodegenerative diseases. Such level fluctuations may be useful for both diagnosis and treatment of these pathological conditions. Beyond steroid administration, enhancing the endogenous production by Translocator Protein (TSPO) targeting has been proposed to restore these altered pathological levels. However, the neurosteroid quantification and the prediction of their final effects are often troublesome, sometimes controversial and context dependent, due to the complexity of neurosteroid biosynthetic pathway and to the low produced amounts. The aim of this review is to report recent advances, and technical limitations, in neurosteroid-related strategies against neuroinflammation.
Collapse
Affiliation(s)
- Elisa Angeloni
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126, Pisa, Italy
| | - Lorenzo Germelli
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126, Pisa, Italy
| | - Barbara Costa
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126, Pisa, Italy
| | - Claudia Martini
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126, Pisa, Italy
| | - Eleonora Da Pozzo
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126, Pisa, Italy.
| |
Collapse
|
4
|
Mrabet HE, Ben Salem H, Ach T, Ben Abdelkarim A, Alaya W. Effects of SGLT-2 inhibitors on clinical and biological hyperandrogenism and menstruation irregularities in patients with polycystic ovary syndrome: A systematic review of randomized trials. SAGE Open Med 2024; 12:20503121241308997. [PMID: 39713268 PMCID: PMC11660270 DOI: 10.1177/20503121241308997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Accepted: 12/05/2024] [Indexed: 12/24/2024] Open
Abstract
Introduction Polycystic ovary syndrome is a common chronic condition characterized by insulin resistance and hyperandrogenism, leading to significant health risks and impaired quality of life. Sodium-glucose transporter type 2 inhibitors have shown promise in improving the metabolic profile of women with polycystic ovary syndrome. However, their impact on hormonal parameters and cycle disorders remains uncertain. Methods This systematic review analyzed randomized clinical trials published up to 1 December 2023, comparing sodium-glucose transporter type 2 inhibitors to metformin, other antidiabetic agents, or placebo in women with polycystic ovary syndrome. The primary outcomes were changes in total testosterone, free androgen index, dehydroepiandrosterone sulfate, delta-4 androstenedione, and cycle disorders. Results Five randomized studies were included, evaluating canagliflozin, dapagliflozin, licogliflozin, or empagliflozin against metformin, exenatide, or placebo, with a total of 214 participants. Improvements in total testosterone and dehydroepiandrosterone sulfatewere observed in some studies, but the effects were inconsistent across drugs and outcomes. Additionally, two studies reported beneficial effects on cycle disorders. Conclusions Sodium-glucose transporter type 2 inhibitors appear to have a potential but variable impact on hormonal parameters in women with polycystic ovary syndrome. However, larger and longer-duration studies are needed to fully elucidate their long-term efficacy in addressing hyperandrogenism and improving overall outcomes in these patients.
Collapse
Affiliation(s)
- Houcem Elomma Mrabet
- Department of Endocrinology, Diabetology, and Internal Medicine, Tahar Sfar University Hospital, Mahdia, Tunisia
| | - Houda Ben Salem
- Department of General Medicine, Eljem Constituency Hospital, Eljem, Mahdia, Tunisia
| | - Taieb Ach
- Department of Endocrinology, Farhat Hachad University Hospital, Sousse, Tunisia
| | - Asma Ben Abdelkarim
- Department of Endocrinology, Farhat Hachad University Hospital, Sousse, Tunisia
| | - Wafa Alaya
- Department of Endocrinology, Diabetology, and Internal Medicine, Tahar Sfar University Hospital, Mahdia, Tunisia
| |
Collapse
|
5
|
Theodorakis N, Feretzakis G, Vamvakou G, Verykios VS, Polymeris A, Nikolaou M. Testosterone therapy for functional hypogonadism in middle-aged and elderly males: current evidence and future perspectives. Hormones (Athens) 2024; 23:801-817. [PMID: 39060901 DOI: 10.1007/s42000-024-00587-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024]
Abstract
Population aging is a global phenomenon driving research focus toward preventing and managing age-related disorders. Functional hypogonadism (FH) has been defined as the combination of low testosterone levels, typically serum total testosterone below 300-350 ng/dL, together with manifestations of hypogonadism, in the absence of an intrinsic pathology of the hypothalamic-pituitary-testicular (HPT) axis. It is usually seen in middle-aged or elderly males as a product of aging and multimorbidity. This age-related decline in testosterone levels has been associated with numerous adverse outcomes. Testosterone therapy (TTh) is the mainstay of treatment for organic hypogonadism with an identifiable intrinsic pathology of the HPT axis. Current guidelines generally make weak recommendations for TTh in patients with FH, mostly in the presence of sexual dysfunction. Concerns about long-term safety have historically limited TTh use in middle-aged and elderly males with FH. However, recent randomized controlled trials and meta-analyses have demonstrated safe long-term outcomes regarding prostatic and cardiovascular health, together with decreases in all-cause mortality and improvements in various domains, including sexual function, body composition, physical strength, bone density, and hematopoiesis. Furthermore, there are numerous insightful studies suggesting additional benefits of TTh, for instance in cardio-renal-metabolic conditions. Specifically, future trials should investigate the role of TTh in improving symptoms and prognosis in various clinical contexts, including sarcopenia, frailty, dyslipidemia, arterial hypertension, diabetes mellitus, fracture risk, heart failure, stable angina, chronic kidney disease, mood disorders, and cognitive dysfunction.
Collapse
Affiliation(s)
- Nikolaos Theodorakis
- Department of Cardiology & 65+ Clinic, Sismanogleio-Amalia Fleming General Hospital, 14, 25th Martiou Str, Melissia, 15127, Greece
- School of Medicine, National and Kapodistrian University of Athens, 75 Mikras Asias, Athens, 11527, Greece
| | - Georgios Feretzakis
- School of Science and Technology, Hellenic Open University, 18 Aristotelous Str, Patras, 26335, Greece.
| | - Georgia Vamvakou
- Department of Cardiology & 65+ Clinic, Sismanogleio-Amalia Fleming General Hospital, 14, 25th Martiou Str, Melissia, 15127, Greece
| | - Vassilios S Verykios
- School of Science and Technology, Hellenic Open University, 18 Aristotelous Str, Patras, 26335, Greece
| | - Antonis Polymeris
- Department of Endocrinology, Metabolism and Diabetes Mellitus, Sismanogleio-Amalia Fleming General Hospital, 14, 25th Martiou Str, Melissia, 15127, Greece
| | - Maria Nikolaou
- Department of Cardiology & 65+ Clinic, Sismanogleio-Amalia Fleming General Hospital, 14, 25th Martiou Str, Melissia, 15127, Greece
| |
Collapse
|
6
|
Lawrence BM, O'Donnell L, Gannon A, Smith S, Curley MK, Darbey A, Mackay R, O'Shaughnessy PJ, Smith LB, Rebourcet D. Compensatory mechanisms that maintain androgen production in mice lacking key androgen biosynthetic enzymes. FASEB J 2024; 38:e70177. [PMID: 39556387 PMCID: PMC11698012 DOI: 10.1096/fj.202402093r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 10/28/2024] [Accepted: 11/01/2024] [Indexed: 11/19/2024]
Abstract
Testosterone and dihydrotestosterone (DHT) are essential for male development and fertility. In the canonical androgen production pathway, testosterone is produced in the testis by HSD17B3; however, adult male Hsd17b3 knockout (KO) mice continue to produce androgens and are fertile, indicating compensatory mechanisms exist. A second, alternate pathway produces DHT from precursors other than testosterone via 5α-reductase (SRD5A) activity. We hypothesized that the alternate pathway contributes to androgen bioactivity in Hsd17b3 KO mice. To investigate contributions arising from and interactions between the canonical and alternate pathways, we pharmacologically inhibited SRD5A and ablated Srd5a1 (the predominant SRD5A in the testis) on the background of Hsd17b3 KO mice. Mice with perturbation of either the canonical or both pathways exhibited increased LH, testicular steroidogenic enzyme expression, and normal reproductive tracts and fertility. In the circulation, alternate pathway steroids were increased in the absence of HSD17B3 but were reduced by co-inhibition of SRD5A1. Mice with perturbations of both pathways produced normal basal levels of intratesticular testosterone, suggesting the action of other unidentified hydroxysteroid dehydrogenase(s). Strikingly, testicular expression of another SRD5A enzyme, Srd5a2, was markedly increased in the absence of Hsd17b3, suggesting a compensatory increase in SRD5A2 to maintain androgen bioactivity during HSD17B3 deficiency. Finally, we observed elevated circulating concentrations of the 11-keto-derivative of DHT, suggesting compensatory extra-gonadal induction of bioactive 11-keto androgen production. Taken together, we conclude that, in the absence of the canonical pathway of androgen production, multiple intra- and extra-gonadal mechanisms cooperate to maintain testosterone and DHT production, supporting male development and fertility.
Collapse
Affiliation(s)
- Ben M. Lawrence
- College of Engineering, Science and EnvironmentThe University of NewcastleCallaghanNew South WalesAustralia
- School of BioSciencesThe University of MelbourneParkvilleVictoriaAustralia
| | - Liza O'Donnell
- Office of the Deputy Vice Chancellor (Research)Griffith UniversitySouthportQueenslandAustralia
| | - Anne‐Louise Gannon
- College of Engineering, Science and EnvironmentThe University of NewcastleCallaghanNew South WalesAustralia
| | - Sarah Smith
- College of Engineering, Science and EnvironmentThe University of NewcastleCallaghanNew South WalesAustralia
| | - Michael K. Curley
- MRC Centre for Reproductive HealthUniversity of Edinburgh, The Queen's Medical Research InstituteEdinburghUK
| | - Annalucia Darbey
- MRC Centre for Reproductive HealthUniversity of Edinburgh, The Queen's Medical Research InstituteEdinburghUK
| | - Rosa Mackay
- MRC Centre for Reproductive HealthUniversity of Edinburgh, The Queen's Medical Research InstituteEdinburghUK
| | - Peter J. O'Shaughnessy
- School of Biodiversity, One Health & Veterinary Medicine, College of Medical, Veterinary and Life SciencesUniversity of Glasgow, Garscube CampusGlasgowUK
| | - Lee B. Smith
- Office of the Deputy Vice Chancellor (Research)Griffith UniversitySouthportQueenslandAustralia
| | - Diane Rebourcet
- College of Engineering, Science and EnvironmentThe University of NewcastleCallaghanNew South WalesAustralia
- Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) ‐ UMR_S 1085Univ RennesRennesFrance
| |
Collapse
|
7
|
Kollerov VV, Timakova TA, Shutov AA, Donova MV. Boldenone and Testosterone Production from Phytosterol via One-Pot Cascade Biotransformations. J Fungi (Basel) 2024; 10:830. [PMID: 39728326 DOI: 10.3390/jof10120830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 11/26/2024] [Accepted: 11/27/2024] [Indexed: 12/28/2024] Open
Abstract
Testosterone (TS) and its 1(2)-dehydrogenated derivative boldenone (BD) are widely used in medicine, veterinary science and as precursors in organic synthesis of many therapeutic steroids. Green production of these compounds is possible from androstenedione (AD) enzymatically, or from phytosterol (PS) using fermentation stages. In this study, the ascomycete Curvularia sp. VKM F-3040 was shown to convert androstadienedione (ADD, 4 and 10 g/L) to yield 97% and 78% (mol/mol) of BD, respectively. Based on its high 17β-hydroxysteroid dehydrogenase (17β-HSD) activity, a novel cascade biotransformation of PS was developed for production of TS and BD. At the first stage, the strains of Mycolicibacterium neoaurum VKM Ac-1815D or M. neoaurum VKM Ac-1816D converted PS (5 or 10 g/L) into AD or ADD (each in a concentration of 2.5 or 5 g/L), respectively. At the second stage, mycelium of the fungus under the revealed optimal conditions reduced AD or ADD with more than 90% efficiency to form TS or BD, respectively. Based on transcriptome analysis, six candidate genes that might encode 17β-HSDs in the Curvularia sp. genome were revealed. Along with 17β-HSDs, the fungus possessed inducible P450cur 7-monooxygenase, which led to the accumulation of 7α-hydroxytestosterone (7α-OH-TS) as a major product from AD (up to 83% within 24 h after mycelium addition at the second stage of cascade biotransformation). The presence of protein synthesis inhibitor cycloheximide (CHX) prevented 7α/β-hydroxylation due to inhibition of de novo synthesis of the enzyme in the fungal cells. The results demonstrate the high biotechnological potential of the Curvularia sp. strain and open up prospects for the synthesis of valuable 17β-reduced and 7-hydroxylated steroids by cascade biotransformations.
Collapse
Affiliation(s)
- Vyacheslav V Kollerov
- Federal Research Center, Pushchino Center for Biological Research of Russian Academy of Sciences, G.K. Skryabin Institute of Biochemistry and Physiology of Microorganisms, Prospekt Nauki, 5, 142290 Pushchino, Moscow Region, Russia
| | - Tatiana A Timakova
- Federal Research Center, Pushchino Center for Biological Research of Russian Academy of Sciences, G.K. Skryabin Institute of Biochemistry and Physiology of Microorganisms, Prospekt Nauki, 5, 142290 Pushchino, Moscow Region, Russia
| | - Andrei A Shutov
- Federal Research Center, Pushchino Center for Biological Research of Russian Academy of Sciences, G.K. Skryabin Institute of Biochemistry and Physiology of Microorganisms, Prospekt Nauki, 5, 142290 Pushchino, Moscow Region, Russia
| | - Marina V Donova
- Federal Research Center, Pushchino Center for Biological Research of Russian Academy of Sciences, G.K. Skryabin Institute of Biochemistry and Physiology of Microorganisms, Prospekt Nauki, 5, 142290 Pushchino, Moscow Region, Russia
| |
Collapse
|
8
|
Sasako T, Ilboudo Y, Liang KYH, Chen Y, Yoshiji S, Richards JB. The Influence of Trinucleotide Repeats in the Androgen Receptor Gene on Androgen-related Traits and Diseases. J Clin Endocrinol Metab 2024; 109:3234-3244. [PMID: 38701087 PMCID: PMC11570371 DOI: 10.1210/clinem/dgae302] [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: 02/09/2024] [Revised: 04/26/2024] [Accepted: 05/01/2024] [Indexed: 05/05/2024]
Abstract
CONTEXT Trinucleotide repeats in the androgen receptor have been proposed to influence testosterone signaling in men, but the clinical relevance of these trinucleotide repeats remains controversial. OBJECTIVE To examine how androgen receptor trinucleotide repeat lengths affect androgen-related traits and disease risks and whether they influence the clinical importance of circulating testosterone levels. METHODS We quantified CAG and GGC repeat lengths in the androgen receptor (AR) gene of European-ancestry male participants in the UK Biobank from whole-genome and whole-exome sequence data using ExpansionHunter and tested associations with androgen-related traits and diseases. We also examined whether the associations between testosterone levels and these outcomes were affected by adjustment for the repeat lengths. RESULTS We successfully quantified the repeat lengths from whole-genome and/or whole-exome sequence data in 181 217 males. Both repeat lengths were shown to be positively associated with circulating total testosterone level and bone mineral density, whereas CAG repeat length was negatively associated with male-pattern baldness, but their effects were relatively small and were not associated with most of the other outcomes. Circulating total testosterone level was associated with various outcomes, but this relationship was not affected by adjustment for the repeat lengths. CONCLUSION In this large-scale study, we found that longer CAG and GGC repeats in the AR gene influence androgen resistance, elevate circulating testosterone level via a feedback loop, and play a role in some androgen-targeted tissues. Generally, however, circulating testosterone level is a more important determinant of androgen action in males than repeat lengths.
