1
|
Gaižauskaitė R, Gladutytė L, Zelionkaitė I, Grikšienė R. Exploring the role of sex, sex steroids, menstrual cycle, and hormonal contraception use in visual working memory: Insights from behavioral and EEG analyses. Int J Psychophysiol 2025; 209:112520. [PMID: 39892467 DOI: 10.1016/j.ijpsycho.2025.112520] [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: 08/05/2024] [Revised: 01/08/2025] [Accepted: 01/28/2025] [Indexed: 02/03/2025]
Abstract
Sex hormones have been shown to influence cognitive and emotional processes, yet their effects on visual working memory (VWM) are poorly understood. This study aimed to investigate the relationship between VWM, sex, and female hormonal status in participants aged 18-35 years. We recruited 32 males (M) and 133 females, categorized into four groups: naturally cycling females in the early follicular (NCF, n = 33) and mid-luteal (NCL, n = 35) phases of the menstrual cycle, oral contraceptive (OC, n = 37), and intrauterine device users (IUD, n = 28). Participants completed a bilateral change detection task while behavioral and EEG data were recorded. We evaluated VWM performance and associated brain electrophysiological responses, specifically Contralateral Delay Activity (CDA). Salivary levels of testosterone, progesterone, and estradiol were assessed. We found no systematic differences in VWM task performance or CDA between groups, nor any correlations with hormone levels. However, an exception to this was that NCF females performed worse than OC users when recalling four items. Age emerged as a significant covariate, with greater age being linked to poorer performance. An interaction between age and group in memory capacity highlighted differential patterns of age-related cognitive decline across sexes and female hormonal status groups. These findings provide valuable insights into the broader relationship between sex, sex hormones, and cognition. They suggest that in studies employing a between-subject design, hormone-dependent differences in more complex processes, such as visuospatial performance, are unlikely to stem from the role of sex hormones in VWM and may instead arise from other factors.
Collapse
Affiliation(s)
- Rimantė Gaižauskaitė
- Department of Neurobiology and Biophysics, Vilnius University, Saulėtekio ave. 7, 10257 Vilnius, Lithuania.
| | - Lina Gladutytė
- Department of Neurobiology and Biophysics, Vilnius University, Saulėtekio ave. 7, 10257 Vilnius, Lithuania
| | - Ingrida Zelionkaitė
- Department of Neurobiology and Biophysics, Vilnius University, Saulėtekio ave. 7, 10257 Vilnius, Lithuania
| | - Ramunė Grikšienė
- Department of Neurobiology and Biophysics, Vilnius University, Saulėtekio ave. 7, 10257 Vilnius, Lithuania
| |
Collapse
|
2
|
Kim GW, Lee M, Lee HS, Park K, Jeong GW. Hippocampal subfield volume differences between female-to-male transgender individuals with cross-sex hormone therapy and cisgender women. Front Neurosci 2025; 19:1477725. [PMID: 40084137 PMCID: PMC11903734 DOI: 10.3389/fnins.2025.1477725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 02/12/2025] [Indexed: 03/16/2025] Open
Abstract
Introduction The prevalence of female-to-male (FtM) transgender individuals has risen recently, yet the effects of cross-sex hormone therapy on volumetric differences in the hippocampal subfields remain poorly understood. This study aimed to evaluate the differences of gray matter (GM) volume, especially focusing on the hippocampal subfields, in FtM transgender individuals and premenopausal cisgender women. Methods Seventeen FtM transgender individuals who had undergone hysterectomies and were receiving testosterone therapy before participating in this study, along with 20 premenopausal women, underwent a single session of T1-weighted magnetic resonance imaging (MRI). Results FtM transgender group had significantly higher free-testosterone (free-T) levels and lower estradiol levels compared with premenopausal women group (p < 0.001). In voxel-wise analysis, FtM transgender individuals showed significantly larger GM volumes in the caudate nucleus, hypothalamus, and thalamus compared with premenopausal women (p < 0.01, FWE-corrected). More specifically, the right hippocampal subiculum volume was larger in FtM transgender individuals (p < 0.05, Bonferroni-corrected), and these volumes were positively correlated with the free-T levels (r = 0.34, p = 0.04). This study revealed the specific hippocampal subfield volume differences in the testosterone-treated FtM transgender group when compared to cisgender premenopausal women group. Discussion These findings might help elucidate the morphological variation of the specific cerebral regions associated with testosterone therapy in FtM transgender individuals and contribute to our understanding of the effects of gender-affirming hormone treatments as well.
Collapse
Affiliation(s)
- Gwang-Won Kim
- Advanced Institute of Aging Science, Chonnam National University, Gwangju, Republic of Korea
| | - Mina Lee
- Department of Urology, Chonnam National University Hospital, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Hyun-Suk Lee
- Department of Urology, Chonnam National University Hospital, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Kwangsung Park
- Advanced Institute of Aging Science, Chonnam National University, Gwangju, Republic of Korea
- Department of Urology, Chonnam National University Hospital, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Gwang-Woo Jeong
- Department of Radiology, Chonnam National University Hospital, Chonnam National University Medical School, Gwangju, Republic of Korea
| |
Collapse
|
3
|
Tian Y, Qiao H, Odamah K, Zhu LQ, Man HY. Role of androgen receptors in sexually dimorphic phenotypes in UBE3A-dependent autism spectrum disorder. iScience 2025; 28:111868. [PMID: 39991542 PMCID: PMC11847089 DOI: 10.1016/j.isci.2025.111868] [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: 01/11/2024] [Revised: 07/04/2024] [Accepted: 01/20/2025] [Indexed: 02/25/2025] Open
Abstract
Autism spectrum disorders (ASDs) involve social, communication, and behavioral challenges. ASDs display a remarkable sex difference with a 4:1 male to female prevalence ratio; however, the underlying mechanism remains largely unknown. Using the UBE3A-overexpressing mouse model for ASD, we studied sexually dimorphic changes at behavioral, genetic, and molecular levels. We found that male mice with extra copies of Ube3A exhibited greater impairments in social communication, long-term memory, and pain sensitivity compared to females. UBE3A-mediated degradation reduced androgen receptor (AR) levels in both sexes but only male mice showed significant dysregulation in the expression of AR target genes. Importantly, restoring AR levels in the brain normalized levels of AR target genes, and rescued the deficits in social preference, grooming, and memory in male UBE3A-overexpressing mice, without affecting females. These findings reveal the critical role of AR signaling in sex-specific changes linked to UBE3A-dependent ASD.
Collapse
Affiliation(s)
- Yuan Tian
- Department of Biology, Boston University, 5 Cummington Mall, Boston, MA 02215, USA
| | - Hui Qiao
- Department of Biology, Boston University, 5 Cummington Mall, Boston, MA 02215, USA
| | - KathrynAnn Odamah
- Department of Biology, Boston University, 5 Cummington Mall, Boston, MA 02215, USA
| | - Ling-Qiang Zhu
- Department of Pathophysiology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Heng-Ye Man
- Department of Biology, Boston University, 5 Cummington Mall, Boston, MA 02215, USA
- Department of Pharmacology, Physiology & Biophysics, Boston University School of Medicine, 72 East Concord St., Boston, MA 02118, USA
- Center for Systems Neuroscience, Boston University, 610 Commonwealth Avenue, Boston, MA 02215, USA
| |
Collapse
|
4
|
Wang Y, Tong X, Xiao Y, Wang Y, Hu W, Lu W, Chen Y, Li J, Gao W, Gao H, Tian Y, Dai S, Feng Y. Regulating Integrin β1 to Restore Gonadotropin-Releasing Hormone-Tanycyte Unit Function in Polycystic Ovary Syndrome-Related Hypothalamic Dysregulation. RESEARCH (WASHINGTON, D.C.) 2025; 8:0619. [PMID: 39975575 PMCID: PMC11836200 DOI: 10.34133/research.0619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 01/27/2025] [Accepted: 01/29/2025] [Indexed: 02/21/2025]
Abstract
Excessive gonadotropin-releasing hormone (GnRH) is considered to be an initiating factor in the etiology of polycystic ovary syndrome (PCOS). GnRH neuronal axons terminate at the hypothalamic arcuate nucleus and median eminence, where tanycytes, specialized glial cells, have been proposed to modulate GnRH secretion through plasticity. However, the precise role of the "GnRH-tanycyte unit" during the pathological state of PCOS has not been thoroughly explored. In this study, we demonstrated the architecture and distribution of GnRH neurons and tanycytes. In PCOS-like mice, retracted tanycyte processes and dysregulated GnRH-tanycyte unit may create an environment conducive to the excessive secretion of GnRH and subsequent reproductive endocrine dysfunction. Mechanistically, excessive androgens impair hypothalamic neuroglial homeostasis by acting through the androgen receptor (AR) and its downstream target integrin β1 (Itgb1), thereby suppressing the FAK/TGF-βR1/Smad2 signaling pathway. Both selective deletion of AR and overexpression of Itgb1 in tanycytes counteracted the detrimental effects of androgens, alleviating endocrine dysfunction. Collectively, this study highlights the alterations in the GnRH-tanycyte unit mediated by androgen/AR/Itgb1 signaling and provides a novel perspective for developing therapies for hypothalamic hormone secretion disorders by maintaining solid neuroglial structures in the brain.
Collapse
Affiliation(s)
- Yu Wang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science,
Fudan University, Shanghai 200032, China
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Shanghai Institute of Acupuncture and Moxibustion, Shanghai 200433, China
| | - Xiaoyu Tong
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science,
Fudan University, Shanghai 200032, China
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Shanghai Institute of Acupuncture and Moxibustion, Shanghai 200433, China
| | - Yan Xiao
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science,
Fudan University, Shanghai 200032, China
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Shanghai Institute of Acupuncture and Moxibustion, Shanghai 200433, China
| | - Yicong Wang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science,
Fudan University, Shanghai 200032, China
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Shanghai Institute of Acupuncture and Moxibustion, Shanghai 200433, China
| | - Wei Hu
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science,
Fudan University, Shanghai 200032, China
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Shanghai Institute of Acupuncture and Moxibustion, Shanghai 200433, China
| | - Wenhan Lu
- Department of Ophthalmology & Visual Science, Eye & ENT Hospital, Shanghai Medical College,
Fudan University, Shanghai, China
| | - Yuning Chen
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science,
Fudan University, Shanghai 200032, China
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Shanghai Institute of Acupuncture and Moxibustion, Shanghai 200433, China
| | - Jiajia Li
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science,
Fudan University, Shanghai 200032, China
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Shanghai Institute of Acupuncture and Moxibustion, Shanghai 200433, China
| | - Wenhao Gao
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science,
Fudan University, Shanghai 200032, China
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Shanghai Institute of Acupuncture and Moxibustion, Shanghai 200433, China
| | - Hongru Gao
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science,
Fudan University, Shanghai 200032, China
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Shanghai Institute of Acupuncture and Moxibustion, Shanghai 200433, China
| | - Yicheng Tian
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science,
Fudan University, Shanghai 200032, China
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Shanghai Institute of Acupuncture and Moxibustion, Shanghai 200433, China
| | - Sizhe Dai
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science,
Fudan University, Shanghai 200032, China
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Shanghai Institute of Acupuncture and Moxibustion, Shanghai 200433, China
| | - Yi Feng
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science,
Fudan University, Shanghai 200032, China
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Shanghai Institute of Acupuncture and Moxibustion, Shanghai 200433, China
| |
Collapse
|
5
|
Matsukawa A, Yanagisawa T, Rajwa P, Fazekas T, Miszczyk M, Tsuboi I, Parizi MK, Laukhtina E, Klemm J, Chiujdea S, Mancon S, Mori K, Kimura S, Karakiewicz PI, Miki J, Kimura T, Shariat SF. Central Nervous System Toxicity in Prostate Cancer Patients Treated with Androgen Receptor Signaling Inhibitors: A Systematic Review, Meta-analysis, and Network Meta-analysis. Clin Genitourin Cancer 2025; 23:102251. [PMID: 39571519 DOI: 10.1016/j.clgc.2024.102251] [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: 08/19/2024] [Revised: 10/17/2024] [Accepted: 10/21/2024] [Indexed: 01/30/2025]
Abstract
BACKGROUND Androgen-receptor signaling inhibitors (ARSIs) significantly improve survival in systemic therapy for advanced/metastatic prostate cancer (PCa) patients; however possible central nervous system (CNS) toxicity is an unaddressed concern. We aimed to assess and compare the incidence of CNS-related adverse events (AEs) secondary to the treatment of PCa patients with different ARSIs. MATERIALS In August 2023, a comprehensive seach was conducted in three databases for randomized controlled trials (RCTs) of PCa patients receiving ARSIs plus ADT. The primary endpoints included mental impairment, cognitive impairment, seizure, fatigue, and falls. RESULTS Twenty-six RCTs, comprising 20,328 patients, were included in meta-analyses and network meta-analyses (NMAs). ARSIs increased the risk of mental impairment (RR: 1.72; 95% CI, 1.09-2.71), cognitive impairment (RR: 2.25; 95% CI, 1.78-2.86), seizure (RR: 2.20, 95% CI, 1.09-4.45), fatigue (RR: 1.31, 95% CI, 1.20-1.43), and falls (RR: 2.07, 95% CI, 1.60-2.67) compared to standard of care (SOC). Based on NMAs, Enzalutamide showed a significant increase in risk for all assessed CNS-related AEs, while Abiraterone demonstrated significant risk increases in cognitive impairment, fatigue, and falls. Conversely, Darolutamide did not exhibit significant increases in risk for any CNS-related AEs, except for fatigue. CONCLUSIONS The addition of ARSIs to ADT increased all examined CNS-related AEs compared to SOC. Each ARSI is associated with a distinct profile of CNS-related AEs. Careful patient selection and monitoring for CNS sequelae is necessary to achieve the best quality of life in patients on ARSI + ADT for PCa.
Collapse
Affiliation(s)
- Akihiro Matsukawa
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Department of Urology, The Jikei University School of Medicine, Tokyo, Japan
| | - Takafumi Yanagisawa
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Department of Urology, The Jikei University School of Medicine, Tokyo, Japan
| | - Pawel Rajwa
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Second Department of Urology, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Tamás Fazekas
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Department of Urology, Semmelweis University, Budapest, Hungary
| | - Marcin Miszczyk
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Collegium Medicum - Faculty of Medicine, WSB University, Dąbrowa Górnicza, Poland
| | - Ichiro Tsuboi
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Department of Urology, Shimane University Faculty of Medicine, Shimane, Japan
| | - Mehdi Kardoust Parizi
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Department of Urology, Shariati Hospital, Tehran University of Medical Science, Tehran, Iran
| | - Ekaterina Laukhtina
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Institute for Urology and Reproductive Health, Sechenov University, Moscow, Russia
| | - Jakob Klemm
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Department of Urology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sever Chiujdea
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Department of Urology, Spitalul Clinic Judetean Murures, University of Medicine, Pharmacy, Science, and Technology of Targu Mures, Mures, Romania
| | - Stefano Mancon
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| | - Keiichiro Mori
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Department of Urology, The Jikei University School of Medicine, Tokyo, Japan
| | - Shoji Kimura
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Department of Urology, The Jikei University School of Medicine, Tokyo, Japan
| | - Pierre I Karakiewicz
- Cancer Prognostics and Health Outcomes Unit, Division of Urology, University of Montréal Health Center, Montréal, QC, Canada
| | - Jun Miki
- Department of Urology, The Jikei University School of Medicine, Tokyo, Japan
| | - Takahiro Kimura
- Department of Urology, The Jikei University School of Medicine, Tokyo, Japan
| | - Shahrokh F Shariat
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Hourani Center for Applied Scientific Research, Al-Ahliyya Amman University, Amman, Jordan; Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Urology, Second Faculty of Medicine, Charles University, Prague, Czech Republic; Karl Landsteiner Institute of Urology and Andrology, Vienna, Austria; Department of Urology, Research Center for Evidence Medicine, Tabriz University of Medical Science, Tabriz, Iran.
| |
Collapse
|
6
|
Pierret ACS, Patel AH, Daniels E, Comninos AN, Dhillo WS, Abbara A. Kisspeptin as a test of hypothalamic dysfunction in pubertal and reproductive disorders. Andrology 2025. [PMID: 39834030 DOI: 10.1111/andr.13843] [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: 10/28/2024] [Revised: 12/30/2024] [Accepted: 01/14/2025] [Indexed: 01/22/2025]
Abstract
The hypothalamic-pituitary-gonadal axis is regulated by the gonadotropin-releasing hormone pulse generator in the hypothalamus. This is comprised of neurons that secrete kisspeptin in a pulsatile manner to stimulate the release of GnRH, and, in turn, downstream gonadotropins from the pituitary gland, and subsequently sex steroids and gametogenesis from the gonads. Many reproductive disorders in both males and females are characterized by hypothalamic dysfunction, including functional disorders (such as age-related hypogonadism, obesity-related secondary hypogonadism, hyperprolactinemia, functional hypothalamic amenorrhea and polycystic ovary syndrome), structural pathologies (such as craniopharyngiomas or radiation or surgery-related hypothalamic dysfunction), and pubertal disorders (constitutional delay of growth and puberty and congenital hypogonadotropic hypogonadism). However, in many of these conditions, the relative contribution of hypothalamic dysfunction to the observed hypogonadism is unclear; as to date, there is no direct method of evaluating hypothalamic reproductive function in humans. Indeed, it is not possible to directly measure gonadotropin-releasing hormone levels in the hypothalamo-pituitary portal vessels, such that secondary (i.e., pituitary dysfunction) and tertiary (i.e., hypothalamic dysfunction) hypogonadism are often conflated as one entity. In this review, we examine the evidence for the use of kisspeptin as a method of directly evaluating hypothalamic reproductive dysfunction, and deliberate its potential future role in the evaluation of pubertal and reproductive disorders.
