1
|
Deng H, Chen Y, Xing J, Zhang N, Xu L. Systematic low-grade chronic inflammation and intrinsic mechanisms in polycystic ovary syndrome. Front Immunol 2024; 15:1470283. [PMID: 39749338 PMCID: PMC11693511 DOI: 10.3389/fimmu.2024.1470283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 12/03/2024] [Indexed: 01/04/2025] Open
Abstract
Polycystic ovary syndrome (PCOS) is a prevalent endocrine and metabolic disorder affecting 6-20% of women of childbearing age worldwide. Immune cell imbalance and dysregulation of inflammatory factors can lead to systematic low-grade chronic inflammation (SLCI), which plays a pivotal role in the pathogenesis of PCOS. A significant higher infiltration of immune cells such as macrophages and lymphocytes and pro-inflammatory factors IL-6 and TNF-α has been detected in PCOS organ systems, impacting not only the female reproductive system but also other organs such as the cardiovascular, intestine, liver, thyroid, brain and other organs. Obesity, insulin resistance (IR), steroid hormones imbalance and intestinal microecological imbalance, deficiencies in vitamin D and selenium, as well as hyperhomocysteinemia (HHcy) can induce systematic imbalance between pro-inflammatory and anti-inflammatory cells and molecules. The pro-inflammatory cells and cytokines also interact with obesity, steroid hormones imbalance and IR, leading to increased metabolic imbalance and reproductive-endocrine dysfunction in PCOS patients. This review aims to summarize the dysregulation of immune response in PCOS organ system and the intrinsic mechanisms affecting SLCI in PCOS to provide new insights for the systemic inflammatory treatment of PCOS in the future.
Collapse
Affiliation(s)
- Hongxia Deng
- Reproductive Endocrinology and Regulation Laboratory, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
| | - Yan Chen
- Reproductive Endocrinology and Regulation Laboratory, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Jilong Xing
- Division of Renal and Endocrinology, Qin Huang Hospital, Xi’an, China
| | - Nannan Zhang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
- National Center for Birth Defect Monitoring, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Liangzhi Xu
- Reproductive Endocrinology and Regulation Laboratory, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
2
|
Mokhosoev IM, Astakhov DV, Terentiev AA, Moldogazieva NT. Human Cytochrome P450 Cancer-Related Metabolic Activities and Gene Polymorphisms: A Review. Cells 2024; 13:1958. [PMID: 39682707 DOI: 10.3390/cells13231958] [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: 10/26/2024] [Revised: 11/15/2024] [Accepted: 11/18/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND Cytochromes P450 (CYPs) are heme-containing oxidoreductase enzymes with mono-oxygenase activity. Human CYPs catalyze the oxidation of a great variety of chemicals, including xenobiotics, steroid hormones, vitamins, bile acids, procarcinogens, and drugs. FINDINGS In our review article, we discuss recent data evidencing that the same CYP isoform can be involved in both bioactivation and detoxification reactions and convert the same substrate to different products. Conversely, different CYP isoforms can convert the same substrate, xenobiotic or procarcinogen, into either a more or less toxic product. These phenomena depend on the type of catalyzed reaction, substrate, tissue type, and biological species. Since the CYPs involved in bioactivation (CYP3A4, CYP1A1, CYP2D6, and CYP2C8) are primarily expressed in the liver, their metabolites can induce hepatotoxicity and hepatocarcinogenesis. Additionally, we discuss the role of drugs as CYP substrates, inducers, and inhibitors as well as the implication of nuclear receptors, efflux transporters, and drug-drug interactions in anticancer drug resistance. We highlight the molecular mechanisms underlying the development of hormone-sensitive cancers, including breast, ovarian, endometrial, and prostate cancers. Key players in these mechanisms are the 2,3- and 3,4-catechols of estrogens, which are formed by CYP1A1, CYP1A2, and CYP1B1. The catechols can also produce quinones, leading to the formation of toxic protein and DNA adducts that contribute to cancer progression. However, 2-hydroxy- and 4-hydroxy-estrogens and their O-methylated derivatives along with conjugated metabolites play cancer-protective roles. CYP17A1 and CYP11A1, which are involved in the biosynthesis of testosterone precursors, contribute to prostate cancer, whereas conversion of testosterone to 5α-dihydrotestosterone as well as sustained activation and mutation of the androgen receptor are implicated in metastatic castration-resistant prostate cancer (CRPC). CYP enzymatic activities are influenced by CYP gene polymorphisms, although a significant portion of them have no effects. However, CYP polymorphisms can determine poor, intermediate, rapid, and ultrarapid metabolizer genotypes, which can affect cancer and drug susceptibility. Despite limited statistically significant data, associations between CYP polymorphisms and cancer risk, tumor size, and metastatic status among various populations have been demonstrated. CONCLUSIONS The metabolic diversity and dual character of biological effects of CYPs underlie their implications in, preliminarily, hormone-sensitive cancers. Variations in CYP activities and CYP gene polymorphisms are implicated in the interindividual variability in cancer and drug susceptibility. The development of CYP inhibitors provides options for personalized anticancer therapy.
Collapse
Affiliation(s)
| | - Dmitry V Astakhov
- Department of Biochemistry, I.M. Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia
| | - Alexander A Terentiev
- Department of Biochemistry and Molecular Biology, N.I. Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | | |
Collapse
|
3
|
Mohan EC, Savarraj JPJ, Colpo GD, Morales D, Finger CE, McAlister A, Ahnstedt H, Choi H, McCullough LD, Manwani B. Aromatase, testosterone, TMPRSS2: determinants of COVID-19 severity. Biol Sex Differ 2024; 15:84. [PMID: 39449074 PMCID: PMC11515603 DOI: 10.1186/s13293-024-00658-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 10/04/2024] [Indexed: 10/26/2024] Open
Abstract
BACKGROUND Male sex has been identified as a risk factor for worse COVID-19 outcomes. This sex difference has been mostly attributed to the complex role of sex hormones. Cell surface entry of SARS-CoV-2 is mediated by the transmembrane protease serine 2 (TMPRSS2) which is under transcriptional regulation by androgens. P450 aromatase enzyme converts androgens to estrogens. This study measured concentrations of aromatase enzyme, testosterone, estradiol, and TMPRSS-2 in plasma of hospitalized COVID-19 patients to elucidate the dynamics of sex-linked disparity in COVID-19 and correlate them with disease severity and mortality. METHODS In this prospective cohort study, a total of 265 patients (41% women), age 18 years and older, who had a positive COVID-19 PCR test and were hospitalized for COVID-19 at Memorial Hermann Hospital in Houston, (between May 2020 and May 2021) were enrolled in the study if met inclusion criteria. Plasma concentrations of Testosterone, aromatase, TMPRSS-2, and estradiol were measured by ELISA. COVID-19 patients were dichotomized based on disease severity into moderate-severe (n = 146) or critical (n = 119). Mann Whitney U and logistic regression were used to correlate the analytes with disease severity and mortality. RESULTS TMPRSS2 (2.5 ± 0.31 vs. 1.73 ± 0.21 ng/mL, p < 0.01) and testosterone (1.2 ± 0.1 vs. 0.44 ± 0.12 ng/mL, p < 0.01) were significantly higher in men as compared to women with COVID-19 after adjusting for age in a multivariate model. There was no sex difference seen in the level of estradiol and aromatase in COVID-19 patients. TMPRSS2 and aromatase were higher, while testosterone was lower in patients with increased COVID-19 severity. They were independently associated with COVID-19 severity, after adjusting for several baseline risk factors in a multivariate logistic regression model. In terms of mortality, TMPRRS2 and aromatase levels were significantly higher in non-survivors. CONCLUSIONS Our study demonstrates that testosterone, aromatase, and TMPRSS2 are markers of COVID-19 severity. Estradiol levels do not change with disease severity in COVID-19. In terms of mortality prediction, higher aromatase and TMPRSS-2 levels can be used to predict mortality from COVID-19 in hospitalized patients. COVID-19 has caused over a million deaths in the U.S., with men often getting sicker than women. Testosterone, a male hormone, helps control a protein called TMPRSS-2, which allows the COVID-19 virus to spread more easily in the body. A protein called aromatase converts the male hormone testosterone into the female hormone estrogen. It is thought that female hormone estrogen helps protect women from getting seriously ill from COVID-19. To understand the role of these hormones in COVID-19 and sex differences, we measured levels of testosterone, estrogen, aromatase (which turns testosterone into estrogen), and TMPRSS-2 in hospitalized COVID-19 patients. We also checked how this level might reflect the severity of the disease. We found that critically ill COVID-19 patients (the ones in ICU) had higher levels of TMPRSS-2 and aromatase, and lower testosterone levels. When we used these hormone levels to predict death in hospitalized COVID-19 patients, higher levels of TMPRSS-2 and aromatase were linked to a lower chance of survival.
Collapse
Affiliation(s)
- Eric C Mohan
- Department of Neurology, University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, 77030, USA
| | - Jude P J Savarraj
- Vivian L. Smith Department of Neurosurgery, University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, 77030, USA
| | - Gabriela D Colpo
- Department of Neurology, University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, 77030, USA
| | - Diego Morales
- Department of Neurology, University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, 77030, USA
| | - Carson E Finger
- Department of Neurology, University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, 77030, USA
| | - Alexis McAlister
- Department of Neurology, University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, 77030, USA
| | - Hilda Ahnstedt
- Department of Neurology, University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, 77030, USA
| | - HuiMahn Choi
- Vivian L. Smith Department of Neurosurgery, University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, 77030, USA
| | - Louise D McCullough
- Department of Neurology, University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, 77030, USA
| | - Bharti Manwani
- Department of Neurology, University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, 77030, USA.
| |
Collapse
|
4
|
Di Stasi V, Contaldo A, Birtolo LI, Shahini E. Interplay of Cardiometabolic Syndrome and Biliary Tract Cancer: A Comprehensive Analysis with Gender-Specific Insights. Cancers (Basel) 2024; 16:3432. [PMID: 39410050 PMCID: PMC11476000 DOI: 10.3390/cancers16193432] [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: 08/24/2024] [Revised: 10/05/2024] [Accepted: 10/09/2024] [Indexed: 10/20/2024] Open
Abstract
BTC overall incidence is globally increasing. CCA, including its subtypes, is a form of BTC. MetS, obesity, MASLD, and diabetes are all linked to CCA in interconnected ways. The link between obesity and CCA is less well-defined in Eastern countries as compared to Western. Although more research is needed to determine the relationship between MASLD and extrahepatic CCA (eCCA), MASLD may be a concurrent risk factor for intrahepatic CCA, particularly in populations with established or unidentified underlying liver disease. Interestingly, the risk of biliary tract cancer (BTC) seemed to be higher in patients with shorter diabetes durations who were not treated with insulin. Therefore, early detection and prevention of chronic liver disease, as well as additional intervention studies, will undoubtedly be required to determine whether improvements to MetS, weight loss, and diabetes therapy can reduce the risk and progression of BTC. However, further studies are needed to understand how reproductive hormones are involved in causing BTC and to develop consistent treatment for patients. Finally, it is critical to carefully assess the cardiological risk in BTC patients due to their increased intrinsic cardiovascular risk, putting them at risk for thrombotic complications, cardiovascular death, cardiac metastasis, and nonbacterial thrombotic endocarditis. This review aimed to provide an updated summary of the relation between the abovementioned cardio-metabolic conditions and BTC.
Collapse
Affiliation(s)
- Vincenza Di Stasi
- Center of Nutrition for the Research and the Care of Obesity and Metabolic Diseases, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, Castellana Grotte, 70013 Bari, Italy;
| | - Antonella Contaldo
- Gastroenterology Unit, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, Castellana Grotte, 70013 Bari, Italy;
| | - Lucia Ilaria Birtolo
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Umberto I Hospital, Sapienza University of Rome, 00185 Rome, Italy;
| | - Endrit Shahini
- Gastroenterology Unit, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, Castellana Grotte, 70013 Bari, Italy;
| |
Collapse
|
5
|
Effah W, Khalil M, Hwang DJ, Miller DD, Narayanan R. Advances in the understanding of androgen receptor structure and function and in the development of next-generation AR-targeted therapeutics. Steroids 2024; 210:109486. [PMID: 39111362 PMCID: PMC11380798 DOI: 10.1016/j.steroids.2024.109486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 07/29/2024] [Accepted: 08/01/2024] [Indexed: 08/10/2024]
Abstract
Androgen receptor (AR) and its ligand androgens are important for development and physiology of various tissues. AR and its ligands also play critical role in the development of various diseases, making it a valuable therapeutic target. AR ligands, both agonists and antagonists, are being widely used to treat pathological conditions, including prostate cancer and hypogonadism. Despite AR being studied widely over the last five decades, the last decade has seen striking advances in the knowledge on AR and discoveries that have the potential to translate to the clinic. This review provides an overview of the advances in AR biology, AR molecular mechanisms of action, and next generation molecules that are currently in development. Several of the areas described in the review are just unraveling and the next decade will bring more clarity on these developments that will put AR at the forefront of both basic biology and drug development.
Collapse
Affiliation(s)
- Wendy Effah
- Department of Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Marjana Khalil
- Department of Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Dong-Jin Hwang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Duane D Miller
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Ramesh Narayanan
- Department of Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States; UTHSC Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN, United States.
| |
Collapse
|
6
|
Valentini Neto J, Almeida Bastos A, Rogero MM, Fisberg RM, Lima Ribeiro SM. Lifestyle aspects are associated with common mental disorders in women over 40 years older in a population-based study. Clin Nutr ESPEN 2024; 64:149-155. [PMID: 39349105 DOI: 10.1016/j.clnesp.2024.09.021] [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: 03/05/2024] [Revised: 09/17/2024] [Accepted: 09/24/2024] [Indexed: 10/02/2024]
Abstract
BACKGROUND & AIMS Common mental disorders (CMD) are more prevalent in women, as well as noncommunicable diseases. Diet and physical activity are lifestyle modifiable factors that might help on managing these conditions. METHODS This study aimed to investigate the association between lifestyle aspects (diet and physical activity) and common mental disorders in women aged 40+ years. METHODS This is a cross-sectional, population-based study (2015 ISA-Nutrition) with a representative sample of São Paulo-SP-Brazil urban residents. The present study was conducted with data from 467 women aged 40+ years old. Common Mental Disorders (dependent variable) were investigated by the Self-Reporting Questionnaire-20 (SRQ-20); the independent variables of interest were: the inflammatory potential of the diet was evaluated by dietary inflammatory index (DII), and physical activity level (evaluated by IPAQ) adopting the leisure dimension of physical activity. We considered as adjusting variables the presence of self-reported Noncommunicable Diseases (NCD), the age intervals (defined as 40-45, 46-50, 51-55, 56-60, 61+ years old), schooling (according to years of formal education), Body Mass Index (BMI); and ethnicity (self-declared skin color). Simple and multi-adjusted logistic regression models were performed to investigate the associations. CONCLUSIONS The main findings indicate that the prevalence of CMD was 32.2 %, and the frequency of having one or more NCD was 67.2 %. In the final regression model, CMD was associated with the highest tertile of the DII (OR = 2.215; p = 0.003) and having three, and four or more NCD (OR = 6.735; p < 0.001, and OR = 3.874; p = 0.033, respectively). Altogether, our results indicate that dietary inflammatory characteristics, and physical activity, along with NCD, are associated with CMD, in women aged 40+ years old, in different dimensions.