Collapse
Affiliation(s)
- Takayoshi Sasako
- McGill University, Montréal, Québec H3T 1E2, Canada
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montréal, Québec H3T 1E2, Canada
- Tanaka Diabetes Clinic Omiya, Saitama 330-0846, Japan
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo 113-0033, Japan
| | - Yann Ilboudo
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montréal, Québec H3T 1E2, Canada
| | - Kevin Y H Liang
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montréal, Québec H3T 1E2, Canada
- Quantitative Life Sciences Program, McGill University, Montréal, Québec H3T 1E2, Canada
| | - Yiheng Chen
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montréal, Québec H3T 1E2, Canada
- Department of Human Genetics, McGill University, Montréal, Québec H3T 1E2, Canada
| | - Satoshi Yoshiji
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montréal, Québec H3T 1E2, Canada
- Department of Human Genetics, McGill University, Montréal, Québec H3T 1E2, Canada
- Kyoto-McGill International Collaborative Program in Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
- Japan Society for the Promotion of Science, Tokyo 102-0083, Japan
| | - J Brent Richards
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montréal, Québec H3T 1E2, Canada
- Department of Human Genetics, McGill University, Montréal, Québec H3T 1E2, Canada
- Five Prime Sciences Inc, Montréal, Québec H3Y 2W4, Canada
- Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montréal, Québec H3T 1E2, Canada
- Department of Twin Research, King's College London, London WC2R 2LS, UK
| |
Collapse
|
9
|
Hsu SH, Chen LR, Chen KH. Primary Osteoporosis Induced by Androgen and Estrogen Deficiency: The Molecular and Cellular Perspective on Pathophysiological Mechanisms and Treatments. Int J Mol Sci 2024; 25:12139. [PMID: 39596206 PMCID: PMC11593909 DOI: 10.3390/ijms252212139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 11/02/2024] [Accepted: 11/05/2024] [Indexed: 11/28/2024] Open
Abstract
Primary osteoporosis is closely linked to hormone deficiency, which disrupts the balance of bone remodeling. It affects postmenopausal women but also significantly impacts older men. Estrogen can promote the production of osteoprotegerin, a decoy receptor for RANKL, thereby preventing RANKL from activating osteoclasts. Furthermore, estrogen promotes osteoblast survival and function via activation of the Wnt signaling pathway. Likewise, androgens play a critical role in bone metabolism, primarily through their conversion to estrogen in men. Estrogen deficiency accelerates bone resorption through a rise in pro-inflammatory cytokines (IL-1, IL-6, TNF-α) and RANKL, which promote osteoclastogenesis. In the classic genomic pathway, estrogen binds to estrogen receptors in the cytoplasm, forming a complex that migrates to the nucleus and binds to estrogen response elements on DNA, regulating gene transcription. Androgens can be defined as high-affinity ligands for the androgen receptor; their combination can serve as a ligand-inducible transcription factor. Hormone replacement therapy has shown promise but comes with associated risks and side effects. In contrast, the non-genomic pathway involves rapid signaling cascades initiated at the cell membrane, influencing cellular functions without directly altering gene expression. Therefore, the ligand-independent actions and rapid signaling pathways of estrogen and androgen receptors can be harnessed to develop new drugs that provide bone protection without the side effects of traditional hormone therapies. To manage primary osteoporosis, other pharmacological treatments (bisphosphonates, teriparatide, RANKL inhibitors, sclerostin inhibitors, SERMs, and calcitonin salmon) can ameliorate osteoporosis and improve BMD via actions on different pathways. Non-pharmacological treatments include nutritional support and exercise, as well as the dietary intake of antioxidants and natural products. The current study reviews the processes of bone remodeling, hormone actions, hormone receptor status, and therapeutic targets of primary osteoporosis. However, many detailed cellular and molecular mechanisms underlying primary osteoporosis seem complicated and unexplored and warrant further investigation.
Collapse
Affiliation(s)
- Shao-Heng Hsu
- Department of Medical Education, Taipei Tzu-Chi Hospital, The Buddhist Tzu-Chi Medical Foundation, New Taipei City 231, Taiwan;
| | - Li-Ru Chen
- Department of Physical Medicine and Rehabilitation, Mackay Memorial Hospital, Taipei 104, Taiwan;
- Department of Mechanical Engineering, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - Kuo-Hu Chen
- Department of Obstetrics and Gynecology, Taipei Tzu-Chi Hospital, The Buddhist Tzu-Chi Medical Foundation, New Taipei City 231, Taiwan
- School of Medicine, Tzu-Chi University, Hualien 970, Taiwan
| |
Collapse
|
10
|
Page ST. Synthetic androgens for male contraception. Contraception 2024:110735. [PMID: 39515746 DOI: 10.1016/j.contraception.2024.110735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 10/25/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024]
Abstract
High global rates of unintended pregnancy (>40% of all pregnancies) are relatively stagnant despite a broad array of female contraceptive options. Data suggest many men are enthusiastic to utilize novel, reversible, male contraceptives, particularly those that may be administered orally. Hormonal male contraception utilizes androgens or androgen-progestin combinations to exploit endocrine negative feedback, interrupting the hypothalamic-pituitary-testicular axis to suppress spermatogenesis. Modifications of testosterone allow for tailoring of steroid activity and show promise as single agent hormonal male contraceptives. The addition of progestin activity by molecular modification of testosterone could enhance the degree and speed of sperm suppression compared to testosterone alone using a single, exogenous steroid. Moreover, modifications that block reduction of testosterone to dihydrotestosterone could diminish androgenic stimulation of prostate tissue. This review summarizes data in the development of modified androgens for male hormonal contraception and suggest a path forward for these molecules in helping to fill the gap in contraceptive technologies for men.
Collapse
Affiliation(s)
- Stephanie T Page
- University of Washington, Division of Metabolism, Endocrinology and Nutrition, Seattle, WA, United States.
| |
Collapse
|
11
|
Draskau MK, Rosenmai AK, Bouftas N, Johansson HKL, Panagiotou EM, Holmer ML, Elmelund E, Zilliacus J, Beronius A, Damdimopoulou P, van Duursen M, Svingen T. AOP Report: An Upstream Network for Reduced Androgen Signaling Leading to Altered Gene Expression of Androgen Receptor-Responsive Genes in Target Tissues. ENVIRONMENTAL TOXICOLOGY AND CHEMISTRY 2024; 43:2329-2337. [PMID: 39206816 DOI: 10.1002/etc.5972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 07/16/2024] [Indexed: 09/04/2024]
Abstract
Adverse outcome pathways (AOPs) can aid with chemical risk assessment by providing plausible links between chemical activity at the molecular level and effect outcomes in intact organisms. Because AOPs can be used to infer causality between upstream and downstream events in toxicological pathways, the AOP framework can also facilitate increased uptake of alternative methods and new approach methodologies to help inform hazard identification. However, a prevailing challenge is the limited number of fully developed and endorsed AOPs, primarily due to the substantial amount of work required by AOP developers and reviewers. Consequently, a more pragmatic approach to AOP development has been proposed where smaller units of knowledge are developed and reviewed independent of full AOPs. In this context, we have developed an upstream network comprising key events (KEs) and KE relationships related to decreased androgen signaling, converging at a nodal KE that can branch out to numerous adverse outcomes (AOs) relevant to androgen-sensitive toxicological pathways. Androgen signaling represents an extensively studied pathway for endocrine disruption. It is linked to numerous disease outcomes and can be affected by many different endocrine-disrupting chemicals. Still, pathways related to disrupted androgen signaling remain underrepresented in the AOP-wiki, and endorsed AOPs are lacking. Given the pivotal role of androgen signaling in development and function across vertebrate taxa and life stages of both sexes, this upstream AOP network serves as a foundational element for developing numerous AOPs. By connecting the upstream network with various downstream AOs, encompassing different species, it can also facilitate cross-species extrapolations for hazard and risk assessment of chemicals. Environ Toxicol Chem 2024;43:2329-2337. © 2024 The Author(s). Environmental Toxicology and Chemistry published by Wiley Periodicals LLC on behalf of SETAC.
Collapse
Affiliation(s)
- Monica K Draskau
- National Food Institute, Technical University of Denmark, Lyngby, Denmark
| | - Anna K Rosenmai
- National Food Institute, Technical University of Denmark, Lyngby, Denmark
| | - Nora Bouftas
- Environmental Health and Toxicology, Amsterdam Institute for Life and Environment, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | | | - Eleftheria M Panagiotou
- Division of Obstetrics and Gynaecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
- Department of Gynecology and Reproductive Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Marie L Holmer
- National Food Institute, Technical University of Denmark, Lyngby, Denmark
| | - Emilie Elmelund
- National Food Institute, Technical University of Denmark, Lyngby, Denmark
| | - Johanna Zilliacus
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Anna Beronius
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Pauliina Damdimopoulou
- Division of Obstetrics and Gynaecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
- Department of Gynecology and Reproductive Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Majorie van Duursen
- Environmental Health and Toxicology, Amsterdam Institute for Life and Environment, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Terje Svingen
- National Food Institute, Technical University of Denmark, Lyngby, Denmark
| |
Collapse
|
12
|
Effah W, Khalil M, Hwang DJ, Miller DD, Narayanan R. Advances in the understanding of androgen receptor structure and function and in the development of next-generation AR-targeted therapeutics. Steroids 2024; 210:109486. [PMID: 39111362 PMCID: PMC11380798 DOI: 10.1016/j.steroids.2024.109486] [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: 05/23/2024] [Revised: 07/29/2024] [Accepted: 08/01/2024] [Indexed: 08/10/2024]
Abstract
Androgen receptor (AR) and its ligand androgens are important for development and physiology of various tissues. AR and its ligands also play critical role in the development of various diseases, making it a valuable therapeutic target. AR ligands, both agonists and antagonists, are being widely used to treat pathological conditions, including prostate cancer and hypogonadism. Despite AR being studied widely over the last five decades, the last decade has seen striking advances in the knowledge on AR and discoveries that have the potential to translate to the clinic. This review provides an overview of the advances in AR biology, AR molecular mechanisms of action, and next generation molecules that are currently in development. Several of the areas described in the review are just unraveling and the next decade will bring more clarity on these developments that will put AR at the forefront of both basic biology and drug development.
Collapse
Affiliation(s)
- Wendy Effah
- Department of Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Marjana Khalil
- Department of Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Dong-Jin Hwang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Duane D Miller
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Ramesh Narayanan
- Department of Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States; UTHSC Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN, United States.
| |
Collapse
|
13
|
Sinha A, Deb VK, Datta A, Yadav S, Phulkar A, Adhikari S. Evaluation of structural features of anabolic-androgenic steroids: entanglement for organ-specific toxicity. Steroids 2024; 212:109518. [PMID: 39322097 DOI: 10.1016/j.steroids.2024.109518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 09/16/2024] [Accepted: 09/20/2024] [Indexed: 09/27/2024]
Abstract
Anabolic-androgenic steroids (AASs), more correctly termed "steroidal androgens", are a broad category of compounds including both synthetic derivatives and endogenously produced androgens like testosterone, which have long been employed as performance-enhancing substances, primarily among recreational athletes and some professionals. While their short-term effects on muscle physiology are well-documented, the long-term health consequences remain inadequately understood. A key finding is the disruption of hormone production, leading to reversible and irreversible changes, particularly with prolonged use. While debate exists over the prevalence of adverse effects, studies suggest a spectrum of somatic and psychiatric consequences, highlighting the need for improved understanding and prevention strategies. AASs are not only affect muscle structure but also influence mood, behavior, and body image, potentially exacerbating substance dependence and psychological distress. Liver alterations are a prominent concern, with oxidative stress implicated in AAS-induced hepatotoxicity. Reproductive complications, including gonadal atrophy and infertility, are common, alongside virilization and feminization effects in both genders. Cardiovascular effects are particularly worrisome, with AASs implicated in hypertension, dyslipidemia, and increased thrombotic risk, contributing to cardiovascular morbidity and mortality. Moreover, AASs may enhance cancer risks, potentially accelerating carcinogenesis in various tissues, including the prostate. The review emphasizes the need for comprehensive public health initiatives to mitigate harm, including harm minimization strategies, routine health screenings, and targeted interventions for AAS users. Understanding the complex interplay of biological mechanisms and systemic effects is crucial for informing clinical management and preventive measures. This review also examines the biological impact of AASs on human muscles, detailing mechanisms of action, chemistry, and associated health risks such as liver damage, cardiovascular disease, and endocrine dysfunction.
Collapse
Affiliation(s)
- Ankan Sinha
- Department of Physical Education, Govt. Degree College, Dharmanagar, Tripura(N) 799253, India.
| | - Vishal Kumar Deb
- School of Health Sciences and Technology, UPES, Dehradun 248007, Uttarakhand, India
| | - Abhijit Datta
- Department of Botany, Ambedkar College, Fatikroy, Unakoti 799290 Tripura, India
| | - Satpal Yadav
- Department of Sports Biomechanics, LNIPE, NERC, Guwahati 782402 Assam, India
| | - Ashish Phulkar
- Department of Sports Management and Coaching, LNIPE, Gwalior 474002, Madhya Pradesh, India
| | - Suman Adhikari
- Department of Chemistry, Govt. Degree College, Dharmanagar, Tripura(N) 799253, India.
| |
Collapse
|
14
|
Xu G, Dai G, Huang Z, Guan Q, Du C, Xu X. The Etiology and Pathogenesis of Benign Prostatic Hyperplasia: The Roles of Sex Hormones and Anatomy. Res Rep Urol 2024; 16:205-214. [PMID: 39345801 PMCID: PMC11430843 DOI: 10.2147/rru.s477396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 09/09/2024] [Indexed: 10/01/2024] Open
Abstract
Benign prostatic hyperplasia (BPH) mainly causes lower urinary tract symptoms in ageing men, but its exact etiology and pathogenesis have not been established. The objective of this review was to design an update on the advances of human BPH research. We undertook a literature search for identifying studies of the roles of sex hormones (androgens and estrogens) in the onset and development of human BPH using the Pubmed database. In literature, many studies have indicated that ageing and obesity are the factors for preceding the onset of BPH. No evidence for the role of testosterone (T) or dihydrotestosterone (DHT) is found in BPH initiation. Since BPH exclusively occurs in the transitional zone (TZ) surrounding the urethra, it is postulated that years of exposure to uncharacterized urinary toxins could disrupt the homeostasis of the stroma and/or epithelium of this prostatic zone that are typically occurring in ageing men. After cellular damage and subsequent inflammation generated, the intraprostatic DHT produced mainly from T by 5α-reductase promotes BPH development. Further, estrogens could take part in the nodular proliferation of stromal cells in some BPH patients. The confounding of BPH may attenuate the development of prostate tumor in the TZ. In conclusion, evidence in literature suggests that androgens are not etiological factors for BPH, and intraprostatic DHT along with chronic inflammation are mainly responsible for nodular proliferation of stromal and/or epithelial cells in prostatic TZ. The urinary factors for the etiology of BPH and BPH as a prediction of PCa progression still need further investigation.
Collapse
Affiliation(s)
- Ganzhe Xu
- Shanghai Medical College, Fudan University, Shanghai, People’s Republic of China
| | - Guoyu Dai
- Department of Biomedical Engineering, Guoke Ningbo Life Science and Health Industry Research Institute, Ningbo, Zhejiang, People’s Republic of China
| | - Zhongli Huang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Qiunong Guan
- Department of Biomedical Engineering, Guoke Ningbo Life Science and Health Industry Research Institute, Ningbo, Zhejiang, People’s Republic of China
| | - Caigan Du
- Department of Biomedical Engineering, Guoke Ningbo Life Science and Health Industry Research Institute, Ningbo, Zhejiang, People’s Republic of China
| | - Xiaoming Xu
- Department of Urology, Ningbo No. 2 Hospital, Ningbo, Zhejiang, People’s Republic of China
| |
Collapse
|
15
|
Ko SH. Effects of Heat Stress-Induced Sex Hormone Dysregulation on Reproduction and Growth in Male Adolescents and Beneficial Foods. Nutrients 2024; 16:3032. [PMID: 39275346 PMCID: PMC11397449 DOI: 10.3390/nu16173032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 09/03/2024] [Accepted: 09/07/2024] [Indexed: 09/16/2024] Open
Abstract
Heat stress due to climate warming can significantly affect the synthesis of sex hormones in male adolescents, which can impair the ability of the hypothalamus to secrete gonadotropin-releasing hormone on the hypothalamic-pituitary-gonadal axis, which leads to a decrease in luteinizing hormone and follicle-stimulating hormone, which ultimately negatively affects spermatogenesis and testosterone synthesis. For optimal spermatogenesis, the testicular temperature should be 2-6 °C lower than body temperature. Heat stress directly affects the testes, damaging them and reducing testosterone synthesis. Additionally, chronic heat stress abnormally increases the level of aromatase in Leydig cells, which increases estradiol synthesis while decreasing testosterone, leading to an imbalance of sex hormones and spermatogenesis failure. Low levels of testosterone in male adolescents lead to delayed puberty and incomplete sexual maturation, negatively affect height growth and bone mineral density, and can lead to a decrease in lean body mass and an increase in fat mass. In order for male adolescents to acquire healthy reproductive capacity, it is recommended to provide sufficient nutrition and energy, avoid exposure to heat stress, and provide foods and supplements to prevent or repair testosterone reduction, germ cell damage, and sperm count reduction caused by heat stress so that they can enter a healthy adulthood.