Collapse
Affiliation(s)
- Aureliane C S Pierret
- Section of Endocrinology and Investigative Medicine, Imperial College London, London, UK
| | - Aaran H Patel
- Section of Endocrinology and Investigative Medicine, Imperial College London, London, UK
| | - Elisabeth Daniels
- Section of Endocrinology and Investigative Medicine, Imperial College London, London, UK
| | - Alexander N Comninos
- Section of Endocrinology and Investigative Medicine, Imperial College London, London, UK
- Department of Endocrinology, Imperial College Healthcare Trust, London, UK
| | - Waljit S Dhillo
- Section of Endocrinology and Investigative Medicine, Imperial College London, London, UK
- Department of Endocrinology, Imperial College Healthcare Trust, London, UK
| | - Ali Abbara
- Section of Endocrinology and Investigative Medicine, Imperial College London, London, UK
- Department of Endocrinology, Imperial College Healthcare Trust, London, UK
| |
Collapse
|
7
|
Xiao H, Qin T, Wen Y, Ai L, Chen N, Cao L, Wu L. A 13-years pharmacovigilance analysis of novel hormonal agents in prostate cancer using the FDA adverse event reporting system database. Expert Opin Drug Saf 2024:1-10. [PMID: 39668443 DOI: 10.1080/14740338.2024.2442514] [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: 07/19/2024] [Revised: 11/17/2024] [Accepted: 11/18/2024] [Indexed: 12/14/2024]
Abstract
BACKGROUND Novel hormonal agents (NHAs), such as abiraterone, apalutamide, enzalutamide, and darolutamide, play a pivotal role in prostate cancer treatment. However, the toxicity spectrum of NHAs in the real-world has not been fully investigated. This study aims to analyze the adverse events (AEs) linked to NHAs. METHODS The safety profiles of NHAs were evaluated by using the United States FDA Adverse Event Reporting System (FAERS) database. Disproportionality analysis methods were applied to compare the safety signals of AEs associated with NHAs. RESULTS Our research has identified significant safety signals for all NHAs in vascular disorders, particularly noting elevated risk in the combined treatment involving docetaxel or poly ADP-ribose polymerase (PARP) inhibitors, specifically in blood and lymphatic system. Apalutamide, enzalutamide, and darolutamide displayed notable safety signals related to nervous system disorders, whereas abiraterone did not. Apalutamide exhibited specific AEs primarily linked to skin and subcutaneous tissue disorders, with severe AEs (SAEs) also concentrated in this category. Enzalutamide's specific AEs were predominantly associated with gastrointestinal disorders. Meanwhile, abiraterone's SAEs were mainly related to cardiac and hepatobiliary disorders. CONCLUSION Our study offers a comprehensive overview of safety signals associated with NHAs, providing a valuable reference for drug selection and future prospective research.
Collapse
Affiliation(s)
- Haiwei Xiao
- Department of Traditional Chinese Medicine, Shenshan Medical Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Shanwei, Guangdong Province, China
| | - Tao Qin
- Department of Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
- Department of Oncology, Shenshan Medical Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Shanwei, Guangdong Province, China
| | - Yanping Wen
- Department of Medical Oncology, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong Province, China
| | - Liang Ai
- Department of Traditional Chinese Medicine, Shenshan Medical Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Shanwei, Guangdong Province, China
- Department of Traditional Chinese Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Nabei Chen
- Department of Traditional Chinese Medicine, Shenshan Medical Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Shanwei, Guangdong Province, China
| | - Linhui Cao
- Department of Traditional Chinese Medicine, Shenshan Medical Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Shanwei, Guangdong Province, China
- Department of Traditional Chinese Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Lisha Wu
- Department of Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
- Department of Oncology, Shenshan Medical Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Shanwei, Guangdong Province, China
| |
Collapse
|
8
|
Peng J, Li Q, Liu L, Gao P, Xing L, Chen L, Liu H, Liu Z. Exploring the material basis and molecular targets of Changma Xifeng tablet in treating Tourette syndrome: an integrative approach of network pharmacology and miRNA analysis. Metab Brain Dis 2024; 39:1573-1590. [PMID: 39436634 DOI: 10.1007/s11011-024-01408-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 08/05/2024] [Indexed: 10/23/2024]
Abstract
This study was to investigate the mechanism of Changma Xifeng tablet, a traditional Chinese medicine in the treatment of Tourette syndrome. Network pharmacology was utilized to pinpoint blood-entering constituents of Changma Xifeng and explore their potential targets. Additionally, differential microRNA expression analysis was conducted to predict Tourette syndrome-associated targets, complemented by molecular docking and dynamics simulations to support the interactions of the active compounds with these targets. The study identified 98 common targets between Changma Xifeng and Tourette syndrome, which may be involved in the treatment process. A protein-protein interaction network and a drug-active ingredient-disease target network highlighted the formulation's multi-component, multi-target therapeutic approach. Eight pivotal targets-AR, GRM5, MET, RORA, HTR2A, CNR1, PDE4B, and TOP1-were identified at the intersection of microRNA and drug targets. Molecular docking revealed 12 complexes with favorable binding energies below - 7 kcal/mol, specifically: AR with Alfacalcidol, TOP1 with Albiflorin, GRM5 with Arachidic Acid, GRM5 with Palmitic Acid, AR with Arachidic Acid, AR with 2-Hydroxyoctadecanoic Acid, RORA with Pinellic Acid, RORA with Palmitic Acid, AR with Acoronene, AR with Epiacoronene, AR with 4,4'-Methylenediphenol, and HTR2A with Calycosin. Our molecular docking and molecular dynamics simulations suggest potential stable interactions between the formulation's active components and target proteins. These computational methods provide a preliminary theoretical framework that will guide our future experimental work. The study provides a scientific rationale for the use of traditional Chinese medicine in Tourette syndrome management and offers new insights for drug development.
Collapse
Affiliation(s)
- Jing Peng
- Department of Pharmacy, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 100 Xianggang Road, Jiang'an District, Wuhan, Hubei, 430016, China.
| | - Qiaoling Li
- Department of Pharmacy, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 100 Xianggang Road, Jiang'an District, Wuhan, Hubei, 430016, China
| | - Linhui Liu
- Department of Pharmacy, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 100 Xianggang Road, Jiang'an District, Wuhan, Hubei, 430016, China
| | - Ping Gao
- Department of Pharmacy, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 100 Xianggang Road, Jiang'an District, Wuhan, Hubei, 430016, China
| | - Lipeng Xing
- Department of Pharmacy, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 100 Xianggang Road, Jiang'an District, Wuhan, Hubei, 430016, China
| | - Li Chen
- Department of Pharmacy, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 100 Xianggang Road, Jiang'an District, Wuhan, Hubei, 430016, China
| | - Hui Liu
- Department of Pharmacy, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 100 Xianggang Road, Jiang'an District, Wuhan, Hubei, 430016, China
| | - Zhisheng Liu
- Department of Neurology, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
9
|
Alewel DI, Kodavanti UP. Neuroendocrine contribution to sex-related variations in adverse air pollution health effects. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART B, CRITICAL REVIEWS 2024; 27:287-314. [PMID: 39075643 PMCID: PMC12032588 DOI: 10.1080/10937404.2024.2383637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/31/2024]
Abstract
Air pollution exposure is ranked as a leading environmental risk factor for not only cardiopulmonary diseases but also for systemic health ailments including diabetes, reproductive abnormalities, and neuropsychiatric disorders, likely mediated by central neural stress mechanisms. Current experimental evidence links many air pollution health outcomes with activation of neuroendocrine sympathetic-adrenal-medullary and hypothalamic-pituitary-adrenal (HPA) stress axes associated with resultant increases in adrenal-derived hormone levels acting as circulating mediators of multi-organ stress reactions. Epidemiological and experimental investigations also demonstrated sex-specific responses to air pollutant inhalation, which may be attributed to hormonal interactions within the stress and reproductive axes. Sex hormones (androgens and estrogens) interact with neuroendocrine functions to influence hypothalamic responses, subsequently augmenting stress-mediated metabolic and immune changes. These neurohormonal interactions may contribute to innate sex-specific responses to inhaled irritants, inducing differing individual susceptibility. The aim of this review was to: (1) examine neuroendocrine co-regulation of the HPA axis by gonadal hormones, (2) provide experimental evidence demonstrating sex-specific respiratory and systemic effects attributed to air pollutant inhalation exposure, and (3) postulate proposed mechanisms of stress and sex hormone interactions during air pollution-related stress.
Collapse
Affiliation(s)
- Devin I. Alewel
- Oak Ridge Institute for Science and Education Research Participation Program, U.S. Environmental Protection Agency, Research Triangle Park, NC, United States of America
| | - Urmila P. Kodavanti
- Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina, United States of America
| |
Collapse
|
10
|
Lengyel K, Rudra M, Berghof TVL, Leitão A, Frankl-Vilches C, Dittrich F, Duda D, Klinger R, Schleibinger S, Sid H, Trost L, Vikkula H, Schusser B, Gahr M. Unveiling the critical role of androgen receptor signaling in avian sexual development. Nat Commun 2024; 15:8970. [PMID: 39419984 PMCID: PMC11487053 DOI: 10.1038/s41467-024-52989-w] [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: 06/30/2023] [Accepted: 09/24/2024] [Indexed: 10/19/2024] Open
Abstract
Gonadal hormone activities mediated by androgen and estrogen receptors, along with cell-autonomous mechanisms arising from the absence of sex-chromosome dosage compensation, are key factors in avian sexual development. In this study, we generate androgen receptor (AR) knockout chickens (AR-/-) to explore the role of androgen signaling in avian sexual development. Despite developing sex-typical gonads and gonadal hormone production, AR-/- males and females are infertile. While few somatic sex-specific traits persist (body size, spurs, and tail feathers), crucial sexual attributes such as comb, wattles and sexual behaviors remain underdeveloped in both sexes. Testosterone treatment of young AR-/- males fails to induce crow behavior, comb development, or regression of the bursa of Fabricius, which are testosterone-dependent phenotypes. These findings highlight the significance of androgen receptor mechanisms in fertility and sex-specific traits in chickens, challenging the concept of a default sex in birds and emphasizing the dominance of androgen signaling in avian sexual development.
Collapse
Affiliation(s)
- Kamila Lengyel
- Department of Behavioural Neurobiology, Max Planck Institute for Biological Intelligence, Seewiesen, Germany
- Reproductive Biotechnology, TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Mekhla Rudra
- Department of Behavioural Neurobiology, Max Planck Institute for Biological Intelligence, Seewiesen, Germany
| | - Tom V L Berghof
- Reproductive Biotechnology, TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Albertine Leitão
- Department of Behavioural Neurobiology, Max Planck Institute for Biological Intelligence, Seewiesen, Germany
| | - Carolina Frankl-Vilches
- Department of Behavioural Neurobiology, Max Planck Institute for Biological Intelligence, Seewiesen, Germany
| | - Falk Dittrich
- Department of Behavioural Neurobiology, Max Planck Institute for Biological Intelligence, Seewiesen, Germany
| | - Denise Duda
- Reproductive Biotechnology, TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Romina Klinger
- Reproductive Biotechnology, TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Sabrina Schleibinger
- Reproductive Biotechnology, TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Hicham Sid
- Reproductive Biotechnology, TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Lisa Trost
- Department of Behavioural Neurobiology, Max Planck Institute for Biological Intelligence, Seewiesen, Germany
| | - Hanna Vikkula
- Reproductive Biotechnology, TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Benjamin Schusser
- Reproductive Biotechnology, TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Manfred Gahr
- Department of Behavioural Neurobiology, Max Planck Institute for Biological Intelligence, Seewiesen, Germany.
| |
Collapse
|
11
|
Zhang T, Pan W, Tan X, Yu J, Cheng S, Wei S, Fan K, Wang L, Luo H, Hu X. A novel L-shaped ortho-quinone analog suppresses glioblastoma progression by targeting acceleration of AR degradation and regulating PI3K/AKT pathway. Biochem Pharmacol 2024; 226:116398. [PMID: 38944395 DOI: 10.1016/j.bcp.2024.116398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 06/14/2024] [Accepted: 06/26/2024] [Indexed: 07/01/2024]
Abstract
Glioblastoma (GBM) is a primary intracranial malignant tumor with the highest mortality and morbidity among all malignant central nervous system tumors. Tanshinone IIA is a fat-soluble active ingredient obtained from Salvia miltiorrhiza, which has an inhibitory effect against various cancers. We designed and synthesized a novel L-shaped ortho-quinone analog TE5 with tanshinone IIA as the lead compound and tested its antitumor activity against GBM. The results indicated that TE5 effectively inhibited the proliferation, migration, and invasion of GBM cells, and demonstrated low toxicity in vitro. We found that TE5 may bind to androgen receptors and promote their degradation through the proteasome. Inhibition of the PI3K/AKT signaling pathway was also observed in TE5 treated GBM cells. Additionally, TE5 arrested the cell cycle at the G2/M phase and induced mitochondria-dependent apoptosis. In vivo experiments further confirmed the anti-tumor activity, safety, and effect on androgen receptor level of TE5 in animal models of GBM. Our results suggest that TE5 may be a potential therapeutic drug to treat GBM.
Collapse
Affiliation(s)
- Tao Zhang
- GuiZhou University Medical College, Guiyang 550025, Guizhou Province, China; State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, Guizhou Province, China; Natural Products Research Center of Guizhou Province, Guiyang 550014, Guizhou Province, China; Department of Neurology, Guizhou Provincial People's Hospital, Guiyang 550002, Guizhou Province, China
| | - Weidong Pan
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, Guizhou Province, China; Natural Products Research Center of Guizhou Province, Guiyang 550014, Guizhou Province, China
| | - Xin Tan
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, Guizhou Province, China; Natural Products Research Center of Guizhou Province, Guiyang 550014, Guizhou Province, China
| | - Jia Yu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, Guizhou Province, China; Natural Products Research Center of Guizhou Province, Guiyang 550014, Guizhou Province, China
| | - Sha Cheng
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, Guizhou Province, China; Natural Products Research Center of Guizhou Province, Guiyang 550014, Guizhou Province, China
| | - Shinan Wei
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, Guizhou Province, China; Natural Products Research Center of Guizhou Province, Guiyang 550014, Guizhou Province, China
| | - Kuan Fan
- Department of Neurology, Guizhou Provincial People's Hospital, Guiyang 550002, Guizhou Province, China
| | - Lu Wang
- Department of Neurology, Guizhou Provincial People's Hospital, Guiyang 550002, Guizhou Province, China
| | - Heng Luo
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, Guizhou Province, China; Natural Products Research Center of Guizhou Province, Guiyang 550014, Guizhou Province, China.
| | - Xiao Hu
- GuiZhou University Medical College, Guiyang 550025, Guizhou Province, China; Department of Neurology, Guizhou Provincial People's Hospital, Guiyang 550002, Guizhou Province, China.
| |
Collapse
|
12
|
Torres T, Adam N, Mhaouty-Kodja S, Naulé L. Reproductive function and behaviors: an update on the role of neural estrogen receptors alpha and beta. Front Endocrinol (Lausanne) 2024; 15:1408677. [PMID: 38978624 PMCID: PMC11228153 DOI: 10.3389/fendo.2024.1408677] [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: 03/28/2024] [Accepted: 05/29/2024] [Indexed: 07/10/2024] Open
Abstract
Infertility is becoming a major public health problem, with increasing frequency due to medical, environmental and societal causes. The increasingly late age of childbearing, growing exposure to endocrine disruptors and other reprotoxic products, and increasing number of medical reproductive dysfunctions (endometriosis, polycystic ovary syndrome, etc.) are among the most common causes. Fertility relies on fine-tuned control of both neuroendocrine function and reproductive behaviors, those are critically regulated by sex steroid hormones. Testosterone and estradiol exert organizational and activational effects throughout life to establish and activate the neural circuits underlying reproductive function. This regulation is mediated through estrogen receptors (ERs) and androgen receptor (AR). Estradiol acts mainly via nuclear estrogen receptors ERα and ERβ. The aim of this review is to summarize the genetic studies that have been undertaken to comprehend the specific contribution of ERα and ERβ in the neural circuits underlying the regulation of the hypothalamic-pituitary-gonadal axis and the expression of reproductive behaviors, including sexual and parental behavior. Particular emphasis will be placed on the neural role of these receptors and the underlying sex differences.