Collapse
Affiliation(s)
- João Valentini Neto
- Department of Nutrition, Public Health School, University of Sao Paulo (USP), São Paulo, SP, Brazil.
| | - Amália Almeida Bastos
- Department of Nutrition, Public Health School, University of Sao Paulo (USP), São Paulo, SP, Brazil
| | - Marcelo Macedo Rogero
- Department of Nutrition, Public Health School, University of Sao Paulo (USP), São Paulo, SP, Brazil
| | - Regina Mara Fisberg
- Department of Nutrition, Public Health School, University of Sao Paulo (USP), São Paulo, SP, Brazil
| | - Sandra Maria Lima Ribeiro
- Department of Nutrition, Public Health School, University of Sao Paulo (USP), São Paulo, SP, Brazil; School of Arts, Sciences and Humanities, University of Sao Paulo (USP), São Paulo, SP, Brazil
| |
Collapse
|
7
|
Johnson ML, Hay BA, Maselko M. Altering traits and fates of wild populations with Mendelian DNA sequence modifying Allele Sails. Nat Commun 2024; 15:6665. [PMID: 39138152 PMCID: PMC11322531 DOI: 10.1038/s41467-024-50992-9] [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: 04/05/2024] [Accepted: 07/26/2024] [Indexed: 08/15/2024] Open
Abstract
Population-scale genome modification can alter the composition or fate of wild populations. Synthetic gene drives provide one set of tools, but their use is complicated by scientific, regulatory, and social issues associated with transgene persistence and flow. Here we propose an alternative approach. An Allele Sail consists of a genome editor (the Wind) that introduces DNA sequence edits, and is inherited in a Mendelian fashion. Meanwhile, the edits (the Sail) experience an arithmetic, Super-Mendelian increase in frequency. We model this system and identify contexts in which a single, low frequency release of an editor brings edits to a very high frequency. We also identify conditions in which manipulation of sex determination can bring about population suppression. In regulatory frameworks that distinguish between transgenics (GMO) and their edited non-transgenic progeny (non-GMO) Allele Sails may prove useful since the spread and persistence of the GM component can be limited.
Collapse
Affiliation(s)
- Michelle L Johnson
- Division of Biology and Biological Engineering, California Institute of Technology, 1200 East California Boulevard, MC156-29, Pasadena, CA, 91125, USA
| | - Bruce A Hay
- Division of Biology and Biological Engineering, California Institute of Technology, 1200 East California Boulevard, MC156-29, Pasadena, CA, 91125, USA.
| | - Maciej Maselko
- Applied BioSciences, Macquarie University, North Ryde, NSW, 2109, Australia.
| |
Collapse
|
8
|
Zhang Y, Zhao L, Liu Y, Zhang J, Zheng L, Zheng M. Adverse Event Profiles of the Third-Generation Aromatase Inhibitors: Analysis of Spontaneous Reports Submitted to FAERS. Biomedicines 2024; 12:1708. [PMID: 39200174 PMCID: PMC11351598 DOI: 10.3390/biomedicines12081708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 07/17/2024] [Accepted: 07/27/2024] [Indexed: 09/02/2024] Open
Abstract
The third-generation aromatase inhibitors (AIs), represented by letrozole, anastrozole, and exemestane, have been used as a standard first-line adjuvant therapy for postmenopausal breast cancer patients with positive hormone receptor. However, their safety in the real world has not been systematically analyzed. We used the U.S. Food and Drug Administration Adverse Event Reporting System (FAERS) to investigate adverse event (AE) profiles of the three AIs, covering the period from Q1 2004 to Q3 2023. The time-to-event onset profiles and cumulative incidence were analyzed by Weibull shape parameter test and Kaplan-Meier method, respectively. The disproportionality analysis was utilized to assess drug toxicity risk. Based on the FAERS database, 18,035, 8242, and 7011 reports listing letrozole, anastrozole, and exemestane as primary suspected drugs were extracted, respectively. AEs associated with anastrozole displayed the latest onset (p < 0.0001); meanwhile, WSP test showed that all three AIs had early failure-type profiles. At the preferred term level, we acquired 95, 59, and 42 significant signals associated with letrozole, anastrozole, and exemestane, which involved 18, 13, and 15 system organ classes, respectively. The three AIs all reported that their strongest AE signal was trigger finger. Neutropenia was the most frequent AE for letrozole, while the highest occurrences of anastrozole and exemestane were arthralgia. We also found that interstitial lung disease, a rare but serious AE, showed strong signal intensity in all three AIs. Additionally, letrozole was also associated with lots of other rare but serious AEs in hematologic, respiratory, and hepatic systems, which were not recorded in the instructions. Our analysis of safety warning signals of the third-generation AIs from the FAERS database provided reference for clinical safe and rational drug use.
Collapse
Affiliation(s)
| | | | | | | | | | - Min Zheng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou 310003, China
| |
Collapse
|
9
|
Zhao X, Fan C, Qie T, Fu X, Chen X, Wang Y, Wu Y, Fu X, Shi K, Yan W, Yu H. Diaph1 knockout inhibits mouse primordial germ cell proliferation and affects gonadal development. Reprod Biol Endocrinol 2024; 22:82. [PMID: 39010074 PMCID: PMC11247884 DOI: 10.1186/s12958-024-01257-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 07/09/2024] [Indexed: 07/17/2024] Open
Abstract
BACKGROUND Exploring the molecular mechanisms of primordial germ cell (PGC) migration and the involvement of gonadal somatic cells in gonad development is valuable for comprehending the origins and potential treatments of reproductive-related diseases. METHODS Diaphanous related formin 1 (Diaph1, also known as mDia1) was screened by analyzing publicly available datasets (ATAC-seq, DNase-seq, and RNA-seq). Subsequently, the CRISPR-Cas9 technology was used to construct Diaph1 knockout mice to investigate the role of Diaph1 in gonad development. RESULTS Based on data from public databases, a differentially expressed gene Diaph1, was identified in the migration of mouse PGC. Additionally, the number of PGCs was significantly reduced in Diaph1 knockout mice compared to wild type mice, and the expression levels of genes related to proliferation (Dicer1, Mcm9), adhesion (E-cadherin, Cdh1), and migration (Cxcr4, Hmgcr, Dazl) were significantly decreased. Diaph1 knockout also inhibited Leydig cell proliferation and induced apoptosis in the testis, as well as granulosa cell apoptosis in the ovary. Moreover, the sperm count in the epididymal region and the count of ovarian follicles were significantly reduced in Diaph1 knockout mice, resulting in decreased fertility, concomitant with lowered levels of serum testosterone and estradiol. Further research found that in Diaph1 knockout mice, the key enzymes involved in testosterone synthesis (CYP11A1, 3β-HSD) were decreased in Leydig cells, and the estradiol-associated factor (FSH receptor, AMH) in granulosa cells were also downregulated. CONCLUSIONS Overall, our findings indicate that the knockout of Diaph1 can disrupt the expression of factors that regulate sex hormone production, leading to impaired secretion of sex hormones, ultimately resulting in damage to reproductive function. These results provide a new perspective on the molecular mechanisms underlying PGC migration and gonadal development, and offer valuable insights for further research on the causes, diagnosis, and treatment of related diseases.
Collapse
Affiliation(s)
- Xin Zhao
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, 010020, Inner Mongolia, China
| | - Chunbiao Fan
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, 010020, Inner Mongolia, China
| | - Tongtong Qie
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, 010020, Inner Mongolia, China
| | - Xinrui Fu
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, 010020, Inner Mongolia, China
| | - Xiaoshuang Chen
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, 010020, Inner Mongolia, China
| | - Yujia Wang
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, 010020, Inner Mongolia, China
| | - Yuan Wu
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, 010020, Inner Mongolia, China
| | - Xinyao Fu
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, 010020, Inner Mongolia, China
| | - Kesong Shi
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, 010020, Inner Mongolia, China
| | - Wenlong Yan
- School of Biology and Agriculture, Shaoguan University, Shaoguan, 512005, Guangdong Province, China.
| | - Haiquan Yu
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, 010020, Inner Mongolia, China.
| |
Collapse
|
10
|
Hetemäki N, Robciuc A, Vihma V, Haanpää M, Hämäläinen E, Tikkanen MJ, Mikkola TS, Savolainen-Peltonen H. Adipose Tissue Sex Steroids in Postmenopausal Women with and without Menopausal Hormone Therapy. J Clin Endocrinol Metab 2024:dgae458. [PMID: 38986008 DOI: 10.1210/clinem/dgae458] [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: 02/11/2024] [Revised: 06/15/2024] [Accepted: 07/02/2024] [Indexed: 07/12/2024]
Abstract
CONTEXT The decrease in serum estrogens after menopause is associated with a shift from a gynoid to an android adipose tissue (AT) distribution. Menopausal hormone therapy (HT) mitigates this change and accompanying metabolic dysfunction, but its effects on AT sex steroid metabolism have not been characterized. OBJECTIVE We studied effects of HT on subcutaneous and visceral AT estrogen and androgen concentrations and metabolism in postmenopausal women. DESIGN, SETTING, PATIENTS, AND INTERVENTIONS Serum and subcutaneous and visceral AT from 63 postmenopausal women with (n=50) and without (n=13) per oral HT were analyzed for estrone, estradiol, progesterone, testosterone, androstenedione, dehydroepiandrosterone, and serum estrone sulfate using liquid chromatography-tandem mass spectrometry. Steroid sulfatase activity was measured using radiolabeled precursors. mRNA expression of genes encoding sex steroid-metabolizing enzymes and receptors was performed using real-time reverse transcription quantitative polymerase chain reaction. RESULTS HT users had 4- to 7-fold higher concentrations of estrone and estradiol in subcutaneous and visceral AT, and 30% lower testosterone in visceral AT compared to non-users. Estrogen-to-androgen ratios were 4- to 12-fold higher in AT of users compared to non-users of HT. In visceral AT, estrogen-to-androgen ratios increased with HT estradiol dose. AT to serum ratios of estrone and estradiol remained high in HT users. CONCLUSIONS Higher local estrogen to androgen ratios and high AT to serum ratios of estrogen concentrations in HT users suggest that HT may significantly influence intracrine sex steroid metabolism in AT, and these local changes could be involved in the preventive effect of HT on menopause-associated abdominal adiposity.
Collapse
Affiliation(s)
- Natalia Hetemäki
- Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, FIN-00029 HUS, Helsinki, Finland
| | - Alexandra Robciuc
- Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, FIN-00029 HUS, Helsinki, Finland
| | - Veera Vihma
- Department of General Practice and Primary Health Care, University of Helsinki, FIN-00014, Helsinki, Finland
| | - Mikko Haanpää
- HUSLAB, Helsinki University Hospital, FIN-00029 HUS, Helsinki, Finland
| | - Esa Hämäläinen
- Departments of Clinical Chemistry, University of Helsinki, FIN-00029 HUS, Helsinki, and University of Eastern Finland, FIN-70210, Kuopio, Finland
| | - Matti J Tikkanen
- Heart and Lung Center, University of Helsinki and Helsinki University Hospital, FIN-00029 HUS, Helsinki, Finland
| | - Tomi S Mikkola
- Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, FIN-00029 HUS, Helsinki, Finland
| | - Hanna Savolainen-Peltonen
- Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, FIN-00029 HUS, Helsinki, Finland
| |
Collapse
|
11
|
Fedeli V, Unfer V, Dinicola S, Laganà AS, Canipari R, Monti N, Querqui A, Galante E, Laurenzi G, Bizzarri M. Inositol Restores Appropriate Steroidogenesis in PCOS Ovaries Both In Vitro and In Vivo Experimental Mouse Models. Cells 2024; 13:1171. [PMID: 39056753 PMCID: PMC11275052 DOI: 10.3390/cells13141171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 06/29/2024] [Accepted: 07/05/2024] [Indexed: 07/28/2024] Open
Abstract
Androgen excess is a key feature of several clinical phenotypes of polycystic ovary syndrome (PCOS). However, the presence of FSH receptor (FSHR) and aromatase (CYP19A1) activity responses to physiological endocrine stimuli play a critical role in the pathogenesis of PCOS. Preliminary data suggest that myo-Inositol (myo-Ins) and D-Chiro-Inositol (D-Chiro-Ins) may reactivate CYP19A1 activity. We investigated the steroidogenic pathway of Theca (TCs) and Granulosa cells (GCs) in an experimental model of murine PCOS induced in CD1 mice exposed for 10 weeks to a continuous light regimen. The effect of treatment with different combinations of myo-Ins and D-Chiro-Ins on the expression of Fshr, androgenic, and estrogenic enzymes was analyzed by real-time PCR in isolated TCs and GCs and in ovaries isolated from healthy and PCOS mice. Myo-Ins and D-Chiro-Ins, at a ratio of 40:1 at pharmacological and physiological concentrations, positively modulate the steroidogenic activity of TCs and the expression of Cyp19a1 and Fshr in GCs. Moreover, in vivo, inositols (40:1 ratio) significantly increase Cyp19a1 and Fshr. These changes in gene expression are mirrored by modifications in hormone levels in the serum of treated animals. Myo-Ins and D-Chiro-Ins in the 40:1 formula efficiently rescued PCOS features by up-regulating aromatase and FSHR levels while down-regulating androgen excesses produced by TCs.
Collapse
Affiliation(s)
- Valeria Fedeli
- Department of Experimental Medicine, University La Sapienza, 00185 Rome, Italy; (N.M.); (A.Q.); (E.G.)
- Systems Biology Group Lab, University La Sapienza, 00185 Rome, Italy;
- The Experts Group on Inositol in Basic and Clinical Research, and on PCOS (EGOI-PCOS), 00161 Rome, Italy;
| | - Vittorio Unfer
- The Experts Group on Inositol in Basic and Clinical Research, and on PCOS (EGOI-PCOS), 00161 Rome, Italy;
- Dept. of Gynaecology, UniCamillus—Saint Camillus International University of Health Sciences, 00131 Rome, Italy
| | - Simona Dinicola
- Systems Biology Group Lab, University La Sapienza, 00185 Rome, Italy;
- The Experts Group on Inositol in Basic and Clinical Research, and on PCOS (EGOI-PCOS), 00161 Rome, Italy;
| | - Antonio Simone Laganà
- Unit of Obstetrics and Gynecology, “Paolo Giaccone” Hospital, Department of Health Promotion, Mother and ChildCare, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90127 Palermo, Italy;
| | - Rita Canipari
- Department of Anatomy, Histology, Forensic Medicine and Orthopedic, Unit of Histology and Medical Embryology, Sapienza, University of Rome, 00185 Rome, Italy; (R.C.); (G.L.)
| | - Noemi Monti
- Department of Experimental Medicine, University La Sapienza, 00185 Rome, Italy; (N.M.); (A.Q.); (E.G.)
- Systems Biology Group Lab, University La Sapienza, 00185 Rome, Italy;
| | - Alessandro Querqui
- Department of Experimental Medicine, University La Sapienza, 00185 Rome, Italy; (N.M.); (A.Q.); (E.G.)
- Systems Biology Group Lab, University La Sapienza, 00185 Rome, Italy;
| | - Emanuele Galante
- Department of Experimental Medicine, University La Sapienza, 00185 Rome, Italy; (N.M.); (A.Q.); (E.G.)
- Systems Biology Group Lab, University La Sapienza, 00185 Rome, Italy;
| | - Gaia Laurenzi
- Department of Anatomy, Histology, Forensic Medicine and Orthopedic, Unit of Histology and Medical Embryology, Sapienza, University of Rome, 00185 Rome, Italy; (R.C.); (G.L.)
| | - Mariano Bizzarri
- Department of Experimental Medicine, University La Sapienza, 00185 Rome, Italy; (N.M.); (A.Q.); (E.G.)
