1
|
Zheng G, Chen Y, Wu G, Song T, Zou X, Nie Q, Zhang P. Review of the Hazards and Contraindications of Etomidate. Int J Toxicol 2025; 44:245-253. [PMID: 39501904 DOI: 10.1177/10915818241297073] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2025]
Abstract
Etomidate, an ultrashort-acting non-barbiturate sedative derived from imidazole, exerts potent inhibitory effects on the central nervous system. It is commonly employed for the induction of intravenous general anaesthesia or assisted anaesthesia. Recently, etomidate has emerged as an alternative to narcotics and novel psychoactive substances, leading to an increasing trend of abuse. Chronic overdose of etomidate can result in irreversible brain damage and various mental disorders. Severe cases may manifest as mental disturbances, behavioural disorders, self-mutilation and even death. The toxicological mechanisms of etomidate remain poorly understood. Additionally, there is limited information on the clinical symptoms and histopathological changes associated with etomidate poisoning and standardized detection methods for etomidate in blood, urine and hair are lacking. Consequently, further research on toxicological pathology and the development of reliable testing methods is crucial. This study reviews the metabolism, distribution, adverse reactions, poisoning manifestations, toxicology mechanisms and testing methods of etomidate.
Collapse
Affiliation(s)
- Gaolin Zheng
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Department of Forensic Medicine, Hainan Provincial Tropical Forensic Engineering Research Center, Hainan Medical University, Haikou, China
| | - Yinyu Chen
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Department of Forensic Medicine, Hainan Provincial Tropical Forensic Engineering Research Center, Hainan Medical University, Haikou, China
| | - Guangmei Wu
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Department of Forensic Medicine, Hainan Provincial Tropical Forensic Engineering Research Center, Hainan Medical University, Haikou, China
| | - Tao Song
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Department of Forensic Medicine, Hainan Provincial Tropical Forensic Engineering Research Center, Hainan Medical University, Haikou, China
| | - Xing Zou
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Department of Forensic Medicine, Hainan Provincial Tropical Forensic Engineering Research Center, Hainan Medical University, Haikou, China
| | - Qianyun Nie
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Department of Forensic Medicine, Hainan Provincial Tropical Forensic Engineering Research Center, Hainan Medical University, Haikou, China
- Department of Pathology, School of Basic Medicine and Life Sciences, Hainan Medical University, Haikou, China
- Department of Pathology, The First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Peng Zhang
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Department of Forensic Medicine, Hainan Provincial Tropical Forensic Engineering Research Center, Hainan Medical University, Haikou, China
| |
Collapse
|
2
|
Yan M, Yang J, Zhu H, Zou Q, Zhao H, Sun H. Volatile organic compound exposure in relation to lung cancer: Insights into mechanisms of action through metabolomics. JOURNAL OF HAZARDOUS MATERIALS 2024; 480:135856. [PMID: 39298956 DOI: 10.1016/j.jhazmat.2024.135856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 09/11/2024] [Accepted: 09/14/2024] [Indexed: 09/22/2024]
Abstract
Volatile organic compounds (VOCs) have proven to be hazardous to the human respiratory system. However, the underlying biological mechanisms remain poorly understood. Therefore, targeted determination of eleven VOC metabolites (mVOCs) along with the nontargeted metabolomic analysis was performed on urine samples collected from lung cancer patients and healthy individuals. Nine mVOCs mainly derived from aldehydes, alkenes, amides, and aromatics were detected in > 90 % of the urine samples, suggesting that the participants were ubiquitously exposed to these typical VOCs. A molecular gatekeeper discovery workflow was employed to link the exposure biomarkers with correlated clusters of endogenous metabolites. As a result, multiple metabolic pathways, including amino acid metabolism, steroid hormone biosynthesis and metabolism, and fatty acid β-oxidation were connected with VOC exposure. Furthermore, 16 of 73 molecular gatekeepers were associated with lung cancer and pointed to a few disrupted metabolic pathways related to hydroxysteroids and acylcarnitine. The shift in molecular profiles was validated in rat model post VOC administration. Thereinto, the up-regulation of enzymes involved in acylcarnitine synthesis and transport in rat lung tissues highlighted that the mitochondrial dysfunction may be a potential carcinogenic mechanism. Our findings provide new insights into the mechanisms underlying lung cancer induced by VOC exposure.