Collapse
Affiliation(s)
| | | | | | - Lydie Naulé
- Sorbonne Université, CNRS UMR8246, INSERM U1130, Neuroscience Paris Seine – Institut de Biologie Paris Seine, Paris, France
| |
Collapse
|
13
|
Tian Y, Qiao H, Zhu LQ, Man HY. Sexually dimorphic phenotypes and the role of androgen receptors in UBE3A-dependent autism spectrum disorder. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.02.592248. [PMID: 38746146 PMCID: PMC11092617 DOI: 10.1101/2024.05.02.592248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Autism spectrum disorders (ASDs) are characterized by social, communication, and behavioral challenges. UBE3A is one of the most common ASD genes. ASDs display a remarkable sex difference with a 4:1 male to female prevalence ratio; however, the underlying mechanism remains largely unknown. Using the UBE3A-overexpressing mouse model for ASD, we studied sex differences at behavioral, genetic, and molecular levels. We found that male mice with extra copies of Ube3A exhibited greater impairments in social interaction, repetitive self-grooming behavior, memory, and pain sensitivity, whereas female mice with UBE3A overexpression displayed greater olfactory defects. Social communication was impaired in both sexes, with males making more calls and females preferring complex syllables. At the molecular level, androgen receptor (AR) levels were reduced in both sexes due to enhanced degradation mediated by UBE3A. However, AR reduction significantly dysregulated AR target genes only in male, not female, UBE3A-overexpressing mice. Importantly, restoring AR levels in the brain effectively normalized the expression of AR target genes, and rescued the deficits in social preference, grooming behavior, and memory in male UBE3A-overexpressing mice, without affecting females. These findings suggest that AR and its signaling cascade play an essential role in mediating the sexually dimorphic changes in UBE3A-dependent ASD.
Collapse
Affiliation(s)
- Yuan Tian
- Department of Biology, Boston University, 5 Cummington Mall, Boston, MA 02215, USA
| | - Hui Qiao
- Department of Biology, Boston University, 5 Cummington Mall, Boston, MA 02215, USA
| | - Ling-Qiang Zhu
- Department of Pathophysiology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Heng-Ye Man
- Department of Biology, Boston University, 5 Cummington Mall, Boston, MA 02215, USA
- Department of Pharmacology, Physiology & Biophysics, Boston University School of Medicine, 72 East Concord St., Boston, MA 02118, USA
- Center for Systems Neuroscience, Boston University, 610 Commonwealth Ave, Boston, MA 02215, USA
| |
Collapse
|
14
|
Turniak-Kusy M, Studzian M, Szpakowski P, Kuchta P, Smietanka K, Mattern C, Pulaski L, Bielecki B. Testosterone Inhibits Secretion of the Pro-Inflammatory Chemokine CXCL1 from Astrocytes. Curr Issues Mol Biol 2024; 46:2105-2118. [PMID: 38534751 DOI: 10.3390/cimb46030135] [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: 07/21/2023] [Revised: 02/25/2024] [Accepted: 03/01/2024] [Indexed: 03/28/2024] Open
Abstract
Astrocytes play an important role in the regulation of the inflammatory response in the CNS, e.g., in demyelinating diseases. Since the chemokine CXCL1 is known to be secreted by astrocytes and to have a pro-inflammatory effect on immune cells in the CNS, we verified the effect of testosterone on its secretion in vitro (in the astrocytic cell line DI TNC1). Testosterone reduced the increase in CXCL1 production caused by the pro-inflammatory agent lysophosphatidylcholine and restored the basal production level of CXCL1. The androgen receptor (present and functional in the studied cell line) was strongly suggested to mediate this effect-its non-steroid ligand flutamide exerted an agonist-like effect, mimicking the activity of testosterone itself on CXCL1 secretion. This novel mechanism has important implications for the known immunomodulatory effect of testosterone and potentially other androgenic hormones. It provides a potential explanation on the molecular level and shows that astrocytes are important players in inflammatory homeostasis in the CNS and its hormonal regulation. Therefore, it suggests new directions for the development of the therapeutic intervention.
Collapse
Affiliation(s)
| | - Maciej Studzian
- Department of Oncobiology and Epigenetics, Faculty of Biology and Environmental Protection, University of Lodz, 90-237 Lodz, Poland
- Laboratory of Transcriptional Regulation, Institute of Medical Biology, Polish Academy of Sciences, 90-364 Lodz, Poland
| | - Piotr Szpakowski
- Department of Neurology and Stroke, Medical University of Lodz, 90-549 Lodz, Poland
| | - Piotr Kuchta
- Faculty of Medicine, Medical University of Lodz, 90-419 Lodz, Poland
| | - Kaja Smietanka
- Department of Neurology and Stroke, Medical University of Lodz, 90-549 Lodz, Poland
| | - Claudia Mattern
- Oceanographic Center, Nova Southeastern University, Fort Lauderdale, FL 33314, USA
- M&P Pharma AG, 6376 Emmetten, Switzerland
| | - Lukasz Pulaski
- Department of Oncobiology and Epigenetics, Faculty of Biology and Environmental Protection, University of Lodz, 90-237 Lodz, Poland
- Laboratory of Transcriptional Regulation, Institute of Medical Biology, Polish Academy of Sciences, 90-364 Lodz, Poland
| | - Bartosz Bielecki
- Department of Neurology, Laboratory of Neuroimmunology, Medical University of Lodz, 90-153 Lodz, Poland
| |
Collapse
|
15
|
Ågmo A. Androgen receptors and sociosexual behaviors in mammals: The limits of generalization. Neurosci Biobehav Rev 2024; 157:105530. [PMID: 38176634 DOI: 10.1016/j.neubiorev.2023.105530] [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: 10/18/2023] [Revised: 12/27/2023] [Accepted: 12/29/2023] [Indexed: 01/06/2024]
Abstract
Circulating testosterone is easily aromatized to estradiol and reduced to dihydrotestosterone in target tissues and elsewhere in the body. Thus, the actions of testosterone can be mediated either by the estrogen receptors, the androgen receptor or by simultaneous action at both receptors. To determine the role of androgens acting at the androgen receptor, we need to eliminate actions at the estrogen receptors. Alternatively, actions at the androgen receptor itself can be eliminated. In the present review, I will analyze the specific role of androgen receptors in male and female sexual behavior as well as in aggression. Some comments about androgen receptors and social recognition are also made. It will be shown that there are important differences between species, even between strains within a species, concerning the actions of the androgen receptor on the behaviors mentioned. This fact makes generalizations from one species to another or from one strain to another very risky. The existence of important species differences is often ignored, leading to many misunderstandings and much confusion.
Collapse
Affiliation(s)
- Anders Ågmo
- Department of Psychology, University of Tromsø, Norway.
| |
Collapse
|
16
|
Nguyen Hoai B, Hoang L, Nguyen Cao T, Pham Minh Q, A Jannini E. Testosterone and aging male, a perspective from a developing country. Aging Male 2023; 26:2223712. [PMID: 37335039 DOI: 10.1080/13685538.2023.2223712] [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: 04/16/2023] [Revised: 05/19/2023] [Accepted: 06/06/2023] [Indexed: 06/21/2023] Open
Abstract
PURPOSE Hypogonadism is associated with a wide range of physical and psychological symptoms that can affect the overall health of men. However, in a developing country, there are several imposing challenges in the diagnosis and treatment of hypogonadism, including a lack of awareness and understanding of the condition among healthcare providers and patients, limited resources and the high cost of treatment. This review aimed to examine the potential benefits and risks of testosterone replacement therapy (TRT) and provides a perspective of a developing country on the topic. MATERIALS AND METHODS A comprehensive literature review was conducted to gather relevant information on the impact of testosterone deficiency on ageing males and the effectiveness of TRT for treating hypogonadism. Published peer-reviewed articles were analyzed to evaluate the benefits and risks of TRT. Additionally, the unique challenges faced in the diagnosis and treatment of hypogonadism in a developing country were considered. RESULTS Testosterone replacement therapy has been shown to be an effective treatment for hypogonadism, particularly in symptomatic men with low testosterone levels. It offers potential benefits such as improvements in symptoms and overall quality of life. However, there are associated risks and side effects that need to be considered. In a developing country, challenges such as limited awareness and understanding of hypogonadism, resource constraints, and high treatment costs pose additional barriers to accessing TRT and comprehensive care. CONCLUSION In conclusion, TRT holds promise as a treatment for hypogonadism, but its implementation and accessibility face significant challenges in a developing country. Addressing these challenges, including raising awareness, allocating resources, and finding cost-effective solutions, is crucial for ensuring that men with hypogonadism in such settings receive appropriate diagnosis and treatment. Further research and efforts are needed to improve the management of hypogonadism in developing countries and optimize the potential benefits of TRT for affected individuals.
Collapse
Affiliation(s)
- Bac Nguyen Hoai
- Department of Andrology and Sexual Medicine, Hanoi Medical University's Hospital, Hanoi, Vietnam
| | - Long Hoang
- Department of Urology, Hanoi Medical University's Hospital, Hanoi, Vietnam
| | - Thang Nguyen Cao
- Department of Andrology and Sexual Medicine, Hanoi Medical University's Hospital, Hanoi, Vietnam
| | - Quan Pham Minh
- Department of Andrology and Sexual Medicine, Hanoi Medical University's Hospital, Hanoi, Vietnam
| | - Emmanuele A Jannini
- Chair of Endocrinology and Sexual Medicine (ENDOSEX), University of Rome Tor Vergata, Rome, Italy
| |
Collapse
|
17
|
Ventura-Aquino E, Ågmo A. The elusive concept of sexual motivation: can it be anchored in the nervous system? Front Neurosci 2023; 17:1285810. [PMID: 38046659 PMCID: PMC10691110 DOI: 10.3389/fnins.2023.1285810] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 10/16/2023] [Indexed: 12/05/2023] Open
Abstract
Sexual motivation is an abstract concept referring to the mechanisms determining the responsivity to sexually relevant stimuli. This responsivity determines the likelihood of producing a sexual response and the intensity of that response. Both responsivity to stimuli and the likelihood of making a response as well as the intensity of response are characteristics of an individual. Therefore, we need to assume that the concept of sexual motivation materializes in physiological mechanisms within the individual. The aim of the present communication is to analyze the requisites for the endeavor to materialize sexual motivation. The first requisite is to provide an operational definition, making the concept quantifiable. We show that parameters of copulatory behavior are inappropriate. We argue that the intensity of sexual approach behaviors provides the best estimate of sexual motivation in non-human animals, whereas the magnitude of genital responses is an exquisite indicator of human sexual motivation. Having assured how to quantify sexual motivation, we can then proceed to the search for physiological or neurobiological underpinnings. In fact, sexual motivation only manifests itself in animals exposed to appropriate amounts of gonadal hormones. In female rats, the estrogen receptor α in the ventrolateral part of the ventromedial nucleus of the hypothalamus is necessary for the expression of sexual approach behaviors. In male rats, androgen receptors within the medial preoptic area are crucial. Thus, in rats sexual motivation can be localized to specific brain structures, and even to specific cells within these structures. In humans, it is not even known if sexual motivation is materialized in the brain or in peripheral structures. Substantial efforts have been made to determine the relationship between the activity of neurotransmitters and the intensity of sexual motivation, particularly in rodents. The results of this effort have been meager. Likewise, efforts of finding drugs to stimulate sexual motivation, particularly in women complaining of low sexual desire, have produced dismal results. In sum, it appears that the abstract concept of sexual motivation can be reliably quantified, and the neurobiological bases can be described in non-human animals. In humans, objective quantification is feasible, but the neurobiological substrate remains enigmatic.
Collapse
Affiliation(s)
- Elisa Ventura-Aquino
- Escuela Nacional de Estudios Superiores, Unidad Juriquilla, UNAM, Juriquilla, Mexico
| | - Anders Ågmo
- Department of Psychology, University of Tromsø, Tromsø, Norway
| |
Collapse
|
18
|
Bey C, Abdelghaffar W, Ach T, Haloui N, Bouzid MI, Rafrafi R. A case of acute psychosis triggered by anabolic steroid abuse (trenbolone). Therapie 2023; 78:775-778. [PMID: 37100702 DOI: 10.1016/j.therap.2023.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 03/10/2023] [Accepted: 04/06/2023] [Indexed: 04/28/2023]
Affiliation(s)
- Cyrine Bey
- Department of Psychiatry, University Hospital of Mongi Slim, 2070 La Marsa, Tunisia; University of Sousse, Faculty of Medicine of Sousse, 4051 Sousse, Tunisia.
| | - Wafa Abdelghaffar
- Department of Psychiatry, University Hospital of Mongi Slim, 2070 La Marsa, Tunisia; University of Tunis El Manar, Faculty of Medicine of Tunis, 1029 Tunis, Tunisia
| | - Taieb Ach
- University of Sousse, Faculty of Medicine of Sousse, 4051 Sousse, Tunisia; Department of Endocrinology, University Hospital of Farhat Hached, 4051 Sousse, Tunisia
| | - Nadia Haloui
- Department of Psychiatry, University Hospital of Mongi Slim, 2070 La Marsa, Tunisia; University of Tunis El Manar, Faculty of Medicine of Tunis, 1029 Tunis, Tunisia
| | - Mariem Inès Bouzid
- Department of Psychiatry, University Hospital of Mongi Slim, 2070 La Marsa, Tunisia; University of Tunis El Manar, Faculty of Medicine of Tunis, 1029 Tunis, Tunisia
| | - Rym Rafrafi
- Department of Psychiatry, University Hospital of Mongi Slim, 2070 La Marsa, Tunisia; University of Tunis El Manar, Faculty of Medicine of Tunis, 1029 Tunis, Tunisia
| |
Collapse
|
19
|
Nowakowska MK, Ortega RM, Wehner MR, Nead KT. Association of Second-generation Antiandrogens With Cognitive and Functional Toxic Effects in Randomized Clinical Trials: A Systematic Review and Meta-analysis. JAMA Oncol 2023; 9:930-937. [PMID: 37227736 PMCID: PMC10214180 DOI: 10.1001/jamaoncol.2023.0998] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 02/13/2023] [Indexed: 05/26/2023]
Abstract
Importance The use of second-generation antiandrogens (AAs) in the treatment of prostate cancer is increasing. Retrospective evidence suggests an association between second-generation AAs and adverse cognitive and functional outcomes, but further data from prospective trials are needed. Objective To examine whether evidence from randomized clinical trials (RCTs) in prostate cancer supports an association between second-generation AAs and cognitive or functional toxic effects. Data Sources PubMed, EMBASE, and Scopus (inception to September 12, 2022). Study Selection Randomized clinical trials of second-generation AAs (abiraterone, apalutamide, darolutamide, or enzalutamide) among individuals with prostate cancer that reported cognitive toxic effects, asthenic toxic effects (eg, fatigue, weakness), or falls were evaluated. Data Extraction and Synthesis Study screening, data abstraction, and bias assessment were completed independently by 2 reviewers following the Preferred Reporting Items for Systematic Reviews and Meta-analyses and Enhancing the Quality and Transparency of Health Research reporting guidelines. Tabular counts for all-grade toxic effects were determined to test the hypothesis formulated before data collection. Main Outcomes and Measures Risk ratios (RRs) and SEs were calculated for cognitive toxic effects, asthenic toxic effects, and falls. Because fatigue was the asthenic toxic effect extracted from all studies, data on fatigue are specified in the results. Meta-analysis and meta-regression were used to generate summary statistics. Results The systematic review included 12 studies comprising 13 524 participants. Included studies had a low risk of bias. An increased risk of cognitive toxic effects (RR, 2.10; 95% CI, 1.30-3.38; P = .002) and fatigue (RR, 1.34; 95% CI, 1.16-1.54; P < .001) was noted among individuals treated with second-generation AAs vs those in the control arms. The findings were consistent in studies that included traditional hormone therapy in both treatment arms for cognitive toxic effects (RR, 1.77; 95% CI, 1.12-2.79; P = .01) and fatigue (RR, 1.32; 95% CI, 1.10-1.58; P = .003). Meta-regression supported that, across studies, increased age was associated with a greater risk of fatigue with second-generation AAs (coefficient, 0.75; 95% CI, 0.04-0.12; P < .001). In addition, the use of second-generation AAs was associated with an increased risk of falls (RR, 1.87; 95% CI, 1.27-2.75; P = .001). Conclusions and Relevance The findings of this systematic review and meta-analysis suggest that second-generation AAs carry an increased risk of cognitive and functional toxic effects, including when added to traditional forms of hormone therapy.