Collapse
Affiliation(s)
- Seong-Hee Ko
- Major in Food Science and Nutrition, College of Human Ecology, Sookmyung Women's University, Seoul 04310, Republic of Korea
| |
Collapse
|
16
|
Safi R, Wardell SE, Watkinson P, Qin X, Lee M, Park S, Krebs T, Dolan EL, Blattler A, Tsuji T, Nayak S, Khater M, Fontanillo C, Newlin MA, Kirkland ML, Xie Y, Long H, Fink EC, Fanning SW, Runyon S, Brown M, Xu S, Owzar K, Norris JD, McDonnell DP. Androgen receptor monomers and dimers regulate opposing biological processes in prostate cancer cells. Nat Commun 2024; 15:7675. [PMID: 39227594 PMCID: PMC11371910 DOI: 10.1038/s41467-024-52032-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Accepted: 08/23/2024] [Indexed: 09/05/2024] Open
Abstract
Most prostate cancers express the androgen receptor (AR), and tumor growth and progression are facilitated by exceptionally low levels of systemic or intratumorally produced androgens. Thus, absolute inhibition of the androgen signaling axis remains the goal of current therapeutic approaches to treat prostate cancer (PCa). Paradoxically, high dose androgens also exhibit considerable efficacy as a treatment modality in patients with late-stage metastatic PCa. Here we show that low levels of androgens, functioning through an AR monomer, facilitate a non-genomic activation of the mTOR signaling pathway to drive proliferation. Conversely, high dose androgens facilitate the formation of AR dimers/oligomers to suppress c-MYC expression, inhibit proliferation and drive a transcriptional program associated with a differentiated phenotype. These findings highlight the inherent liabilities in current approaches used to inhibit AR action in PCa and are instructive as to strategies that can be used to develop new therapeutics for this disease and other androgenopathies.
Collapse
Affiliation(s)
- Rachid Safi
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, USA
| | - Suzanne E Wardell
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, USA
| | - Paige Watkinson
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, USA
| | - Xiaodi Qin
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC, USA
| | - Marissa Lee
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC, USA
| | - Sunghee Park
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, USA
| | - Taylor Krebs
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, USA
| | - Emma L Dolan
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, USA
| | - Adam Blattler
- Oncogenesis Thematic Research Center, Bristol Myers Squibb, San Diego, CA, USA
| | - Toshiya Tsuji
- Oncogenesis Thematic Research Center, Bristol Myers Squibb, San Diego, CA, USA
| | - Surendra Nayak
- Oncogenesis Thematic Research Center, Bristol Myers Squibb, San Diego, CA, USA
| | - Marwa Khater
- Informatics and Predictive Sciences, Bristol Myers Squibb, San Diego, CA, USA
| | - Celia Fontanillo
- Informatics and Predictive Sciences, Bristol Myers Squibb, San Diego, CA, USA
| | - Madeline A Newlin
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, USA
| | - Megan L Kirkland
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, USA
| | | | - Henry Long
- Dana-Farber Cancer Institute, Boston, MA, USA
| | - Emma C Fink
- Department of Cancer Biology, Loyola University, Maywood, IL, USA
| | - Sean W Fanning
- Department of Cancer Biology, Loyola University, Maywood, IL, USA
| | - Scott Runyon
- RTI International, Research Triangle Park, NC, USA
| | - Myles Brown
- Dana-Farber Cancer Institute, Boston, MA, USA
| | - Shuichan Xu
- Oncogenesis Thematic Research Center, Bristol Myers Squibb, San Diego, CA, USA
| | - Kouros Owzar
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC, USA
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC, USA
| | - John D Norris
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, USA
| | - Donald P McDonnell
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
17
|
Lakshmikanth T, Consiglio C, Sardh F, Forlin R, Wang J, Tan Z, Barcenilla H, Rodriguez L, Sugrue J, Noori P, Ivanchenko M, Piñero Páez L, Gonzalez L, Habimana Mugabo C, Johnsson A, Ryberg H, Hallgren Å, Pou C, Chen Y, Mikeš J, James A, Dahlqvist P, Wahlberg J, Hagelin A, Holmberg M, Degerblad M, Isaksson M, Duffy D, Kämpe O, Landegren N, Brodin P. Immune system adaptation during gender-affirming testosterone treatment. Nature 2024; 633:155-164. [PMID: 39232147 PMCID: PMC11374716 DOI: 10.1038/s41586-024-07789-z] [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: 04/06/2023] [Accepted: 07/04/2024] [Indexed: 09/06/2024]
Abstract
Infectious, inflammatory and autoimmune conditions present differently in males and females. SARS-CoV-2 infection in naive males is associated with increased risk of death, whereas females are at increased risk of long COVID1, similar to observations in other infections2. Females respond more strongly to vaccines, and adverse reactions are more frequent3, like most autoimmune diseases4. Immunological sex differences stem from genetic, hormonal and behavioural factors5 but their relative importance is only partially understood6-8. In individuals assigned female sex at birth and undergoing gender-affirming testosterone therapy (trans men), hormone concentrations change markedly but the immunological consequences are poorly understood. Here we performed longitudinal systems-level analyses in 23 trans men and found that testosterone modulates a cross-regulated axis between type-I interferon and tumour necrosis factor. This is mediated by functional attenuation of type-I interferon responses in both plasmacytoid dendritic cells and monocytes. Conversely, testosterone potentiates monocyte responses leading to increased tumour necrosis factor, interleukin-6 and interleukin-15 production and downstream activation of nuclear factor kappa B-regulated genes and potentiation of interferon-γ responses, primarily in natural killer cells. These findings in trans men are corroborated by sex-divergent responses in public datasets and illustrate the dynamic regulation of human immunity by sex hormones, with implications for the health of individuals undergoing hormone therapy and our understanding of sex-divergent immune responses in cisgender individuals.
Collapse
Affiliation(s)
| | - Camila Consiglio
- Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden
- Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Fabian Sardh
- Center for Molecular Medicine, Department of Medicine, Karolinska Institutet, Solna, Sweden
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Rikard Forlin
- Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden
| | - Jun Wang
- Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden
| | - Ziyang Tan
- Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden
| | - Hugo Barcenilla
- Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden
| | - Lucie Rodriguez
- Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden
| | - Jamie Sugrue
- Translational Immunology Unit, Institut Pasteur, Paris, France
| | - Peri Noori
- Center for Molecular Medicine, Department of Medicine, Karolinska Institutet, Solna, Sweden
| | - Margarita Ivanchenko
- Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden
| | - Laura Piñero Páez
- Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden
| | - Laura Gonzalez
- Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden
| | | | - Anette Johnsson
- Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden
| | - Henrik Ryberg
- Department of Clinical Chemistry, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Internal Medicine and Clinical Nutrition, University of Gothenburg, Gothenburg, Sweden
| | - Åsa Hallgren
- Center for Molecular Medicine, Department of Medicine, Karolinska Institutet, Solna, Sweden
| | - Christian Pou
- Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden
| | - Yang Chen
- Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden
| | - Jaromír Mikeš
- Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden
| | - Anna James
- Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden
| | - Per Dahlqvist
- Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | | | - Anders Hagelin
- ANOVA, Karolinska University Hospital, Stockholm, Sweden
- Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Mats Holmberg
- ANOVA, Karolinska University Hospital, Stockholm, Sweden
- Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Marie Degerblad
- ANOVA, Karolinska University Hospital, Stockholm, Sweden
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Solna, Sweden
| | - Magnus Isaksson
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Darragh Duffy
- Translational Immunology Unit, Institut Pasteur, Paris, France
| | - Olle Kämpe
- Center for Molecular Medicine, Department of Medicine, Karolinska Institutet, Solna, Sweden
- Department of Endocrinology, Metabolism and Diabetes, Karolinska University Hospital, Stockholm, Sweden
| | - Nils Landegren
- Center for Molecular Medicine, Department of Medicine, Karolinska Institutet, Solna, Sweden.
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden.
| | - Petter Brodin
- Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden.
- Medical Research Council, Laboratory of Medical Sciences, London, UK.
- Department of Immunology and Inflammation, Imperial College London, London, UK.
| |
Collapse
|
18
|
Wang J, Su C, Qian M, Wang X, Chen C, Liu Y, Liu W, Xiang Z, Xu B. Subchronic toxic effects of bisphenol A on the gut-liver-hormone axis in rats via intestinal flora and metabolism. Front Endocrinol (Lausanne) 2024; 15:1415216. [PMID: 39268238 PMCID: PMC11390593 DOI: 10.3389/fendo.2024.1415216] [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: 04/10/2024] [Accepted: 07/30/2024] [Indexed: 09/15/2024] Open
Abstract
Background Bisphenol A (BPA), a characteristic endocrine disruptor, is a substance that seriously interferes with the human endocrine system and causes reproductive disorders and developmental abnormalities. However, its toxic effects on the gut-liver-hormone axis are still unclear. Method Male and female rats were exposed to BPA (300 mg/kg) by oral gavage for 60 consecutive days. H&E staining was used for histopathological evaluation, and the serum biochemical indexes were determined using an automatic analyzer. The 16S rRNA gene sequencing was used to detect the intestinal microbial diversity, and the GC-MS was used to analyze the contents of short-chain fatty acids (SCFAs) in colon contents. UPLC-QTOF MS was used to analyze the related metabolites. The ELISA method was used to assess the levels of serum inflammatory factors. Results Histopathological analysis indicated that the liver, heart, and testis were affected by BPA. There was a significant effect on alanine aminotransferase (ALT), triglyceride (TG), total cholesterol (TC), and low-density lipoprotein (LDL) in the male-BPA group (P < 0.05), and globulin (GLB), indirect bilirubin (IBIL), alkaline phosphatase (ALP), ALT, TG, TC, high-density lipoprotein (HDL), and creatinine (Cr) in the female-BPA group (P < 0.05). Metagenomics (16S rRNA gene sequencing) analysis indicated that BPA reduced the diversity and changed the composition of gut microbiota in rats significantly. Compared with the control and blank groups, the contents of caproic acid, isobutyric acid, isovaleric acid, and propanoic acid in the colon contents decreased in the male-BPA group (P < 0.05), and caproic acid, isobutyric acid, isovaleric acid, and valeric acid in the colon contents decreased in the female-BPA group (P < 0.05). Metabolomic analysis of the serum indicated that BPA could regulate bile acid levels, especially ursodeoxycholic acid (UDCA) and its conjugated forms. The contents of amino acids, hormones, and lipids were also significantly affected after exposure to BPA. The increase in interleukin-6 (IL-6), interleukin-23 (IL-23), and transforming growth factor-β (TGF-β) in the serum of the male-BPA group suggests that BPA exposure affects the immune system. Conclusion BPA exposure will cause toxicity to rats via disrupting the gut-liver-hormone axis.
Collapse
Affiliation(s)
- Jiaqi Wang
- School of Pharmaceutical Science, Liaoning University, Shenyang, China
- Shenyang Key Laboratory for Causes and Drug Discovery of Chronic Diseases, Shenyang, China
| | - Ce Su
- Pharmacy Department, Shenyang Tenth People's Hospital, Shenyang, China
| | - Mingqin Qian
- Department of Ultrasound, People's Hospital of Liaoning Province, Shenyang, China
| | - Xin Wang
- School of Pharmaceutical Science, Liaoning University, Shenyang, China
- Shenyang Key Laboratory for Causes and Drug Discovery of Chronic Diseases, Shenyang, China
| | - Changlan Chen
- School of Pharmaceutical Science, Liaoning University, Shenyang, China
| | - Yangcheng Liu
- School of Pharmaceutical Science, Liaoning University, Shenyang, China
- Shenyang Key Laboratory for Causes and Drug Discovery of Chronic Diseases, Shenyang, China
| | - Wei Liu
- School of Pharmaceutical Science, Liaoning University, Shenyang, China
| | - Zheng Xiang
- School of Pharmaceutical Science, Liaoning University, Shenyang, China
- Shenyang Key Laboratory for Causes and Drug Discovery of Chronic Diseases, Shenyang, China
| | - Baoli Xu
- Department of Pharmacy, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| |
Collapse
|
19
|
Planinic A, Maric T, Himelreich Peric M, Jezek D, Katusic Bojanac A. Dynamics of HSD17B3 expression in human fetal testis: implications for the role of Sertoli cells in fetal testosterone biosynthesis. Front Cell Dev Biol 2024; 12:1429292. [PMID: 39139451 PMCID: PMC11319120 DOI: 10.3389/fcell.2024.1429292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 07/09/2024] [Indexed: 08/15/2024] Open
Abstract
Introduction: Androgens play a pivotal role in shaping male sexual characteristics, with testosterone being an essential hormone in orchestrating various developmental processes. Testosterone biosynthesis involves a series of enzymatic reactions, among which the 17β-hydroxysteroid dehydrogenase type 3 (HSD17B3) holds significance. While its role in adult Leydig cells is well established, its localization and importance during the fetal period remain less known, especially in humans. This study aims to delineate the dynamics of HSD17B3 expression in human fetal testes to clarify the contribution of specific cell types to testosterone biosynthesis. Methods: Using immunofluorescence staining, we investigated the expression pattern of HSD17B3 in human fetal and adult testicular tissues. Results and discussion: The findings of this study revealed a distinct temporal and cellular expression pattern of HSD17B3 protein in the fetal period. We detected its expression exclusively in Sertoli cells, the highest during the second trimester. This unique localization suggests the inclusion of fetal Sertoli cells in testosterone production during the critical masculinization-programming window. Furthermore, we demonstrated a shift in HSD17B3 expression from Sertoli cells to Leydig cells in adulthood, corroborating findings from rodent studies. This study sheds light on the intricate, still underexplored regulation of steroidogenesis during fetal development, whose disturbance might lead to testicular dysgenesis. Further research is warranted to elucidate the regulatory pathways governing the expression of HSD17B3 and its transition between Sertoli and Leydig cells, potentially paving the way for novel therapeutic interventions in disorders of sexual development.
Collapse
Affiliation(s)
- Ana Planinic
- Department of Histology and Embryology, University of Zagreb School of Medicine, Zagreb, Croatia
- Scientific Centre of Excellence for Reproductive and Regenerative Medicine, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Tihana Maric
- Scientific Centre of Excellence for Reproductive and Regenerative Medicine, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Marta Himelreich Peric
- Scientific Centre of Excellence for Reproductive and Regenerative Medicine, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Davor Jezek
- Department of Histology and Embryology, University of Zagreb School of Medicine, Zagreb, Croatia
- Scientific Centre of Excellence for Reproductive and Regenerative Medicine, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Ana Katusic Bojanac
- Scientific Centre of Excellence for Reproductive and Regenerative Medicine, University of Zagreb School of Medicine, Zagreb, Croatia
- Department of Medical Biology, University of Zagreb School of Medicine, Zagreb, Croatia
| |
Collapse
|
20
|
Dourson AJ, Darken RS, Baranski TJ, Gereau RW, Ross WT, Nahman-Averbuch H. The role of androgens in migraine pathophysiology. NEUROBIOLOGY OF PAIN (CAMBRIDGE, MASS.) 2024; 16:100171. [PMID: 39498299 PMCID: PMC11532460 DOI: 10.1016/j.ynpai.2024.100171] [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: 07/12/2024] [Revised: 10/01/2024] [Accepted: 10/04/2024] [Indexed: 11/07/2024]
Abstract
Migraine affects ∼12 % of the worldwide population and is more prevalent in females, which suggests a role of sex hormones in migraine pathophysiology. Most studies have focused on estrogen and progesterone, and the involvement of androgens has been less studied. However, due to the recent advances in androgen interventions, which could advance new androgen-based migraine treatments, it is critical to better understand the role of androgens in migraine. Testosterone, the most studied androgen, was found to have an antinociceptive effect in various animal and human pain studies. Thus, it could also have a protective effect related to lower migraine severity and prevalence. In this review, we discuss studies examining the role of androgens on migraine-related symptoms in migraine animal models. Additionally, we summarize the results of human studies comparing androgen levels between patients with migraine and healthy controls, studies assessing the relationships between androgen levels and migraine severity, and intervention studies examining the impact of testosterone treatment on migraine severity. Many of the studies have limitations, however, the results suggest that androgens may have a minor effect on migraine. Still, it is possible that androgens are involved in migraine pathophysiology in a sub-group of patients such as in adolescents or postmenopausal women. We discuss potential mechanisms in which testosterone, as the main androgen tested, can impact migraine. These mechanisms range from the cellular level to systems and behavior and include the effect of testosterone on sensory neurons, the immune and vascular systems, the stress response, brain function, and mood. Lastly, we suggest future directions to advance this line of research.