- Systems Biology Group Lab, University La Sapienza, 00185 Rome, Italy;
- The Experts Group on Inositol in Basic and Clinical Research, and on PCOS (EGOI-PCOS), 00161 Rome, Italy;
| |
Collapse
|
12
|
Costa NDSD, Lima LS, Galiciolli MEA, Ribeiro DHF, Ribeiro MM, Garica GDPJ, Marçal IS, Silva JFD, Pereira ME, Oliveira CS, Guiloski IC. Drug-induced osteoporosis and mechanisms of bone tissue regeneration through trace elements. J Trace Elem Med Biol 2024; 84:127446. [PMID: 38615498 DOI: 10.1016/j.jtemb.2024.127446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 03/30/2024] [Accepted: 04/03/2024] [Indexed: 04/16/2024]
Abstract
Osteoporosis is associated with an imbalance in bone formation, with certain drugs used in disease treatment being implicated in its development. Supplementation with trace elements may contribute to bone regeneration, offering an alternative approach by enhancing bone mineral density (BMD) and thereby thwarting the onset of osteoporosis. This review aims to assess the mechanisms through which trace elements such as copper (Cu), iron (Fe), selenium (Se), manganese (Mn), and zinc (Zn) are linked to increased bone mass, thus mitigating the effects of pharmaceuticals. Our findings underscore that the use of drugs such as aromatase inhibitors (AIs), proton pump inhibitors (PPIs), antiretrovirals, glucocorticoids, opioids, or anticonvulsants can result in decreased BMD, a primary contributor to osteoporosis. Research indicates that essential elements like Cu, Fe, Se, Mn, and Zn, through various mechanisms, can bolster BMD and forestall the onset of the disease, owing to their protective effects. Consequently, our study recommends a minimum daily intake of these essential minerals for patients undergoing treatment with the aforementioned drugs, as the diverse mechanisms governing the effects of trace elements Cu, Fe, Mn, Se, and Zn facilitate bone remodeling.
Collapse
Affiliation(s)
- Nayara de Souza da Costa
- Instituto de Pesquisas Pelé Pequeno Príncipe, Curitiba 80035-000, Brazil; Faculdades Pequeno Príncipe, Curitiba 80230-020, Brazil
| | - Luíza Siqueira Lima
- Instituto de Pesquisas Pelé Pequeno Príncipe, Curitiba 80035-000, Brazil; Faculdades Pequeno Príncipe, Curitiba 80230-020, Brazil
| | - Maria Eduarda Andrade Galiciolli
- Instituto de Pesquisas Pelé Pequeno Príncipe, Curitiba 80035-000, Brazil; Faculdades Pequeno Príncipe, Curitiba 80230-020, Brazil
| | - Deborah Helen Fabiano Ribeiro
- Instituto de Pesquisas Pelé Pequeno Príncipe, Curitiba 80035-000, Brazil; Faculdades Pequeno Príncipe, Curitiba 80230-020, Brazil
| | - Milena Mariano Ribeiro
- Instituto de Pesquisas Pelé Pequeno Príncipe, Curitiba 80035-000, Brazil; Faculdades Pequeno Príncipe, Curitiba 80230-020, Brazil
| | - Gisele de Paula Júlia Garica
- Instituto de Pesquisas Pelé Pequeno Príncipe, Curitiba 80035-000, Brazil; Faculdades Pequeno Príncipe, Curitiba 80230-020, Brazil
| | - Isabela Saragioto Marçal
- Instituto de Pesquisas Pelé Pequeno Príncipe, Curitiba 80035-000, Brazil; Faculdades Pequeno Príncipe, Curitiba 80230-020, Brazil
| | - Juliana Ferreira da Silva
- Instituto de Pesquisas Pelé Pequeno Príncipe, Curitiba 80035-000, Brazil; Faculdades Pequeno Príncipe, Curitiba 80230-020, Brazil
| | - Meire Ellen Pereira
- Instituto de Pesquisas Pelé Pequeno Príncipe, Curitiba 80035-000, Brazil; Faculdades Pequeno Príncipe, Curitiba 80230-020, Brazil
| | - Cláudia Sirlene Oliveira
- Instituto de Pesquisas Pelé Pequeno Príncipe, Curitiba 80035-000, Brazil; Faculdades Pequeno Príncipe, Curitiba 80230-020, Brazil
| | - Izonete Cristina Guiloski
- Instituto de Pesquisas Pelé Pequeno Príncipe, Curitiba 80035-000, Brazil; Faculdades Pequeno Príncipe, Curitiba 80230-020, Brazil.
| |
Collapse
|
13
|
Clinical practice guidelines for full-cycle standardized management of bone health in breast cancer patients. CANCER INNOVATION 2024; 3:e111. [PMID: 38948531 PMCID: PMC11212291 DOI: 10.1002/cai2.111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 01/07/2024] [Accepted: 01/09/2024] [Indexed: 07/02/2024]
Abstract
Bone health management for breast cancer spans the entire cycle of patient care, including the prevention and treatment of bone loss caused by early breast cancer treatment, the adjuvant application of bone-modifying agents to improve prognosis, and the diagnosis and treatment of advanced bone metastases. Making good bone health management means formulating appropriate treatment strategies and dealing with adverse drug reactions, and will help to improve patients' quality of life and survival rates. The Breast Cancer Expert Committee of the National Cancer Center for Quality Control organized relevant experts to conduct an in-depth discussion on the full-cycle management of breast cancer bone health based on evidence-based medicine, and put forward reasonable suggestions to guide clinicians to better deal with health issues in bone health clinics.
Collapse
|
14
|
Gonçalves LT, Costa DTD, Rouver WDN, Santos RLD. Testosterone modulates vasodilation in mesenteric arteries of hypertensive rats. Life Sci 2024; 338:122405. [PMID: 38176584 DOI: 10.1016/j.lfs.2023.122405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 12/22/2023] [Accepted: 12/28/2023] [Indexed: 01/06/2024]
Abstract
AIMS To evaluate the effects of testosterone on endothelium-dependent vasodilation and oxidative stress in mesenteric resistance arteries. MAIN METHODS Spontaneously hypertensive rats (SHR), aged 8 to 10 weeks, were divided into four groups: intact (SHAM), intact treated with testosterone (TTO; 3 mg/kg/day) via subcutaneous route (s.c.), intact treated with testosterone and anastrozole [aromatase enzyme inhibitor (TTO + ANA; 0.1 mg/kg/day, s.c.)] and intact treated with testosterone and finasteride [5 α-reductase enzyme inhibitor (TTO + FIN; 5 mg/kg/day, s.c.)] for four weeks. Concentration-response curves to acetylcholine (ACh, 0.1 nmol/L - 10 μmol/L) were obtained in mesenteric resistance arteries previously contracted with phenylephrine (PE, 3 μmol/L), before and after the use of selective inhibitors. Reactive oxygen species (ROS) levels were assessed in the vessels and the endothelium analyzed by scanning electron microscopy. KEY FINDINGS TTO group showed a lower participation of nitric oxide (NO), increased oxidative stress, and participation of prostanoids and endothelium-dependent hyperpolarization (EDH), possibly to maintain the vasodilator response. Lower participation of NO and prostanoids, combined to an increased participation of EDH, were observed in the TTO + ANA group, in addition to higher levels of ROS and altered endothelial morphology. The vasodilation to ACh was impaired in TTO + FIN, along increased participation of NO, reduction of prostanoids, and greater EDH-dependent vasodilation. SIGNIFICANCE Testosterone contributes to endothelial vasodilation by enhancing EDH through an increased participation of epoxyeicosatrienoic acids. While the decrease in NO appears to involve the participation of dihydrotestosterone, 17 β-estradiol seems to stimulate the action of the NO pathway and prostanoids.
Collapse
Affiliation(s)
- Leticia Tinoco Gonçalves
- Department of Physiological Sciences, Health Sciences Center, Universidade Federal do Espírito Santo (UFES), Vitoria, ES, Brazil
| | - Débora Tacon da Costa
- Department of Physiological Sciences, Health Sciences Center, Universidade Federal do Espírito Santo (UFES), Vitoria, ES, Brazil
| | - Wender do Nascimento Rouver
- Department of Physiological Sciences, Health Sciences Center, Universidade Federal do Espírito Santo (UFES), Vitoria, ES, Brazil
| | - Roger Lyrio Dos Santos
- Department of Physiological Sciences, Health Sciences Center, Universidade Federal do Espírito Santo (UFES), Vitoria, ES, Brazil.
| |
Collapse
|
15
|
Alvarado-Torres JK, Morales-Silva R, Sanabria Ponce de Leon A, Rodriguez-Torres G, Perez-Torres J, Perez-Perez Y, Mueller D, Sepulveda-Orengo MT. Estradiol reduction through aromatase inhibition impairs cocaine seeking in male rats. Front Behav Neurosci 2024; 17:1307606. [PMID: 38292056 PMCID: PMC10824998 DOI: 10.3389/fnbeh.2023.1307606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 12/26/2023] [Indexed: 02/01/2024] Open
Abstract
Introduction Clinical and preclinical research on cocaine use disorder (CUD) has shown that sex differences in drug seeking are influenced by hormonal fluctuations. Estradiol (E2), a sex steroid hormone, has been linked to female drug effects, vulnerability to use/abuse, and psychosocial factors. Preclinical studies show that estradiol in females facilitates the extinction of cocaine-seeking behavior indicating a possible role in regulating extinction learning. Similar to females, males' brains contain the aromatase enzyme which converts testosterone to estradiol. However, it is unclear whether estradiol plays a role in male extinction learning as it does in females. Furthermore, how endogenously aromatized estradiol affects drug addiction in males is unknown. Therefore, this study investigated whether endogenous estradiol regulates cocaine seeking in male rats. We hypothesized that decreased aromatase enzyme activity, resulting in decreased estradiol synthesis in male brains, will impair extinction learning leading to increased cocaine-seeking behavior. Methods This hypothesis was tested using cocaine-conditioned place preference (CPP), and short access self-administration (SA), followed by extinction and reinstatement. Before each extinction session for CPP or SA, male rats received an injection of either 1 (low dose) or 2.5 mg/kg (high dose) of the aromatase inhibitor Fadrozole (FAD), or vehicle. Results FAD groups showed dose-dependent effects on cocaine-seeking behavior compared to the vehicle group during CPP extinction. Specifically, low dose FAD facilitated extinction of cocaine CPP, whereas high dose FAD impaired it. In contrast, neither dose of FAD had any effects on the extinction of cocaine SA. Interestingly, only the low dose FAD group had decreased active lever pressing during cue- and cocaine-primed reinstatement compared to the vehicle group. Neither dose of FAD had an effect on sucrose extinction or reinstatement of sucrose seeking. Discussion These results from CPP experiments suggest that estradiol may impact extinction learning, as a low dose of FAD may strengthen the formation of cocaine extinction memory. Additionally, in male rats undergoing cocaine SA, the same low dose of aromatase inhibitor effectively reduced reinstatement of cocaine-seeking behavior. Thus, estradiol impacts cocaine seeking and extinction in both males and females, and it may also influence the development of sex-specific treatment strategies for CUD.
Collapse
Affiliation(s)
- John K. Alvarado-Torres
- Basic Sciences Department, Ponce Research Institute, Ponce Health Sciences University, Ponce, Puerto Rico
| | - Roberto Morales-Silva
- Basic Sciences Department, Ponce Research Institute, Ponce Health Sciences University, Ponce, Puerto Rico
| | | | - Genesis Rodriguez-Torres
- Basic Sciences Department, Ponce Research Institute, Ponce Health Sciences University, Ponce, Puerto Rico
| | - Joshua Perez-Torres
- Basic Sciences Department, Ponce Research Institute, Ponce Health Sciences University, Ponce, Puerto Rico
| | - Yobet Perez-Perez
- Basic Sciences Department, Ponce Research Institute, Ponce Health Sciences University, Ponce, Puerto Rico
| | - Devin Mueller
- Department of Biological Sciences, Kent State University, Kent, OH, United States
| | - Marian T. Sepulveda-Orengo
- Basic Sciences Department, Ponce Research Institute, Ponce Health Sciences University, Ponce, Puerto Rico
| |
Collapse
|
16
|
Xia M, Zheng J, Chen S, Tang Y, Wang S, Ji Z, Hao T, Li H, Li L, Ge RS, Liu Y. Bisphenol a alternatives suppress human and rat aromatase activity: QSAR structure-activity relationship and in silico docking analysis. Food Chem Toxicol 2024; 183:114257. [PMID: 38040240 DOI: 10.1016/j.fct.2023.114257] [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: 09/20/2023] [Revised: 11/16/2023] [Accepted: 11/21/2023] [Indexed: 12/03/2023]
Abstract
The use of alternative substances to replace bisphenol A (BPA) has been encouraged. The objective of this study was to evaluate the effects of BPA and 9 BPA alternatives on human and rat aromatase (CYP19A1) in human and rat placental microsomes. The results revealed that bisphenol A, AP, B, C, E, F, FL, S, and Z, and 4,4'-thiodiphenol (TDP) inhibited human CYP19A1 and bisphenol A, AP, B, C, FL, Z, and TDP inhibited rat CYP19A1. The IC50 values of human CYP19A1 ranged from 3.3 to 172.63 μM and those of rat CYP19A1 ranged from 2.20 to over 100 μM. BPA alternatives were mixed/competitive inhibitors and inhibited estradiol production in BeWo placental cells. Molecular docking analysis showed that BPA alternatives bind to the domain between heme and steroid and form a hydrogen bond with catalytic residue Met374. Pharmacophore analysis showed that there were one hydrogen bond donor, one hydrophobic region, and one ring aromatic hydrophobic region. Bivariate correlation analysis showed that molecular weight, alkyl atom weight, and LogP of BPA alternatives were inversely correlated with their IC50 values. In conclusion, BPA alternatives can inhibit human and rat CYP19A1 and the lipophilicity and the substituted alkyl size determines their inhibitory strength.
Collapse
Affiliation(s)
- Miaomiao Xia
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China; Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital and Yuying Children's Hospital, Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Jingyi Zheng
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Sailin Chen
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Yunbing Tang
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Shaowei Wang
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Zhongyao Ji
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Ting Hao
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Huitao Li
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Linxi Li
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China; Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital and Yuying Children's Hospital, Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China; Key Laboratory of Environment and Male Reproductive Medicine of Wenzhou, and Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, 325000, Zhejiang Province, China.
| | - Ren-Shan Ge
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China; Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital and Yuying Children's Hospital, Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China; Key Laboratory of Environment and Male Reproductive Medicine of Wenzhou, and Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, 325000, Zhejiang Province, China.
| | - Yi Liu
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China.
| |
Collapse
|
17
|
Hosoi T, Yakabe M, Hashimoto S, Akishita M, Ogawa S. The roles of sex hormones in the pathophysiology of age-related sarcopenia and frailty. Reprod Med Biol 2024; 23:e12569. [PMID: 38476959 PMCID: PMC10927916 DOI: 10.1002/rmb2.12569] [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: 12/30/2023] [Revised: 02/13/2024] [Accepted: 02/26/2024] [Indexed: 03/14/2024] Open
Abstract
Background Sarcopenia is an age-related condition characterized by a progressive and systemic decline in skeletal muscle mass, quality, and strength. The incidence of sarcopenia contains sex-specific aspects, indicating the contribution of sex hormones to its pathophysiology. This review focuses on changing trends in sarcopenia, discusses alterations in definitions and diagnostic criteria, and emphasizes the association between sarcopenia and sex hormones. Methods A literature search was performed on PubMed for related articles published between 1997 and December 2023 using appropriate keywords. Main Findings Results Advances in research have emphasized the significance of muscle quality and strength over muscle mass, resulting in new diagnostic criteria for sarcopenia. Androgens demonstrated anabolic effects on skeletal muscles and played a significant role in the pathophysiology of sarcopenia. In clinical settings, androgen replacement therapy has exhibited certain positive outcomes for treating sarcopenia, despite concerns about potential side effects. Conversely, estrogen is involved in skeletal muscle maintenance, but the detailed mechanisms remain unclear. Moreover, results regarding the clinical application of estrogen replacement therapy for treating sarcopenia remained inconsistent. Conclusion The elucidation of molecular mechanisms that involve sex hormones is eagerly awaited for novel therapeutic interventions for sarcopenia.