Collapse
Affiliation(s)
| | | | - Mackenzie R. Wehner
- Department of Dermatology, The University of Texas MD Anderson Cancer Center, Houston
- Department of Health Services Research, The University of Texas MD Anderson Cancer Center, Houston
| | - Kevin T. Nead
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston
| |
Collapse
|
20
|
Ducroq S, Duplus E, Penalva-Mousset L, Trivelloni F, L’honoré A, Chabat-Courrède C, Nemazanyy I, Grange-Messent V, Petropoulos I, Mhaouty-Kodja S. Behavior, Neural Structure, and Metabolism in Adult Male Mice Exposed to Environmentally Relevant Doses of Di(2-ethylhexyl) Phthalate Alone or in a Phthalate Mixture. ENVIRONMENTAL HEALTH PERSPECTIVES 2023; 131:77008. [PMID: 37458746 PMCID: PMC10351581 DOI: 10.1289/ehp11514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 05/12/2023] [Accepted: 06/23/2023] [Indexed: 07/20/2023]
Abstract
BACKGROUND We have previously shown that chronic exposure of adult male mice to low doses of di(2-ethylhexyl) phthalate (DEHP) altered male sexual behavior and induced down-regulation of the androgen receptor (AR) in the neural circuitry controlling this behavior. OBJECTIVES The cellular mechanisms induced by chronic exposure of adult male mice to low doses of DEHP alone or in an environmental phthalate mixture were studied. METHODS Two-month-old C57BL/6J males were exposed orally for 8 wk to DEHP alone (0, 5, or 50μg/kg/d) or to DEHP (50μg/kg/d) in a phthalate mixture. Behavior, dendritic density per 50-μm length, pre-/postsynaptic markers, synapse ultrastructure, and bioenergetic activity were analyzed. RESULTS Mice exposed to DEHP either alone or in a phthalate mixture differed in mating, emission of ultrasonic vocalizations, and the ability to attract receptive females in urinary preference tests from control mice. Analyses in the medial preoptic area, the key hypothalamic region involved in male sexual behavior, showed lower dendritic spine density and protein levels of glutamate receptors and differences in other postsynaptic components and presynaptic markers between the treated groups. Ultrastructural observation of dendritic synapses by electron microscopy showed comparable morphology between the treated groups. Metabolic analyses highlighted differences in hypothalamic metabolites of males exposed to DEHP alone or in a phthalate mixture compared to control mice. These differences included lower tryptophan and higher NAD+ levels, respectively, a precursor and end product of the kynurenine pathway of tryptophan metabolism. The protein amounts of the xenobiotic aryl hydrocarbon receptor, one of the targets of this metabolic pathway and known negative regulator of the AR, were higher in the medial preoptic area of exposed male mice. DISCUSSION Differences in behavior of male mice exposed to environmental doses of phthalates were associated with differences in neural structure and metabolism, with possibly a key role of the kynurenine pathway of tryptophan metabolism in the effects mediated by these substances. https://doi.org/10.1289/EHP11514.
Collapse
Affiliation(s)
- Suzanne Ducroq
- Sorbonne Université, CNRS UMR 8246, Inserm U1130, Neuroscience Paris Seine – Institut de Biologie Paris Seine, 75005 Paris, France
| | - Eric Duplus
- Sorbonne Université, CNRS UMR 8256, Inserm ERL1164, Biological Adaptation and Ageing – Institut de Biologie Paris-Seine, 75005 Paris, France
| | - Lucille Penalva-Mousset
- Sorbonne Université, CNRS UMR 8256, Inserm ERL1164, Biological Adaptation and Ageing – Institut de Biologie Paris-Seine, 75005 Paris, France
| | - Francesca Trivelloni
- Sorbonne Université, CNRS UMR 8246, Inserm U1130, Neuroscience Paris Seine – Institut de Biologie Paris Seine, 75005 Paris, France
| | - Aurore L’honoré
- Sorbonne Université, CNRS UMR 8256, Inserm ERL1164, Biological Adaptation and Ageing – Institut de Biologie Paris-Seine, 75005 Paris, France
| | - Caroline Chabat-Courrède
- Sorbonne Université, CNRS UMR 8256, Inserm ERL1164, Biological Adaptation and Ageing – Institut de Biologie Paris-Seine, 75005 Paris, France
| | - Ivan Nemazanyy
- Platform for Metabolic Analyses, Structure Fédérative de Recherche Necker, Inserm US24/CNRS UMS 3633, Paris 75015, France
| | - Valérie Grange-Messent
- Sorbonne Université, CNRS UMR 8246, Inserm U1130, Neuroscience Paris Seine – Institut de Biologie Paris Seine, 75005 Paris, France
| | - Isabelle Petropoulos
- Sorbonne Université, CNRS UMR 8256, Inserm ERL1164, Biological Adaptation and Ageing – Institut de Biologie Paris-Seine, 75005 Paris, France
| | - Sakina Mhaouty-Kodja
- Sorbonne Université, CNRS UMR 8246, Inserm U1130, Neuroscience Paris Seine – Institut de Biologie Paris Seine, 75005 Paris, France
| |
Collapse
|
21
|
Meng X, Su S, Wei X, Wang S, Guo T, Li J, Song H, Wang M, Wang Z. Exposure to bisphenol A alternatives bisphenol AF and fluorene-9-bisphenol induces gonadal injuries in male zebrafish. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 253:114634. [PMID: 36801538 DOI: 10.1016/j.ecoenv.2023.114634] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 02/05/2023] [Accepted: 02/08/2023] [Indexed: 06/18/2023]
Abstract
Bisphenol A (BPA), present in many household products, can damage the male reproductive system. Accordingly, we summarized urine samples from 6921 human in National Health and Nutrition Examination Survey and found urinary BPA levels were inversely linked with blood testosterone in the children group. Currently, BPA replacements, such as fluorene-9-bisphenol (BHPF) and Bisphenol AF (BPAF), have been introduced to produce "BPA-free" products. Here we demonstrated that BPAF and BHPF could induce delayed gonadal migration and reduce the number of progenitors of germ cell lineage in zebrafish larvae. A close receptor analysis study reveals that BHPF and BPAF can strongly bind to androgen receptors, leading to the downregulation of meiosis-related genes and the overexpression of inflammatory markers. Furthermore, BPAF and BPHF can induce activation of the gonadal axis via negative feedback, leading to the hypersecretion of some upstream hormones and an increase in the expression of upstream hormone receptors. Our findings call for further research on the toxicological effects of BHPF and BPAF on human health and recommend that BPA replacements be investigated for anti-estrogenic action.
Collapse
Affiliation(s)
- Xiangyu Meng
- Department of Urology, First Affiliated Hospital of Nanjing Medical University, No. 300, Guangzhou Street, Nanjing, Jiangsu Province 210029, China
| | - Shifeng Su
- Department of Urology, First Affiliated Hospital of Nanjing Medical University, No. 300, Guangzhou Street, Nanjing, Jiangsu Province 210029, China
| | - Xiyi Wei
- Department of Urology, First Affiliated Hospital of Nanjing Medical University, No. 300, Guangzhou Street, Nanjing, Jiangsu Province 210029, China
| | - Shangqian Wang
- Department of Urology, First Affiliated Hospital of Nanjing Medical University, No. 300, Guangzhou Street, Nanjing, Jiangsu Province 210029, China
| | - Tao Guo
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Junjian Li
- College of Life and Environmental Science, Wenzhou University, Wenzhou, Zhejiang Province, China
| | - Huaidong Song
- The Core Laboratory in Medical Center of Clinical Research, Department of Molecular Diagnostics & Endocrinology, Shanghai Ninth People's Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.
| | - Mengjing Wang
- State Key Laboratory of Marine Pollution, City University of Hong Kong, Kowloon 999077, Hong Kong, China.
| | - Zengjun Wang
- Department of Urology, First Affiliated Hospital of Nanjing Medical University, No. 300, Guangzhou Street, Nanjing, Jiangsu Province 210029, China.
| |
Collapse
|
22
|
Donaldson NM, Prescott M, Ruddenklau A, Campbell RE, Desroziers E. Maternal androgen excess significantly impairs sexual behavior in male and female mouse offspring: Perspective for a biological origin of sexual dysfunction in PCOS. Front Endocrinol (Lausanne) 2023; 14:1116482. [PMID: 36875467 PMCID: PMC9975579 DOI: 10.3389/fendo.2023.1116482] [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: 12/05/2022] [Accepted: 01/31/2023] [Indexed: 02/17/2023] Open
Abstract
INTRODUCTION Polycystic ovary syndrome (PCOS) is the most common infertility disorder worldwide, typically characterised by high circulating androgen levels, oligo- or anovulation, and polycystic ovarian morphology. Sexual dysfunction, including decreased sexual desire and increased sexual dissatisfaction, is also reported by women with PCOS. The origins of these sexual difficulties remain largely unidentified. To investigate potential biological origins of sexual dysfunction in PCOS patients, we asked whether the well-characterized, prenatally androgenized (PNA) mouse model of PCOS exhibits modified sex behaviours and whether central brain circuits associated with female sex behaviour are differentially regulated. As a male equivalent of PCOS is reported in the brothers of women with PCOS, we also investigated the impact of maternal androgen excess on the sex behaviour of male siblings. METHODS Adult male and female offspring of dams exposed to dihydrotestosterone (PNAM/PNAF) or an oil vehicle (VEH) from gestational days 16 to 18 were tested for a suite of sex-specific behaviours. RESULTS PNAM showed a reduction in their mounting capabilities, however, most of PNAM where able to reach ejaculation by the end of the test similar to the VEH control males. In contrast, PNAF exhibited a significant impairment in the female-typical sexual behaviour, lordosis. Interestingly, while neuronal activation was largely similar between PNAF and VEH females, impaired lordosis behaviour in PNAF was unexpectedly associated with decreased neuronal activation in the dorsomedial hypothalamic nucleus (DMH). CONCLUSION Taken together, these data link prenatal androgen exposure that drives a PCOS-like phenotype with altered sexual behaviours in both sexes.
Collapse
|
23
|
Hauger RL, Saelzler UG, Pagadala MS, Panizzon MS. The role of testosterone, the androgen receptor, and hypothalamic-pituitary-gonadal axis in depression in ageing Men. Rev Endocr Metab Disord 2022; 23:1259-1273. [PMID: 36418656 PMCID: PMC9789012 DOI: 10.1007/s11154-022-09767-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/20/2022] [Indexed: 11/25/2022]
Abstract
Considerable research has shown that testosterone regulates many physiological systems, modulates clinical disorders, and contributes to health outcome. However, studies on the interaction of testosterone levels with depression and the antidepressant effect of testosterone replacement therapy in hypogonadal men with depression have been inconclusive. Current findings indicate that low circulating levels of total testosterone meeting stringent clinical criteria for hypogonadism and testosterone deficiency induced by androgen deprivation therapy are associated with increased risk for depression and current depressive symptoms. The benefits of testosterone replacement therapy in men with major depressive disorder and low testosterone levels in the clinically defined hypogonadal range remain uncertain and require further investigation. Important considerations going forward are that major depressive disorder is a heterogeneous phenotype with depressed individuals differing in inherited polygenic determinants, onset and clinical course, symptom complexes, and comorbidities that contribute to potential multifactorial differences in pathophysiology. Furthermore, polygenic mechanisms are likely to be critical to the biological heterogeneity that influences testosterone-depression interactions. A genetically informed precision medicine approach using genes regulating testosterone levels and androgen receptor sensitivity will likely be essential in gaining critical insight into the role of testosterone in depression.
Collapse
Affiliation(s)
- Richard L Hauger
- Center of Excellence for Stress and Mental Health (CESAMH), VA San Diego Healthcare System, San Diego, CA, USA.
- Center for Behavior Genetics of Aging, Department of Psychiatry, School of Medicine, University of California San Diego, La Jolla, CA, USA.
| | - Ursula G Saelzler
- Center for Behavior Genetics of Aging, Department of Psychiatry, School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Meghana S Pagadala
- Medical Scientist Training Program, School of Medicine, University of California San Diego, La Jolla, CA, USA
- Biomedical Science Program, School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Matthew S Panizzon
- Center of Excellence for Stress and Mental Health (CESAMH), VA San Diego Healthcare System, San Diego, CA, USA
- Center for Behavior Genetics of Aging, Department of Psychiatry, School of Medicine, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
24
|
Liu J, Liu S, Hao L, Liu F, Mu S, Wang T. Uncovering the mechanism of Radix Paeoniae Alba in the treatment of restless legs syndrome based on network pharmacology and molecular docking. Medicine (Baltimore) 2022; 101:e31791. [PMID: 36401463 PMCID: PMC9678500 DOI: 10.1097/md.0000000000031791] [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] [Indexed: 12/03/2022] Open
Abstract
Restless legs syndrome (RLS) is a neurological motor disorder with a high prevalence. The treatment efficacy of RLS is unsatisfactory. Radix Paeoniae Alba (RPA) can effectively treat RLS symptoms such as the discomfort of the legs. RPA has great potential for the development of new medications for RLS. Hence, we explored the mechanism of RPA in the treatment of RLS using network pharmacology and molecular docking. The active components and targets of RPA were obtained from the Traditional Chinese Medicine System Pharmacology database and analysis platform and PharmMapper platform. The RLS-related targets were found in GeneCards, OMIM, DrugBank, and DisGeNET databases. The overlapping targets of RPA and RLS were then collected. The "active components-overlapping targets" network was built, and network topology analysis was performed. Furthermore, Cytoscape 3.9.1 software was used to screen the key components of RPA in the treatment of RLS. Protein-protein interaction was performed using the Search Tool for the Retrieval of Interacting Genes. The gene ontology functions and Kyoto Encyclopedia of Genes and Genomes signaling pathways were analyzed using ClusterProfiler, PathView, and other R packages to reveal the main mechanism of RPA in treating RLS. Component and protein structures were downloaded from the Traditional Chinese Medicine System Pharmacology and Protein Data Bank databases, respectively. The AutoDock 4.2.6 software was used for molecular docking. A total of 12 active components and 109 targets of RPA, as well as 2387 RLS-related targets, were collected. Following that, 47 overlapping targets were obtained. Furthermore, 5 key components and 12 core targets were screened. The results of gene ontology functions were as follows: 2368 biological processes, 264 molecular functions, and 164 cellular components. A total of 207 Kyoto Encyclopedia of Genes and Genomes signaling pathways were obtained, including the lipid and atherosclerosis pathway, the endocrine resistance pathway, the prolactin signaling pathway, and the IL-17 signaling pathway. The components and the core targets completed molecular docking stably. RPA has multi-component, multi-target, and multi-pathway characteristics in treating RLS, which could provide a basis for future research and improve clinical efficacy.