Collapse
Affiliation(s)
- Adam J. Dourson
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA
- Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Rachel S. Darken
- Department of Neurology, Washington University School of Medicine, St. Louis Missouri, USA
| | - Thomas J. Baranski
- Division of Endocrinology, Diabetes and Metabolism Washington University School of Medicine in St. Louis Missouri, USA
| | - Robert W. Gereau
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA
- Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Whitney Trotter Ross
- Division of Minimally Invasive Gynecologic Surgery, Washington University in St. Louis, St. Louis, MO, USA
| | - Hadas Nahman-Averbuch
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA
- Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
21
|
Wu Z, Ma Y, Chen S, Liu Y, Liu X, Cao H, Jin T, Li L, Huang M, Yang F, Dong W. Arginine Biosynthesis Mediates Wulingzhi Extract Resistance to Busulfan-Induced Male Reproductive Toxicity. Int J Mol Sci 2024; 25:6320. [PMID: 38928028 PMCID: PMC11203605 DOI: 10.3390/ijms25126320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 06/01/2024] [Accepted: 06/05/2024] [Indexed: 06/28/2024] Open
Abstract
Busulfan, an indispensable medicine in cancer treatment, can cause serious reproductive system damage to males as a side effect of its otherwise excellent therapeutic results. Its widespread use has also caused its accumulation in the environment and subsequent ecotoxicology effects. As a Chinese medicine, Wulingzhi (WLZ) has the effects of promoting blood circulation and improving female reproductive function. However, the potential effects of WLZ in male reproduction and in counteracting busulfan-induced testis damage, as well as its probable mechanisms, are still ambiguous. In this study, busulfan was introduced in a mouse model to evaluate its production of the testicular damage. The components of different WLZ extracts were compared using an untargeted metabolome to select extracts with greater efficacy, which were further confirmed in vivo. Here, we demonstrate abnormal spermatogenesis and low sperm quality in busulfan-injured testes. The WLZ extracts showed a strong potential to rehabilitate the male reproductive system; this effect was more prominent in room-temperature extracts. Additionally, both water and ethanol WLZ extracts at room temperature alleviated various busulfan-induced adverse effects. In particular, WLZ recovered spermatogenesis, re-activated arginine biosynthesis, and alleviated the increased oxidative stress and inflammation in the testis, ultimately reversing the busulfan-induced testicular injury. Collectively, these results suggest a promising approach to protecting the male reproductive system from busulfan-induced adverse side effects, as well as those of other similar anti-cancer drugs.
Collapse
Affiliation(s)
- Zifang Wu
- College of Animal Science and Technology, Northwest A&F University, Xianyang 712100, China; (Z.W.); (Y.M.); (S.C.); (Y.L.); (H.C.); (T.J.); (L.L.); (M.H.)
| | - Yuxuan Ma
- College of Animal Science and Technology, Northwest A&F University, Xianyang 712100, China; (Z.W.); (Y.M.); (S.C.); (Y.L.); (H.C.); (T.J.); (L.L.); (M.H.)
| | - Shaoxian Chen
- College of Animal Science and Technology, Northwest A&F University, Xianyang 712100, China; (Z.W.); (Y.M.); (S.C.); (Y.L.); (H.C.); (T.J.); (L.L.); (M.H.)
| | - Yuyan Liu
- College of Animal Science and Technology, Northwest A&F University, Xianyang 712100, China; (Z.W.); (Y.M.); (S.C.); (Y.L.); (H.C.); (T.J.); (L.L.); (M.H.)
| | - Xianglin Liu
- College of Forestry, Northwest A&F University, Xianyang 712100, China;
| | - Heran Cao
- College of Animal Science and Technology, Northwest A&F University, Xianyang 712100, China; (Z.W.); (Y.M.); (S.C.); (Y.L.); (H.C.); (T.J.); (L.L.); (M.H.)
| | - Tianqi Jin
- College of Animal Science and Technology, Northwest A&F University, Xianyang 712100, China; (Z.W.); (Y.M.); (S.C.); (Y.L.); (H.C.); (T.J.); (L.L.); (M.H.)
| | - Long Li
- College of Animal Science and Technology, Northwest A&F University, Xianyang 712100, China; (Z.W.); (Y.M.); (S.C.); (Y.L.); (H.C.); (T.J.); (L.L.); (M.H.)
| | - Mengqi Huang
- College of Animal Science and Technology, Northwest A&F University, Xianyang 712100, China; (Z.W.); (Y.M.); (S.C.); (Y.L.); (H.C.); (T.J.); (L.L.); (M.H.)
| | - Fangxia Yang
- College of Forestry, Northwest A&F University, Xianyang 712100, China;
| | - Wuzi Dong
- College of Animal Science and Technology, Northwest A&F University, Xianyang 712100, China; (Z.W.); (Y.M.); (S.C.); (Y.L.); (H.C.); (T.J.); (L.L.); (M.H.)
- College of Forestry, Northwest A&F University, Xianyang 712100, China;
| |
Collapse
|
22
|
Zhao J, Wang Q, Tan AF, Loh CJL, Toh HC. Sex differences in cancer and immunotherapy outcomes: the role of androgen receptor. Front Immunol 2024; 15:1416941. [PMID: 38863718 PMCID: PMC11165033 DOI: 10.3389/fimmu.2024.1416941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Accepted: 05/16/2024] [Indexed: 06/13/2024] Open
Abstract
Across the wide range of clinical conditions, there exists a sex imbalance where biological females are more prone to autoimmune diseases and males to some cancers. These discrepancies are the combinatory consequence of lifestyle and environmental factors such as smoking, alcohol consumption, obesity, and oncogenic viruses, as well as other intrinsic biological traits including sex chromosomes and sex hormones. While the emergence of immuno-oncology (I/O) has revolutionised cancer care, the efficacy across multiple cancers may be limited because of a complex, dynamic interplay between the tumour and its microenvironment (TME). Indeed, sex and gender can also influence the varying effectiveness of I/O. Androgen receptor (AR) plays an important role in tumorigenesis and in shaping the TME. Here, we lay out the epidemiological context of sex disparity in cancer and then review the current literature on how AR signalling contributes to such observation via altered tumour development and immunology. We offer insights into AR-mediated immunosuppressive mechanisms, with the hope of translating preclinical and clinical evidence in gender oncology into improved outcomes in personalised, I/O-based cancer care.
Collapse
Affiliation(s)
- Junzhe Zhao
- Duke-NUS Medical School, Singapore, Singapore
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - Qian Wang
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore
- Department of Medical Oncology Cancer Hospital of China Medical University/Liaoning Cancer Hospital & Institute, Shenyang, Liaoning, China
| | | | - Celestine Jia Ling Loh
- Duke-NUS Medical School, Singapore, Singapore
- Sengkang General Hospital, Singapore, Singapore
| | - Han Chong Toh
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore
| |
Collapse
|
23
|
Liu JL, Chen LJ, Liu Y, Li JH, Zhang KK, Hsu C, Li XW, Yang JZ, Chen L, Zeng JH, Xie XL, Wang Q. The gut microbiota contributes to methamphetamine-induced reproductive toxicity in male mice. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 279:116457. [PMID: 38754198 DOI: 10.1016/j.ecoenv.2024.116457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 04/25/2024] [Accepted: 05/09/2024] [Indexed: 05/18/2024]
Abstract
Methamphetamine (METH) is a psychostimulant drug belonging to the amphetamine-type stimulant class, known to exert male reproductive toxicity. Recent studies suggest that METH can disrupt the gut microbiota. Furthermore, the gut-testis axis concept has gained attention due to the potential link between gut microbiome dysfunction and reproductive health. Nonetheless, the role of the gut microbiota in mediating the impact of METH on male reproductive toxicity remains unclear. In this study, we employed a mouse model exposed to escalating doses of METH to assess sperm quality, testicular pathology, and reproductive hormone levels. The fecal microbiota transplantation method was employed to investigate the effect of gut microbiota on male reproductive toxicity. Transcriptomic, metabolomic, and microbiological analyses were conducted to explore the damage mechanism to the male reproductive system caused by METH. We found that METH exposure led to hormonal disorders, decreased sperm quality, and changes in the gut microbiota and testicular metabolome in mice. Testicular RNA sequencing revealed enrichment of several Gene Ontology terms associated with reproductive processes, as well as PI3K-Akt signaling pathways. FMT conveyed similar reproductive damage from METH-treated mice to healthy recipient mice. The aforementioned findings suggest that the gut microbiota plays a substantial role in facilitating the reproductive toxicity caused by METH, thereby highlighting a prospective avenue for therapeutic intervention in the context of METH-induced infertility.
Collapse
Affiliation(s)
- Jia-Li Liu
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, China
| | - Li-Jian Chen
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, China
| | - Yi Liu
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, China
| | - Jia-Hao Li
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, China
| | - Kai-Kai Zhang
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, China
| | - Clare Hsu
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, China
| | - Xiu-Wen Li
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, China
| | - Jian-Zheng Yang
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, China
| | - Long Chen
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, China
| | - Jia-Hao Zeng
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, China
| | - Xiao-Li Xie
- Department of Toxicology, School of Public Health, Southern Medical University (Guangdong Provincial Key Laboratory of Tropical Disease Research), Guangzhou 510515, China.
| | - Qi Wang
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
24
|
Kaleta M, Oklestkova J, Klíčová K, Kvasnica M, Koníčková D, Menšíková K, Strnad M, Novák O. Simultaneous Determination of Selected Steroids with Neuroactive Effects in Human Serum by Ultrahigh-Performance Liquid Chromatography-Tandem Mass Spectrometry. ACS Chem Neurosci 2024; 15:1990-2005. [PMID: 38655788 PMCID: PMC11099924 DOI: 10.1021/acschemneuro.3c00824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 03/27/2024] [Accepted: 04/15/2024] [Indexed: 04/26/2024] Open
Abstract
Neuroactive steroids are a group of steroid molecules that are involved in the regulation of functions of the nervous system. The nervous system is not only the site of their action, but their biosynthesis can also occur there. Neuroactive steroid levels depend not only on the physiological state of an individual (person's sex, age, diurnal variation, etc.), but they are also affected by various pathological processes in the nervous system (some neurological and psychiatric diseases or injuries), and new knowledge can be gained by monitoring these processes. The aim of our research was to develop and validate a comprehensive method for the simultaneous determination of selected steroids with neuroactive effects in human serum. The developed method enables high throughput and a sensitive quantitative analysis of nine neuroactive steroid substances (pregnenolone, progesterone, 5α-dihydroprogesterone, allopregnanolone, testosterone, 5α-dihydrotestosterone, androstenedione, dehydroepiandrosterone, and epiandrosterone) in 150 μL of human serum by ultrahigh-performance liquid chromatography with tandem mass spectrometry. The correlation coefficients above 0.999 indicated that the developed analytical procedure was linear in the range of 0.90 nmol/L to 28.46 μmol/L in human serum. The accuracy and precision of the method for all analytes ranged from 83 to 118% and from 0.9 to 14.1%, respectively. This described method could contribute to a deeper understanding of the pathophysiology of various diseases. Similarly, it can also be helpful in the search for new biomarkers and diagnostic options or therapeutic approaches.
Collapse
Affiliation(s)
- Michal Kaleta
- Laboratory
of Growth Regulators, Faculty of Science, Palacký University & Institute of Experimental Botany
of the Czech Academy of Sciences, Šlechtitelů 27, Olomouc 783 71, Czech Republic
- Department
of Neurology, Faculty of Medicine and Dentistry, Palacký University, Olomouc 779 00, Czech Republic
| | - Jana Oklestkova
- Laboratory
of Growth Regulators, Faculty of Science, Palacký University & Institute of Experimental Botany
of the Czech Academy of Sciences, Šlechtitelů 27, Olomouc 783 71, Czech Republic
| | - Kateřina Klíčová
- Department
of Neurology, Faculty of Medicine and Dentistry, Palacký University, Olomouc 779 00, Czech Republic
- Department
of Neurology, University Hospital Olomouc, Olomouc 779 00, Czech Republic
| | - Miroslav Kvasnica
- Laboratory
of Growth Regulators, Faculty of Science, Palacký University & Institute of Experimental Botany
of the Czech Academy of Sciences, Šlechtitelů 27, Olomouc 783 71, Czech Republic
| | - Dorota Koníčková
- Department
of Neurology, Faculty of Medicine and Dentistry, Palacký University, Olomouc 779 00, Czech Republic
- Department
of Neurology, University Hospital Olomouc, Olomouc 779 00, Czech Republic
| | - Kateřina Menšíková
- Department
of Neurology, Faculty of Medicine and Dentistry, Palacký University, Olomouc 779 00, Czech Republic
- Department
of Neurology, University Hospital Olomouc, Olomouc 779 00, Czech Republic
| | - Miroslav Strnad
- Laboratory
of Growth Regulators, Faculty of Science, Palacký University & Institute of Experimental Botany
of the Czech Academy of Sciences, Šlechtitelů 27, Olomouc 783 71, Czech Republic
| | - Ondřej Novák
- Laboratory
of Growth Regulators, Faculty of Science, Palacký University & Institute of Experimental Botany
of the Czech Academy of Sciences, Šlechtitelů 27, Olomouc 783 71, Czech Republic
| |
Collapse
|
25
|
Nazzari M, Romitti M, Kip AM, Kamps R, Costagliola S, van de Beucken T, Moroni L, Caiment F. Impact of benzo[a]pyrene, PCB153 and sex hormones on human ESC-Derived thyroid follicles using single cell transcriptomics. ENVIRONMENT INTERNATIONAL 2024; 188:108748. [PMID: 38763096 DOI: 10.1016/j.envint.2024.108748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 04/25/2024] [Accepted: 05/10/2024] [Indexed: 05/21/2024]
Abstract
INTRODUCTION Endocrine disruptors are compounds of manmade origin able to interfere with the endocrine system and constitute an important environmental concern. Indeed, detrimental effects on thyroid physiology and functioning have been described. Differences exist in the susceptibility of human sexes to the incidence of thyroid disorders, like autoimmune diseases or cancer. METHODS To study how different hormonal environments impact the thyroid response to endocrine disruptors, we exposed human embryonic stem cell-derived thyroid organoids to physiological concentrations of sex hormones resembling the serum levels of human females post-ovulation or males of reproductive age for three days. Afterwards, we added 10 µM benzo[a]pyrene or PCB153 for 24 h and analyzed the transcriptome changes via single-cell RNA sequencing with differential gene expression and gene ontology analysis. RESULTS The sex hormones receptors genes AR, ESR1, ESR2 and PGR were expressed at low levels. Among the thyroid markers, only TG resulted downregulated by benzo[a]pyrene or benzo[a]pyrene with the "male" hormones mix. Both hormone mixtures and benzo[a]pyrene alone upregulated ribosomal genes and genes involved in oxidative phosphorylation, while their combination decreased the expression compared to benzo[a]pyrene alone. The "male" mix and benzo[a]pyrene, alone or in combination, upregulated genes involved in lipid transport and metabolism (APOA1, APOC3, APOA4, FABP1, FABP2, FABP6). The combination of "male" hormones and benzo[a]pyrene induced also genes involved in inflammation and NFkB targets. Benzo[a]pyrene upregulated CYP1A1, CYP1B1 and NQO1 irrespective of the hormonal context. The induction was stronger in the "female" mix. Benzo[a]pyrene alone upregulated genes involved in cell cycle regulation, response to reactive oxygen species and apoptosis. PCB153 had a modest effect in presence of "male" hormones, while we did not observe any changes with the "female" mix. CONCLUSION This work shows how single cell transcriptomics can be applied to selectively study the in vitro effects of endocrine disrupters and their interaction with different hormonal contexts.
Collapse
Affiliation(s)
- Marta Nazzari
- Department of Toxicogenomics, GROW School for Oncology and Developmental Biology, Maastricht University, 6229 ER Maastricht, the Netherlands
| | - Mírian Romitti
- Institute of Interdisciplinary Research in Molecular Human Biology (IRIBHM), Université Libre de Bruxelles, 808 Route de Lennik, 1070 Brussels, Belgium
| | - Anna M Kip
- Department of Complex Tissue Regeneration, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University 6229 ER Maastricht, the Netherlands
| | - Rick Kamps
- Department of Toxicogenomics, Maastricht University, 6229 ER Maastricht, the Netherlands
| | - Sabine Costagliola
- Institute of Interdisciplinary Research in Molecular Human Biology (IRIBHM), Université Libre de Bruxelles, 808 Route de Lennik, 1070 Brussels, Belgium
| | - Twan van de Beucken
- Department of Toxicogenomics, Maastricht University, 6229 ER Maastricht, the Netherlands
| | - Lorenzo Moroni
- Department of Complex Tissue Regeneration, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University 6229 ER Maastricht, the Netherlands
| | - Florian Caiment
- Department of Toxicogenomics, GROW School for Oncology and Developmental Biology, Maastricht University, 6229 ER Maastricht, the Netherlands.
| |
Collapse
|
26
|
Dignam JP, Sharma S, Stasinopoulos I, MacLean MR. Pulmonary arterial hypertension: Sex matters. Br J Pharmacol 2024; 181:938-966. [PMID: 37939796 DOI: 10.1111/bph.16277] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 10/10/2023] [Accepted: 10/11/2023] [Indexed: 11/10/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a complex disease of multifactorial origin. While registries have demonstrated that women are more susceptible to the disease, females with PAH have superior right ventricle (RV) function and a better prognosis than their male counterparts, a phenomenon referred to as the 'estrogen paradox'. Numerous pre-clinical studies have investigated the involvement of sex hormones in PAH pathobiology, often with conflicting results. However, recent advances suggest that abnormal estrogen synthesis, metabolism and signalling underpin the sexual dimorphism of this disease. Other sex hormones, such as progesterone, testosterone and dehydroepiandrosterone may also play a role. Several non-hormonal factor including sex chromosomes and epigenetics have also been implicated. Though the underlying pathophysiological mechanisms are complex, several compounds that modulate sex hormones levels and signalling are under investigation in PAH patients. Further elucidation of the estrogen paradox will set the stage for the identification of additional therapeutic targets for this disease.