Collapse
Affiliation(s)
- Tatsuya Hosoi
- Department of Geriatric Medicine, Graduate School of MedicineThe University of TokyoBunkyo‐ku, TokyoJapan
| | - Mitsutaka Yakabe
- Department of Geriatric Medicine, Graduate School of MedicineThe University of TokyoBunkyo‐ku, TokyoJapan
| | - Seiji Hashimoto
- Department of Geriatric Medicine, Graduate School of MedicineThe University of TokyoBunkyo‐ku, TokyoJapan
| | - Masahiro Akishita
- Department of Geriatric Medicine, Graduate School of MedicineThe University of TokyoBunkyo‐ku, TokyoJapan
| | - Sumito Ogawa
- Department of Geriatric Medicine, Graduate School of MedicineThe University of TokyoBunkyo‐ku, TokyoJapan
| |
Collapse
|
18
|
Eissa MA, Gohar EY. Aromatase enzyme: Paving the way for exploring aromatization for cardio-renal protection. Biomed Pharmacother 2023; 168:115832. [PMID: 37931519 PMCID: PMC10843764 DOI: 10.1016/j.biopha.2023.115832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 10/15/2023] [Accepted: 10/31/2023] [Indexed: 11/08/2023] Open
Abstract
Documented male-female differences in the risk of cardiovascular and chronic kidney diseases have been largely attributed to estrogens. The cardiovascular and renal protective effects of estrogens are mediated via the activation of estrogen receptors (ERα and ERβ) and G protein-coupled estrogen receptor, and involve interactions with the renin-angiotensin-aldosterone system. Aromatase, also called estrogen synthase, is a cytochrome P-450 enzyme that plays a pivotal role in the conversion of androgens into estrogens. Estrogens are biosynthesized in gonadal and extra-gonadal sites by the action of aromatase. Evidence suggests that aromatase inhibitors, which are used to treat high estrogen-related pathologies, are associated with the development of cardiovascular events. We review the potential role of aromatization in providing cardio-renal protection and highlight several meta-analysis studies on cardiovascular events associated with aromatase inhibitors. Overall, we present the potential of aromatase enzyme as a fundamental contributor to cardio-renal protection.
Collapse
Affiliation(s)
- Manar A Eissa
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Merit University, New Sohag, Sohag, Egypt
| | - Eman Y Gohar
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States.
| |
Collapse
|
19
|
Sumangala N, Im SC, Valentín-Goyco J, Auchus RJ. Influence of cholesterol on kinetic parameters for human aromatase (P450 19A1) in phospholipid nanodiscs. J Inorg Biochem 2023; 247:112340. [PMID: 37544101 PMCID: PMC11260420 DOI: 10.1016/j.jinorgbio.2023.112340] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 07/20/2023] [Accepted: 07/22/2023] [Indexed: 08/08/2023]
Abstract
Cholesterol, a significant constituent of the endoplasmic reticulum membrane, exerts a substantial effect on the membrane's biophysical and mechanical properties. Cholesterol, however, is often neglected in model systems used to study membrane-bound proteins. For example, the influence of cholesterol on the enzymatic functions of type 2 cytochromes P450, which require a phospholipid bilayer and the redox partner P450-oxidoreductase (POR) for activity, are rarely investigated. Human aromatase (P450 19A1) catalyzes three sequential oxygenations of 19‑carbon steroids to estrogens and is widely expressed across various tissues, which are characterized by varying cholesterol compositions. Our study examined the impact of cholesterol on the functionality of the P450 19A1 complex with POR. Nanodiscs containing P450 19A1 with 20% cholesterol/80% phospholipid had similar rates and affinity of androstenedione binding as phospholipid-only P450 19A1 nanodiscs, and rates of product formation were indistinguishable among these conditions. In contrast, the rate of the first electron transfer from POR to P450 19A1 was 3-fold faster in cholesterol-containing nanodiscs than in phospholipid-only nanodiscs. These results suggest that cholesterol influences some aspects of POR interaction with P450 19A1 and might serve as an additional regulatory mechanism in this catalytic system.
Collapse
Affiliation(s)
- Nirupama Sumangala
- Division of Metabolism, Endocrinology, & Diabetes, Department of Internal Medicine, Ann Arbor, MI 48109, USA; Program in Biophysics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Sang-Choul Im
- Division of Metabolism, Endocrinology, & Diabetes, Department of Internal Medicine, Ann Arbor, MI 48109, USA; Department of Pharmacology, University of Michigan, Ann Arbor, MI 48109, USA; Veterans Affairs Medical Center, Ann Arbor, MI 48105, United States
| | - Juan Valentín-Goyco
- Division of Metabolism, Endocrinology, & Diabetes, Department of Internal Medicine, Ann Arbor, MI 48109, USA; Department of Pharmacology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Richard J Auchus
- Division of Metabolism, Endocrinology, & Diabetes, Department of Internal Medicine, Ann Arbor, MI 48109, USA; Program in Biophysics, University of Michigan, Ann Arbor, MI 48109, USA; Department of Pharmacology, University of Michigan, Ann Arbor, MI 48109, USA; Veterans Affairs Medical Center, Ann Arbor, MI 48105, United States.
| |
Collapse
|
20
|
Widjiati W, Kuntjorodjakti S, Ananda AT, Sajida MVP, Ilmi AF, Adisti MZ, Chou D, Luqman EM. The Effect of Administering Forest Honey to Rats Exposed to Physical Stress on Corticosteroid Levels, Folliculogenesis and the Number of Corpus Luteum. BIOMEDICAL AND PHARMACOLOGY JOURNAL 2023; 16:1431-1440. [DOI: 10.13005/bpj/2721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Livestock that has stress releases glucocorticoids in response to it, and it causes inhibition of the hypothalamus-pituitary-gonadal axis (HPG) signaling pathway so that it reduces reproductive efficiency. Forest honey can reduce corticosteroid levels as a stress response from physical stress induction which is expected to increase reproductive efficiency including folliculogenesis and the formation of the corpus luteum. This study aims to determine the effect of forest honey on rats (Rattus novergicus) exposed to physical stress on corticosteroid levels, folliculogenesis, and the number of corpus luteum. This study is an experimental laboratory one using 32 rats which were divided into 4 treatment groups; control positive (C) treated with physical stress, treatment 1 (T1) treated with physical stress + honey 2 g/rat/day PO, treatment 2 (T2) treated with physical stress + honey 4 g/rat/day PO and treatment 3 (T3) treated with physical stress + honey 6 g/rat/day PO. All treatments were carried out for 14 days. The results showed that T1 had the lowest corticosteroid level compared to all treatment groups and the corticosteroid level of this group was significantly different (p <0.05) compared to that of C and T3. The folliculogenesis profile showed that the number of primary secondary, tertiary, and Graafian follicles of group T1 was significantly different (p<0.05) compared to that of C, T2, and T3. In terms of the number of corpus luteum, it showed that T1 had the highest number of corpus luteum, and the number of corpus luteum in this group was significantly different (p<0.05) from that of C, T2, and T3. It can be concluded that the administration of forest honey at a dose of 2g/rat/day could reduce corticosteroid levels, improve the folliculogenesis profile, and increase the number of corpus luteum in rats exposed to physical stress. The use of forest honey could reduce corticosteroid levels as a stress response from physical stress induction which was expected to increase reproductive efficiency.
Collapse
Affiliation(s)
- Widjiati Widjiati
- 1Department of Veterinary Science, Faculty of Veterinary Medicine, Universitas Airlangga, Surabaya 60115, Jawa Timur, Indonesia,
| | - Suryo Kuntjorodjakti
- 1Department of Veterinary Science, Faculty of Veterinary Medicine, Universitas Airlangga, Surabaya 60115, Jawa Timur, Indonesia,
| | - Aditya Tri Ananda
- 2Postgraduate Reproductive Biology, Faculty of Veterinary Medicine, Universitas Airlangga, Surabaya 60115, Jawa Timur, Indonesia
| | - Mey Vanda Pusparina Sajida
- 2Postgraduate Reproductive Biology, Faculty of Veterinary Medicine, Universitas Airlangga, Surabaya 60115, Jawa Timur, Indonesia
| | - Alivia Fairuz Ilmi
- 2Postgraduate Reproductive Biology, Faculty of Veterinary Medicine, Universitas Airlangga, Surabaya 60115, Jawa Timur, Indonesia
| | - Meisa Zalfa Adisti
- 3Graduate Student Faculty of Veterinary Medicine, Universitas Airlangga, Surabaya 60115, Jawa Timur, Indonesia,
| | - Dean Chou
- 4Department of Biomedical Engineering National Cheng Kung University, No. 1, Dasyue Rd, East District, Tainan City, Taiwan
| | - Epy Muhammad Luqman
- 1Department of Veterinary Science, Faculty of Veterinary Medicine, Universitas Airlangga, Surabaya 60115, Jawa Timur, Indonesia,
| |
Collapse
|
21
|
Belluti S, Imbriano C, Casarini L. Nuclear Estrogen Receptors in Prostate Cancer: From Genes to Function. Cancers (Basel) 2023; 15:4653. [PMID: 37760622 PMCID: PMC10526871 DOI: 10.3390/cancers15184653] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/01/2023] [Accepted: 09/18/2023] [Indexed: 09/29/2023] Open
Abstract
Estrogens are almost ubiquitous steroid hormones that are essential for development, metabolism, and reproduction. They exert both genomic and non-genomic action through two nuclear receptors (ERα and ERβ), which are transcription factors with disregulated functions and/or expression in pathological processes. In the 1990s, the discovery of an additional membrane estrogen G-protein-coupled receptor augmented the complexity of this picture. Increasing evidence elucidating the specific molecular mechanisms of action and opposing effects of ERα and Erβ was reported in the context of prostate cancer treatment, where these issues are increasingly investigated. Although new approaches improved the efficacy of clinical therapies thanks to the development of new molecules targeting specifically estrogen receptors and used in combination with immunotherapy, more efforts are needed to overcome the main drawbacks, and resistance events will be a challenge in the coming years. This review summarizes the state-of-the-art on ERα and ERβ mechanisms of action in prostate cancer and promising future therapies.
Collapse
Affiliation(s)
- Silvia Belluti
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (S.B.); (C.I.)
| | - Carol Imbriano
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (S.B.); (C.I.)
| | - Livio Casarini
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Ospedale di Baggiovara, 41126 Modena, Italy
| |
Collapse
|
22
|
Bardhi O, Palmer BF, Clegg DJ. The evolutionary impact and influence of oestrogens on adipose tissue structure and function. Philos Trans R Soc Lond B Biol Sci 2023; 378:20220207. [PMID: 37482787 PMCID: PMC10363706 DOI: 10.1098/rstb.2022.0207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 06/06/2023] [Indexed: 07/25/2023] Open
Abstract
Oestrogens are sex steroid hormones that have gained prominence over the years owing to their crucial roles in human health and reproduction functions which have been preserved throughout evolution. One of oestrogens actions, and the focus of this review, is their ability to determine adipose tissue distribution, function and adipose tissue 'health'. Body fat distribution is sexually dimorphic, affecting males and females differently. These differences are also apparent in the development of the metabolic syndrome and other chronic conditions where oestrogens are critical. In this review, we summarize the different molecular mechanisms, pathways and resulting pathophysiology which are a result of oestrogens actions in and on adipose tissues. This article is part of a discussion meeting issue 'Causes of obesity: theories, conjectures and evidence (Part I)'.
Collapse
Affiliation(s)
- Olgert Bardhi
- Department of Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Biff F. Palmer
- Division of Nephrology, Department of Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Deborah J. Clegg
- Vice President for Research, Texas Tech Health Sciences Center, El Paso, TX 75390, USA
| |
Collapse
|
23
|
Jones TH, Dobs AS, Randeva H, Moore W, Parkin JM. Leflutrozole in male obesity-associated hypogonadotropic hypogonadism: Ph 2b double-blind randomised controlled trial. Eur J Endocrinol 2023; 189:297-308. [PMID: 37579053 DOI: 10.1093/ejendo/lvad099] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 03/27/2023] [Accepted: 05/24/2023] [Indexed: 08/16/2023]
Abstract
OBJECTIVE Assessment of the efficacy and safety/tolerability of the aromatase inhibitor leflutrozole to normalise testosterone in Obesity-associated Hypogonadotropic Hypogonadism (OHH). DESIGN Placebo-controlled, double-blind, RCT, in 70 sites in Europe/USA. METHODS Patient inclusion criteria: men with BMI of 30-50 kg/m2, morning total testosterone (TT) < 10.41 nmol/L, and two androgen deficiency symptoms (at least one of sexual dysfunction). Patients randomised to weekly leflutrozole (0.1/0.3/1.0 mg) or placebo for 24 weeks. Primary endpoint: normalisation of TT levels in ≥75% of patients after 24 weeks. Secondary endpoints (included): time to TT normalisation and change in LH/FSH. Safety was assessed through adverse events and laboratory monitoring. RESULTS AND CONCLUSIONS Of 2103 screened, 271 were randomised, 81 discontinued. Demographic characteristics were similar across groups. Mean BMI was 38.1 kg/m2 and TT 7.97 nmol/L. The primary endpoint was achieved in all leflutrozole-treated groups by 24 weeks with a dose-tiered response; mean TT 15.89; 17.78; 20.35 nmol/L, for leflutrozole 0.1 mg, 0.3 mg, and 1.0 mg groups respectively, vs 8.04 nmol/L for placebo. LH/FSH significantly increased in leflutrozole vs placebo groups. No improvements in body composition or sexual dysfunction were observed. Semen volume/total motile sperm count improved with leflutrozole vs placebo. Treatment-emergent adverse events, more common in leflutrozole-treated groups included, raised haematocrit, hypertension, increased PSA, and headache. Some reduction in lumbar bone density was observed with leflutrozole (mean -1.24%, -1.30%, -2.09%) and 0.66% for 0.1 mg, 0.3 mg, 1.0 mg, and placebo, respectively, without change at the hip. This RCT of leflutrozole in OHH demonstrated normalisation of TT in obese men. FSH/LH and semen parameter changes support that leflutrozole may preserve/improve testicular function. CLINICAL TRIAL REGISTRATION NUMBER NCT02730169.
Collapse
Affiliation(s)
- T Hugh Jones
- Centre for Diabetes and Endocrinology, Barnsley Hospital NHS Trust, Barnsley, United Kingdom
- Department of Oncology and Metabolism, University of Sheffield Medical School, Sheffield, United Kingdom
| | - Adrian S Dobs
- Division of Endocrinology and Metabolism, The Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Harpal Randeva
- Clinical Sciences Research Laboratories, Warwick Medical School, University of Warwick, Coventry, United Kingdom
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM) and the Human Metabolism Research Unit (HMRU), Coventry, United Kingdom
| | | | | |
Collapse
|
24
|
Szukiewicz D. Insight into the Potential Mechanisms of Endocrine Disruption by Dietary Phytoestrogens in the Context of the Etiopathogenesis of Endometriosis. Int J Mol Sci 2023; 24:12195. [PMID: 37569571 PMCID: PMC10418522 DOI: 10.3390/ijms241512195] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 07/25/2023] [Accepted: 07/27/2023] [Indexed: 08/13/2023] Open
Abstract
Phytoestrogens (PEs) are estrogen-like nonsteroidal compounds derived from plants (e.g., nuts, seeds, fruits, and vegetables) and fungi that are structurally similar to 17β-estradiol. PEs bind to all types of estrogen receptors, including ERα and ERβ receptors, nuclear receptors, and a membrane-bound estrogen receptor known as the G protein-coupled estrogen receptor (GPER). As endocrine-disrupting chemicals (EDCs) with pro- or antiestrogenic properties, PEs can potentially disrupt the hormonal regulation of homeostasis, resulting in developmental and reproductive abnormalities. However, a lack of PEs in the diet does not result in the development of deficiency symptoms. To properly assess the benefits and risks associated with the use of a PE-rich diet, it is necessary to distinguish between endocrine disruption (endocrine-mediated adverse effects) and nonspecific effects on the endocrine system. Endometriosis is an estrogen-dependent disease of unknown etiopathogenesis, in which tissue similar to the lining of the uterus (the endometrium) grows outside of the uterus with subsequent complications being manifested as a result of local inflammatory reactions. Endometriosis affects 10-15% of women of reproductive age and is associated with chronic pelvic pain, dysmenorrhea, dyspareunia, and infertility. In this review, the endocrine-disruptive actions of PEs are reviewed in the context of endometriosis to determine whether a PE-rich diet has a positive or negative effect on the risk and course of endometriosis.