Collapse
Affiliation(s)
- Jun Liu
- Department of Gastroenterology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Suxian Liu
- Department of Gastroenterology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Liansheng Hao
- Department 2 of Bone Trauma, Liaocheng Hospital of Traditional Chinese Medicine, Liaocheng, China
| | - Fangfang Liu
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shengkai Mu
- Department 2 of Bone Trauma, Liaocheng Hospital of Traditional Chinese Medicine, Liaocheng, China
| | - Tengteng Wang
- Department of Acupuncture, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- * Correspondence: Tengteng Wang, Department of Acupuncture, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China (e-mail: )
| |
Collapse
|
25
|
Rosato E, Sciarra F, Anastasiadou E, Lenzi A, Venneri MA. Revisiting the physiological role of androgens in women. Expert Rev Endocrinol Metab 2022; 17:547-561. [PMID: 36352537 DOI: 10.1080/17446651.2022.2144834] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 11/03/2022] [Indexed: 11/09/2022]
Abstract
INTRODUCTION Extensive research underlines the critical functions of androgens in females. Nevertheless, the precise mechanisms of their action are poorly understood. Here, we review the existing literature regarding the physiological role of androgens in women throughout life. AREAS COVERED Several studies show that androgen receptors (ARs) are broadly expressed in numerous female tissues. They are essential for many physiological processes, including reproductive, sexual, cardiovascular, bone, muscle, and brain health. They are also involved in adipose tissue and liver function. Androgen levels change with the menstrual cycle and decrease in the first decades of life, independently of menopause. EXPERT OPINION To date, studies are limited by including small numbers of women, the difficulty of dosing androgens, and their cyclical variations. In particular, whether androgens play any significant role in regulating the establishment of pregnancy is poorly understood. The neural functions of ARs have also been investigated less thoroughly, although it is expressed at high levels in brain structures. Moreover, the mechanism underlying the decline of dehydroepiandrosterone (DHEA) and dehydroepiandrosterone sulfate (DHEAS) with age is unclear. Other factors, including estrogen's effect on adrenal androgen production, reciprocal regulation of ARs, and non-classical effects of androgens, remain to be determined.
Collapse
Affiliation(s)
- Elena Rosato
- Department of Experimental Medicine, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Francesca Sciarra
- Department of Experimental Medicine, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Eleni Anastasiadou
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Andrea Lenzi
- Department of Experimental Medicine, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Mary Anna Venneri
- Department of Experimental Medicine, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| |
Collapse
|
26
|
Sengupta M, Pluciennik A, Merry DE. The role of ubiquitination in spinal and bulbar muscular atrophy. Front Mol Neurosci 2022; 15:1020143. [PMID: 36277484 PMCID: PMC9583669 DOI: 10.3389/fnmol.2022.1020143] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
Spinal and bulbar muscular atrophy (SBMA) is a neurodegenerative and neuromuscular genetic disease caused by the expansion of a polyglutamine-encoding CAG tract in the androgen receptor (AR) gene. The AR is an important transcriptional regulator of the nuclear hormone receptor superfamily; its levels are regulated in many ways including by ubiquitin-dependent degradation. Ubiquitination is a post-translational modification (PTM) which plays a key role in both AR transcriptional activity and its degradation. Moreover, the ubiquitin-proteasome system (UPS) is a fundamental component of cellular functioning and has been implicated in diseases of protein misfolding and aggregation, including polyglutamine (polyQ) repeat expansion diseases such as Huntington's disease and SBMA. In this review, we discuss the details of the UPS system, its functions and regulation, and the role of AR ubiquitination and UPS components in SBMA. We also discuss aspects of the UPS that may be manipulated for therapeutic effect in SBMA.
Collapse
Affiliation(s)
| | | | - Diane E. Merry
- Department of Biochemistry and Molecular Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, United States
| |
Collapse
|
27
|
Proaño B, Casani-Cubel J, Benlloch M, Rodriguez-Mateos A, Navarro-Illana E, Lajara-Romance JM, de la Rubia Ortí JE. Is Dutasteride a Therapeutic Alternative for Amyotrophic Lateral Sclerosis? Biomedicines 2022; 10:biomedicines10092084. [PMID: 36140184 PMCID: PMC9495995 DOI: 10.3390/biomedicines10092084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 08/17/2022] [Accepted: 08/23/2022] [Indexed: 11/24/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease that is characterized by the loss of upper and lower motor neurons (MNs) in the cerebral cortex, brainstem and spinal cord, with consequent weakness, atrophy and the progressive paralysis of all muscles. There is currently no medical cure, and riluzole and edaravone are the only two known approved drugs for treating this condition. However, they have limited efficacy, and hence there is a need to find new molecules. Dutasteride, a dual inhibitor of type 1 and type 2 5α-reductase (5AR) enzymes, the therapeutic purposes of which, to date, are the treatment of benign prostatic hyperplasia and androgenic alopecia, shows great anti-ALS properties by the molecular-topology methodology. Based on this evidence, this review aims to assess the effects of dutasteride on testosterone (T), progesterone (PROG) and 17β-estradiol (17BE) as a therapeutic alternative for the clinical improvement of ALS, based on the hormonal, metabolic and molecular pathways related to the pathogenesis of the disease. According to the evidence found, dutasteride shows great neuroprotective, antioxidant and anti-inflammatory effects. It also appears effective against glutamate toxicity, and it is capable of restoring altered dopamine activity (DA). These effects are achieved both directly and through steroid hormones. Therefore, dutasteride seems to be a promising molecule for the treatment of ALS, although clinical studies are required for confirmation.
Collapse
Affiliation(s)
- Belén Proaño
- Doctoral Degree School, Health Sciences, Catholic University of Valencia San Vicente Mártir, 46001 Valencia, Spain
| | - Julia Casani-Cubel
- School of Medicine and Health Sciences, Catholic University San Vicente Mártir, 46001 Valencia, Spain
- Correspondence: (J.C.-C.); (M.B.)
| | - María Benlloch
- Department Nursing, Catholic University San Vicente Mártir, 46001 Valencia, Spain
- Correspondence: (J.C.-C.); (M.B.)
| | - Ana Rodriguez-Mateos
- Department of Nutritional Sciences, King’s College London, Franklin Wilkins Building, London SE1 9NH, UK
| | | | | | | |
Collapse
|
28
|
Oliveira JM, Oliveira IM, Sleiman HK, Dal Forno GO, Romano MA, Romano RM. Consumption of soy isoflavones during the prepubertal phase delays puberty and causes hypergonadotropic hypogonadism with disruption of hypothalamic-pituitary gonadotropins regulation in male rats. Toxicol Lett 2022; 369:1-11. [PMID: 35963426 DOI: 10.1016/j.toxlet.2022.08.004] [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: 01/27/2022] [Revised: 07/17/2022] [Accepted: 08/08/2022] [Indexed: 10/15/2022]
Abstract
Isoflavones are phytoestrogens with recognized estrogenic activity but may also affect testosterone, corticosterone and thyroid hormone levels in experimental models. However, the molecular mechanisms involved in these alterations are still unclear. Isoflavones are present in soy-based infant formula, in breast milk after the consumption of soy by the mother and are widely used for the preparation of beverages consumed by toddlers and teenagers. In this sense, we proposed to investigate the effects of soy isoflavone exposure during the prepubertal period, a recognized window of sensitivity for endocrine disruption, over the hypothalamic-pituitary-testicular (HPT) axis. For this, 42 3-week-old male Wistar rats were exposed to 0.5, 5 or 50 mg of soy isoflavones/kg from postnatal day (PND) 23 to PND60. We evaluated body growth, age at puberty, serum concentrations of LH, FSH, testosterone and estradiol, and the expression of the transcripts (mRNA) of genes encoding key genes controlling the hypothalamic-pituitary-testicular (HPT) axis. In the hypothalamus, we observed an increase in Esr1 mRNA expression (0.5 and 5 mg). In the pituitary, we observed an increase in Gnrhr mRNA expression (50 mg), a reduction in Lhb mRNA expression (0.5 mg), and a reduction in Ar mRNA expression. In the testis, we observed an increase in Lhcgr mRNA expression (50 mg) and a reduction in Star mRNA expression (0.5 and 5 mg). The serum levels of LH (5 and 50 mg) and FSH (0.5 mg) were increased, while testosterone and estradiol were reduced. Puberty was delayed in all groups. Taken together, these results suggest that prepubertal consumption of relevant levels of soy isoflavones disrupts the HPT axis, causing hypergonadotropic hypogonadism and altered expression levels of key genes regulating the axis.
Collapse
Affiliation(s)
- Jeane Maria Oliveira
- Laboratory of Reproductive Toxicology, Department of Medicine, State University of Centro-Oeste (UNICENTRO), Rua Simeão Camargo Varela de Sa, 03, Zip-Code 85040-080, Parana, Brazil.
| | - Isabela Medeiros Oliveira
- Laboratory of Reproductive Toxicology, Department of Medicine, State University of Centro-Oeste (UNICENTRO), Rua Simeão Camargo Varela de Sa, 03, Zip-Code 85040-080, Parana, Brazil.
| | - Hanan Khaled Sleiman
- Laboratory of Reproductive Toxicology, Department of Medicine, State University of Centro-Oeste (UNICENTRO), Rua Simeão Camargo Varela de Sa, 03, Zip-Code 85040-080, Parana, Brazil.
| | - Gonzalo Ogliari Dal Forno
- Laboratory of Reproductive Toxicology, Department of Medicine, State University of Centro-Oeste (UNICENTRO), Rua Simeão Camargo Varela de Sa, 03, Zip-Code 85040-080, Parana, Brazil.
| | - Marco Aurelio Romano
- Laboratory of Reproductive Toxicology, Department of Medicine, State University of Centro-Oeste (UNICENTRO), Rua Simeão Camargo Varela de Sa, 03, Zip-Code 85040-080, Parana, Brazil.
| | - Renata Marino Romano
- Laboratory of Reproductive Toxicology, Department of Medicine, State University of Centro-Oeste (UNICENTRO), Rua Simeão Camargo Varela de Sa, 03, Zip-Code 85040-080, Parana, Brazil.
| |
Collapse
|
29
|
Sex Steroid Receptors in Polycystic Ovary Syndrome and Endometriosis: Insights from Laboratory Studies to Clinical Trials. Biomedicines 2022; 10:biomedicines10071705. [PMID: 35885010 PMCID: PMC9312843 DOI: 10.3390/biomedicines10071705] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 07/12/2022] [Accepted: 07/13/2022] [Indexed: 12/13/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) and endometriosis are reproductive disorders that may cause infertility. The pathology of both diseases has been suggested to be associated with sex steroid hormone receptors, including oestrogen receptors (ER), progesterone receptors (PRs) and androgen receptors (ARs). Therefore, with this review, we aim to provide an update on the available knowledge of these receptors and how their interactions contribute to the pathogenesis of PCOS and endometriosis. One of the main PCOS-related medical conditions is abnormal folliculogenesis, which is associated with the downregulation of ER and AR expression in the ovaries. In addition, metabolic disorders in PCOS are caused by dysregulation of sex steroid hormone receptor expression. Furthermore, endometriosis is related to the upregulation of ER and the downregulation of PR expression. These receptors may serve as therapeutic targets for the treatment of PCOS-related disorders and endometriosis, considering their pathophysiological roles. Receptor agonists may be applied to increase the expression of a specific receptor and treat endometriosis or metabolic disorders. In contrast, receptor antagonist functions to reduce receptor expression and can be used to treat endometriosis and induce ovulation. Understanding PCOS and the pathological roles of endometriosis sex steroid receptors is crucial for developing potential therapeutic strategies to treat infertility in both conditions. Therefore, research should be continued to fill the knowledge gap regarding the subject.
Collapse
|
30
|
LaDage LD. Seasonal variation in gonadal hormones, spatial cognition, and hippocampal attributes: More questions than answers. Horm Behav 2022; 141:105151. [PMID: 35299119 DOI: 10.1016/j.yhbeh.2022.105151] [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: 10/01/2021] [Revised: 02/25/2022] [Accepted: 02/27/2022] [Indexed: 11/04/2022]
Abstract
A large body of research has been dedicated to understanding the factors that modulate spatial cognition and attributes of the hippocampus, a highly plastic brain region that underlies spatial processing abilities. Variation in gonadal hormones impacts spatial memory and hippocampal attributes in vertebrates, although the direction of the effect has not been entirely consistent. To add complexity, individuals in the field must optimize fitness by coordinating activities with the appropriate environmental cues, and many of these behaviors are correlated tightly with seasonal variation in gonadal hormone release. As such, it remains unclear if the relationship among systemic gonadal hormones, spatial cognition, and the hippocampus also exhibits seasonal variation. This review presents an overview of the relationship among gonadal hormones, the hippocampus, and spatial cognition, and how the seasonal release of gonadal hormones correlates with seasonal variation in spatial cognition and hippocampal attributes. Additionally, this review presents other neuroendocrine mechanisms that may be involved in modulating the relationship among seasonality, gonadal hormone release, and the hippocampus and spatial cognition, including seasonal rhythms of steroid hormone binding globulins, neurosteroids, sex steroid hormone receptor expression, and hormone interactions. Here, endocrinology, ecology, and behavioral neuroscience are brought together to present an overview of the research demonstrating the mechanistic effects of systemic gonadal hormones on spatial cognition and the hippocampus, while, at a functional level, superimposing seasonal effects to examine ecologically-relevant circannual changes in gonadal hormones and spatial behaviors.
Collapse
Affiliation(s)
- Lara D LaDage
- Penn State Altoona, Division of Mathematics & Natural Sciences, 3000 Ivyside Dr., Altoona, PA 16601, USA.
| |
Collapse
|
31
|
Ahmadpour D, Mhaouty-Kodja S, Grange-Messent V. Effects and underlying cellular pathway involved in the impairment of the neurovascular unit following exposure of adult male mice to low doses of di(2-ethylhexyl) phthalate alone or in an environmental phthalate mixture. ENVIRONMENTAL RESEARCH 2022; 207:112235. [PMID: 34678253 DOI: 10.1016/j.envres.2021.112235] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 10/15/2021] [Accepted: 10/16/2021] [Indexed: 06/13/2023]
Abstract
We have previously shown that adult male mice exposure to low doses of di (2-ethylhexyl)phthalate (DEHP) impacts the blood-brain barrier (BBB) integrity and surrounding parenchyma in the medial preoptic area (mPOA), a key hypothalamic area involved in the male sexual behavior. BBB leakage was associated with a decrease in the endothelial tight junction accessory protein, zona occludens-1, and caveolae protein Cav-1, added to an inflammatory profile including glial activation accompanied by enhanced expression of inducible nitric oxide synthase. As this failure of BBB functionality in the mPOA could participate, at least in part, in reported alteration of sexual behavior following DEHP exposure, we explored the cellular pathway connecting cerebral capillaries and neurons. Two-month-old C57BL/6J male mice were orally exposed for 6 weeks to DEHP alone (5 and 50 μg/kg/day) or to DEHP (5 μg/kg/day) in an environmental phthalate mixture. The presence of androgen receptor (AR) and estrogen receptor-α (ERα) were first evidenced in brain capillaries. Protein levels of AR but not of ERα were reduced in cerebral capillaries after phthalate exposure. The amounts of basement membrane and cell-matrix interaction components were decreased, while matrix metalloprotease MMP-2 and MMP-9 activities were increased. Fluorojade® labelling suggested that exposure to phthalates also lead to a neurodegenerative process in the mPOA. Altogether, the data suggest that environmental exposure to endocrine disruptors such as phthalates, could alter AR/Cav-1 interaction, impacting a Cav-1/nitric oxide/MMP pathway. This would lead to disruption of the glio-neurovascular coupling which is essential to neuronal functioning.