Collapse
Affiliation(s)
- Joshua P Dignam
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, Scotland, UK
| | - Smriti Sharma
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, Scotland, UK
| | - Ioannis Stasinopoulos
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, Scotland, UK
- Mass Spectrometry Core, Edinburgh Clinical Research Facility, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, Scotland, UK
| | - Margaret R MacLean
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, Scotland, UK
| |
Collapse
|
27
|
Chen J, Ma W, Yue S, Li D, Chen L, Zhang C, Guan Y, Li C, Jiang C, Liao G, Liang C, Wang H, Tai S. Dual deficiency of melatonin and dihydrotestosterone promotes stromal cell damage and mediates prostatitis via the cGAS-STING pathway in sleep-deprived mice. Cell Commun Signal 2024; 22:183. [PMID: 38491517 PMCID: PMC10941623 DOI: 10.1186/s12964-024-01554-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Accepted: 03/02/2024] [Indexed: 03/18/2024] Open
Abstract
PURPOSE Prostatitis is a highly prevalent condition that seriously affects men's physical and mental health. Although epidemiological investigations have provided evidence of a correlation between insufficient sleep and prostatitis, the pathogenesis of prostatitis remains unclear. We sought to identify the underlying mechanism involved and identify a promising therapeutic target. METHODS Sleep deprivation (SD) was utilized to establish a mouse model of insufficient sleep in a special device. Prostatitis was observed at different time points post-SD. The degree of prostatitis was evaluated by pathological section and behavioural tests. Using immunofluorescence, western blot, and proteomic analyses, the underlying mechanism of SD-related prostatitis was investigated, and the development and therapeutic target of prostatitis were elucidated. RESULTS SD, as an initial pathological trigger, resulted in a reduction in dihydrotestosterone and melatonin levels. Proteomic analysis revealed that the cGAS-STING pathway may play a significant role in inducing prostatitis. The subsequent results illustrated that the dual reduction in dihydrotestosterone and melatonin led to an accumulation of reactive oxygen species and the release of mitochondrial DNA (mt-DNA). The accumulation of mt-DNA activated the cGAS-STING pathway, which recruited inflammatory cells into the prostatic stroma through the secretion of interferon-β. Consequently, an inflammatory microenvironment was formed, ultimately promoting the development of prostatitis. Notably, mice with SD-induced prostatitis gradually recovered to a normal state within 7 days of recovery sleep. However, after being subjected to SD again, these mice tended to have a more pronounced manifestation of prostatitis within a shorter timeframe, which suggested that prostatitis is prone to relapse. CONCLUSIONS The cGAS-STING pathway activated by dual deficiency of dihydrotestosterone and melatonin plays a comprehensive inflammatory role in SD-related prostatitis. This research provides valuable insights into the pathogenesis, therapeutic targets, and prevention strategies of prostatitis.
Collapse
Affiliation(s)
- Jia Chen
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Institute of Urology, Anhui Medical University, Anhui Province Key Laboratory of Genitourinary Diseases, Hefei, 230022, P.R. China
| | - Wenming Ma
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Institute of Urology, Anhui Medical University, Anhui Province Key Laboratory of Genitourinary Diseases, Hefei, 230022, P.R. China
| | - Shaoyu Yue
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Institute of Urology, Anhui Medical University, Anhui Province Key Laboratory of Genitourinary Diseases, Hefei, 230022, P.R. China
| | - Dongsheng Li
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Institute of Urology, Anhui Medical University, Anhui Province Key Laboratory of Genitourinary Diseases, Hefei, 230022, P.R. China
| | - Lei Chen
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Institute of Urology, Anhui Medical University, Anhui Province Key Laboratory of Genitourinary Diseases, Hefei, 230022, P.R. China
| | - Cheng Zhang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Institute of Urology, Anhui Medical University, Anhui Province Key Laboratory of Genitourinary Diseases, Hefei, 230022, P.R. China
| | - Yu Guan
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Institute of Urology, Anhui Medical University, Anhui Province Key Laboratory of Genitourinary Diseases, Hefei, 230022, P.R. China
| | - Chun Li
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Institute of Urology, Anhui Medical University, Anhui Province Key Laboratory of Genitourinary Diseases, Hefei, 230022, P.R. China
| | - Changqin Jiang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Institute of Urology, Anhui Medical University, Anhui Province Key Laboratory of Genitourinary Diseases, Hefei, 230022, P.R. China
| | - Guiyi Liao
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Institute of Urology, Anhui Medical University, Anhui Province Key Laboratory of Genitourinary Diseases, Hefei, 230022, P.R. China
| | - Chaozhao Liang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Institute of Urology, Anhui Medical University, Anhui Province Key Laboratory of Genitourinary Diseases, Hefei, 230022, P.R. China.
| | - Hui Wang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Institute of Urology, Anhui Medical University, Anhui Province Key Laboratory of Genitourinary Diseases, Hefei, 230022, P.R. China.
| | - Sheng Tai
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Institute of Urology, Anhui Medical University, Anhui Province Key Laboratory of Genitourinary Diseases, Hefei, 230022, P.R. China.
| |
Collapse
|
28
|
Zhang YY, Xie N, Sun XD, Nice EC, Liou YC, Huang C, Zhu H, Shen Z. Insights and implications of sexual dimorphism in osteoporosis. Bone Res 2024; 12:8. [PMID: 38368422 PMCID: PMC10874461 DOI: 10.1038/s41413-023-00306-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 11/04/2023] [Accepted: 11/27/2023] [Indexed: 02/19/2024] Open
Abstract
Osteoporosis, a metabolic bone disease characterized by low bone mineral density and deterioration of bone microarchitecture, has led to a high risk of fatal osteoporotic fractures worldwide. Accumulating evidence has revealed that sexual dimorphism is a notable feature of osteoporosis, with sex-specific differences in epidemiology and pathogenesis. Specifically, females are more susceptible than males to osteoporosis, while males are more prone to disability or death from the disease. To date, sex chromosome abnormalities and steroid hormones have been proven to contribute greatly to sexual dimorphism in osteoporosis by regulating the functions of bone cells. Understanding the sex-specific differences in osteoporosis and its related complications is essential for improving treatment strategies tailored to women and men. This literature review focuses on the mechanisms underlying sexual dimorphism in osteoporosis, mainly in a population of aging patients, chronic glucocorticoid administration, and diabetes. Moreover, we highlight the implications of sexual dimorphism for developing therapeutics and preventive strategies and screening approaches tailored to women and men. Additionally, the challenges in translating bench research to bedside treatments and future directions to overcome these obstacles will be discussed.
Collapse
Affiliation(s)
- Yuan-Yuan Zhang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Na Xie
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Xiao-Dong Sun
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Edouard C Nice
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, 3800, Australia
| | - Yih-Cherng Liou
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore, 117543, Republic of Singapore
| | - Canhua Huang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Huili Zhu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, Department of Reproductive Medicine, West China Second University Hospital of Sichuan University, Chengdu, China.
| | - Zhisen Shen
- Department of Otorhinolaryngology and Head and Neck Surgery, The Affiliated Lihuili Hospital, Ningbo University, 315040, Ningbo, Zhejiang, China.
| |
Collapse
|
29
|
Liu H, Tu M, Yin Z, Zhang D, Ma J, He F. Unraveling the complexity of polycystic ovary syndrome with animal models. J Genet Genomics 2024; 51:144-158. [PMID: 37777062 DOI: 10.1016/j.jgg.2023.09.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 09/17/2023] [Accepted: 09/20/2023] [Indexed: 10/02/2023]
Abstract
Polycystic ovary syndrome (PCOS) is a highly familial and heritable endocrine disorder. Over half of the daughters born to women with PCOS may eventually develop their own PCOS-related symptoms. Progress in the treatment of PCOS is currently hindered by the complexity of its clinical manifestations and incomplete knowledge of its etiopathogenesis. Various animal models, including experimentally induced, naturally occurring, and spontaneously arising ones, have been established to emulate a wide range of phenotypical and pathological traits of human PCOS. These studies have led to a paradigm shift in understanding the genetic, developmental, and evolutionary origins of this disorder. Furthermore, emerging evidence suggests that animal models are useful in evaluating state-of-the-art drugs and treatments for PCOS. This review aims to provide a comprehensive summary of recent studies of PCOS in animal models, highlighting the power of these disease models in understanding the biology of PCOS and aiding high-throughput approaches.
Collapse
Affiliation(s)
- Huanju Liu
- Center for Genetic Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Mixue Tu
- Key Laboratory of Women's Reproductive Health of Zhejiang Province and Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China
| | - Zhiyong Yin
- Center for Genetic Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Dan Zhang
- Key Laboratory of Women's Reproductive Health of Zhejiang Province and Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China; Zhejiang Provincial Clinical Research Center for Child Health, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China; Clinical Research Center on Birth Defect Prevention and Intervention of Zhejiang Province, Hangzhou, Zhejiang 310006, China.
| | - Jun Ma
- Center for Genetic Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China; Key Laboratory of Women's Reproductive Health of Zhejiang Province and Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China; Institute of Genetics, Zhejiang University International School of Medicine, Hangzhou, Zhejiang 310058, China; Zhejiang Provincial Key Laboratory of Genetic and Developmental Disorder, Hangzhou, Zhejiang 310058, China.
| | - Feng He
- Center for Genetic Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China; Key Laboratory of Women's Reproductive Health of Zhejiang Province and Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China; Institute of Genetics, Zhejiang University International School of Medicine, Hangzhou, Zhejiang 310058, China; Zhejiang Provincial Key Laboratory of Genetic and Developmental Disorder, Hangzhou, Zhejiang 310058, China.
| |
Collapse
|
30
|
Daniels JP, Mirocha J, Adjei M, Moreira D, Freedland SJ. Serum Testosterone and Dihydrotestosterone and Incidence and Progression of Lower Urinary Tract Symptoms: Results From the REDUCE Study. J Urol 2024; 211:101-110. [PMID: 37873943 PMCID: PMC10842939 DOI: 10.1097/ju.0000000000003738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 09/25/2023] [Indexed: 10/25/2023]
Abstract
PURPOSE Though the pathogenesis of benign prostatic hyperplasia is unclear, it was previously believed that increasing androgen levels contributed, though not all data support this idea. We tested if elevated serum testosterone or dihydrotestosterone were risk factors for lower urinary tract symptoms incidence in asymptomatic men and for lower urinary tract symptoms progression in symptomatic men. MATERIALS AND METHODS A post hoc analysis of REDUCE was performed in 3009 asymptomatic men and in 2145 symptomatic men. REDUCE was a randomized trial of dutasteride for prostate cancer prevention in men with an elevated prostate-specific antigen and negative prestudy biopsy. We estimated multivariable adjusted hazard ratios and 95% confidence intervals using Cox models to test the association between quintiles of serum testosterone and dihydrotestosterone at baseline and lower urinary tract symptoms incidence and progression and tested for interaction by treatment arm (dutasteride vs placebo). RESULTS In asymptomatic men, there was no evidence serum testosterone or dihydrotestosterone were related to lower urinary tract symptoms incidence (P = .9, P = .4). In symptomatic men, there was no evidence serum testosterone or dihydrotestosterone were related to lower urinary tract symptoms progression (P = .9, P = .7). Results were similar in both placebo and dutasteride arms (all P interaction ≥ .3). CONCLUSIONS In REDUCE, higher serum testosterone and higher serum dihydrotestosterone were not associated with either lower urinary tract symptoms incidence in asymptomatic men or lower urinary tract symptoms progression in symptomatic men. These data do not support the hypothesis that serum androgens in middle-aged men are associated with lower urinary tract symptoms.
Collapse
Affiliation(s)
| | - James Mirocha
- Cedars-Sinai Medical Center, Los Angeles, California
| | - Michie Adjei
- Cedars-Sinai Medical Center, Los Angeles, California
| | - Daniel Moreira
- The University of Illinois College of Medicine, Chicago, Illinois
| | - Stephen J Freedland
- Cedars-Sinai Medical Center, Los Angeles, California
- Durham VA Medical Center, Durham, North Carolina
| |
Collapse
|
31
|
Lee MS, Bunin DI, Furimsky AM, Nguyen D, Parman T, Kim K, Rausch L, Lin MT, Gupta P, Brown JE, Kroopnick JM, Blithe DL. Novel progestogenic androgens for male contraception: design, synthesis, and activity of C7 α-substituted testosterone†. Biol Reprod 2023; 109:851-863. [PMID: 37669128 PMCID: PMC10724455 DOI: 10.1093/biolre/ioad111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 08/08/2023] [Accepted: 08/30/2023] [Indexed: 09/07/2023] Open
Abstract
Male contraceptive development has included use of testosterone (T) with or without a progestin or the use of a single molecule such as progestogenic androgens (PA) for suppression of testicular T production. Expanding upon the vast amount of data accumulated from nortestosterone (NT), NT analogs, and their prodrugs, a new series of PA, the C7 methyl, and ethyl α-substituted T analogs 7α-Methyltestosterone (7α-MT) and 7α-Ethyltestosterone (7α-ET), respectively, were hypothesized and designed to have superior androgenic and progestogenic activities when compared with parent T. Results from androgen receptor and progesterone receptor competitive binding and transcriptional activation assays showed favorable activities for these T analogs. Additionally, 7α-MT and 7α-ET were shown to be active substrates for aromatase in vitro, mitigating a potential negative impact on bone mineral density with long-term use. In conjunction with this observation, the diminished metabolism of these T analogs by 5α-reductase may reduce potential concerns for prostatic growth. In the Hershberger in vivo rat bioassay, 7α-MT and 7α-ET showed superior androgenic and anabolic activities as compared with T. These C7 α-substituted T analogs also showed clear progestogenic activity in the McPhail bioassay which evaluated endometrial glandular arborization in a rabbit model. The discovery of aromatizable molecules with reduced metabolism by 5α-reductase that have androgenic, anabolic, and progestogenic properties indicates that the core and/or prodrugs of 7α-MT and 7α-ET are promising molecules for further development as male contraceptive PAs.
Collapse
Affiliation(s)
- Min S Lee
- Contraceptive Development Program, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institute of Health, Bethesda, MD, USA
| | - Deborah I Bunin
- Biosciences Division, SRI International, Menlo Park, CA, USA
| | - Anna M Furimsky
- Biosciences Division, SRI International, Menlo Park, CA, USA
| | - Donna Nguyen
- Biosciences Division, SRI International, Menlo Park, CA, USA
| | | | - Kyuri Kim
- Jazz Pharmaceuticals, Palo Alto, CA, USA
| | - Linda Rausch
- Biosciences Division, SRI International, Menlo Park, CA, USA
| | | | | | - Jill E Brown
- Department of Gynecologic Surgery & Obstetrics, Uniformed Services University, Bethesda, MD, USA
| | - Jeffrey M Kroopnick
- Contraceptive Development Program, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institute of Health, Bethesda, MD, USA
| | - Diana L Blithe
- Contraceptive Development Program, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institute of Health, Bethesda, MD, USA
| |
Collapse
|
32
|
Darjani A, Alizadeh N, Gharaei Nejad K, Eftekhari H, Rafiei R, Kazemi H, Rafiei E. Testosterone or dihydrotestosterone: what should be evaluated in hirsutism? Ir J Med Sci 2023; 192:2823-2827. [PMID: 37043171 DOI: 10.1007/s11845-023-03366-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 04/03/2023] [Indexed: 04/13/2023]
Abstract
PURPOSE Dihydrotestosterone is a more potent androgen derived from testosterone and androstenediones, but its measurement has not been routinely recommended in women with hirsutism, and there is limited information in this regard with equivocal findings. This study aimed to evaluate serum dihydrotestosterone level in patients with hirsutism compared to women without hirsutism. METHODS In this case-control study (during 2021-2022), serum levels of total testosterone, free testosterone, and dihydrotestosterone were evaluated in 101 women with hirsutism and 101 healthy women. Hormonal levels were measured with chemiluminescent immunoassay method. Age and hormonal levels in each group, body mass index, menstrual status, complaint of decreased scalp hair density, and ovarian ultrasound findings in hirsutism group were collected and analyzed. RESULTS There was significant difference in free testosterone and dihydrotestosterone levels (P < 0.001) and no significant difference in total testosterone level between two groups (P = 0.628). Dihydrotestosterone level was significantly higher in women with hirsutism with menses irregularity, complaint of decreased scalp hair density, and presence of polycystic ovary on ultrasound (P < 0.05). CONCLUSIONS Measuring dihydrotestosterone level is not considered in routine evaluation of hirsutism, but we think that this significant difference shows that elevated level of dihydrotestosterone hormone in women with hirsutism is an important factor.