Collapse
Affiliation(s)
- Dariusz Szukiewicz
- Department of Biophysics, Physiology & Pathophysiology, Faculty of Health Sciences, Medical University of Warsaw, 02-004 Warsaw, Poland
| |
Collapse
|
25
|
Olasore HSA, Oyedeji TA, Olawale MO, Ogundele OI, Faleti JOO. Relationship between testosterone-estradiol ratio and some anthropometric and metabolic parameters among Nigerian men. Metabol Open 2023; 18:100249. [PMID: 37396673 PMCID: PMC10313505 DOI: 10.1016/j.metop.2023.100249] [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: 05/09/2023] [Revised: 05/29/2023] [Accepted: 06/02/2023] [Indexed: 07/04/2023] Open
Abstract
Background Alterations in sex hormone levels are implicated in the regulation of metabolic processes in men. In recent years, the prevalence of metabolic disorders, such as obesity, insulin resistance, and type 2 diabetes, has risen in Nigeria. In men, these disorders may be associated with the ratio of serum testosterone to estradiol levels. Therefore, we investigated the relationship between the testosterone-estradiol (T/E2) ratio, anthropometry, and metabolic parameters in Nigerian men. Method Eighty-five adult men were recruited for this study. Participants' data such as age, weight, height, BMI, and waist circumference were collected. Plasma total testosterone and estradiol levels, as well as metabolic parameters such as fasting blood sugar, creatinine, urea, HDL cholesterol, total cholesterol, and triglycerides levels, were determined. The data were analyzed using SPSS version 25 software. Results Anthropometric parameters such as weight, height, BMI, and waist circumference showed a negative correlation with plasma T/E2 (r = -0.265, -0.288, -0.106, -0.204; p = 0.007, 0.004, 0.167, 0.061 respectively). However, the T/E2 ratio showed a positive correlation with the metabolic parameters such as fasting blood sugar, HDL cholesterol levels, plasma creatinine, and urea (r = 0.219, 0.096, 0.992, 0.152; p = 0.022, 0.192, <0.001, 0.082 respectively), while there were negative correlations with total cholesterol and triglycerides levels (r = -0.200, -0.083; p = 0.034, 0.226 respectively). Conclusion These findings show that there are significant correlations between the T/E2 ratio and weight, height, fasting blood sugar, creatinine, and urea, while there are no significant correlations between T/E2 ratio and BMI, waist circumference, HDL-cholesterol, and triglycerides.
Collapse
Affiliation(s)
- Holiness Stephen Adedeji Olasore
- Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine of the University of Lagos, Idi Araba, Lagos, Nigeria
| | - Tolulope Adejoke Oyedeji
- Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine of the University of Lagos, Idi Araba, Lagos, Nigeria
| | - Matthew Olamide Olawale
- Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine of the University of Lagos, Idi Araba, Lagos, Nigeria
| | | | - Joseph Ogo-Oluwa Faleti
- Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine of the University of Lagos, Idi Araba, Lagos, Nigeria
| |
Collapse
|
26
|
Patel J, Chen S, Katzmeyer T, Pei YA, Pei M. Sex-dependent variation in cartilage adaptation: from degeneration to regeneration. Biol Sex Differ 2023; 14:17. [PMID: 37024929 PMCID: PMC10077643 DOI: 10.1186/s13293-023-00500-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 03/20/2023] [Indexed: 04/08/2023] Open
Abstract
Despite acknowledgement in the scientific community of sex-based differences in cartilage biology, the implications for study design remain unclear, with many studies continuing to arbitrarily assign demographics. Clinically, it has been well-established that males and females differ in cartilage degeneration, and accumulating evidence points to the importance of sex differences in the field of cartilage repair. However, a comprehensive review of the mechanisms behind this trend and the influence of sex on cartilage regeneration has not yet been presented. This paper aims to summarize current findings regarding sex-dependent variation in knee anatomy, sex hormones' effect on cartilage, and cartilaginous degeneration and regeneration, with a focus on stem cell therapies. Findings suggest that the stem cells themselves, as well as their surrounding microenvironment, contribute to sex-based differences. Accordingly, this paper underscores the contribution of both stem cell donor and recipient sex to sex-related differences in treatment efficacy. Cartilage regeneration is a field that needs more research to optimize strategies for better clinical results; taking sex into account could be a big factor in developing more effective and personalized treatments. The compilation of this information emphasizes the importance of investing further research in sex differences in cartilage biology.
Collapse
Affiliation(s)
- Jhanvee Patel
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, 64 Medical Center Drive, PO Box 9196, Morgantown, WV, 26506-9196, USA
| | - Song Chen
- Department of Orthopaedics, The General Hospital of Western Theater Command, Chengdu, 610083, Sichuan, China
| | - Torey Katzmeyer
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, 64 Medical Center Drive, PO Box 9196, Morgantown, WV, 26506-9196, USA
| | - Yixuan Amy Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, 64 Medical Center Drive, PO Box 9196, Morgantown, WV, 26506-9196, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, 64 Medical Center Drive, PO Box 9196, Morgantown, WV, 26506-9196, USA.
- WVU Cancer Institute, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV, 26506, USA.
| |
Collapse
|
27
|
Cobos-Ontiveros LA, Romero-Hernández LL, Mastranzo-Sánchez EB, Colín-Lozano B, Puerta A, Padrón JM, Merino-Montiel P, Vega Baez JL, Montiel-Smith S. Synthesis, antiproliferative evaluation and in silico studies of a novel steroidal spiro morpholinone. Steroids 2023; 192:109173. [PMID: 36621620 DOI: 10.1016/j.steroids.2023.109173] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 12/20/2022] [Accepted: 12/23/2022] [Indexed: 01/07/2023]
Abstract
Estrogens play a pivotal role in the development of estrogen-dependent breast cancer and other hormone-dependent disorders. A common strategy to overcome the pathological effects of estrogens is the use of aromatase inhibitors (AIs), which bind to the enzyme and prevent the union with the natural substrate, decreasing the amount of estrogens produced. Several AIs have been developed, including inhibitors with a steroidal backbone and a nitrogen heterocycle in their structure. Encouraged by the notable results presented by current and clinical steroidal drugs, herein we present the synthesis of a steroidal spiro morpholinone derivative as a plausible aromatase inhibitor. The morpholinone derivative was synthesized over a six-step methodology starting from estrone. The title compound and its hydroxychloroacetamide derivative precursor were evaluated for their antiproliferative profile against estrogen-dependent and independent solid tumor cell lines: A549, HBL-100, HeLa, SW1573, T-47D and WiDr. Both compounds exhibited a potent antiproliferative activity in the micromolar range against the six cancer cell lines, with the hydroxychloroacetamide derivative precursor being a more potent inhibitor (GI50 = 0.25-2.4 µM) than the morpholinone derivative (GI50 = 2.0-11 µM). Furthermore, both compounds showed, in almost all cases, better GI50 values than the steroidal anticancer drugs abiraterone and galeterone. Docking simulations of the derivatives were performed in order to explain the experimental biological activity. The results showed interactions with the iron heme (derivative 3) and important residues of the steroidal binding-site (Met374) for the inhibition of human aromatase. A correlation was found between in vitro assays and the score obtained from the molecular docking study.
Collapse
Affiliation(s)
- Luis A Cobos-Ontiveros
- Facultad de Ciencias Químicas, Ciudad Universitaria, Benemérita Universidad Autónoma de Puebla, 72570 Puebla, Puebla, Mexico
| | - Laura L Romero-Hernández
- Facultad de Ciencias Químicas, Ciudad Universitaria, Benemérita Universidad Autónoma de Puebla, 72570 Puebla, Puebla, Mexico.
| | - Eduardo B Mastranzo-Sánchez
- Facultad de Ciencias Químicas, Ciudad Universitaria, Benemérita Universidad Autónoma de Puebla, 72570 Puebla, Puebla, Mexico
| | - Blanca Colín-Lozano
- Facultad de Ciencias Químicas, Ciudad Universitaria, Benemérita Universidad Autónoma de Puebla, 72570 Puebla, Puebla, Mexico
| | - Adrián Puerta
- BioLab, Instituto Universitario de Bio-Orgánica "Antonio González" (IUBO-AG), Universidad de La Laguna, c/ Astrofísico Francisco Sánchez 2, 38206 La Laguna, Spain
| | - José M Padrón
- BioLab, Instituto Universitario de Bio-Orgánica "Antonio González" (IUBO-AG), Universidad de La Laguna, c/ Astrofísico Francisco Sánchez 2, 38206 La Laguna, Spain
| | - Penélope Merino-Montiel
- Facultad de Ciencias Químicas, Ciudad Universitaria, Benemérita Universidad Autónoma de Puebla, 72570 Puebla, Puebla, Mexico.
| | - Jose Luis Vega Baez
- Facultad de Ciencias Químicas, Ciudad Universitaria, Benemérita Universidad Autónoma de Puebla, 72570 Puebla, Puebla, Mexico
| | - Sara Montiel-Smith
- Facultad de Ciencias Químicas, Ciudad Universitaria, Benemérita Universidad Autónoma de Puebla, 72570 Puebla, Puebla, Mexico
| |
Collapse
|
28
|
Wang S, Zhang A, Pan Y, Liu L, Niu S, Zhang F, Liu X. Association between COVID-19 and Male Fertility: Systematic Review and Meta-Analysis of Observational Studies. World J Mens Health 2023; 41:311-329. [PMID: 36326165 PMCID: PMC10042646 DOI: 10.5534/wjmh.220091] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 07/29/2022] [Accepted: 08/03/2022] [Indexed: 11/06/2022] Open
Abstract
PURPOSE Whether COVID-19 reduces male fertility remains requires further investigation. This meta-analysis and systematic review evaluated the impact of COVID-19 on male fertility. MATERIALS AND METHODS The literature in PubMed, Embase, MEDLINE, Web of Science, and Cochrane Library up to January 01, 2022 was systematically searched, and a meta-analysis was conducted to investigate the effect of COVID-19 on male fertility. Totally 17 studies with a total of 1,627 patients and 1,535 control subjects were included in our meta-analysis. RESULTS Regarding sperm quality, COVID-19 decreased the total sperm count (p=0.012), sperm concentration (p=0.001), total motility (p=0.001), progressive sperm motility (p=0.048), and viability (p=0.031). Subgroup analyses showed that different control group populations did not change the results. It was found that during the illness stage of COVID-19, semen volume decreased, and during the recovery stage of COVID-19, sperm concentration and total motility decreased <90 days. We found that sperm concentration and total motility decreased during recovery for ≥90 days. Fever because of COVID-19 significantly reduced sperm concentration and progressive sperm motility, and COVID-19 without fever ≥90 days, the sperm total motility and progressive sperm motility decreased. Regarding disease severity, the moderate type of COVID-19 significantly reduced sperm total motility, but not the mild type. Regarding sex hormones, COVID-19 increased prolactin and estradiol. Subgroup analyses showed that during the illness stage, COVID-19 decreased testosterone (T) levels and increased luteinizing hormone levels. A potential publication bias may have existed in our meta-analysis. CONCLUSIONS COVID-19 in men significantly reduced sperm quality and caused sex hormone disruption. COVID-19 had long-term effects on sperm quality, especially on sperm concentration and total motility. It is critical to conduct larger multicenter studies to determine the consequences of COVID-19 on male fertility.
Collapse
Affiliation(s)
- Shangren Wang
- Department of Urology, Tianjin Medical University General Hospital, Tianjin, China
| | - Aiqiao Zhang
- Department of Neonatology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Department of Neonatology, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Yang Pan
- Department of Urology, Tianjin Medical University General Hospital, Tianjin, China
| | - Li Liu
- Department of Urology, Tianjin Medical University General Hospital, Tianjin, China
| | - Shuai Niu
- Department of Urology, Tianjin Medical University General Hospital, Tianjin, China
| | - Fujun Zhang
- Department of Neonatology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Department of Neonatology, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Xiaoqiang Liu
- Department of Urology, Tianjin Medical University General Hospital, Tianjin, China.
| |
Collapse
|
29
|
Khaw YM, Anwar S, Zhou J, Kawano T, Lin P, Otero A, Barakat R, Drnevich J, Takahashi T, Ko CJ, Inoue M. Estrogen receptor alpha signaling in dendritic cells modulates autoimmune disease phenotype in mice. EMBO Rep 2023; 24:e54228. [PMID: 36633157 PMCID: PMC9986829 DOI: 10.15252/embr.202154228] [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: 10/26/2021] [Revised: 11/23/2022] [Accepted: 12/16/2022] [Indexed: 01/13/2023] Open
Abstract
Estrogen is a disease-modifying factor in multiple sclerosis (MS) and its animal model experimental autoimmune encephalomyelitis (EAE) via estrogen receptor alpha (ERα). However, the mechanisms by which ERα signaling contributes to changes in disease pathogenesis have not been completely elucidated. Here, we demonstrate that ERα deletion in dendritic cells (DCs) of mice induces severe neurodegeneration in the central nervous system in a mouse EAE model and resistance to interferon beta (IFNβ), a first-line MS treatment. Estrogen synthesized by extragonadal sources is crucial for controlling disease phenotypes. Mechanistically, activated ERα directly interacts with TRAF3, a TLR4 downstream signaling molecule, to degrade TRAF3 via ubiquitination, resulting in reduced IRF3 nuclear translocation and transcription of membrane lymphotoxin (mLT) and IFNβ components. Diminished ERα signaling in DCs generates neurotoxic effector CD4+ T cells via mLT-lymphotoxin beta receptor (LTβR) signaling. Lymphotoxin beta receptor antagonist abolished EAE disease symptoms in the DC-specific ERα-deficient mice. These findings indicate that estrogen derived from extragonadal sources, such as lymph nodes, controls TRAF3-mediated cytokine production in DCs to modulate the EAE disease phenotype.