Collapse
Affiliation(s)
- Delnia Ahmadpour
- Sorbonne Université, CNRS, INSERM, Neuroscience Paris-Seine, Institut de Biologie Paris-Seine, 75005, Paris, France
| | - Sakina Mhaouty-Kodja
- Sorbonne Université, CNRS, INSERM, Neuroscience Paris-Seine, Institut de Biologie Paris-Seine, 75005, Paris, France
| | - Valérie Grange-Messent
- Sorbonne Université, CNRS, INSERM, Neuroscience Paris-Seine, Institut de Biologie Paris-Seine, 75005, Paris, France.
| |
Collapse
|
32
|
Trouillet AC, Ducroq S, Naulé L, Capela D, Parmentier C, Radovick S, Hardin-Pouzet H, Mhaouty-Kodja S. Deletion of neural estrogen receptor alpha induces sex differential effects on reproductive behavior in mice. Commun Biol 2022; 5:383. [PMID: 35444217 PMCID: PMC9021208 DOI: 10.1038/s42003-022-03324-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Accepted: 03/25/2022] [Indexed: 11/20/2022] Open
Abstract
Estrogen receptor (ER) α is involved in several estrogen-modulated neural and peripheral functions. To determine its role in the expression of female and male reproductive behavior, a mouse line lacking the ERα in the nervous system was generated. Mutant females did not exhibit sexual behavior despite normal olfactory preference, and had a reduced number of progesterone receptor-immunoreactive neurons in the ventromedial hypothalamus. Mutant males displayed a moderately impaired sexual behavior and unaffected fertility, despite evidences of altered organization of sexually dimorphic populations in the preoptic area. In comparison, males deleted for both neural ERα and androgen receptor (AR) displayed greater sexual deficiencies. Thus, these data highlight a predominant role for neural ERα in females and a complementary role with the AR in males in the regulation of sexual behavior, and provide a solid background for future analyses of neuronal versus glial implication of these signaling pathways in both sexes. Neural deletion of the estrogen receptor, ERα, inhibits sexual behavior in female mice, but only has moderately effect in male mice. These results contrast with previous studies using global ERα knockouts, which found that ERα is mandatory for reproductive behavior in both sexes.
Collapse
Affiliation(s)
- Anne-Charlotte Trouillet
- Sorbonne Université, CNRS, INSERM, Neuroscience Paris Seine - Institut de Biologie Paris Seine, 75005, Paris, France
| | - Suzanne Ducroq
- Sorbonne Université, CNRS, INSERM, Neuroscience Paris Seine - Institut de Biologie Paris Seine, 75005, Paris, France
| | - Lydie Naulé
- Sorbonne Université, CNRS, INSERM, Neuroscience Paris Seine - Institut de Biologie Paris Seine, 75005, Paris, France
| | - Daphné Capela
- Sorbonne Université, CNRS, INSERM, Neuroscience Paris Seine - Institut de Biologie Paris Seine, 75005, Paris, France
| | - Caroline Parmentier
- Sorbonne Université, CNRS, INSERM, Neuroscience Paris Seine - Institut de Biologie Paris Seine, 75005, Paris, France
| | - Sally Radovick
- Unit of Pediatric Endocrinology, Department of Pediatrics, Rutgers-Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Hélène Hardin-Pouzet
- Sorbonne Université, CNRS, INSERM, Neuroscience Paris Seine - Institut de Biologie Paris Seine, 75005, Paris, France
| | - Sakina Mhaouty-Kodja
- Sorbonne Université, CNRS, INSERM, Neuroscience Paris Seine - Institut de Biologie Paris Seine, 75005, Paris, France.
| |
Collapse
|
33
|
Helsen C, Nguyen T, Vercruysse T, Wouters S, Daelemans D, Voet A, Claessens F. The T850D Phosphomimetic Mutation in the Androgen Receptor Ligand Binding Domain Enhances Recruitment at Activation Function 2. Int J Mol Sci 2022; 23:ijms23031557. [PMID: 35163481 PMCID: PMC8836279 DOI: 10.3390/ijms23031557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 01/26/2022] [Accepted: 01/27/2022] [Indexed: 02/04/2023] Open
Abstract
Several key functions of the androgen receptor (AR) such as hormone recognition and co-regulator recruitment converge in the ligand binding domain (LBD). Loss- or gain-of-function of the AR contributes to pathologies such as the androgen insensitivity syndrome and prostate cancer. Here, we describe a gain-of-function mutation of the surface-exposed threonine at position 850, located at the amino-terminus of Helix 10 (H10) in the AR LBD. Since T850 phosphorylation was reported to affect AR function, we created the phosphomimetic mutation T850D. The AR T850D variant has a 1.5- to 2-fold increased transcriptional activity with no effect on ligand affinity. In the androgen responsive LNCaP cell line grown in medium with low androgen levels, we observed a growth advantage for cells in which the endogenous AR was replaced by AR T850D. Despite the distance to the AF2 site, the AR T850D LBD displayed an increased affinity for coactivator peptides as well as the 23FQNLF27 motif of AR itself. Molecular Dynamics simulations confirm allosteric transmission of the T850D mutation towards the AF2 site via extended hydrogen bond formation between coactivator peptide and AF2 site. This mechanistic study thus confirms the gain-of-function character of T850D and T850 phosphorylation for AR activity and reveals details of the allosteric communications within the LBD.
Collapse
Affiliation(s)
- Christine Helsen
- Laboratory of Molecular Endocrinology, Department of Cellular and Molecular Medicine, KU Leuven, ON I, 3000 Leuven, Belgium;
- Correspondence: ; Tel.: +32-16377388
| | - Tien Nguyen
- Laboratory of Biomolecular Modelling and Design, Department of Chemistry, KU Leuven, Celestijnenlaan 200G, 3001 Leuven, Belgium; (T.N.); (S.W.); (A.V.)
| | - Thomas Vercruysse
- Laboratory of Virology and Chemotherapy, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, 3000 Leuven, Belgium; (T.V.); (D.D.)
| | - Staf Wouters
- Laboratory of Biomolecular Modelling and Design, Department of Chemistry, KU Leuven, Celestijnenlaan 200G, 3001 Leuven, Belgium; (T.N.); (S.W.); (A.V.)
| | - Dirk Daelemans
- Laboratory of Virology and Chemotherapy, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, 3000 Leuven, Belgium; (T.V.); (D.D.)
| | - Arnout Voet
- Laboratory of Biomolecular Modelling and Design, Department of Chemistry, KU Leuven, Celestijnenlaan 200G, 3001 Leuven, Belgium; (T.N.); (S.W.); (A.V.)
| | - Frank Claessens
- Laboratory of Molecular Endocrinology, Department of Cellular and Molecular Medicine, KU Leuven, ON I, 3000 Leuven, Belgium;
| |
Collapse
|
34
|
Estrogens and Androgens in Plants: The Last 20 Years of Studies. PLANTS 2021; 10:plants10122783. [PMID: 34961254 PMCID: PMC8705621 DOI: 10.3390/plants10122783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 12/13/2021] [Accepted: 12/13/2021] [Indexed: 11/16/2022]
Abstract
Although the only known steroid hormones in plants are brassinosteroids, interestingly, mammalian steroid hormones such as androgens or estrogens are also part of the plant metabolic profile. This presented review is focused on the progress that has been made in this matter during the last two decades. The presence of testosterone, 17β-estradiol, and other androgens/estrogens in plants (particularly those that can be measured using more advanced techniques) is described. The physiological activity of androgens and estrogens, especially in plants’ stress response, are discussed, together with some possible mechanisms of their action. The current knowledge indicates that although androgens and estrogens do not have the status of hormones in plants, they are physiologically active and can serve as regulators that support the activity of classic hormones in (1) regulating the various processes connected with plant growth and development and (2) the interaction of plants with their environment.
Collapse
|
35
|
Nowakowska MK, Lei X, Wehner MR, Corn PG, Giordano SH, Nead KT. Association of Second-generation Antiandrogens With Depression Among Patients With Prostate Cancer. JAMA Netw Open 2021; 4:e2140803. [PMID: 34940861 PMCID: PMC8703250 DOI: 10.1001/jamanetworkopen.2021.40803] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
IMPORTANCE Previous studies have shown a consistent association between hormone therapy (HT), such as androgen deprivation therapy, to treat prostate cancer and depression risk. However, the association between second-generation antiandrogens (AAs) and depression is unknown. OBJECTIVE To test the a priori hypothesis that second-generation AAs are associated with an increased risk of depression, including compared with traditional forms of HT. DESIGN, SETTING, AND PARTICIPANTS This retrospective cohort study analyzed patients aged 66 years and older who were diagnosed with prostate cancer without a second cancer in 12 months from January 2011 to December 2015. Patients with continuous Medicare Parts A, B, and D coverage were included. Individuals who received any form of HT prior to prostate cancer diagnosis and those previously diagnosed with depression were excluded. Data were collected from the Surveillance, Epidemiology, and End Results-Medicare and Texas Cancer Registry-Medicare linked databases. Data were analyzed from February to May 2021. EXPOSURES The following treatment groups were compared: (1) no HT group, (2) traditional HT group (HT without second-generation AA exposure), and (3) second-generation AA group. MAIN OUTCOMES AND MEASURES Risk of depression in the second-generation AA group compared with the no HT and traditional HT groups, determined prior to data collection, stratified by diagnosis stage. RESULTS Of 210 804 patients diagnosed with prostate cancer during the study window, 30 069 men (11 484 [38%] aged 66-70 years; 22 594 [75%] White) who met inclusion criteria were identified. Overall, 17 710 (59%) received no HT, 11 311 (38%) received traditional HT only, and 1048 (3%) received a second-generation AA. Those receiving a second-generation AA were more likely to be older (aged ≥81 years: second-generation AA group, 246 [24%]; traditional HT group, 1997 [18%]; no HT group, 1173 [7%]) and present with advanced disease (eg, distant disease: second-generation AA group, 562 [24%]; traditional HT group, 876 [8%]; no HT group, 129 [0.7%]). Multivariable Cox proportional hazards analysis showed that the second-generation AA group had an increased risk of depression compared with the no HT group (hazard ratio [HR], 2.15; 95% CI, 1.79-2.59; P < .001) and the traditional HT group (HR, 2.26; 95% CI, 1.88-2.73; P < .001), including specifically among those with metastatic disease at diagnosis (HR, 2.40; 95% CI, 1.38-4.15; P = .002). CONCLUSIONS AND RELEVANCE In this cohort study, patients with prostate cancer who received a second-generation AA had a large and clinically significant increased risk of depression compared with patients who received traditional HT alone or no HT, including when limiting our analysis to individuals with metastatic disease at diagnosis.
Collapse
Affiliation(s)
| | - Xiudong Lei
- Department of Health Services Research, University of Texas MD Anderson Cancer Center, Houston
| | - Mackenzie R. Wehner
- Department of Health Services Research, University of Texas MD Anderson Cancer Center, Houston
- Department of Dermatology, University of Texas MD Anderson Cancer Center, Houston
| | - Paul G. Corn
- Department of Genitourinary Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Sharon H. Giordano
- Department of Health Services Research, University of Texas MD Anderson Cancer Center, Houston
- Department of Breast Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Kevin T. Nead
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston
- Department of Epidemiology, University of Texas MD Anderson Cancer Center, Houston
| |
Collapse
|
36
|
Reiss AB, Saeedullah U, Grossfeld DJ, Glass AD, Pinkhasov A, Katz AE. Prostate cancer treatment and the relationship of androgen deprivation therapy to cognitive function. Clin Transl Oncol 2021; 24:733-741. [PMID: 34743290 DOI: 10.1007/s12094-021-02727-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 10/23/2021] [Indexed: 02/01/2023]
Abstract
Prostate cancer is the second most common form of cancer in men. For advanced, high risk prostate cancer, androgen deprivation therapy (ADT) is the preferred treatment and can induce remission, but resistance to ADT brings biochemical recurrence and progression of cancer. ADT brings adverse effects such as erectile dysfunction, decreased libido, and diminished physical strength. It is estimated that between 25 and 50% of men on ADT manifest some form of cognitive dysfunction that may be self-reported or reported by a family member. There is concern that impaired cognitive function with ADT is due to loss of testosterone support. Testosterone and its metabolites are known to possess neuroprotective properties. While a direct causal relationship between ADT and cognitive decline in prostate cancer patients has not been established, this review describes the controversy surrounding the possible connection between ADT and neurocognitive deterioration. The cellular and molecular mechanisms believed to underlie the protection of neuronal integrity by androgens are discussed. Results from animal models and human clinical studies are presented. Finally, we call attention to lifestyle modifications that may minimize cognitive issues in prostate cancer patients.
Collapse
Affiliation(s)
- A B Reiss
- Biomedical Research Institute, NYU Long Island School of Medicine, 101 Mineola Boulevard, Suite 4-004, Mineola, NY, 11501, USA.
| | - U Saeedullah
- Biomedical Research Institute, NYU Long Island School of Medicine, 101 Mineola Boulevard, Suite 4-004, Mineola, NY, 11501, USA
| | - D J Grossfeld
- Biomedical Research Institute, NYU Long Island School of Medicine, 101 Mineola Boulevard, Suite 4-004, Mineola, NY, 11501, USA
| | - A D Glass
- Biomedical Research Institute, NYU Long Island School of Medicine, 101 Mineola Boulevard, Suite 4-004, Mineola, NY, 11501, USA
| | - A Pinkhasov
- Biomedical Research Institute, NYU Long Island School of Medicine, 101 Mineola Boulevard, Suite 4-004, Mineola, NY, 11501, USA
| | - A E Katz
- Biomedical Research Institute, NYU Long Island School of Medicine, 101 Mineola Boulevard, Suite 4-004, Mineola, NY, 11501, USA
| |
Collapse
|
37
|
Ahmadpour D, Mhaouty-Kodja S, Grange-Messent V. Disruption of the blood-brain barrier and its close environment following adult exposure to low doses of di(2-ethylhexyl)phthalate alone or in an environmental phthalate mixture in male mice. CHEMOSPHERE 2021; 282:131013. [PMID: 34090004 DOI: 10.1016/j.chemosphere.2021.131013] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 04/14/2021] [Accepted: 05/26/2021] [Indexed: 05/20/2023]
Abstract
We have previously shown that adult male mice exposure to low doses of di(2-ethylhexyl)phthalate (DEHP) alters neural function and behaviour. Whether such exposure also affects the integrity and function of the blood-brain barrier (BBB) remained to be explored. The impact of adult exposure to low doses of DEHP alone or in an environmental phthalate mixture on the BBB integrity and surrounding parenchyma was studied in male mice. Two-month-old C57BL/6J males were orally exposed for 6 weeks to DEHP alone (5, and 50 μg/kg/day) or to DEHP (5 μg/kg/day) in an environmental phthalate mixture. BBB permeability, glial activation and neuroinflammation were investigated in the hypothalamic medial preoptic area (mPOA) and hippocampus involved, respectively on the reproductive and cognitive functions. Exposure to DEHP alone or in a phthalate mixture increased BBB permeability and affected the endothelial accessory tight junction protein zona occludens-1 and caveolae protein Cav-1 in the mPOA and the hippocampal CA1 and CA3 areas. This was associated with an inflammatory profile including astrocyte activation accompanied by enhanced expression of inducible nitric oxide synthase in the mPOA, and a microglial activation in the mPOA and the hippocampal CA1 and CA3 areas. The protein levels of the inflammatory molecule cyclooxygenase-2 were increased in activated microglial cells of the exposed mPOA. None of the major effects induced by DEHP alone or in a mixture was detected in the hippocampal dendate gyrus. The data highlight that environmental exposure to endocrine disruptors such as phthalates, could represent a risk factor for the cerebrovascular function.