Collapse
Affiliation(s)
- Abbas Darjani
- Skin Research Center, Department of Dermatology, Razi Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Narges Alizadeh
- Skin Research Center, Department of Dermatology, Razi Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Kaveh Gharaei Nejad
- Skin Research Center, Department of Dermatology, Razi Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Hojat Eftekhari
- Skin Research Center, Department of Dermatology, Razi Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Rana Rafiei
- Skin Research Center, Department of Dermatology, Razi Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran.
| | - Hasan Kazemi
- Skin Research Center, Department of Dermatology, Razi Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Elahe Rafiei
- Razi Clinical Research Development Unit, Guilan University of Medical Sciences, Rasht, Iran
| |
Collapse
|
33
|
Chen X, Wang Z, Su J, Li H, Xiong J, Fu K, Wang Z, Yuan X, Shi Z, Miao X, Yang M, Yang Y, Shi Z. Altitude-dependent metabolite biomarkers reveal the mechanism of plateau pika adaptation to high altitudes. Integr Zool 2023; 18:1041-1055. [PMID: 36880690 DOI: 10.1111/1749-4877.12710] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2023]
Abstract
The harsh environment in the Tibetan plateau, the highest place in the world, poses thermoregulatory challenges and hypoxic stress to animals. The impacts of plateau environment on animal physiology and reproduction include external factors such as strong ultraviolet radiation and low temperature, and internal factors such as animal metabolites and gut microbiota. However, it remains unclear how plateau pika adapt to high altitudes through the combination of serum metabolites and gut microbiota. To this end, we captured 24 wild plateau pikas at the altitudes of 3400, 3600, or 3800 m a.s.l. in a Tibetan alpine grassland. Using the machine learning algorithms (random forest), we identified five biomarkers of serum metabolites indicative of the altitudes, that is, dihydrotestosterone, homo-l-arginine, alpha-ketoglutaric-acid, serotonin, and threonine, which were related to body weight, reproduction, and energy metabolism of pika. Those metabolic biomarkers were positively correlated with Lachnospiraceae_ Agathobacter, Ruminococcaceae, or Prevotellaceae_Prevotella, suggesting the close relationship between metabolites and gut microbiota. By identifying the metabolic biomarkers and gut microbiota analysis, we reveal the mechanisms of adaptation to high altitudes in plateau pika.
Collapse
Affiliation(s)
- Xi Chen
- State Key Laboratory of Herbage Improvement and Grassland Agro-ecosystems, Center for Grassland Microbiome, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou, China
| | - Zaiwei Wang
- State Key Laboratory of Herbage Improvement and Grassland Agro-ecosystems, Center for Grassland Microbiome, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou, China
| | - Junhu Su
- College of Grassland Science, Key Laboratory of Grassland Ecosystem (Ministry of Education), Gansu Agricultural University, Lanzhou, China
| | - Huan Li
- State Key Laboratory of Herbage Improvement and Grassland Agro-ecosystems, Center for Grassland Microbiome, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou, China
| | - Jinbo Xiong
- School of Marine Sciences, Ningbo University, Ningbo, China
| | - Keyi Fu
- State Key Laboratory of Herbage Improvement and Grassland Agro-ecosystems, Center for Grassland Microbiome, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou, China
| | - Zilong Wang
- State Key Laboratory of Herbage Improvement and Grassland Agro-ecosystems, Center for Grassland Microbiome, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou, China
| | - Xuefeng Yuan
- State Key Laboratory of Herbage Improvement and Grassland Agro-ecosystems, Center for Grassland Microbiome, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou, China
| | - Ziyue Shi
- State Key Laboratory of Herbage Improvement and Grassland Agro-ecosystems, Center for Grassland Microbiome, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou, China
| | - Xiumei Miao
- State Key Laboratory of Herbage Improvement and Grassland Agro-ecosystems, Center for Grassland Microbiome, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou, China
| | - Mei Yang
- State Key Laboratory of Herbage Improvement and Grassland Agro-ecosystems, Center for Grassland Microbiome, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou, China
| | - Yunfeng Yang
- State Key Laboratory of Herbage Improvement and Grassland Agro-ecosystems, Center for Grassland Microbiome, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou, China
- State Key Joint Laboratory of Environment Simulation and Pollution Control, School of Environment, Tsinghua University, China
| | - Zunji Shi
- State Key Laboratory of Herbage Improvement and Grassland Agro-ecosystems, Center for Grassland Microbiome, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou, China
| |
Collapse
|
34
|
Mitrović-Ajtić O, Đikić D, Subotički T, Bižić-Radulović S, Beleslin-Čokić B, Dragojević T, Živković E, Miljatović S, Vukotić M, Stanisavljević D, Santibanez J, Čokić VP. Sex Differences and Cytokine Profiles among Patients Hospitalized for COVID-19 and during Their Recovery: The Predominance of Adhesion Molecules in Females and Oxidative Stress in Males. Vaccines (Basel) 2023; 11:1560. [PMID: 37896963 PMCID: PMC10610714 DOI: 10.3390/vaccines11101560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 09/22/2023] [Accepted: 09/29/2023] [Indexed: 10/29/2023] Open
Abstract
The severity and mortality of coronavirus disease 2019 (COVID-19) are greater in males than in females, though the infection rate is the same in the two sexes. We investigated sex hormone differences associated with the hyperinflammatory immune response to SARS-CoV-2 on the basis of patients' cytokine profiles and vaccination statuses. Clinical and laboratory data of 117 patients with COVID-19 were collected to examine sex differences associated with oxidative stress markers, neutrophil extracellular traps (NETs), and plasma cytokine levels up to 5 months from hospital admission. The testosterone and free testosterone levels were low in male patients with COVID-19 and returned to normal values after recovery from the disease. The dihydrotestosterone (DHT) levels were transiently reduced, while the sex hormone-binding globulin levels were decreased in post-COVID-19 male patients. The levels of the inflammatory cytokines interleukin-6 (IL-6) and IL-10 appeared generally increased at diagnosis and decreased in post-COVID-19 patients. In females, the concentration of tumor necrosis factor-alpha was increased by four times at diagnosis. The levels of the coagulation markers intercellular adhesion molecule-1 (ICAM-1) and E-selectin were consistently upregulated in post-COVID-19 female patients, in contrast to those of vascular cell adhesion molecule-1 (VCAM-1), P-selectin, and chemokine IL-8. DHT increased the levels of reactive oxygen species in the neutrophils of male patients, while estradiol decreased them in females. Markers for NET, such as circulating DNA and myeloperoxidase, were significantly more abundant in the patients' plasma. Sex hormones have a potential protective role during SARS-CoV-2 infection, which is weakened by impaired testosterone synthesis in men.
Collapse
Affiliation(s)
- Olivera Mitrović-Ajtić
- Institute for Medical Research, University of Belgrade, National Institute of Republic of Serbia, Dr. Subotica starijeg 4, 11129 Belgrade, Serbia; (O.M.-A.); (D.Đ.); (T.S.); (T.D.); (E.Ž.); (M.V.); (J.S.)
| | - Dragoslava Đikić
- Institute for Medical Research, University of Belgrade, National Institute of Republic of Serbia, Dr. Subotica starijeg 4, 11129 Belgrade, Serbia; (O.M.-A.); (D.Đ.); (T.S.); (T.D.); (E.Ž.); (M.V.); (J.S.)
| | - Tijana Subotički
- Institute for Medical Research, University of Belgrade, National Institute of Republic of Serbia, Dr. Subotica starijeg 4, 11129 Belgrade, Serbia; (O.M.-A.); (D.Đ.); (T.S.); (T.D.); (E.Ž.); (M.V.); (J.S.)
| | - Sandra Bižić-Radulović
- Clinic of Hematology, University Clinical Center of Serbia, Dr. Koste Todorovica 2, 11000 Belgrade, Serbia;
| | - Bojana Beleslin-Čokić
- Clinic of Endocrinology, Diabetes and Metabolic Diseases, University Clinical Center of Serbia, Dr. Subotica starijeg 13, 11000 Belgrade, Serbia;
| | - Teodora Dragojević
- Institute for Medical Research, University of Belgrade, National Institute of Republic of Serbia, Dr. Subotica starijeg 4, 11129 Belgrade, Serbia; (O.M.-A.); (D.Đ.); (T.S.); (T.D.); (E.Ž.); (M.V.); (J.S.)
| | - Emilija Živković
- Institute for Medical Research, University of Belgrade, National Institute of Republic of Serbia, Dr. Subotica starijeg 4, 11129 Belgrade, Serbia; (O.M.-A.); (D.Đ.); (T.S.); (T.D.); (E.Ž.); (M.V.); (J.S.)
| | - Sanja Miljatović
- Clinic for Infectious and Tropical Diseases, University Clinical Center of Serbia, Bulevar oslobođenja 16, 11000 Belgrade, Serbia
| | - Milica Vukotić
- Institute for Medical Research, University of Belgrade, National Institute of Republic of Serbia, Dr. Subotica starijeg 4, 11129 Belgrade, Serbia; (O.M.-A.); (D.Đ.); (T.S.); (T.D.); (E.Ž.); (M.V.); (J.S.)
| | - Dejana Stanisavljević
- Institute for Medical Statistics and Informatics, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
| | - Juan Santibanez
- Institute for Medical Research, University of Belgrade, National Institute of Republic of Serbia, Dr. Subotica starijeg 4, 11129 Belgrade, Serbia; (O.M.-A.); (D.Đ.); (T.S.); (T.D.); (E.Ž.); (M.V.); (J.S.)
- Centro Integrativo de Biología y Química Aplicada (CIBQA), Universidad Bernardo O’Higgins, Santiago 8370854, Chile
| | - Vladan P. Čokić
- Institute for Medical Research, University of Belgrade, National Institute of Republic of Serbia, Dr. Subotica starijeg 4, 11129 Belgrade, Serbia; (O.M.-A.); (D.Đ.); (T.S.); (T.D.); (E.Ž.); (M.V.); (J.S.)
| |
Collapse
|
35
|
Ayele HT, Reynier P, Azoulay L, Platt RW, Benayoun S, Filion KB. The Cardiovascular Safety of Five-Alpha-Reductase Inhibitors Among Men with Benign Prostatic Hyperplasia: A Population-Based Cohort Study. Am J Med 2023; 136:1000-1010.e7. [PMID: 37481022 DOI: 10.1016/j.amjmed.2023.06.021] [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: 11/09/2022] [Revised: 06/16/2023] [Accepted: 06/19/2023] [Indexed: 07/24/2023]
Abstract
BACKGROUND Five-alpha reductase inhibitors (5αRIs) are used to treat benign prostatic hyperplasia (BPH). However, the cardiovascular effects of 5αRIs remain poorly understood. The study objective was to compare the rate of hospitalization for heart failure among men with BPH prescribed 5αRIs to that of men with BPH not prescribed BPH medications. METHODS Using the Clinical Practice Research Datalink linked with hospitalization and vital statistics data, we conducted a population-based cohort study among patients newly diagnosed with BPH. We defined exposure as the current use of 5αRIs, current use of alpha-blockers, and no current use of BPH medications in a time-varying approach. The primary endpoint was hospitalization for heart failure, and secondary endpoints were myocardial infarction, stroke, and cardiovascular death. We used time-dependent Cox-proportional hazards models to estimate adjusted hazard ratios (HRs) and 95% confidence intervals (CIs). RESULTS Our cohort included 94,440 men with incident BPH. A total of 3893 hospitalizations for heart failure occurred over 527,660 person-years of follow-up (incidence rate 7.38; 95% CI, 7.15-7.61, per 1000 person-years). Compared with no current use of BPH medications, current use of 5αRIs was not associated with an increased risk of hospitalization for heart failure (HR 0.94; 95% CI, 0.86-1.03), myocardial infarction (HR 0.92; 95% CI, 0.81-1.05), stroke (HR 0.94; 95% CI, 0.85-1.05), or cardiovascular death (HR 0.89; 95% CI, 0.80-0.99). CONCLUSIONS The use of 5αRIs was not associated with an increased risk of hospitalization for heart failure, myocardial infarction, stroke, or cardiovascular death compared with non-use.
Collapse
Affiliation(s)
- Henok Tadesse Ayele
- Department of Epidemiology, Biostatistics, Occupational Health, McGill University, Montreal, Quebec, Canada; Center for Clinical Epidemiology, Lady Davis Institute, Jewish General Hospital, Montreal, Quebec, Canada
| | - Pauline Reynier
- Center for Clinical Epidemiology, Lady Davis Institute, Jewish General Hospital, Montreal, Quebec, Canada
| | - Laurent Azoulay
- Department of Epidemiology, Biostatistics, Occupational Health, McGill University, Montreal, Quebec, Canada; Center for Clinical Epidemiology, Lady Davis Institute, Jewish General Hospital, Montreal, Quebec, Canada; Gerald Bronfman Department of Oncology, McGill University, Montreal, Quebec, Canada
| | - Robert W Platt
- Department of Epidemiology, Biostatistics, Occupational Health, McGill University, Montreal, Quebec, Canada; Center for Clinical Epidemiology, Lady Davis Institute, Jewish General Hospital, Montreal, Quebec, Canada; Department of Pediatrics, McGill University, Montreal, Quebec, Canada
| | - Serge Benayoun
- Department of Surgery, Urology Unit, Hôpital Maisonneuve-Rosemont, Montreal, Canada
| | - Kristian B Filion
- Department of Epidemiology, Biostatistics, Occupational Health, McGill University, Montreal, Quebec, Canada; Center for Clinical Epidemiology, Lady Davis Institute, Jewish General Hospital, Montreal, Quebec, Canada; Department of Medicine, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
36
|
Equey T, Salamin O, Ponzetto F, Nicoli R, Kuuranne T, Saugy J, Saugy M, Aikin R, Baume N. Longitudinal Profiling of Endogenous Steroids in Blood Using the Athlete Biological Passport Approach. J Clin Endocrinol Metab 2023; 108:1937-1946. [PMID: 36794909 DOI: 10.1210/clinem/dgad085] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 02/02/2023] [Accepted: 02/09/2023] [Indexed: 02/17/2023]
Abstract
CONTEXT Detection of endogenous anabolic androgenic steroids (EAAS), like testosterone (T), as doping agents has been improved with the launch of the Steroidal Module of the Athlete Biological Passport (ABP) in urine samples. OBJECTIVE To target doping practices with EAAS, particularly in individuals with low level of biomarkers excreted in urine, by including new target compounds measured in blood. DESIGN T and T/androstenedione (T/A4) distributions were obtained from 4 years of anti-doping data and applied as priors to analyze individual profiles from 2 T administration studies in female and male subjects. SETTING Anti-doping laboratory. Elite athletes (n = 823) and male and female clinical trials subjects (n = 19 and 14, respectively). INTERVENTION(S) Two open-label administration studies were carried out. One involved a control phase period followed by patch and then oral T administration in male volunteers and the other followed female volunteers during 3 menstrual cycles with 28 days of daily transdermal T application during the second month. MAIN OUTCOME MEASURE(S) Serum samples were analyzed for T and A4 and the performance of a longitudinal ABP-based approach was evaluated for T and T/A4. RESULTS An ABP-based approach set at a 99% specificity flagged all female subjects during the transdermal T application period and 44% of subjects 3 days after the treatment. T showed the best sensitivity (74%) in response to transdermal T application in males. CONCLUSIONS Inclusion of T and T/A4 as markers in the Steroidal Module can improve the performance of the ABP to identify T transdermal application, particularly in females.