Collapse
Affiliation(s)
- Yee Ming Khaw
- Department of Comparative BiosciencesUniversity of Illinois at Urbana‐ChampaignUrbanaILUSA
- Neuroscience ProgramUniversity of Illinois at Urbana‐ChampaignUrbanaILUSA
| | - Shehata Anwar
- Department of Comparative BiosciencesUniversity of Illinois at Urbana‐ChampaignUrbanaILUSA
- Department of Pathology, Faculty of Veterinary MedicineBeni‐Suef University (BSU)Beni‐SuefEgypt
| | - Jinyan Zhou
- Department of Comparative BiosciencesUniversity of Illinois at Urbana‐ChampaignUrbanaILUSA
- Neuroscience ProgramUniversity of Illinois at Urbana‐ChampaignUrbanaILUSA
| | - Tasuku Kawano
- Department of Comparative BiosciencesUniversity of Illinois at Urbana‐ChampaignUrbanaILUSA
- Division of Pathophysiology, Department of Pharmaceutical Sciences, Faculty of Pharmaceutical SciencesTohoku Medical and Pharmaceutical UniversitySendaiJapan
| | - Po‐Ching Lin
- Department of Comparative BiosciencesUniversity of Illinois at Urbana‐ChampaignUrbanaILUSA
| | - Ashley Otero
- Department of Comparative BiosciencesUniversity of Illinois at Urbana‐ChampaignUrbanaILUSA
- Neuroscience ProgramUniversity of Illinois at Urbana‐ChampaignUrbanaILUSA
| | - Radwa Barakat
- Department of Comparative BiosciencesUniversity of Illinois at Urbana‐ChampaignUrbanaILUSA
- Department of Toxicology and Forensic MedicineCollege of Veterinary Medicine, Benha UniversityQalyubiaEgypt
| | - Jenny Drnevich
- Roy J. Carver Biotechnology CenterUniversity of Illinois Urbana‐ChampaignUrbanaILUSA
| | - Tomoko Takahashi
- Division of Pathophysiology, Department of Pharmaceutical Sciences, Faculty of Pharmaceutical SciencesTohoku Medical and Pharmaceutical UniversitySendaiJapan
| | - CheMyong Jay Ko
- Department of Comparative BiosciencesUniversity of Illinois at Urbana‐ChampaignUrbanaILUSA
- Neuroscience ProgramUniversity of Illinois at Urbana‐ChampaignUrbanaILUSA
| | - Makoto Inoue
- Department of Comparative BiosciencesUniversity of Illinois at Urbana‐ChampaignUrbanaILUSA
- Neuroscience ProgramUniversity of Illinois at Urbana‐ChampaignUrbanaILUSA
- Beckman Institute for Advanced Science and TechnologyUrbanaILUSA
| |
Collapse
|
30
|
Dubol M, Immenschuh J, Jonasson M, Takahashi K, Niwa T, Hosoya T, Roslin S, Wikström J, Antoni G, Watanabe Y, Lubberink M, Biegon A, Sundström-Poromaa I, Comasco E. Acute nicotine exposure blocks aromatase in the limbic brain of healthy women: A [ 11C]cetrozole PET study. Compr Psychiatry 2023; 123:152381. [PMID: 36905856 DOI: 10.1016/j.comppsych.2023.152381] [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: 10/14/2022] [Revised: 02/07/2023] [Accepted: 02/28/2023] [Indexed: 03/13/2023] Open
Abstract
BACKGROUND Of interest to women's mental health, a wealth of studies suggests sex differences in nicotine addiction and treatment response, but their psychoneuroendocrine underpinnings remain largely unknown. A pathway involving sex steroids could indeed be involved in the behavioural effects of nicotine, as it was found to inhibit aromatase in vitro and in vivo in rodents and non-human primates, respectively. Aromatase regulates the synthesis of oestrogens and, of relevance to addiction, is highly expressed in the limbic brain. METHODS The present study sought to investigate in vivo aromatase availability in relation to exposure to nicotine in healthy women. Structural magnetic resonance imaging and two [11C]cetrozole positron emission tomography (PET) scans were performed to assess the availability of aromatase before and after administration of nicotine. Gonadal hormones and cotinine levels were measured. Given the region-specific expression of aromatase, a ROI-based approach was employed to assess changes in [11C]cetrozole non-displaceable binding potential. RESULTS The highest availability of aromatase was found in the right and left thalamus. Upon nicotine exposure, [11C]cetrozole binding in the thalamus was acutely decreased bilaterally (Cohen's d = -0.99). In line, cotinine levels were negatively associated with aromatase availability in the thalamus, although as non-significant trend. CONCLUSIONS These findings indicate acute blocking of aromatase availability by nicotine in the thalamic area. This suggests a new putative mechanism mediating the effects of nicotine on human behaviour, particularly relevant to sex differences in nicotine addiction.
Collapse
Affiliation(s)
- Manon Dubol
- Department of Women's and Children's Health, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Jana Immenschuh
- Department of Women's and Children's Health, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - My Jonasson
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Kayo Takahashi
- RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Takashi Niwa
- RIKEN Center for Biosystems Dynamics Research, Kobe, Japan; Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo, Japan
| | - Takamitsu Hosoya
- RIKEN Center for Biosystems Dynamics Research, Kobe, Japan; Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo, Japan
| | - Sara Roslin
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Johan Wikström
- Department of Surgical Sciences, Neuroradiology, Uppsala University, Sweden
| | - Gunnar Antoni
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | | | - Mark Lubberink
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Anat Biegon
- Departments of Radiology and Neurology, Stony Brook University School of Medicine, Stony Brook, NY, USA
| | | | - Erika Comasco
- Department of Women's and Children's Health, Science for Life Laboratory, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
31
|
Kim SY, Park GI, Park SY, Lee EH, Choi H, Koh JT, Han S, Choi MH, Park EK, Kim IS, Kim JE. Gulp1 deficiency augments bone mass in male mice by affecting osteoclasts due to elevated 17β-estradiol levels. J Cell Physiol 2023; 238:1006-1019. [PMID: 36870066 DOI: 10.1002/jcp.30987] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 01/19/2023] [Accepted: 02/14/2023] [Indexed: 03/05/2023]
Abstract
The engulfment adaptor phosphotyrosine-binding domain containing 1 (GULP1) is an adaptor protein involved in the engulfment of apoptotic cells via phagocytosis. Gulp1 was first found to promote the phagocytosis of apoptotic cells by macrophages, and its role in various tissues, including neurons and ovaries, has been well studied. However, the expression and function of GULP1 in bone tissue are poorly understood. Consequently, to determine whether GULP1 plays a role in the regulation of bone remodeling in vitro and in vivo, we generated Gulp1 knockout (KO) mice. Gulp1 was expressed in bone tissue, mainly in osteoblasts, while its expression is very low in osteoclasts. Microcomputed tomography and histomorphometry analysis in 8-week-old male Gulp1 KO mice revealed a high bone mass in comparison with male wild-type (WT) mice. This was a result of decreased osteoclast differentiation and function in vivo and in vitro as confirmed by a reduced actin ring and microtubule formation in osteoclasts. Gas chromatography-mass spectrometry analysis further showed that both 17β-estradiol (E2) and 2-hydroxyestradiol levels, and the E2/testosterone metabolic ratio, reflecting aromatase activity, were also higher in the bone marrow of male Gulp1 KO mice than in male WT mice. Consistent with mass spectrometry analysis, aromatase enzymatic activity was significantly higher in the bone marrow of male Gulp1 KO mice. Altogether, our results suggest that GULP1 deficiency decreases the differentiation and function of osteoclasts themselves and increases sex steroid hormone-mediated inhibition of osteoclast differentiation and function, rather than affecting osteoblasts, resulting in a high bone mass in male mice. To the best of our knowledge, this is the first study to explore the direct and indirect roles of GULP1 in bone remodeling, providing new insights into its regulation.
Collapse
Affiliation(s)
- Soon-Young Kim
- Department of Molecular Medicine, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, Republic of Korea.,BK21 Four KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Science, Kyungpook National University, Daegu, Republic of Korea
| | - Gun-Il Park
- Department of Molecular Medicine, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Seung-Yoon Park
- Department of Biochemistry, School of Medicine, Dongguk University, Gyeongju, Republic of Korea
| | - Eun-Hye Lee
- Department of Molecular Medicine, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Hyuck Choi
- Department of Pharmacology and Dental Therapeutics, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea
| | - Jeong-Tae Koh
- Department of Pharmacology and Dental Therapeutics, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea
| | - Soyun Han
- Molecular Recognition Research Center, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Man Ho Choi
- Molecular Recognition Research Center, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Eui Kyun Park
- Department of Oral Pathology and Regenerative Medicine, School of Dentistry, Kyungpook National University, Daegu, Republic of Korea
| | - In-San Kim
- Center for Theragnosis, Biomedical Research Institute, Korea Institute Science and Technology, Seoul, Republic of Korea.,KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea
| | - Jung-Eun Kim
- Department of Molecular Medicine, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, Republic of Korea.,BK21 Four KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Science, Kyungpook National University, Daegu, Republic of Korea
| |
Collapse
|
32
|
Szukiewicz D. Aberrant epigenetic regulation of estrogen and progesterone signaling at the level of endometrial/endometriotic tissue in the pathomechanism of endometriosis. VITAMINS AND HORMONES 2023; 122:193-235. [PMID: 36863794 DOI: 10.1016/bs.vh.2022.11.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Endometriosis is a term referring to a condition whereby the endometrial tissue is found outside the uterine cavity. This progressive and debilitating condition affects up to 15% of women of reproductive age. Due to the fact that endometriosis cells may express estrogen receptors (ERα, Erβ, GPER) and progesterone (P4) receptors (PR-A, PR-B), their growth, cyclic proliferation, and breakdown are similar to the processes occurring in the endometrium. The underlying etiology and pathogenesis of endometriosis are still not fully explained. The retrograde transport of viable menstrual endometrial cells with the retained ability to attach within the pelvic cavity, proliferate, differentiate and invade into the surrounding tissue explains the most widely accepted implantation theory. Endometrial stromal cells (EnSCs) with clonogenic potential constitute the most abundant population of cells within endometrium that resemble the properties of mesenchymal stem cells (MSCs). Accordingly, formation of the endometriotic foci in endometriosis may be due to a kind of EnSCs dysfunction. Increasing evidence indicates the underestimated role of epigenetic mechanisms in the pathogenesis of endometriosis. Hormone-mediated epigenetic modifications of the genome in EnSCs or even MSCs were attributed an important role in the etiopathogenesis of endometriosis. The roles of excess estrogen exposure and P4 resistance were also found to be crucial in the development of epigenetic homeostasis failure. Therefore, the aim of this review was to consolidate the current knowledge regarding the epigenetic background of EnSCs and MSCs and the changed properties due to estrogen/P4 imbalances in the context of the etiopathogenesis of endometriosis.
Collapse
Affiliation(s)
- Dariusz Szukiewicz
- Department of Biophysics, Physiology & Pathophysiology, Faculty of Health Sciences, Medical University of Warsaw, Warsaw, Poland.
| |
Collapse
|
33
|
Trinkner P, Günther S, Monsef I, Kerschbaum E, von Bergwelt-Baildon M, Cordas Dos Santos DM, Theurich S. Survival and immunotoxicities in association with sex-specific body composition patterns of cancer patients undergoing immune-checkpoint inhibitor therapy - A systematic review and meta-analysis. Eur J Cancer 2023; 184:151-171. [PMID: 36931074 DOI: 10.1016/j.ejca.2023.01.030] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 01/28/2023] [Accepted: 01/30/2023] [Indexed: 02/19/2023]
Abstract
BACKGROUND Imbalanced body composition is mechanistically connected to dysregulated immune activities. Whether overweight/obesity or sarcopenia has an impact on treatment results in cancer patients undergoing immune checkpoint inhibitor (ICI) therapy is currently under debate. We aimed to answer if survival rates and occurrence of immune-related adverse events (irAEs) were different in obese or sarcopenic patients. METHODS A systematic search was conducted in PubMed, Embase and CENTRAL for all records published until July 2022 using specific search terms for body composition in combination with terms for ICI regimens. Two authors screened independently. All studies that reported on body mass index or sarcopenia measures were selected for further analysis. RESULTS 48 studies reporting on overweight/obesity comprising of 19,767 patients, and 32 studies reporting on sarcopenia comprising of 3193 patients fulfilled the inclusion criteria. In the entire cohort, overweight/obesity was significantly associated with better progression-free survival (PFS; p = 0.009) and overall survival (OS; p <0.00001). Subgroup analyses stratified by sex revealed that overweight/obese males had the strongest survival benefit (PFS: p = 0.05; OS: p = 0.0005), and overweight/obese female patients did not show any. However, overweight/obese patients of both sexes had a higher risk to develop irAEs grade ≥3 (p = 0.0009). Sarcopenic patients showed significantly shorter PFS (p <0.0001) and OS (p <0.0001). The frequency of irAEs did not differ between sarcopenic and non-sarcopenic patients. CONCLUSION This meta-analysis suggests that body composition is associated in a sex-specific manner with survival and irAEs in cancer patients undergoing ICI treatment.
Collapse
Affiliation(s)
- Paul Trinkner
- Department of Medicine III, University Hospital, LMU Munich, Munich, Germany; Cancer- and Immunometabolism Research Group, Gene Center, LMU Munich, Munich, Germany
| | - Sophie Günther
- Department of Medicine III, University Hospital, LMU Munich, Munich, Germany; Cancer- and Immunometabolism Research Group, Gene Center, LMU Munich, Munich, Germany
| | - Ina Monsef
- Evidence-based Medicine, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany
| | - Eva Kerschbaum
- Comprehensive Cancer Center Munich (CCCM), Munich, Germany
| | - Michael von Bergwelt-Baildon
- Department of Medicine III, University Hospital, LMU Munich, Munich, Germany; Comprehensive Cancer Center Munich (CCCM), Munich, Germany; German Cancer Consortium (DKTK), Partner Site Munich, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - David M Cordas Dos Santos
- Department of Medicine III, University Hospital, LMU Munich, Munich, Germany; Cancer- and Immunometabolism Research Group, Gene Center, LMU Munich, Munich, Germany; German Cancer Consortium (DKTK), Partner Site Munich, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sebastian Theurich
- Department of Medicine III, University Hospital, LMU Munich, Munich, Germany; Cancer- and Immunometabolism Research Group, Gene Center, LMU Munich, Munich, Germany; German Cancer Consortium (DKTK), Partner Site Munich, and German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
34
|
Saito M, Okamatsu-Ogura Y. Thermogenic Brown Fat in Humans: Implications in Energy Homeostasis, Obesity and Metabolic Disorders. World J Mens Health 2023:41.e26. [PMID: 36792089 DOI: 10.5534/wjmh.220224] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 11/08/2022] [Indexed: 01/27/2023] Open
Abstract
In mammals including humans, there are two types of adipose tissue, white and brown adipose tissues (BATs). White adipose tissue is the primary site of energy storage, while BAT is a specialized tissue for non-shivering thermogenesis to dissipate energy as heat. Although BAT research has long been limited mostly in small rodents, the rediscovery of metabolically active BAT in adult humans has dramatically promoted the translational studies on BAT in health and diseases. It is now established that BAT, through its thermogenic and energy dissipating activities, plays a role in the regulation of body temperature, whole-body energy expenditure, and body fatness. Moreover, increasing evidence has demonstrated that BAT secretes various paracrine and endocrine factors, which influence other peripheral tissues and control systemic metabolic homeostasis, suggesting BAT as a metabolic regulator, other than for thermogenesis. In fact, clinical studies have revealed an association of BAT not only with metabolic disorders such as insulin resistance, diabetes, dyslipidemia, and fatty liver, but also with cardiovascular diseases including hypertension and atherosclerosis. Thus, BAT is an intriguing tissue combating obesity and related metabolic diseases. In this review, we summarize current knowledge on human BAT, focusing its patho-physiological roles in energy homeostasis, obesity and related metabolic disorders. The effects of aging and sex on BAT are also discussed.
Collapse
Affiliation(s)
- Masayuki Saito
- Laboratory of Biochemistry, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan.
| | - Yuko Okamatsu-Ogura
- Laboratory of Biochemistry, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
35
|
Ketchem JM, Bowman EJ, Isales CM. Male sex hormones, aging, and inflammation. Biogerontology 2023; 24:1-25. [PMID: 36596999 PMCID: PMC9810526 DOI: 10.1007/s10522-022-10002-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 11/15/2022] [Indexed: 01/05/2023]
Abstract
Adequate levels of androgens (eugonadism), and specifically testosterone, are vital compounds for male quality of life, longevity, and positive health outcomes. Testosterone exerts its effects by binding to the androgen receptor, which is expressed in numerous tissues throughout the body. Significant research has been conducted on the impact of this steroid hormone on skeletal, muscle and adipose tissues and on the cardiovascular, immune, and nervous systems. Testosterone levels have also been studied in relation to the impact of diseases, aging, nutrition and the environment on its circulating levels. Conversely, the impact of testosterone on health has also been evaluated with respect to its cardiac and vascular protective effects, body composition, autoimmunity and all-cause mortality. The male aging process results in decreasing testosterone levels over time. The exact mechanisms and impact of these changes in testosterone levels with age on health- and life-span are still not completely clear. Further research is needed to determine the optimal testosterone and androgen levels to protect from chronic age-related conditions such as frailty and osteoporosis.