Collapse
Affiliation(s)
- Delnia Ahmadpour
- Sorbonne Université, CNRS, INSERM, Neuroscience Paris-Seine, Institut de Biologie Paris-Seine, 75005 Paris, France
| | - Sakina Mhaouty-Kodja
- Sorbonne Université, CNRS, INSERM, Neuroscience Paris-Seine, Institut de Biologie Paris-Seine, 75005 Paris, France
| | - Valérie Grange-Messent
- Sorbonne Université, CNRS, INSERM, Neuroscience Paris-Seine, Institut de Biologie Paris-Seine, 75005 Paris, France.
| |
Collapse
|
38
|
Kırgezen T, Yücetaş U, Server EA, Övünç O, Yiğit Ö. Possible effects of low testosterone levels on olfactory function in males. Braz J Otorhinolaryngol 2021; 87:702-710. [PMID: 32345533 PMCID: PMC9422496 DOI: 10.1016/j.bjorl.2020.03.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Revised: 01/05/2020] [Accepted: 03/08/2020] [Indexed: 11/21/2022] Open
Abstract
INTRODUCTION Functions attributed to androgens have increased, ranging from the role in hypothalamic-pituitary-gonadal axis and reproductive behaviors to modulation of cognition, mood and some other functions. Sex differences and changes in circulating sex hormones affect human sensory function. In the literature, authors reported this kind of influence for olfaction predominantly in females. OBJECTIVE To investigate the effects of low testosterone levels on olfactory functions in males, in this prospective clinical study. METHODS Male patients diagnosed with prostate cancer were included. Thirty-nine patients with prostate cancer whose testosterone levels were lower than 50ng/dL due to castration, were the study group. Thirty-one patients with prostate cancer who were not castrated with testosterone levels higher than 50ng/dL were selected as the control group. Acoustic rhinometry and peak nasal inspiratory flow tests were performed for all participants; and for evaluation of olfactory function, both groups completed the Connecticut chemosensory clinical research center olfactory test. RESULTS The mean ages of the patients and controls were 69.6±7.2 (57-89) and 66.3±5.8 (50-78) years, respectively (p=0.039). There was a significant difference between groups in terms of testosterone levels (p<0.0001). The multivariate logistic regression revealed testosterone level as the only predictive factor determining the difference between the groups. In terms of olfactory parameters, all scores were lower in the emasculated group (butanol threshold test p=0.019, identification p=0.059, and Connecticut center score p=0.029) There was a significant correlation between testosterone levels and olfactory parameters (p=0.023; p=0.025 for identification and Connecticut center scores, respectively). CONCLUSION Low testosterone levels in males have negative effects on olfactory functions. Further molecular research is required to understand the connection between testosterone and olfaction.
Collapse
Affiliation(s)
- Tolga Kırgezen
- University of Health Sciences, Istanbul Training and Research Hospital, Department of Otorhinolaryngology/Head and Neck Surgery, Istanbul, Turkey.
| | - Uğur Yücetaş
- University of Health Sciences, Istanbul Training and Research Hospital, Department of Urology, Istanbul, Turkey
| | - Ela Araz Server
- University of Health Sciences, Istanbul Training and Research Hospital, Department of Otorhinolaryngology/Head and Neck Surgery, Istanbul, Turkey
| | - Okan Övünç
- University of Health Sciences, Istanbul Training and Research Hospital, Department of Otorhinolaryngology/Head and Neck Surgery, Istanbul, Turkey
| | - Özgür Yiğit
- University of Health Sciences, Istanbul Training and Research Hospital, Department of Otorhinolaryngology/Head and Neck Surgery, Istanbul, Turkey
| |
Collapse
|
39
|
Luo D, Han L, Gao S, Xiao Z, Zhou Q, Cheng X, Zhang Y, Zhou W. LINCS Dataset-Based Repositioning of Dutasteride as an Anti-Neuroinflammation Agent. Brain Sci 2021; 11:1411. [PMID: 34827410 PMCID: PMC8615696 DOI: 10.3390/brainsci11111411] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/17/2021] [Accepted: 10/22/2021] [Indexed: 12/21/2022] Open
Abstract
Neuroinflammation is often accompanied by central nervous system (CNS) injury seen in various CNS diseases, with no specific treatment. Drug repurposing is a strategy of finding new uses for approved or investigational drugs, and can be enabled by the Library of Integrated Network-based Cellular Signatures (LINCS), a large drug perturbation database. In this study, the signatures of Lipopolysaccharide (LPS) were compared with the signatures of compounds contained in the LINCS dataset. To the top 100 compounds obtained, the Quantitative Structure-Activity Relationship (QSAR)-based tool admetSAR was used to identify the top 10 candidate compounds with relatively high blood-brain barrier (BBB) penetration. Furthermore, the seventh-ranked compound, dutasteride, a 5-α-reductase inhibitor, was selected for in vitro and in vivo validation of its anti-neuroinflammation activity. The results showed that dutasteride significantly reduced the levels of IL-6 and TNF-α in the supernatants of LPS-stimulated BV2 cells, and decreased the levels of IL-6 in the hippocampus and plasma, and the number of activated microglia in the brain of LPS administration mice. Furthermore, dutasteride also attenuated the cognitive impairment caused by LPS stimulation in mice. Taken together, this study demonstrates that the LINCS dataset-based drug repurposing strategy is an effective approach, and the predicted candidate, dutasteride, has the potential to ameliorate LPS-induced neuroinflammation and cognitive impairment.
Collapse
Affiliation(s)
- Dan Luo
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; (D.L.); (L.H.); (S.G.); (Z.X.); (Q.Z.); (X.C.)
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing 100850, China
| | - Lu Han
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; (D.L.); (L.H.); (S.G.); (Z.X.); (Q.Z.); (X.C.)
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing 100850, China
| | - Shengqiao Gao
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; (D.L.); (L.H.); (S.G.); (Z.X.); (Q.Z.); (X.C.)
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing 100850, China
| | - Zhiyong Xiao
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; (D.L.); (L.H.); (S.G.); (Z.X.); (Q.Z.); (X.C.)
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing 100850, China
| | - Qingru Zhou
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; (D.L.); (L.H.); (S.G.); (Z.X.); (Q.Z.); (X.C.)
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing 100850, China
| | - Xiaorui Cheng
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; (D.L.); (L.H.); (S.G.); (Z.X.); (Q.Z.); (X.C.)
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing 100850, China
| | - Yongxiang Zhang
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; (D.L.); (L.H.); (S.G.); (Z.X.); (Q.Z.); (X.C.)
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing 100850, China
| | - Wenxia Zhou
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; (D.L.); (L.H.); (S.G.); (Z.X.); (Q.Z.); (X.C.)
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing 100850, China
| |
Collapse
|
40
|
Chen YH, Chen YC, Hwang LL, Yang LY, Lu DY. Deficiency in Androgen Receptor Aggravates Traumatic Brain Injury-Induced Pathophysiology and Motor Deficits in Mice. Molecules 2021; 26:6250. [PMID: 34684832 PMCID: PMC8537172 DOI: 10.3390/molecules26206250] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/12/2021] [Accepted: 10/13/2021] [Indexed: 11/16/2022] Open
Abstract
Androgens have been shown to have a beneficial effect on brain injury and lower reactive astrocyte expression after TBI. Androgen receptors (ARs) are known to mediate the neuroprotective effects of androgens. However, whether ARs play a crucial role in TBI remains unknown. In this study, we investigated the role of ARs in TBI pathophysiology, using AR knockout (ARKO) mice. We used the controlled cortical impact model to produce primary and mechanical brain injuries and assessed motor function and brain-lesion volume. In addition, the AR knockout effects on necrosis and autophagy were evaluated after TBI. AR knockout significantly increased TBI-induced expression of the necrosis marker alpha-II-spectrin breakdown product 150 and astrogliosis marker glial fibrillary acidic protein. In addition, the TBI-induced astrogliosis increase in ARKO mice lasted for three weeks after a TBI. The autophagy marker Beclin-1 was also enhanced in ARKO mice compared with wild-type mice after TBI. Our results also indicated that ARKO mice showed a more unsatisfactory performance than wild-type mice in a motor function test following TBI. Further, they were observed to have more severe lesions than wild-type mice after injury. These findings strongly suggest that ARs play a role in TBI.
Collapse
Affiliation(s)
- Yu-Hsin Chen
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (Y.-H.C.); (Y.-C.C.); (L.-L.H.)
| | - Yen-Chou Chen
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (Y.-H.C.); (Y.-C.C.); (L.-L.H.)
| | - Ling-Ling Hwang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (Y.-H.C.); (Y.-C.C.); (L.-L.H.)
| | - Liang-Yo Yang
- Department of Physiology, School of Medicine, College of Medicine, China Medical University, Taichung 40402, Taiwan
- Laboratory of Neural Repair, Department of Medical Research, China Medical University Hospital, Taichung 40447, Taiwan
| | - Dah-Yuu Lu
- Department of Pharmacology, School of Medicine, China Medical University, Taichung 404333, Taiwan
- Department of Photonics and Communication Engineering, Asia University, Taichung 404333, Taiwan
| |
Collapse
|
41
|
Jean A, Mhaouty-Kodja S, Hardin-Pouzet H. Hypothalamic cellular and molecular plasticity linked to sexual experience in male rats and mice. Front Neuroendocrinol 2021; 63:100949. [PMID: 34687674 DOI: 10.1016/j.yfrne.2021.100949] [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: 07/23/2021] [Revised: 09/22/2021] [Accepted: 10/18/2021] [Indexed: 10/20/2022]
Abstract
Male sexual behavior is subject to learning, resulting in increased efficiency of experienced males compared to naive ones. The improvement in behavioral parameters is underpinned by cellular and molecular changes in the neural circuit controlling sexual behavior, particularly in the hypothalamic medial preoptic area. This review provides an update on the mechanisms related to the sexual experience in male rodents, emphasizing the differences between rats and mice.
Collapse
Affiliation(s)
- Arnaud Jean
- Sorbonne Université - Faculté de Sciences et Ingénierie, Neuroplasticité des Comportements de la Reproduction, Neurosciences Paris Seine, UM119 - CNRS UMR 8246 - INSERM UMRS 1130, 7 quai Saint Bernard, 75 005 Paris, France
| | - Sakina Mhaouty-Kodja
- Sorbonne Université - Faculté de Sciences et Ingénierie, Neuroplasticité des Comportements de la Reproduction, Neurosciences Paris Seine, UM119 - CNRS UMR 8246 - INSERM UMRS 1130, 7 quai Saint Bernard, 75 005 Paris, France
| | - Hélène Hardin-Pouzet
- Sorbonne Université - Faculté de Sciences et Ingénierie, Neuroplasticité des Comportements de la Reproduction, Neurosciences Paris Seine, UM119 - CNRS UMR 8246 - INSERM UMRS 1130, 7 quai Saint Bernard, 75 005 Paris, France.
| |
Collapse
|
42
|
Almaguer-Mederos LE, Aguilera-Rodríguez R, Almaguer-Gotay D, Hechavarría-Barzaga K, Álvarez-Sosa A, Chapman-Rodríguez Y, Silva-Ricardo Y, González-Zaldivar Y, Vázquez-Mojena Y, Cuello-Almarales D, Rodríguez-Estupiñán A. Testosterone Levels Are Decreased and Associated with Disease Duration in Male Spinocerebellar Ataxia Type 2 Patients. THE CEREBELLUM 2021; 19:597-604. [PMID: 32440846 DOI: 10.1007/s12311-020-01134-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Spinocerebellar ataxia type 2 (SCA2) is a progressive neurodegenerative disorder due to an unstable expansion of a CAG repeat in the ATXN2 gene. Despite clinical and experimental evidence indicating the relevance of the gonadotropic axis to the prognosis and therapeutics for several late-onset neurodegenerative disorders, its functioning and association with disease severity have not been previously explored in SCA2. To assess serum levels of testosterone, luteinizing hormone (LH), and follicle-stimulating hormone (FSH), and their clinical relevance in SCA2 patients. A case-control study involving 94 Cuban SCA2 patients and 101 gender- and age-matched healthy controls was conducted. Testosterone, LH, and FSH serum levels were determined by radioimmunoassay or immunoradiometric assay systems. Clinical outcomes included age at onset, disease duration, Scale for the Assessment and Rating of Ataxia (SARA) score, and progression rate. Univariate general linear models were generated. Testosterone, LH, and FSH serum levels were significantly reduced in male SCA2 patients relative to control individuals. On average, there was a 35% reduction in testosterone levels in male patients versus male control individuals. Testosterone levels were associated with disease duration (r = 0.383; p = 0.025) and age at onset (r = 0.414; p = 0.011) in male SCA2 patients, but no association was observed between testosterone and CAG expansion size, SARA score, or progression rate. Testosterone levels might be a biomarker of disease progression in male SCA2 patients. Further studies are needed to explore the effects of low testosterone levels on non-motor symptoms, and to assess the potential of testosterone replacement therapy in male SCA2 patients.
Collapse
Affiliation(s)
- Luis E Almaguer-Mederos
- Center for the Investigation and Rehabilitation of Hereditary Ataxias (CIRAH), Holguin, Cuba.
| | - Raúl Aguilera-Rodríguez
- Center for the Investigation and Rehabilitation of Hereditary Ataxias (CIRAH), Holguin, Cuba
| | - Dennis Almaguer-Gotay
- Center for the Investigation and Rehabilitation of Hereditary Ataxias (CIRAH), Holguin, Cuba
| | | | | | | | | | | | - Yaimé Vázquez-Mojena
- Center for the Investigation and Rehabilitation of Hereditary Ataxias (CIRAH), Holguin, Cuba
| | - Dany Cuello-Almarales
- Center for the Investigation and Rehabilitation of Hereditary Ataxias (CIRAH), Holguin, Cuba
| | | |
Collapse
|
43
|
Uncovering Evidence for Endocrine-Disrupting Chemicals That Elicit Differential Susceptibility through Gene-Environment Interactions. TOXICS 2021; 9:toxics9040077. [PMID: 33917455 PMCID: PMC8067468 DOI: 10.3390/toxics9040077] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 03/27/2021] [Accepted: 04/02/2021] [Indexed: 12/17/2022]
Abstract
Exposure to endocrine-disrupting chemicals (EDCs) is linked to myriad disorders, characterized by the disruption of the complex endocrine signaling pathways that govern development, physiology, and even behavior across the entire body. The mechanisms of endocrine disruption involve a complex system of pathways that communicate across the body to stimulate specific receptors that bind DNA and regulate the expression of a suite of genes. These mechanisms, including gene regulation, DNA binding, and protein binding, can be tied to differences in individual susceptibility across a genetically diverse population. In this review, we posit that EDCs causing such differential responses may be identified by looking for a signal of population variability after exposure. We begin by summarizing how the biology of EDCs has implications for genetically diverse populations. We then describe how gene-environment interactions (GxE) across the complex pathways of endocrine signaling could lead to differences in susceptibility. We survey examples in the literature of individual susceptibility differences to EDCs, pointing to a need for research in this area, especially regarding the exceedingly complex thyroid pathway. Following a discussion of experimental designs to better identify and study GxE across EDCs, we present a case study of a high-throughput screening signal of putative GxE within known endocrine disruptors. We conclude with a call for further, deeper analysis of the EDCs, particularly the thyroid disruptors, to identify if these chemicals participate in GxE leading to differences in susceptibility.
Collapse
|
44
|
de Wit AE, Giltay EJ, de Boer MK, Bosker FJ, Cohn AY, Nolen WA, Kaiser UB, Joffe H, Penninx BW, Schoevers RA. Plasma androgens and the presence and course of depression in a large cohort of women. Transl Psychiatry 2021; 11:124. [PMID: 33579903 PMCID: PMC7881099 DOI: 10.1038/s41398-021-01249-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 01/07/2021] [Accepted: 01/14/2021] [Indexed: 01/05/2023] Open
Abstract
Major depressive disorder (MDD) has a higher prevalence in women with supraphysiologic androgen levels. Whether there is also an association between depression and androgen levels in the physiological range, is unknown. This study examined if women with current MDD have higher androgen levels compared to women who have never had MDD, and if androgen levels are associated with onset and remission of MDD. In 1659 women (513 current MDD, 754 remitted MDD, and 392 never MDD), baseline plasma levels of total testosterone, 5α-dihydrotestosterone, and androstenedione were determined with liquid chromatography-tandem mass spectrometry, and dehydroepiandrosterone-sulfate and sex hormone binding globulin (SHBG) with radioimmunoassays. Free testosterone was calculated. MDD status was assessed at baseline, and at 2 and 4 years follow-up. Women were aged between 18 and 65 years (mean age 41) with total testosterone levels in the physiological range (geometric mean 0.72 nmol/L [95% CI 0.27-1.93]). After adjusting for covariates and multiple testing, women with current MDD had a higher mean free testosterone than women who never had MDD (adjusted geometric mean 8.50 vs. 7.55 pmol/L, p = 0.0005), but this difference was not large enough to be considered clinically meaningful as it was consistent with statistical equivalence. Levels of other androgens and SHBG did not differ and were also statistically equivalent between the groups. None of the androgens or SHBG levels predicted onset or remission of MDD. Our findings support the idea that plasma androgens within the physiological range have no or only limited effects on depressive disorders in women.