Collapse
Affiliation(s)
- Tristan Equey
- World Anti-Doping Agency (WADA), Montreal, Quebec H4Z 1B7, Canada
| | - Olivier Salamin
- Swiss Laboratory for Doping Analyses, University Center of Legal Medicine, Genève and Lausanne, Centre Hospitalier Universitaire Vaudois and University of Lausanne, 1066 Epalinges, Switzerland
- Research and Expertise in anti-Doping Sciences (REDs), Institute of Sport Sciences, University of Lausanne, 1015 Lausanne, Switzerland
| | - Federico Ponzetto
- Endocrinology, Diabetes and Metabolism, Department of Medical Sciences, City of Health and Science University Hospital, University of Turin, 10126 Turin, Italy
| | - Raul Nicoli
- Swiss Laboratory for Doping Analyses, University Center of Legal Medicine, Genève and Lausanne, Centre Hospitalier Universitaire Vaudois and University of Lausanne, 1066 Epalinges, Switzerland
| | - Tiia Kuuranne
- Swiss Laboratory for Doping Analyses, University Center of Legal Medicine, Genève and Lausanne, Centre Hospitalier Universitaire Vaudois and University of Lausanne, 1066 Epalinges, Switzerland
| | - Jonas Saugy
- Research and Expertise in anti-Doping Sciences (REDs), Institute of Sport Sciences, University of Lausanne, 1015 Lausanne, Switzerland
| | - Martial Saugy
- Research and Expertise in anti-Doping Sciences (REDs), Institute of Sport Sciences, University of Lausanne, 1015 Lausanne, Switzerland
| | - Reid Aikin
- World Anti-Doping Agency (WADA), Montreal, Quebec H4Z 1B7, Canada
| | - Norbert Baume
- World Anti-Doping Agency (WADA), Montreal, Quebec H4Z 1B7, Canada
| |
Collapse
|
37
|
Lee HI, Kim S, Kim SW, Lee M, Song K, Suh J, Lee YS, Chae HW, Kim HS, Han S, Kwon A. Effects of Androgen Treatment on Growth in Patients with 5-α-Reductase Type 2 Deficiency. J Pers Med 2023; 13:992. [PMID: 37373981 DOI: 10.3390/jpm13060992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 05/31/2023] [Accepted: 06/12/2023] [Indexed: 06/29/2023] Open
Abstract
BACKGROUND Patients with 5-α-reductase type 2 deficiency (5αRD2) require androgen treatment for the growth of normal male external genitalia. Since limited research has been conducted on the effects of androgen treatment on height in individuals with 5αRD2, we investigated the effect of androgen treatment on bone age (BA) and the height status in children with 5αRD2. METHODS Of the 19 participants who were followed up for an average of 10.6 years, 12 received androgen treatment. BA and height standard deviation scores (SDS) were compared between the treatment and non-treatment groups, as well as between the dihydrotestosterone (DHT) and testosterone enanthate (TE) treatment groups. RESULTS Despite the above-average height of the 19 patients with 5αRD2, the height SDS relative to BA (htSDS-BA) was below average, particularly in the androgen treatment group. DHT treatment did not lead to an increase in BA or htSDS-BA, whereas TE treatment resulted in BA advancement and decreased htSDS-BA, especially in the prepubertal period. CONCLUSIONS DHT treatment is more favorable for height than TE treatment in patients with 5αRD2, particularly during the prepubertal period. Therefore, age and the type of androgen used should be carefully considered to minimize the risk of height reduction in these patient groups.
Collapse
Affiliation(s)
- Hae In Lee
- Department of Pediatrics, CHA Gangnam Medical Center, CHA University, Seoul 03722, Republic of Korea
| | - Sujin Kim
- Department of Pediatrics, Severance Children's Hospital, Institute of Endocrinology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Sang-Woon Kim
- Department of Urology, Urological Science Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Myeongseob Lee
- Department of Pediatrics, Severance Children's Hospital, Institute of Endocrinology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Kyungchul Song
- Department of Pediatrics, Severance Children's Hospital, Institute of Endocrinology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Junghwan Suh
- Department of Pediatrics, Severance Children's Hospital, Institute of Endocrinology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Yong Seung Lee
- Department of Urology, Urological Science Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Hyun Wook Chae
- Department of Pediatrics, Severance Children's Hospital, Institute of Endocrinology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Ho-Seong Kim
- Department of Pediatrics, Severance Children's Hospital, Institute of Endocrinology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Sangwon Han
- Department of Urology, CHA Gangnam Medical Center, CHA University, Seoul 03722, Republic of Korea
| | - Ahreum Kwon
- Department of Pediatrics, Severance Children's Hospital, Institute of Endocrinology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| |
Collapse
|
38
|
Song S, Gu H, Li J, Yang P, Qi X, Liu J, Zhou J, Li Y, Shu P. Identification of immune-related gene signature for predicting prognosis in uterine corpus endometrial carcinoma. Sci Rep 2023; 13:9255. [PMID: 37286702 DOI: 10.1038/s41598-023-35655-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 05/22/2023] [Indexed: 06/09/2023] Open
Abstract
The objective of this study is to develop a gene signature related to the immune system that can be used to create personalized immunotherapy for Uterine Corpus Endometrial Carcinoma (UCEC). To classify the UCEC samples into different immune clusters, we utilized consensus clustering analysis. Additionally, immune correlation algorithms were employed to investigate the tumor immune microenvironment (TIME) in diverse clusters. To explore the biological function, we conducted GSEA analysis. Next, we developed a Nomogram by integrating a prognostic model with clinical features. Finally, we performed experimental validation in vitro to verify our prognostic risk model. In our study, we classified UCEC patients into three clusters using consensus clustering. We hypothesized that cluster C1 represents the immune inflammation type, cluster C2 represents the immune rejection type, and cluster C3 represents the immune desert type. The hub genes identified in the training cohort were primarily enriched in the MAPK signaling pathway, as well as the PD-L1 expression and PD-1 checkpoint pathway in cancer, all of which are immune-related pathways. Cluster C1 may be a more suitable for immunotherapy. The prognostic risk model showed a strong predictive ability. Our constructed risk model demonstrated a high level of accuracy in predicting the prognosis of UCEC, while also effectively reflecting the state of TIME.
Collapse
Affiliation(s)
- Siyuan Song
- Jiangsu Provincial Hospital of Chinese Medicine, Nanjing, 210029, Jiangsu Province, China
- Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu Province, China
| | - Haoqing Gu
- Jiangsu Provincial Hospital of Chinese Medicine, Nanjing, 210029, Jiangsu Province, China
- Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu Province, China
| | - Jingzhan Li
- Jiangsu Provincial Hospital of Chinese Medicine, Nanjing, 210029, Jiangsu Province, China
- Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu Province, China
| | - Peipei Yang
- Jiangsu Provincial Hospital of Chinese Medicine, Nanjing, 210029, Jiangsu Province, China
- Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu Province, China
| | - Xiafei Qi
- Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu Province, China
| | - Jiatong Liu
- Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu Province, China
| | - Jiayu Zhou
- Jiangsu Provincial Hospital of Chinese Medicine, Nanjing, 210029, Jiangsu Province, China
- Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu Province, China
| | - Ye Li
- Jiangsu Provincial Hospital of Chinese Medicine, Nanjing, 210029, Jiangsu Province, China
- Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu Province, China
| | - Peng Shu
- Jiangsu Provincial Hospital of Chinese Medicine, Nanjing, 210029, Jiangsu Province, China.
- Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu Province, China.
| |
Collapse
|
39
|
Wróbel TM, Jørgensen FS, Pandey AV, Grudzińska A, Sharma K, Yakubu J, Björkling F. Non-steroidal CYP17A1 Inhibitors: Discovery and Assessment. J Med Chem 2023; 66:6542-6566. [PMID: 37191389 DOI: 10.1021/acs.jmedchem.3c00442] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
CYP17A1 is an enzyme that plays a major role in steroidogenesis and is critically involved in the biosynthesis of steroid hormones. Therefore, it remains an attractive target in several serious hormone-dependent cancer diseases, such as prostate cancer and breast cancer. The medicinal chemistry community has been committed to the discovery and development of CYP17A1 inhibitors for many years, particularly for the treatment of castration-resistant prostate cancer. The current Perspective reflects upon the discovery and evaluation of non-steroidal CYP17A1 inhibitors from a medicinal chemistry angle. Emphasis is placed on the structural aspects of the target, key learnings from the presented chemotypes, and design guidelines for future inhibitors.
Collapse
Affiliation(s)
- Tomasz M Wróbel
- Department of Synthesis and Chemical Technology of Pharmaceutical Substances, Faculty of Pharmacy, Medical University of Lublin, Chodźki 4a, 20093 Lublin, Poland
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark
| | - Flemming Steen Jørgensen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark
| | - Amit V Pandey
- Pediatric Endocrinology, Department of Pediatrics, University Children's Hospital, Inselspital, Bern and Translational Hormone Research Program, Department of Biomedical Research, University of Bern, Freiburgstrasse 15, 3010 Bern, Switzerland
| | - Angelika Grudzińska
- Department of Synthesis and Chemical Technology of Pharmaceutical Substances, Faculty of Pharmacy, Medical University of Lublin, Chodźki 4a, 20093 Lublin, Poland
| | - Katyayani Sharma
- Pediatric Endocrinology, Department of Pediatrics, University Children's Hospital, Inselspital, Bern and Translational Hormone Research Program, Department of Biomedical Research, University of Bern, Freiburgstrasse 15, 3010 Bern, Switzerland
| | - Jibira Yakubu
- Pediatric Endocrinology, Department of Pediatrics, University Children's Hospital, Inselspital, Bern and Translational Hormone Research Program, Department of Biomedical Research, University of Bern, Freiburgstrasse 15, 3010 Bern, Switzerland
| | - Fredrik Björkling
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark
| |
Collapse
|
40
|
Zhao YC, Wang CC, Yang JY, Li XY, Yanagita T, Xue CH, Zhang TT, Wang YM. N-3 PUFA Deficiency from Early Life to Adulthood Exacerbated Susceptibility to Reactive Oxygen Species-Induced Testicular Dysfunction in Adult Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:6908-6919. [PMID: 37098125 DOI: 10.1021/acs.jafc.2c07328] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Homeostasis of reactive oxygen species is required to maintain sperm maturation and capacitation. Docosahexaenoic acid (DHA) is accumulated in testicles and spermatozoa and has the ability to manipulate the redox status. The effects of dietary n-3 polyunsaturated fatty acid (n-3 PUFA) deficiency from early life to adulthood on the physiological and functional properties of males under the redox imbalance of testicular tissue deserve attention. The consecutive injection of hydrogen peroxide (H2O2) and tert-butyl hydroperoxide (t-BHP) for 15 days to induce oxidative stress in testicular tissue was used to elucidate the consequences of testicular n-3 PUFA deficiency. The results indicated that reactive oxygen species treatment in adult male mice with DHA deficiency in the testis could reduce spermatogenesis and disrupt sex hormone production, as well as trigger testicular lipid peroxidation and tissue damage. N-3 PUFA deficiency from early life to adulthood resulted in higher susceptibility to testicular dysfunction in the germinal function of supplying germ cells and the endocrine role of secreting hormones through the mechanism of aggravating mitochondria-mediated apoptosis and destruction of blood testicular barrier under oxidative stress, which might provide a basis for humans to reduce susceptibility to chronic disease and maintain reproductive health in adulthood through dietary interventions of n-3 PUFAs.
Collapse
Affiliation(s)
- Ying-Cai Zhao
- College of Food Science and Engineering, Ocean University of China, Qingdao 266003, Shandong, P. R. China
| | - Cheng-Cheng Wang
- College of Food Science and Engineering, Ocean University of China, Qingdao 266003, Shandong, P. R. China
| | - Jin-Yue Yang
- College of Food Science and Engineering, Ocean University of China, Qingdao 266003, Shandong, P. R. China
| | - Xiao-Yue Li
- College of Food Science and Engineering, Ocean University of China, Qingdao 266003, Shandong, P. R. China
| | - Teruyoshi Yanagita
- Laboratory of Nutrition Biochemistry, Department of Applied Biochemistry and Food Science, Saga University, Saga 840-8502, Japan
| | - Chang-Hu Xue
- College of Food Science and Engineering, Ocean University of China, Qingdao 266003, Shandong, P. R. China
- Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao 266237 Shandong Province, P. R. China
| | - Tian-Tian Zhang
- College of Food Science and Engineering, Ocean University of China, Qingdao 266003, Shandong, P. R. China
| | - Yu-Ming Wang
- College of Food Science and Engineering, Ocean University of China, Qingdao 266003, Shandong, P. R. China
- Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao 266237 Shandong Province, P. R. China
| |
Collapse
|
41
|
Ogawa K, Tanida T. Mixed-Culture Propagation of Uterine-Tissue-Resident Macrophages and Their Expression Properties of Steroidogenic Molecules. Biomedicines 2023; 11:biomedicines11030985. [PMID: 36979964 PMCID: PMC10046189 DOI: 10.3390/biomedicines11030985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/09/2023] [Accepted: 03/19/2023] [Indexed: 03/30/2023] Open
Abstract
Tissue-resident macrophages (Mø) play tissue/organ-specific roles, and the physiological/pathological implications of uterine Mø in fertility and infertility are not yet fully understood. Herein, we report a simple propagation method for tissue-resident Mø by mixed culture with the respective tissue/organ-residing cells as the niche. We successfully propagated mouse uterine Mø by mixed culture with fibroblastic cells that exhibited properties of endometrial stromal cells. Propagated mouse uterine Mø were CD206- and arginase-1-positive; iNOS- and MHC-II-negative, indicating M2 polarization; and highly phagocytic, similar to endometrial Mø. Furthermore, uterine Mø were observed to express steroidogenic molecules including SRD5A1 and exhibited gap junction formation, likely with endometrial stromal cells. Accordingly, uterine Mø propagated by mixed culture may provide a new tool for studying immune-endocrine interactions related to fertility and infertility, particularly androgen's intracrine actions in preparing the uterine tissue environment to support implantation and pregnancy as well as in the etiology of endometriosis.
Collapse
Affiliation(s)
- Kazushige Ogawa
- Laboratory of Veterinary Anatomy, Graduate School of Veterinary Science, Osaka Metropolitan University, 1-58 Rinku-Ourai-Kita, Izumisano, Osaka 598-8531, Japan
| | - Takashi Tanida
- Laboratory of Veterinary Anatomy, Graduate School of Veterinary Science, Osaka Metropolitan University, 1-58 Rinku-Ourai-Kita, Izumisano, Osaka 598-8531, Japan
| |
Collapse
|
42
|
Zhang R, Singh S, Pan C, Xu B, Kindblom J, Eng KH, Krolewski JJ, Nastiuk KL. Rate of castration-induced prostate stroma regression is reduced in a mouse model of benign prostatic hyperplasia. AMERICAN JOURNAL OF CLINICAL AND EXPERIMENTAL UROLOGY 2023; 11:12-26. [PMID: 36923722 PMCID: PMC10009314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Accepted: 02/25/2023] [Indexed: 03/18/2023]
Abstract
Benign prostatic hyperplasia (BPH) is a non-neoplastic proliferative disease producing lower urinary tract symptoms related to the resulting enlarged prostate. BPH is pathologically characterized by hyperplastic growth in both epithelial and stromal compartments. Androgen signaling is essential for prostate function and androgen blockade is the second-line medical therapy to relieve symptoms of BPH. Here we examined the prostates of probasin promoter-driven prolactin (Pb-PRL) transgenic mice, a robust model of BPH that spontaneously develops prostate enlargement, to investigate prostate regression in response to surgical castration. Serial ultrasound imaging demonstrated very uniform self-limited growth of Pb-PRL prostate volume that is consistent with the benign, limited cellular proliferation characteristic of BPH and that contrasts with the highly variable, exponential growth of murine prostate cancer models. Castration elicited only a partial reduction in prostate volume, relative to castration-induced regression of the normal prostate gland. The anti-androgen finasteride induced a diminished reduction of Pb-PRL prostate volume versus castration. The limited extent of Pb-PRL mouse prostate volume regression correlated with the initial volume of the stromal compartment, suggesting a differential sensitivity of the epithelial and stromal compartments to androgen withdrawal. Indeed, two-dimensional morphometric analyses revealed a distinctly reduced rate of regression for the stromal compartment in Pb-PRL mice. The myofibroblast component of the Pb-PRL prostate stroma appeared normal, but the stromal compartment contained more fibroblasts and extracellular collagen deposition. Like normal prostate, the rate of regression of the Pb-PRL prostate was partially dependent on TGFß and TNF signaling, but unlike the normal prostate, the extent of castration-induced regression was not affected by TGFß or TNF blockade. Our studies show that androgen deprivation can effectively reduce the overall volume of hyperplastic prostate, but the stromal compartment is relatively resistant, suggesting additional therapies might be required to offer an effective treatment for the clinical manifestations of BPH.