Collapse
Affiliation(s)
- Justin M. Ketchem
- grid.410427.40000 0001 2284 9329Medical College of Georgia at Augusta University, Augusta, GA 30912 USA
| | | | - Carlos M. Isales
- grid.410427.40000 0001 2284 9329Departments of Medicine, Neuroscience and Regenerative Medicine, Augusta University, Augusta, GA 30912 USA
| |
Collapse
|
36
|
Convissar S, Bennett-Toomey J, Stocco C. Insulin-like growth factor 1 enhances follicle-stimulating hormone-induced phosphorylation of GATA4 in rat granulosa cells. Mol Cell Endocrinol 2023; 559:111807. [PMID: 36279967 PMCID: PMC10041677 DOI: 10.1016/j.mce.2022.111807] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 10/05/2022] [Accepted: 10/17/2022] [Indexed: 11/06/2022]
Abstract
Preovulatory granulosa cell (GC) differentiation is essential for the maturation and release of oocytes from the ovary. We have previously demonstrated that follicle-stimulating hormone (FSH) and insulin-like growth factors (IGFs) closely interact to control GC function. Similarly, we showed that GATA4 mediates FSH actions and it is required for preovulatory follicle formation. This report aimed to determine in vivo the effect of FSH on GATA4 phosphorylation and to investigate whether FSH and IGF1 interact to regulate GATA4 activity. In rat ovaries, treatment with equine chorionic gonadotropin (eCG) increased the phosphorylation of GATA4, which was confined to the nucleus of GCs. Using primary rat GCs, we observed that GATA4 phosphorylation at serine 105 increases the transcriptional activity of this transcription factor. Like FSH, IGF1 stimulated GATA4 phosphorylation at serine 105. Interestingly, GATA4 phosphorylation was significantly higher in cells cotreated with FSH and IGF1 when compared to FSH or IGF1 alone, suggesting that IGF1 augments the effects of FSH on GATA4. It was also found that the enhancing effect of IGF1 requires AKT activity and is mimicked by the inhibition of glycogen synthase kinase-3 β (GSK3β), suggesting that AKT inhibition of GSK3β may play a role in the regulation of GATA4 phosphorylation. The data support an important role of the IGF1/AKT/GSK3β signaling pathway in the regulation of GATA4 transcriptional activity and provide new insights into the mechanisms by which FSH and IGF1 regulate GC differentiation. Our findings suggest that GATA4 transcriptional activation may, at least partially, mediate AKT actions in GCs.
Collapse
Affiliation(s)
- Scott Convissar
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Jill Bennett-Toomey
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Carlos Stocco
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL, 60612, USA.
| |
Collapse
|
37
|
Qiu Y, Lv Y, Zhang M, Ji S, Wu B, Zhao F, Qu Y, Sun Q, Guo Y, Zhu Y, Lin X, Zheng X, Li Z, Fu H, Li Y, Song H, Wei Y, Ding L, Chen G, Zhu Y, Cao Z, Shi X. Cadmium exposure is associated with testosterone levels in men: A cross-sectional study from the China National Human Biomonitoring. CHEMOSPHERE 2022; 307:135786. [PMID: 35872064 DOI: 10.1016/j.chemosphere.2022.135786] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 06/21/2022] [Accepted: 07/18/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Sex hormone disorders can cause adverse health consequences. While experimental data suggests that cadmium (Cd) disrupts the endocrine system, little is known about the link between Cd exposure and sex hormones in men. METHODS We measured blood cadmium (B-Cd), urine cadmium (U-Cd), serum testosterone and serum estradiol in men aged ≥18 years old participating in the China National Human Biomonitoring program, from 2017 to 2018. Urine cadmium adjusted for creatinine (Ucr-Cd) and the serum testosterone to serum estradiol ratio (T/E2) were calculated. The association of Cd exposure to serum testosterone and T/E2 in men was analyzed with multiple linear regression models. RESULTS Among Chinese men ≥18 years old, the weighted geometric mean (95% CI) of B-Cd and Ucr-Cd levels were 1.23 (1.12-1.35) μg/L and 0.53 (0.47-0.59) μg/g, respectively. The geometric means (95% CI) of serum testosterone and T/E2 were 18.56 (17.92-19.22) nmol/L and 143.86 (137.24-150.80). After adjusting for all covariates, each doubling of B-Cd level was associated with a 5.04% increase in serum testosterone levels (β = 0.071; 95%CI: 0.057-0.086) and a 4.03% increase in T/E2 (β = 0.057; 95%CI: 0.040-0.075); similar findings were found in Ucr-Cd. CONCLUSIONS In Chinese men, Cd may be an endocrine disruptor, which is positively associated with serum testosterone and T/E2.
Collapse
Affiliation(s)
- Yidan Qiu
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China; Department of Big Data in Health Science, School of Public Health, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yuebin Lv
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Miao Zhang
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Saisai Ji
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Bing Wu
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China; Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Feng Zhao
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Yingli Qu
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Qi Sun
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Yanbo Guo
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China; Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, Jilin, China
| | - Yuanduo Zhu
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Xiao Lin
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Xulin Zheng
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China; Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zheng Li
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Hui Fu
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Yawei Li
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Haocan Song
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Yuan Wei
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China; Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, Jilin, China
| | - Liang Ding
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Guangdi Chen
- Institute of Environmental Health, School of Public Health, and Bioelectromagnetics Laboratory, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Ying Zhu
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Zhaojin Cao
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Xiaoming Shi
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China; Department of Big Data in Health Science, School of Public Health, Zhejiang University, Hangzhou, Zhejiang, China.
| |
Collapse
|
38
|
Kamal DAM, Ibrahim SF, Ugusman A, Zaid SSM, Mokhtar MH. Kelulut Honey Improves Folliculogenesis, Steroidogenic, and Aromatase Enzyme Profiles and Ovarian Histomorphology in Letrozole-Induced Polycystic Ovary Syndrome Rats. Nutrients 2022; 14:4364. [PMID: 36297046 PMCID: PMC9612175 DOI: 10.3390/nu14204364] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/09/2022] [Accepted: 10/14/2022] [Indexed: 11/16/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) has been linked to aberrant folliculogenesis and abnormalities in the aromatase enzyme (Cyp19a1) and the steroidogenic enzyme, 17-alpha-hydroxylase (Cyp17a1) expression. It has been demonstrated that Kelulut honey (KH) improves both female and male reproductive system anomalies in animal studies. Here, we examined the effects of isolated and combined KH, metformin, and clomiphene in improving folliculogenesis, aromatase, and steroidogenic enzyme profiles and ovarian histomorphology in letrozole-induced PCOS rats. Letrozole (1 mg/kg/day) was administered to female Sprague-Dawley (SD) rats for 21 days to induce PCOS. PCOS rats were subsequently divided into six experimental groups: untreated, treatment with metformin (500 mg/kg/day), clomiphene (2 mg/kg/day), KH (1 g/kg/day), combined KH (1 g/kg/day) and metformin (500 mg/kg/day), and combined KH (1 g/kg/day) and clomiphene (2 mg/kg/day). All treatments were given orally for 35 days. We found that KH was comparable with clomiphene and metformin in improving the expression of Cyp17a1 and Cyp19a1, apart from enhancing folliculogenesis both histologically and through the expression of folliculogenesis-related genes. Besides, the combination of KH with clomiphene was the most effective treatment in improving the ovarian histomorphology of PCOS rats. The effectiveness of KH in restoring altered folliculogenesis, steroidogenic, and aromatase enzyme profiles in PCOS warrants a future clinical trial to validate its therapeutic effect clinically.
Collapse
Affiliation(s)
- Datu Agasi Mohd Kamal
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Malaysia Sabah, Kota Kinabalu 88400, Malaysia
| | - Siti Fatimah Ibrahim
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Azizah Ugusman
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Siti Sarah Mohamad Zaid
- Department of Environment, Faculty of Forestry and Environment, Universiti Putra Malaysia, Serdang 43400, Malaysia
| | - Mohd Helmy Mokhtar
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| |
Collapse
|
39
|
Alemany M. The Roles of Androgens in Humans: Biology, Metabolic Regulation and Health. Int J Mol Sci 2022; 23:11952. [PMID: 36233256 PMCID: PMC9569951 DOI: 10.3390/ijms231911952] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Revised: 09/26/2022] [Accepted: 09/27/2022] [Indexed: 11/17/2022] Open
Abstract
Androgens are an important and diverse group of steroid hormone molecular species. They play varied functional roles, such as the control of metabolic energy fate and partition, the maintenance of skeletal and body protein and integrity and the development of brain capabilities and behavioral setup (including those factors defining maleness). In addition, androgens are the precursors of estrogens, with which they share an extensive control of the reproductive mechanisms (in both sexes). In this review, the types of androgens, their functions and signaling are tabulated and described, including some less-known functions. The close interrelationship between corticosteroids and androgens is also analyzed, centered in the adrenal cortex, together with the main feedback control systems of the hypothalamic-hypophysis-gonads axis, and its modulation by the metabolic environment, sex, age and health. Testosterone (T) is singled out because of its high synthesis rate and turnover, but also because age-related hypogonadism is a key signal for the biologically planned early obsolescence of men, and the delayed onset of a faster rate of functional losses in women after menopause. The close collaboration of T with estradiol (E2) active in the maintenance of body metabolic systems is also presented Their parallel insufficiency has been directly related to the ravages of senescence and the metabolic syndrome constellation of disorders. The clinical use of T to correct hypoandrogenism helps maintain the functionality of core metabolism, limiting excess fat deposition, sarcopenia and cognoscitive frailty (part of these effects are due to the E2 generated from T). The effectiveness of using lipophilic T esters for T replacement treatments is analyzed in depth, and the main problems derived from their application are discussed.
Collapse
Affiliation(s)
- Marià Alemany
- Facultat de Biologia, Universitat de Barcelona, Av. Diagonal, 635, 08028 Barcelona, Catalonia, Spain;
- Institut de Biomedicina, Universitat de Barcelona, 08028 Barcelona, Catalonia, Spain
| |
Collapse
|
40
|
Almutlaq RN, Newell-Fugate AE, Evans LC, Fatima H, Gohar EY. Aromatase inhibition increases blood pressure and markers of renal injury in female rats. Am J Physiol Renal Physiol 2022; 323:F349-F360. [PMID: 35900340 PMCID: PMC9423724 DOI: 10.1152/ajprenal.00055.2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 06/22/2022] [Accepted: 07/13/2022] [Indexed: 11/22/2022] Open
Abstract
Aromatase is a monooxygenase that catalyzes the rate-limiting step of estrogen biosynthesis from androgens. Aromatase inhibitors are widely used for the treatment of patients with hormone receptor-positive breast cancer. However, the effects of aromatase inhibitors on cardiovascular and renal health in females are understudied. Given that estrogen is protective against cardiovascular and kidney diseases, we hypothesized that aromatase inhibition elevates blood pressure and induces kidney injury in female Sprague-Dawley rats. Twelve-week-old female rats were implanted with radiotelemetry transmitters to continuously monitor blood pressure. After baseline blood pressure recording, rats were randomly assigned to treatment with the aromatase inhibitor anastrozole (ASZ) or vehicle (Veh) in drinking water. Twenty days after treatment initiation, rats were shifted from a normal-salt (NS) diet to a high-salt (HS) diet for an additional 40 days. Rats were euthanized 60 days after treatment initiation. Body weight increased in both groups over the study period, but the increase was greater in the ASZ-treated group than in the Veh-treated group. Mean arterial pressure increased in ASZ-treated rats during the NS and HS diet phases but remained unchanged in Veh-treated rats. In addition, urinary excretion of albumin and kidney injury marker-1 and plasma urea were increased in response to aromatase inhibition. Furthermore, histological assessment revealed that ASZ treatment increased morphological evidence of renal tubular injury and proximal tubular brush border loss. In conclusion, chronic aromatase inhibition in vivo with ASZ increases blood pressure and markers of renal proximal tubular injury in female Sprague-Dawley rats, suggesting an important role for aromatization in the maintenance cardiovascular and renal health in females.NEW & NOTEWORTHY Aromatase enzyme catalyzes the rate-limiting step in estrogen biosynthesis. Aromatase inhibitors are clinically used for the treatment of patients with breast cancer; however, the impact of inhibiting aromatization on blood pressure and renal function is incompletely understood. The present findings demonstrate that systemic anastrozole treatment increases blood pressure and renal tubular injury markers in female rats fed a high-salt diet, suggesting an important role for aromatization in preserving cardiovascular and renal health in females.
Collapse
Affiliation(s)
- Rawan N Almutlaq
- Cardiorenal Physiology and Medicine Section, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Annie E Newell-Fugate
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas
| | - Louise C Evans
- Department of Surgery, Medical School, University of Minnesota, Minneapolis, Minnesota
| | - Huma Fatima
- Division of Anatomic and Clinical Pathology, Department of Pathology, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Eman Y Gohar
- Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
41
|
Moin ASM, Sathyapalan T, Atkin SL, Butler AE. Inflammatory Markers in Non-Obese Women with Polycystic Ovary Syndrome Are Not Elevated and Show No Correlation with Vitamin D Metabolites. Nutrients 2022; 14:nu14173540. [PMID: 36079796 PMCID: PMC9459875 DOI: 10.3390/nu14173540] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 08/22/2022] [Accepted: 08/24/2022] [Indexed: 11/19/2022] Open
Abstract
Introduction. Chronic low-grade inflammation is a characteristic of women with polycystic ovary syndrome (PCOS), although this may be obesity-driven rather than an intrinsic facet of PCOS; furthermore, vitamin D deficiency, another common feature of PCOS, is reported to have an association with increased inflammation. Therefore, circulating inflammatory protein levels and circulating levels of vitamin D may be linked in PCOS, though it is unclear which vitamin D metabolites may be important. Methods. We measured plasma levels of 24 inflammatory proteins and 12 matrix metalloproteinases (proteins modulated by the inflammatory process) by slow off-rate modified aptamer (SOMA)-scan plasma protein measurement in weight and aged-matched non-obese non-insulin resistant PCOS (n = 24) and control (n = 24) women. Inflammatory proteins and matrix metalloproteinases were correlated to 25-hydroxy vitamin D3 (25(OH)D3), its epimer 25-hydroxy-3epi-vitamin D (3epi25(OH)D) and the active 1,25-dihydroxyvitamin D3 (1,25(OH)2D3) as measured by gold standard isotope-dilution liquid chromatography tandem mass spectrometry. Results. PCOS women had both an elevated free androgen index and circulating anti-mullerian hormone, though insulin resistance was comparable to controls. C-reactive protein, as a standard circulatory marker of inflammation, was comparable between cohorts. Levels of circulating inflammatory proteins and matrix metalloproteinases were not different between the PCOS and control women, with no correlation of 25(OH)D3, 1,25(OH)2D3 or 3epi25(OH)D with any of the inflammatory proteins. Conclusion. In a non-obese PCOS population matched for age and insulin resistance, circulating inflammatory proteins and matrix metalloproteinases were not elevated and did not correlate with 25(OH)D3, its epimer 3epi25(OH)D or 1,25(OH)2D3 in either control or PCOS women, indicating that the inflammatory response is absent and the vitamin D-metabolite independent in non-obese women with PCOS.
Collapse
Affiliation(s)
| | - Thozhukat Sathyapalan
- Academic Endocrinology, Diabetes and Metabolism, Hull York Medical School, Hull YO10 5DD, UK
| | - Stephen L. Atkin
- Royal College of Surgeons in Ireland Bahrain, Adliya 15503, Bahrain
| | - Alexandra E. Butler
- Royal College of Surgeons in Ireland Bahrain, Adliya 15503, Bahrain
- Correspondence: or ; Tel.: +973-32360292
| |
Collapse
|
42
|
Mateus D, Sebastião AI, Carrascal MA, do Carmo A, Matos AM, Cruz MT. Crosstalk between estrogen, dendritic cells, and SARS-CoV-2 infection. Rev Med Virol 2022; 32:e2290. [PMID: 34534372 PMCID: PMC8646421 DOI: 10.1002/rmv.2290] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 08/17/2021] [Accepted: 08/18/2021] [Indexed: 12/11/2022]
Abstract
The novel coronavirus disease 2019 (Covid-19) first appeared in Wuhan and has so far killed more than four million people worldwide. Men are more affected than women by Covid-19, but the cellular and molecular mechanisms behind these differences are largely unknown. One plausible explanation is that differences in sex hormones could partially account for this distinct prevalence in both sexes. Accordingly, several papers have reported a protective role of 17β-estradiol during Covid-19, which might help explain why women appear less likely to die from Covid-19 than men. 17β-estradiol is the predominant and most biologically active endogenous estrogen, which signals through estrogen receptor α, estrogen receptor β, and G protein-coupled estrogen receptor 1. These receptors are expressed in mature cells from the innate and the adaptive immune system, particularly on dendritic cells (DCs), suggesting that estrogens could modulate their effector functions. DCs are the most specialized and proficient antigen-presenting cells, acting at the interface of innate and adaptive immunity with a powerful capacity to prime antigen-specific naive CD8+ T cells. DCs are richly abundant in the lung where they respond to viral infection. A relative increase of mature DCs in broncho-alveolar lavage fluids from Covid-19 patients has already been reported. Here we will describe how SARS-CoV-2 acts on DCs, the role of estrogen on DC immunobiology, summarise the impact of sex hormones on the immune response against Covid-19, and explore clinical trials regarding Covid-19.