Collapse
Affiliation(s)
- Anouk E. de Wit
- grid.4494.d0000 0000 9558 4598University of Groningen, University Medical Center Groningen, Department of Psychiatry, Groningen, The Netherlands
| | - Erik J. Giltay
- grid.10419.3d0000000089452978Leiden University Medical Center, Department of Psychiatry, Leiden, The Netherlands
| | - Marrit K. de Boer
- grid.4494.d0000 0000 9558 4598University of Groningen, University Medical Center Groningen, Department of Psychiatry, Groningen, The Netherlands
| | - Fokko J. Bosker
- grid.4494.d0000 0000 9558 4598University of Groningen, University Medical Center Groningen, Department of Psychiatry, Groningen, The Netherlands
| | - Aviva Y. Cohn
- Brigham and Women’s Hospital, Harvard Medical School, Department of Medicine, Boston, MA USA
| | - Willem A. Nolen
- grid.4494.d0000 0000 9558 4598University of Groningen, University Medical Center Groningen, Department of Psychiatry, Groningen, The Netherlands
| | - Ursula B. Kaiser
- Brigham and Women’s Hospital, Harvard Medical School, Department of Medicine, Boston, MA USA
| | - Hadine Joffe
- Brigham and Women’s Hospital, Harvard Medical School, Department of Psychiatry, Boston, MA USA ,Brigham and Women’s Hospital, Harvard Medical School, Connors Center for Women’s Health and Gender Biology, Boston, MA USA
| | - Brenda W.J.H. Penninx
- grid.12380.380000 0004 1754 9227Department of Psychiatry, Amsterdam UMC, VU University, Amsterdam, The Netherlands
| | - Robert A. Schoevers
- grid.4494.d0000 0000 9558 4598University of Groningen, University Medical Center Groningen, Department of Psychiatry, Groningen, The Netherlands
| |
Collapse
|
45
|
Transcriptome Sequencing in the Preoptic Region of Rat Dams Reveals a Role of Androgen Receptor in the Control of Maternal Behavior. Int J Mol Sci 2021; 22:ijms22041517. [PMID: 33546359 PMCID: PMC7913516 DOI: 10.3390/ijms22041517] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/24/2021] [Accepted: 01/27/2021] [Indexed: 01/17/2023] Open
Abstract
(1) Background: Preoptic region of hypothalamus is responsible to control maternal behavior, which was hypothesized to be associated with gene expressional changes. (2) Methods: Transcriptome sequencing was first applied in the preoptic region of rat dams in comparison to a control group of mothers whose pups were taken away immediately after parturition and did not exhibit caring behavior 10 days later. (3) Results: Differentially expressed genes were found and validated by quantitative RT-PCR, among them NACHT and WD repeat domain containing 1 (Nwd1) is known to control androgen receptor (AR) protein levels. The distribution of Nwd1 mRNA and AR was similar in the preoptic area. Therefore, we focused on this steroid hormone receptor and found its reduced protein level in rat dams. To establish the function of AR in maternal behavior, its antagonist was administered intracerebroventricularly into mother rats and increased pup-directed behavior of the animals. (4) Conclusions: AR levels are suppressed in the preoptic area of mothers possibly mediated by altered Nwd1 expression in order to allow sustained high-level care for the pups. Thus, our study first implicated the AR in the control of maternal behaviors.
Collapse
|
46
|
Ahmadpour D, Grange-Messent V. Involvement of Testosterone Signaling in the Integrity of the Neurovascular Unit in the Male: Review of Evidence, Contradictions, and Hypothesis. Neuroendocrinology 2021; 111:403-420. [PMID: 32512571 DOI: 10.1159/000509218] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 06/08/2020] [Indexed: 11/19/2022]
Abstract
Age-related central nervous system function decline and increased susceptibility of females compared to males with respect to prevalence of several neurodegenerative and neuropsychiatric diseases are both based on the principle that hormonal factors could be involved. These cerebral disorders are characterized by an alteration of blood-brain barrier (BBB) properties and chronic neuroinflammation, which lead to disease progression. Neuroinflammation, in turn, contributes to BBB dysfunction. The BBB and its environment, called the neurovascular unit (NVU), are crucial for cerebral homeostasis and neuronal function. Interestingly, sex steroids influence BBB properties and modulate neuroinflammatory responses. To date however, the majority of work reported has focused on the effects of estrogens on BBB function and neuroinflammation in female mammals. In contrast, the effects of testosterone signaling on the NVU in males are still poorly studied. The aim of this review was to summarize and discuss the literature, providing insights and contradictions to highlight hypothesis and the need for further investigations.
Collapse
Affiliation(s)
- Delnia Ahmadpour
- Sorbonne Université, INSERM U1130, CNRS UMR 8246, Neuroscience Paris-Seine, Institut de Biologie Paris-Seine, Paris, France
| | - Valérie Grange-Messent
- Sorbonne Université, INSERM U1130, CNRS UMR 8246, Neuroscience Paris-Seine, Institut de Biologie Paris-Seine, Paris, France,
| |
Collapse
|
47
|
Chang KF, Huang XF, Chang JT, Huang YC, Weng JC, Tsai NM. Cedrol suppresses glioblastoma progression by triggering DNA damage and blocking nuclear translocation of the androgen receptor. Cancer Lett 2020; 495:180-190. [DOI: 10.1016/j.canlet.2020.09.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 08/19/2020] [Accepted: 09/06/2020] [Indexed: 12/20/2022]
|
48
|
Hoffman JR, Zuckerman A, Ram O, Sadot O, Cohen H. Changes in Hippocampal Androgen Receptor Density and Behavior in Sprague-Dawley Male Rats Exposed to a Low-Pressure Blast Wave. Brain Plast 2020; 5:135-145. [PMID: 33282677 PMCID: PMC7685673 DOI: 10.3233/bpl-200107] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Objective The purpose of this study was to examine the effect of exposure of a low-intensity blast wave on androgen receptor (AR) density in the hippocampus and the potential influence on behavioral and cognitive responses. Methods Sprague-Dawley rats were randomly assigned to either a blast exposed group (n = 27) or an unexposed (control) group (n = 10). Animals were treated identically, except that rats within the control group were not exposed to any of the characteristics of the blast wave. Behavior measures were conducted on day seven post-exposure. The rats were initially assessed in the elevated plus maze followed by the acoustic startle response paradigm. Spatial memory performance using the Morris water-maze test was assessed at 8-days post-exposure, for seven consecutive days. Following all behavioral tests AR immunofluorescence staining was performed in different hippocampal subregions. Results A significant elevation in anxiety index (p < 0.001) and impaired learning (p < 0.015) and spatial memory (p < 0.0015) were noted in exposed rats. In addition, a significant attenuation of the AR was noted in the CA1 (p = 0.006) and dentate gyrus (p = 0.031) subregions of the hippocampus in blast exposed animals. Correlational analyses revealed significant associations between AR and both anxiety index (r = -.36, p = 0.031) and memory (r = -0.38, p = 0.019). Conclusions The results of this study demonstrate that exposure to a low-pressure blast wave resulted in a decrease in AR density, which was associated with significant behavioral and cognitive changes.
Collapse
Affiliation(s)
- Jay R Hoffman
- Department of Physical Therapy, Ariel University, Ariel, Israel
| | - Amitai Zuckerman
- Anxiety and Stress Research Unit, Beer-Sheva Mental Health Center, Faculty of Health Sciences, Division of Psychiatry, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Omri Ram
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Oren Sadot
- Department of Mechanical Engineering, Ben-Gurion University, Israel
| | - Hagit Cohen
- Anxiety and Stress Research Unit, Beer-Sheva Mental Health Center, Faculty of Health Sciences, Division of Psychiatry, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| |
Collapse
|
49
|
Wu Y, Zhang Y, Ou J, Hu Y, Zilioli S. Exogenous testosterone increases the audience effect in healthy males: evidence for the social status hypothesis. Proc Biol Sci 2020; 287:20200976. [PMID: 32673552 DOI: 10.1098/rspb.2020.0976] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Several studies have implicated testosterone in the modulation of altruistic behaviours instrumental to advancing social status. Independent studies have also shown that people tend to behave more altruistically when being watched (i.e. audience effect). To date, little is known about whether testosterone could modulate the audience effect. In the current study, we tested the effect of testosterone on altruistic behaviour using a donation task, wherein participants were asked to either accept or reject a monetary transfer to a charity organization accompanying a personal cost either in the presence or absence of an observer. We administered testosterone gel or placebo to healthy young men (n = 140) in a double-blind, placebo-controlled, mixed design. Our results showed that participants were more likely to accept the monetary transfer to the charity when being observed compared to when they completed the task alone. More importantly, this audience effect was amplified among people receiving testosterone versus placebo. Our findings suggest that testosterone administration increases the audience effect and further buttress the social status hypothesis, according to which testosterone promotes status-seeking behaviour in a context-dependent manner.
Collapse
Affiliation(s)
- Yin Wu
- School of Psychology, Shenzhen University, Shenzhen 518060, People's Republic of China.,Shenzhen Key Laboratory of Affective and Social Cognitive Science, Shenzhen University, Shenzhen 518060, People's Republic of China
| | - Yinhua Zhang
- School of Psychology, Shenzhen University, Shenzhen 518060, People's Republic of China.,Shenzhen Key Laboratory of Affective and Social Cognitive Science, Shenzhen University, Shenzhen 518060, People's Republic of China
| | - Jianxin Ou
- School of Psychology, Shenzhen University, Shenzhen 518060, People's Republic of China.,Shenzhen Key Laboratory of Affective and Social Cognitive Science, Shenzhen University, Shenzhen 518060, People's Republic of China
| | - Yang Hu
- School of Psychological and Cognitive Sciences, Peking University, Beijing 100871, People's Republic of China
| | - Samuele Zilioli
- Department of Psychology, Wayne State University, Detroit, MI 48202, USA.,Department of Family Medicine and Public Health Sciences, Wayne State University, Detroit, MI 48202, USA
| |
Collapse
|
50
|
Rusmini P, Cristofani R, Tedesco B, Ferrari V, Messi E, Piccolella M, Casarotto E, Chierichetti M, Cicardi ME, Galbiati M, Geroni C, Lombardi P, Crippa V, Poletti A. Enhanced Clearance of Neurotoxic Misfolded Proteins by the Natural Compound Berberine and Its Derivatives. Int J Mol Sci 2020; 21:ijms21103443. [PMID: 32414108 PMCID: PMC7279252 DOI: 10.3390/ijms21103443] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/08/2020] [Accepted: 05/11/2020] [Indexed: 12/17/2022] Open
Abstract
Background: Accumulation of misfolded proteins is a common hallmark of several neurodegenerative disorders (NDs) which results from a failure or an impairment of the protein quality control (PQC) system. The PQC system is composed by chaperones and the degradative systems (proteasome and autophagy). Mutant proteins that misfold are potentially neurotoxic, thus strategies aimed at preventing their aggregation or at enhancing their clearance are emerging as interesting therapeutic targets for NDs. Methods: We tested the natural alkaloid berberine (BBR) and some derivatives for their capability to enhance misfolded protein clearance in cell models of NDs, evaluating which degradative pathway mediates their action. Results: We found that both BBR and its semisynthetic derivatives promote degradation of mutant androgen receptor (ARpolyQ) causative of spinal and bulbar muscular atrophy, acting mainly via proteasome and preventing ARpolyQ aggregation. Overlapping effects were observed on other misfolded proteins causative of amyotrophic lateral sclerosis, frontotemporal-lobar degeneration or Huntington disease, but with selective and specific action against each different mutant protein. Conclusions: BBR and its analogues induce the clearance of misfolded proteins responsible for NDs, representing potential therapeutic tools to counteract these fatal disorders.
Collapse
Affiliation(s)
- Paola Rusmini
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Dipartimento di Eccellenza 2018-2022, Centro di Eccellenza sulle Malattie Neurodegenerative, Università degli Studi di Milano, 20133 Milan, Italy; (P.R.); (R.C.); (B.T.); (V.F.); (E.M.); (M.P.); (E.C.); (M.C.); (M.E.C.); (M.G.); (V.C.)
| | - Riccardo Cristofani
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Dipartimento di Eccellenza 2018-2022, Centro di Eccellenza sulle Malattie Neurodegenerative, Università degli Studi di Milano, 20133 Milan, Italy; (P.R.); (R.C.); (B.T.); (V.F.); (E.M.); (M.P.); (E.C.); (M.C.); (M.E.C.); (M.G.); (V.C.)
| | - Barbara Tedesco
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Dipartimento di Eccellenza 2018-2022, Centro di Eccellenza sulle Malattie Neurodegenerative, Università degli Studi di Milano, 20133 Milan, Italy; (P.R.); (R.C.); (B.T.); (V.F.); (E.M.); (M.P.); (E.C.); (M.C.); (M.E.C.); (M.G.); (V.C.)
| | - Veronica Ferrari
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Dipartimento di Eccellenza 2018-2022, Centro di Eccellenza sulle Malattie Neurodegenerative, Università degli Studi di Milano, 20133 Milan, Italy; (P.R.); (R.C.); (B.T.); (V.F.); (E.M.); (M.P.); (E.C.); (M.C.); (M.E.C.); (M.G.); (V.C.)
| | - Elio Messi
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Dipartimento di Eccellenza 2018-2022, Centro di Eccellenza sulle Malattie Neurodegenerative, Università degli Studi di Milano, 20133 Milan, Italy; (P.R.); (R.C.); (B.T.); (V.F.); (E.M.); (M.P.); (E.C.); (M.C.); (M.E.C.); (M.G.); (V.C.)
| | - Margherita Piccolella
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Dipartimento di Eccellenza 2018-2022, Centro di Eccellenza sulle Malattie Neurodegenerative, Università degli Studi di Milano, 20133 Milan, Italy; (P.R.); (R.C.); (B.T.); (V.F.); (E.M.); (M.P.); (E.C.); (M.C.); (M.E.C.); (M.G.); (V.C.)
| | - Elena Casarotto
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Dipartimento di Eccellenza 2018-2022, Centro di Eccellenza sulle Malattie Neurodegenerative, Università degli Studi di Milano, 20133 Milan, Italy; (P.R.); (R.C.); (B.T.); (V.F.); (E.M.); (M.P.); (E.C.); (M.C.); (M.E.C.); (M.G.); (V.C.)
| | - Marta Chierichetti
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Dipartimento di Eccellenza 2018-2022, Centro di Eccellenza sulle Malattie Neurodegenerative, Università degli Studi di Milano, 20133 Milan, Italy; (P.R.); (R.C.); (B.T.); (V.F.); (E.M.); (M.P.); (E.C.); (M.C.); (M.E.C.); (M.G.); (V.C.)
| | - Maria Elena Cicardi
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Dipartimento di Eccellenza 2018-2022, Centro di Eccellenza sulle Malattie Neurodegenerative, Università degli Studi di Milano, 20133 Milan, Italy; (P.R.); (R.C.); (B.T.); (V.F.); (E.M.); (M.P.); (E.C.); (M.C.); (M.E.C.); (M.G.); (V.C.)
- Jefferson Weinberg ALS Center, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Mariarita Galbiati
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Dipartimento di Eccellenza 2018-2022, Centro di Eccellenza sulle Malattie Neurodegenerative, Università degli Studi di Milano, 20133 Milan, Italy; (P.R.); (R.C.); (B.T.); (V.F.); (E.M.); (M.P.); (E.C.); (M.C.); (M.E.C.); (M.G.); (V.C.)
| | - Cristina Geroni
- Naxospharma srl, Novate Milanese, 20026 Milan, Italy; (C.G.); (P.L.)
| | - Paolo Lombardi
- Naxospharma srl, Novate Milanese, 20026 Milan, Italy; (C.G.); (P.L.)
| | - Valeria Crippa
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Dipartimento di Eccellenza 2018-2022, Centro di Eccellenza sulle Malattie Neurodegenerative, Università degli Studi di Milano, 20133 Milan, Italy; (P.R.); (R.C.); (B.T.); (V.F.); (E.M.); (M.P.); (E.C.); (M.C.); (M.E.C.); (M.G.); (V.C.)
| | - Angelo Poletti
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Dipartimento di Eccellenza 2018-2022, Centro di Eccellenza sulle Malattie Neurodegenerative, Università degli Studi di Milano, 20133 Milan, Italy; (P.R.); (R.C.); (B.T.); (V.F.); (E.M.); (M.P.); (E.C.); (M.C.); (M.E.C.); (M.G.); (V.C.)
- Correspondence:
| |
Collapse
|