Collapse
Affiliation(s)
- Renyuan Zhang
- Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer CenterBuffalo, NY 14263 USA
| | - Shalini Singh
- Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer CenterBuffalo, NY 14263 USA
| | - Chunliu Pan
- Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer CenterBuffalo, NY 14263 USA
| | - Bo Xu
- Pathology and Laboratory Medicine, Roswell Park Comprehensive Cancer CenterBuffalo, NY 14263 USA
| | - Jon Kindblom
- Department of Oncology, University of GothenburgGoteborg 41345, Sweden
| | - Kevin H Eng
- Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer CenterBuffalo, NY 14263 USA
- Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer CenterBuffalo, NY 14263 USA
- Bristol Myers SquibbPrinceton, NJ, USA
| | - John J Krolewski
- Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer CenterBuffalo, NY 14263 USA
- Department of Biology and Interdisciplinary Unit, Data Science and Analytics, Buffalo State College, State University of New YorkNew York, NY 14263, USA
| | - Kent L Nastiuk
- Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer CenterBuffalo, NY 14263 USA
- Urology, Roswell Park Comprehensive Cancer CenterBuffalo, NY 14263, USA
| |
Collapse
|
43
|
Wei L, Gao H, Yu J, Zhang H, Nguyen TTL, Gu Y, Passow MR, Carter JM, Qin B, Boughey JC, Goetz MP, Weinshilboum RM, Ingle JN, Wang L. Pharmacological Targeting of Androgen Receptor Elicits Context-Specific Effects in Estrogen Receptor-Positive Breast Cancer. Cancer Res 2023; 83:456-470. [PMID: 36469363 PMCID: PMC9896025 DOI: 10.1158/0008-5472.can-22-1016] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 10/04/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022]
Abstract
Androgen receptor (AR) is expressed in 80% to 90% of estrogen receptor α-positive (ER+) breast cancers. Accumulated evidence has shown that AR is a tumor suppressor and that its expression is associated with improved prognosis in ER+ breast cancer. However, both a selective AR agonist (RAD140) and an AR inhibitor (enzalutamide, ENZ) have shown a therapeutic effect on ER+ breast cancer, so the potential for clinical application of AR-targeting therapy for ER+ breast cancer is still in dispute. In this study, we evaluated the efficacy of ENZ and RAD140 in vivo and in vitro in AR+/ER+ breast cancer models, characterizing the relationship of AR and ER levels to response to AR-targeting drugs and investigating the alterations of global gene expression and chromatin binding of AR and ERα after ENZ treatment. In the AR-low setting, ENZ directly functioned as an ERα antagonist. Cell growth inhibition by ENZ in breast cancer with low AR expression was independent of AR and instead dependent on ER. In AR-high breast cancer models, AR repressed ERα signaling and ENZ promoted ERα signaling by antagonizing AR. In contrast, RAD140 activated AR signaling and suppressed AR-high tumor growth by deregulating ERα expression and blocking ERα function. Overall, analysis of the dynamic efficacies and outcomes of AR agonist, and antagonist in the presence of different AR and ERα levels reveals regulators of response and supports the clinical investigation of ENZ in selected ER+ tumors with a low AR/ER ratio and AR agonists in tumors with a high AR/ER ratio. SIGNIFICANCE The ratio of androgen receptor to estrogen receptor in breast cancer dictates the response to AR-targeted therapies, providing guidelines for developing AR-directed treatment strategies for patients with breast cancer.
Collapse
Affiliation(s)
- Lixuan Wei
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota
| | - Huanyao Gao
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota
| | - Jia Yu
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota
| | - Huan Zhang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota
| | - Thanh Thanh L. Nguyen
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota
| | - Yayun Gu
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota
| | - Marie R. Passow
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Jodi M. Carter
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Bo Qin
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota
- Department of Oncology, Mayo Clinic, Rochester, Minnesota
| | | | - Matthew P. Goetz
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota
- Department of Oncology, Mayo Clinic, Rochester, Minnesota
| | - Richard M. Weinshilboum
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota
| | - James N. Ingle
- Department of Oncology, Mayo Clinic, Rochester, Minnesota
| | - Liewei Wang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
44
|
Söderqvist G, Naessén S. Androgens impact on psychopathological variables according to CPRS, and EDI 2 scores: In women with bulimia nervosa, and eating disorder not otherwise specified. J Steroid Biochem Mol Biol 2023; 226:106217. [PMID: 36368624 DOI: 10.1016/j.jsbmb.2022.106217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 10/28/2022] [Accepted: 11/06/2022] [Indexed: 11/09/2022]
Abstract
Bulimia nervosa (BN) is characterized by binge eating, compensatory behavior, over-evaluation of weight and shape, which often co-occur with symptoms of anxiety and depression. Depression is the most common comorbid diagnosis in women with eating disorders. The role of androgens in the pathophysiology of depression has been recognized in recent years. However, the research on psychopathological comorbidity and androgen levels in bulimic disease is sparse. This study aimed to investigate, if there were any correlations between the androgens, testosterone (T), dehydroepiandrosterone sulphate (DHEAS), androstenedione (A4), 5α-dihydrotestosterone, (5α-DHT), and test scores of psychopathological variables, in women with bulimia nervosa (BN), eating disorder not otherwise specified of purging subtype (EDNOS-P) assessed by CPRS, and EDI 2. Women with DSM-IV diagnosis of BN (n = 36), EDNOS-P (n = 27), and healthy control subjects (n = 58) evaluated for fifteen psychopathological variables, i.a. depressive symptoms, impulsivity, personal traits, as well as serum androgen levels. All women were euthyroid, and polycystic ovarian syndrome (PCOS) diagnosis was excluded. Although androgen levels were almost equal for all three groups, significant correlations between core psychopathological symptoms (9/15) of bulimia nervosa and the most potent endogenous androgen, 5α-DHT, was found only in the EDNOS-P group. The role of 5α-DHT in women is not fully elucidated. Both animal and human studies have shown that the brain is able to locally synthesize steroids de novo and is a target of steroid hormones. Maybe these results can be interpreted in the light of differences in androgen receptor variability, metabolism and origin of T and 5α-DHT.
Collapse
Affiliation(s)
- Gunnar Söderqvist
- Department of Women's, and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Sabine Naessén
- Department of Women's, and Children's Health, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
45
|
Topical Delivery of Atraric Acid Derived from Stereocaulon japonicum with Enhanced Skin Permeation and Hair Regrowth Activity for Androgenic Alopecia. Pharmaceutics 2023; 15:pharmaceutics15020340. [PMID: 36839662 PMCID: PMC9960134 DOI: 10.3390/pharmaceutics15020340] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Revised: 01/13/2023] [Accepted: 01/17/2023] [Indexed: 01/21/2023] Open
Abstract
Atraric acid (AA) is a phenolic compound isolated from Stereocaulon japonicum that has demonstrated anti-androgen properties and was used to design an alternative formulation for the treatment of alopecia. This new topical formulation was designed using a solvent mixture system composed of ethanol as a volatile vehicle, oleic acid as a permeation enhancer, and water for skin hydration. The ideal topical AA formulation (AA-TF#15) exhibited an 8.77-fold higher human skin flux and a 570% increase in dermal drug deposition, compared to 1% (w/w) AA in ethanol. In addition, compared to other formulations, AA-TF#15 (1% [w/w] AA) activated keratinocytes and human dermal papilla cell proliferation at a concentration of 50 µM AA, which is equivalent to 50 µM minoxidil. Moreover, AA-TF#15 treatment produced a significant increase in hair regrowth by 58.0% and 41.9% compared to the 1% (w/w) minoxidil and oral finasteride (1 mg/kg)-treated mice. In addition, AA-TF#15 showed a higher expression level of aldehyde dehydrogenase 1, β-catenin, cyclin D1, and pyruvate kinase M2 proteins in the skin of AA-TF#15-treated mice compared to that of those treated with minoxidil and oral finasteride. These findings suggest AA-TF#15 is an effective formulation for the treatment of scalp androgenic alopecia.
Collapse
|
46
|
Hargrove-Wiley E, Fingleton B. Sex Hormones in Breast Cancer Immunity. Cancer Res 2023; 83:12-19. [PMID: 36279153 DOI: 10.1158/0008-5472.can-22-1829] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 09/22/2022] [Accepted: 10/18/2022] [Indexed: 02/03/2023]
Abstract
Sex hormones, such as estrogens and androgens, regulate genomic and cellular processes that contribute to sex-specific disparities in the pathophysiology of various cancers. Sex hormones can modulate the immune signals and activities of tumor cells and tumor-associated leukocytes to support or suppress cancer progression. Therefore, hormonal differences between males and females play a crucial role in cancer immunity and in the response to therapies that exploit the intrinsic immune system to eliminate malignant cells. In this review, we summarize the impact of sex hormones in the breast cancer microenvironment, with a focus on how the hormonal environment affects tumor immunity. We also discuss the potential benefits of endocrine therapy used in combination with immunotherapy to strengthen the antitumor immune response.
Collapse
Affiliation(s)
- Ebony Hargrove-Wiley
- Program in Cancer Biology, Department of Pharmacology, Vanderbilt University, Nashville, Tennessee
| | - Barbara Fingleton
- Program in Cancer Biology, Department of Pharmacology, Vanderbilt University, Nashville, Tennessee
| |
Collapse
|
47
|
Schafstedde M, Nordmeyer S. The role of androgens in pressure overload myocardial hypertrophy. Front Endocrinol (Lausanne) 2023; 14:1112892. [PMID: 36817598 PMCID: PMC9929540 DOI: 10.3389/fendo.2023.1112892] [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: 11/30/2022] [Accepted: 01/06/2023] [Indexed: 02/04/2023] Open
Abstract
Pressure overload hypertrophy of the left ventricle is a common result of many cardiovascular diseases. Androgens show anabolic effects in skeletal muscles, but also in myocardial hypertrophy. We carefully reviewed literature regarding possible effects of androgens on specific left ventricular hypertrophy in pressure overload conditions excluding volume overload conditions or generel sex differences.
Collapse
Affiliation(s)
- Marie Schafstedde
- Department of Congenital Heart Disease – Pediatric Cardiology, Deutsches Herzzentrum der Charité – Medical Heart Center of Charité and German Heart Institute Berlin, Berlin, Germany
- Institute of Computer-Assisted Cardiovascular Medicine, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Partner Site Berlin, German Center for Cardiovascular Research (DZHK), Berlin, Germany
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Sarah Nordmeyer
- Department of Congenital Heart Disease – Pediatric Cardiology, Deutsches Herzzentrum der Charité – Medical Heart Center of Charité and German Heart Institute Berlin, Berlin, Germany
- Institute of Computer-Assisted Cardiovascular Medicine, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Partner Site Berlin, German Center for Cardiovascular Research (DZHK), Berlin, Germany
- *Correspondence: Sarah Nordmeyer,
| |
Collapse
|
48
|
Pruett JE, Romero DG, Yanes Cardozo LL. Obesity-associated cardiometabolic complications in polycystic ovary syndrome: The potential role of sodium-glucose cotransporter-2 inhibitors. Front Endocrinol (Lausanne) 2023; 14:951099. [PMID: 36875461 PMCID: PMC9974663 DOI: 10.3389/fendo.2023.951099] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 01/26/2023] [Indexed: 02/17/2023] Open
Abstract
Polycystic Ovary Syndrome (PCOS) is the most common endocrine disorder in reproductive-age women. PCOS is characterized by androgen excess, oligo/anovulation, and polycystic appearance of the ovaries. Women with PCOS have an increased prevalence of multiple cardiovascular risk factors such as insulin resistance, hypertension, renal injury, and obesity. Unfortunately, there is a lack of effective, evidence-based pharmacotherapeutics to target these cardiometabolic complications. Sodium-glucose cotransporter-2 (SGLT2) inhibitors provide cardiovascular protection in patients with and without type 2 diabetes mellitus. Although the exact mechanisms of how SGLT2 inhibitors confer cardiovascular protection remains unclear, numerous mechanistic hypotheses for this protection include modulation of the renin-angiotensin system and/or the sympathetic nervous system and improvement in mitochondrial function. Data from recent clinical trials and basic research show a potential role for SGLT2 inhibitors in treating obesity-associated cardiometabolic complications in PCOS. This narrative review discusses the mechanisms of the beneficial effect of SGLT2 inhibitors in cardiometabolic diseases in PCOS.
Collapse
Affiliation(s)
- Jacob E. Pruett
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS, United States
| | - Damian G. Romero
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS, United States
- Mississippi Center of Excellence in Perinatal Research, University of Mississippi Medical Center, Jackson, MS, United States
- Women’s Health Research Center, University of Mississippi Medical Center, Jackson, MS, United States
- Cardiovascular-Renal Research Center, University of Mississippi Medical Center, Jackson, MS, United States
| | - Licy L. Yanes Cardozo
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS, United States
- Mississippi Center of Excellence in Perinatal Research, University of Mississippi Medical Center, Jackson, MS, United States
- Women’s Health Research Center, University of Mississippi Medical Center, Jackson, MS, United States
- Cardiovascular-Renal Research Center, University of Mississippi Medical Center, Jackson, MS, United States
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, United States
- *Correspondence: Licy L. Yanes Cardozo,
| |
Collapse
|
49
|
Huang G, Bhasin S, Pencina K, Cheng M, Jasuja R. Circulating dihydrotestosterone, testosterone, and free testosterone levels and dihydrotestosterone-to-testosterone ratios in healthy women across the menstrual cycle. Fertil Steril 2022; 118:1150-1158. [PMID: 36371319 DOI: 10.1016/j.fertnstert.2022.09.011] [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: 06/09/2022] [Revised: 08/30/2022] [Accepted: 09/06/2022] [Indexed: 11/10/2022]
Abstract
OBJECTIVE To characterize the circulating androgen levels across the menstrual cycle in healthy women using highly sensitive and accurate methods and report sex differences in the relative levels of dihydrotestosterone (DHT) to testosterone (T) levels. DESIGN Prospective cohort study. SETTING Research clinic, academic teaching hospital. PATIENT(S) Twenty-one healthy premenopausal women, aged 19-40 years, with regular menstrual cycles. INTERVENTION(S) Not applicable. MAIN OUTCOME MEASURE(S) Serum total T and DHT levels measured using liquid chromatography-tandem mass spectrometry, free T levels measured using a standardized equilibrium dialysis method coupled with measurement of the T levels in the dialysate using liquid chromatography-tandem mass spectrometry, and comparison of the DHT-to-T ratio between healthy women and age-matched healthy men. RESULT(S) The serum total and free T levels increased across the follicular phase and peaked at midcycle (total T, 43.6 ± 16.2 ng/dL; free T, 15.6 ± 11.9 pg/mL) and gradually declined in the luteal phase. The DHT level did not significantly change across the menstrual cycle. The DHT-to-T ratios were 1:4 and 1:13 in women and men, respectively. CONCLUSION(S) In healthy premenopausal women, the total and free T levels varied significantly across the menstrual cycle, whereas the DHT levels did not change; the peak total and free T levels in the midcycle period were higher than previously reported, underscoring the importance of establishing menstrual phase-specific reference ranges to avoid misdiagnosis of hyperandrogenism. Women have significantly higher DHT levels relative to total T than men; the significance of this sex difference in the DHT-to-T ratio needs further investigation.
Collapse
Affiliation(s)
- Grace Huang
- Research Program in Men's Health: Aging and Metabolism, Boston Claude D. Pepper Older Americans Independence Center, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.
| | - Shalender Bhasin
- Research Program in Men's Health: Aging and Metabolism, Boston Claude D. Pepper Older Americans Independence Center, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Karol Pencina
- Research Program in Men's Health: Aging and Metabolism, Boston Claude D. Pepper Older Americans Independence Center, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Ming Cheng
- Research Program in Men's Health: Aging and Metabolism, Boston Claude D. Pepper Older Americans Independence Center, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Ravi Jasuja
- Research Program in Men's Health: Aging and Metabolism, Boston Claude D. Pepper Older Americans Independence Center, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
50
|
Zhang H, Zhang L, Chen S, Yao M, Ma Z, Yuan Y. Yeast-based evolutionary modeling of androgen receptor mutations and natural selection. PLoS Genet 2022; 18:e1010518. [PMID: 36459502 PMCID: PMC9718406 DOI: 10.1371/journal.pgen.1010518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 11/10/2022] [Indexed: 12/05/2022] Open
Abstract
Cancer progression is associated with the evolutionary accumulation of genetic mutations that are biologically significant. Mutations of the androgen receptor (AR) are associated with the development of prostate cancer (PCa) by responding to non-androgenic hormones, and the lack of annotations in their responsiveness to hormone ligands remains a daunting challenge. Here, we have used a yeast reporter system to quickly evaluate the responsiveness of all fifty clinical AR mutations to a variety of steroidal ligands including dihydrotestosterone (DHT), 17β-estradiol (E2), progesterone (PROG), and cyproterone acetate (CPA). Based on an AR-driven reporter that synthesizes histidine, a basic amino acid required for yeast survival and propagation, the yeast reporter system enabling clonal selection was further empowered by combining with a random DNA mutagenesis library to simulate the natural evolution of AR gene under the selective pressures of steroidal ligands. In a time-frame of 1-2 weeks, 19 AR mutants were identified, in which 11 AR mutants were validated for activation by tested steroidal compounds. The high efficiency of our artificial evolution strategy was further evidenced by a sequential selection that enabled the discovery of multipoint AR mutations and evolution directions under the pressure of steroidal ligands. In summary, our designer yeast is a portable reporter module that can be readily adapted to streamline high-throughput AR-compound screening, used as a PCa clinical reference, and combined with additional bioassay systems to further extend its potential.
Collapse
Affiliation(s)
- Haoran Zhang
- Frontier Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin, People’s Republic of China
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, People’s Republic of China
| | - Lu Zhang
- Frontier Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin, People’s Republic of China
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, People’s Republic of China
| | - Shaoyong Chen
- Hematology-Oncology Division, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Mingdong Yao
- Frontier Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin, People’s Republic of China
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, People’s Republic of China
| | - Zhenyi Ma
- School of Basic Medical Sciences, Tianjin Medical University, Tianjin, People’s Republic of China
| | - Yingjin Yuan
- Frontier Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin, People’s Republic of China
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, People’s Republic of China
- * E-mail:
| |
Collapse
|