Collapse
Affiliation(s)
- Daniela Mateus
- Faculty of Pharmacy—FFUCUniversity of CoimbraCoimbraPortugal
| | | | - Mylène A. Carrascal
- Center for Neuroscience and Cell Biology—CNCUniversity of CoimbraCoimbraPortugal
- UpCellsTecnimed GroupSintraPortugal
| | - Anália do Carmo
- Clinical Pathology DepartmentCentro Hospitalar e Universitário de CoimbraCoimbraPortugal
| | - Ana Miguel Matos
- Faculty of Pharmacy—FFUCUniversity of CoimbraCoimbraPortugal
- Chemical Engineering Processes and Forest Products Research Center, CIEPQPFFaculty of Sciences and Technology, University of CoimbraCoimbraPortugal
| | - Maria Teresa Cruz
- Faculty of Pharmacy—FFUCUniversity of CoimbraCoimbraPortugal
- Center for Neuroscience and Cell Biology—CNCUniversity of CoimbraCoimbraPortugal
| |
Collapse
|
43
|
Hashemain Z, Amiri-Yekta A, Khosravifar M, Alvandian F, Shahhosseini M, Hosseinkhani S, Afsharian P. CYP19A1 Promoters Activity in Human Granulosa Cells: A Comparison between PCOS and Normal Subjects. CELL JOURNAL 2022; 24:170-175. [PMID: 35674020 PMCID: PMC9124446 DOI: 10.22074/cellj.2022.7787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 03/01/2021] [Indexed: 11/06/2022]
Abstract
Objective Estrogen, a female hormone maintaining several critical functions in women's physiology, e.g., folliculogenesis and fertility, is predominantly produced by ovarian granulosa cells where aromatase enzyme converts androgen to estrogen. The principal enzyme responsible for this catalytic reaction is encoded by the CYP19A1 gene, with a long regulatory region. Abnormalities in this process cause metabolic disorders in women, one of the most common of which is polycystic ovary syndrome (PCOS). The main purpose of this research was to determine the effect of the promoters on aromatase expression in cells with normal and PCOS characteristics. Materials and Methods In this experimental study, four promoters of the CYP19A1 gene, including PII, I.3, I.4, and PII/ I .3 promoter fragments, were cloned upstream of the luciferase gene and transfected into normal and PCOS granulosa cells. Subsequently, the effect of follicle-stimulating hormone (FSH) on the activity of these regulatory regions was examined in the presence and absence of FSH. Western blotting was used to confirm aromatase expression in all groups. Data analysis was performed using ANOVA and paired sample t test, compared by post-hoc least significant difference (LSD) test. Results Luciferase results confirmed the intense activity of PII promoter in the presence of FSH. Moreover, the study demonstrated reduced activity of PII promoter in normal granulosa cells, possibly due to the regulatory region of I.3 next to PII. Conclusion FSH stimulates transcription of aromatase enzyme by affecting PII promoter, a process regulated by the inhibitory role of the I.3 region in PII activity in granulosa cells. Given the distinct role of these promoters in normal and PCOS granulosa cells, the importance of nuclear factors residing in these regions can be discerned.
Collapse
Affiliation(s)
- Zohreh Hashemain
- Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR,
Tehran, Iran,Human and Animal Cell Bank, Iranian Biological Resource Center (IBRC), ACECR, Tehran, Iran ,Faculty of Sciences and Advanced Technologies in Biology, University of Science and Culture, Tehran, Iran
| | - Amir Amiri-Yekta
- Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR,
Tehran, Iran
| | - Mona Khosravifar
- Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR,
Tehran, Iran
| | - Faezeh Alvandian
- Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR,
Tehran, Iran
| | - Maryam Shahhosseini
- Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR,
Tehran, Iran,Faculty of Sciences and Advanced Technologies in Biology, University of Science and Culture, Tehran, Iran
| | - Saman Hosseinkhani
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran ,P.O.Box: 14115-175Department of BiochemistryFaculty of Biological SciencesTarbiat Modares UniversityTehranIranP.O.Box: 16635-148Department of GeneticsReproductive Biomedicine Research CenterRoyan Institute for Reproductive BiomedicineACECRTehranIran
Emails:,
| | - Parvaneh Afsharian
- Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR,
Tehran, Iran,Faculty of Sciences and Advanced Technologies in Biology, University of Science and Culture, Tehran, Iran ,P.O.Box: 14115-175Department of BiochemistryFaculty of Biological SciencesTarbiat Modares UniversityTehranIranP.O.Box: 16635-148Department of GeneticsReproductive Biomedicine Research CenterRoyan Institute for Reproductive BiomedicineACECRTehranIran
Emails:,
| |
Collapse
|
44
|
Sobhani K, Vaziry A, Farzinpour A, Rostamzadeh J. Relative expression of aromatase in the male goat reproductive organs during different seasons. Reprod Domest Anim 2022; 57:806-812. [DOI: 10.1111/rda.14118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 03/27/2022] [Indexed: 11/26/2022]
Affiliation(s)
- Keyvan Sobhani
- Department of Animal Science Faculty of Agriculture University of Kurdistan Sanandaj Kurdistan Iran
| | - Asaad Vaziry
- Department of Animal Science Faculty of Agriculture University of Kurdistan Sanandaj Kurdistan Iran
| | - Amjad Farzinpour
- Department of Animal Science Faculty of Agriculture University of Kurdistan Sanandaj Kurdistan Iran
| | - Jalal Rostamzadeh
- Department of Animal Science Faculty of Agriculture University of Kurdistan Sanandaj Kurdistan Iran
| |
Collapse
|
45
|
Caballero Alfonso AY, Mora Lagares L, Novic M, Benfenati E, Kumar A. Exploration of structural requirements for azole chemicals towards human aromatase CYP19A1 activity: Classification modeling, structure-activity relationships and read-across study. Toxicol In Vitro 2022; 81:105332. [PMID: 35176449 DOI: 10.1016/j.tiv.2022.105332] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 01/10/2022] [Accepted: 02/10/2022] [Indexed: 01/23/2023]
Abstract
Human aromatase, also called CYP19A1, plays a major role in the conversion of androgens into estrogens. Inhibition of aromatase is an important target for estrogen receptor (ER)-responsive breast cancer therapy. Use of azole compounds as aromatase inhibitors is widespread despite their low selectivity. A toxicological evaluation of commonly used azole-based drugs and agrochemicals with respect to CYP19A1is currently requested by the European Union- Registration, Evaluation, Authorization and Restriction of Chemicals (EU-REACH) regulations due to their potential as endocrine disruptors. In this connection, identification of structural alerts (SAs) is an effective strategy for the toxicological assessment and safe drug design. The present study describes the identification of SAs of azole-based chemicals as guiding experts to predict the aromatase activity. Total 21 SAs associated with aromatase activity were extracted from dataset of 326 azole-based drugs/chemicals obtained from Tox21 library. A cross-validated classification model having high accuracy (error rate 5%) was proposed which can precisely classify azole chemicals into active/inactive toward aromatase. In addition, mechanistic details and toxicological properties (agonism/antagonism) of azoles with respect to aromatase were explored by comparing active and inactive chemicals using structure-activity relationships (SAR). Lastly, few structural alerts were applied to form chemical categories for read-across applications.
Collapse
Affiliation(s)
- Ana Y Caballero Alfonso
- Laboratory of Environmental Chemistry and Toxicology, Department of Environmental Health Sciences, Istituto di RicercheFarmacologiche "Mario Negri"-IRCCS, Milano, Italy; Jozef Stefan International Postgraduate School, Jamova cesta 39, 1000 Ljubljana, Slovenia
| | - Liadys Mora Lagares
- Jozef Stefan International Postgraduate School, Jamova cesta 39, 1000 Ljubljana, Slovenia; Theory Department, Laboratory for Cheminformatics, National Institute of Chemistry, Ljubljana, Slovenia
| | - Marjana Novic
- Theory Department, Laboratory for Cheminformatics, National Institute of Chemistry, Ljubljana, Slovenia
| | - Emilio Benfenati
- Laboratory of Environmental Chemistry and Toxicology, Department of Environmental Health Sciences, Istituto di RicercheFarmacologiche "Mario Negri"-IRCCS, Milano, Italy
| | - Anil Kumar
- Department of Applied Sciences, University Institute of Engineering and Technology, Panjab University, Chandigarh 160014, India.
| |
Collapse
|
46
|
Hager E, Chen J, Zhao L. Minireview: Parabens Exposure and Breast Cancer. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:1873. [PMID: 35162895 PMCID: PMC8834979 DOI: 10.3390/ijerph19031873] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Revised: 02/01/2022] [Accepted: 02/03/2022] [Indexed: 12/21/2022]
Abstract
There is increasing recognition that environmental exposure to chemicals, such as endocrine-disruptive chemicals (EDCs), contributes to the development of breast cancer. Parabens are a group of EDCs commonly found in personal care products, foods, and pharmaceuticals. Systemic exposure to parabens has been confirmed by the ubiquitous detection of parabens in human blood and urine samples. Although evidence from in vivo and epidemiological studies linking parabens exposure to breast cancer is limited, the current evidence suggests that parabens may negatively interfere with some endocrine and intracrine targets relevant to breast carcinogenesis. So far, most studies have focused on a single paraben's effects and the direct modulating effects on estrogen receptors or the androgen receptor in vitro. Recent studies have revealed that parabens can modulate local estrogen-converting enzymes, 17β-hydroxysteroid dehydrogenase 1 and 2 and increase local estrogen levels. Also, parabens can crosstalk with the human epidermal growth factor receptor 2 (HER2) pathway and work with ER signaling to increase pro-oncogenic c-Myc expression in ER+/HER2+ breast cancer cells. Future studies investigating paraben mixtures and their crosstalk with other EDCs or signaling pathways both in vitro and in vivo in the context of breast cancer development are warranted.
Collapse
Affiliation(s)
- Emily Hager
- Department of Nutrition, University of Tennessee, Knoxville, TN 37996, USA;
| | - Jiangang Chen
- Department of Public Health, University of Tennessee, Knoxville, TN 37996, USA
| | - Ling Zhao
- Department of Nutrition, University of Tennessee, Knoxville, TN 37996, USA;
| |
Collapse
|
47
|
Clark TD, Reichelt AC, Ghosh-Swaby O, Simpson SJ, Crean AJ. Nutrition, anxiety and hormones. Why sex differences matter in the link between obesity and behavior. Physiol Behav 2022; 247:113713. [DOI: 10.1016/j.physbeh.2022.113713] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 01/12/2022] [Accepted: 01/19/2022] [Indexed: 12/12/2022]
|
48
|
Fukami M. 11-Oxyandrogens from the viewpoint of pediatric endocrinology. Clin Pediatr Endocrinol 2022; 31:110-115. [PMID: 35928376 PMCID: PMC9297174 DOI: 10.1297/cpe.2022-0029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 05/19/2022] [Indexed: 11/10/2022] Open
Abstract
11-Oxyandrogens, such as 11-ketotestosterone (11-KT), 11-ketodihydrotestosterone
(11-KDHT), 11β-hydroxytestosterone (11-OHT), 11β-hydroxyandrostenedione (11-OHA4), and
11-KA4, are newly specified human androgens. These 11-oxyandrogens are present in the cord
blood and placenta, as well as in the blood of men and women of various ages, and are
produced primarily in the adrenal gland. Accumulating evidence suggests that these
steroids contribute to androgen excess in patients with 21-hydroxylase deficiency or
polycystic ovary syndrome. More importantly, unlike classic androgens, 11-oxyandrogens
produced in maternal tumors can pass through the placenta without being converted into
estrogens, and cause severe virilization of female fetuses. Thus, overproduction of
11-oxyandrogens represents a new mechanism of 46,XX disorders of sex development. On the
other hand, the physiological roles of 11-oxyandrogens remain to be clarified. This
mini-review introduces the current understanding of 11-oxyandrogens, from the perspective
of pediatric endocrinology.
Collapse
Affiliation(s)
- Maki Fukami
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, Tokyo, Japan
| |
Collapse
|
49
|
Cai Z, Zhong J, Jiang Y, Zhang J. Associations between COVID-19 infection and sex steroid hormones. Front Endocrinol (Lausanne) 2022; 13:940675. [PMID: 36303865 PMCID: PMC9592769 DOI: 10.3389/fendo.2022.940675] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 09/14/2022] [Indexed: 12/03/2022] Open
Abstract
AIMS Coronavirus disease 2019 (COVID-19) is caused by infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), and within a few months of the first outbreak, it was declared a global pandemic by the WHO. The lethal virus SARS-CoV-2 is transmitted through respiratory droplets and enters host cells through angiotensin-converting enzyme 2 (ACE-2) receptors. ACE-2 receptors are highly expressed in many tissues, including testes. Therefore, the objective of this study was to summarize the available literature regarding the correlation between sex hormone levels and COVID-19. METHODS The PubMed, Web of Science, Embase, and Cochrane Library databases were reviewed systematically through August 2022 for studies comparing sex hormone levels between different patient groups: COVID-19 versus no COVID-19, more severe versus less severe COVID-19, and non-survivors versus survivors. Various types of clinical research reporting sex hormone levels, including free testosterone (FT), luteinizing hormone (LH), follicle-stimulating hormone (FSH), 17β-oestradiol (E2), the oestradiol-to-testosterone ratio (E2/T), prolactin (PRL), and sex hormone-binding globulin (SHBG), were included. Random- or fixed-effects models were used to calculate weighted mean differences (WMDs) and 95% confidence intervals (CIs). Heterogeneity among the studies was assessed by the I2 index, and data analyses were performed using meta-analysis with Stata version 12.0. RESULTS Twenty-two articles that included 3369 patients were ultimately included in the meta-analysis. According to analysis of the included studies, patients with COVID-19 had significantly low T/LH, FSH/LH, and SHBG levels and high levels of LH, and E2/T, but their levels of FT, FSH, PRL, E2, and progesterone were not affected. Publication bias was not found according to funnel plots and Egger's regression and Begg's rank correlation tests. CONCLUSION Low T/LH, FSH/LH, and SHBG serum levels and high LH, and E2/T levels may increase the risk of COVID-19. Additionally, the greater is the clinical severity of COVID-19, the higher is the probability of increases in LH, and E2/T serum levels and decreases in T/LH, FSH/LH, and SHBG levels. COVID-19 may have unfavourable effects on gonadal functions, which should be taken seriously by clinicians. Routine monitoring of sex hormone levels might help clinicians to evaluate disease severity in patients with COVID-19.
Collapse
Affiliation(s)
- Zixin Cai
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Jiaxin Zhong
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Yingling Jiang
- Department of Metabolism and Endocrinology, The Affiliated Zhuzhou Hospital Xiangya Medical College CSU, Zhuzhou, China
| | - Jingjing Zhang
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
- *Correspondence: Jingjing Zhang,
| |
Collapse
|
50
|
Tharakan T, Minhas S, Jayasena CN. Male Sexual and Reproductive Health. COMPREHENSIVE PHARMACOLOGY 2022:94-124. [DOI: 10.1016/b978-0-12-820472-6.00036-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|