Collapse
Affiliation(s)
- Mengqi Yan
- MOE Key Laboratory of Pollution Processes and Environmental Criteria, College of Environmental Science and Engineering, Nankai University, Tianjin 300350, China
| | - Jintao Yang
- MOE Key Laboratory of Pollution Processes and Environmental Criteria, College of Environmental Science and Engineering, Nankai University, Tianjin 300350, China
| | - Hongkai Zhu
- MOE Key Laboratory of Pollution Processes and Environmental Criteria, College of Environmental Science and Engineering, Nankai University, Tianjin 300350, China.
| | - Qiang Zou
- Department of Interventional Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China.
| | - Hongzhi Zhao
- MOE Key Laboratory of Pollution Processes and Environmental Criteria, College of Environmental Science and Engineering, Nankai University, Tianjin 300350, China
| | - Hongwen Sun
- MOE Key Laboratory of Pollution Processes and Environmental Criteria, College of Environmental Science and Engineering, Nankai University, Tianjin 300350, China
| |
Collapse
|
3
|
Zhang H, Wu A, Nan X, Yang L, Zhang D, Zhang Z, Liu H. The Application and Pharmaceutical Development of Etomidate: Challenges and Strategies. Mol Pharm 2024; 21:5989-6006. [PMID: 39495089 DOI: 10.1021/acs.molpharmaceut.4c00325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2024]
Abstract
Etomidate is a synthetic imidazole anesthetic that exerts hypnotic effects by potentiating the action of the inhibitory neurotransmitter γ-aminobutyric acid (GABA) or directly activating the anionic GABA (GABAA) receptor. It stands out among many anesthetics because of its multiple advantages, such as good hemodynamic stability and minimal inhibition of spontaneous respiration. However, its low water solubility and side effects, such as adrenal cortex inhibition and myoclonus, have limited the clinical application of this drug. To address these issues, extensive research has been conducted on the drug delivery of etomidate in recent decades, which has led to the emergence of different etomidate preparations. Despite so many etomidate preparations, so far some of the toxic side effects have not yet been effectively addressed. Herein we discuss the pharmaceutical design of etomidate that may resolve the above problem. We also propose targeted strategies for future research on etomidate preparations and discuss the feasibility of different administration routes and dosage forms to expand the application of this drug. Through this review, we hope to draw more attention to the potential of etomidate and its application challenges and provide valuable insights into the development of new etomidate preparations.
Collapse
Affiliation(s)
- Hao Zhang
- School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, People's Republic of China
- Department of Pharmacy, Zigong First People's Hospital, Zigong, Sichuan 643000, People's Republic of China
| | - Ailing Wu
- Department of Anesthesiology, Second People's Hospital of Neijiang, Southwest Medical University, Neijiang, Sichuan 641000, People's Republic of China
- Department of Anesthesiology, First People's Hospital of Neijiang, Neijiang, Sichuan 641099, People's Republic of China
| | - Xichen Nan
- School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, People's Republic of China
| | - Luhan Yang
- School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, People's Republic of China
| | - Dan Zhang
- School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, People's Republic of China
| | - Zhuo Zhang
- School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, People's Republic of China
| | - Hao Liu
- School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, People's Republic of China
| |
Collapse
|
4
|
Chen Y, Liu J, Song T, Zou X, Li L, Nie Q, Zhang P. A case of fatal poisoning caused by etomidate: evidence from pathological and toxicological analyses. Forensic Sci Med Pathol 2024; 20:1453-1457. [PMID: 38652218 DOI: 10.1007/s12024-024-00813-8] [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] [Accepted: 04/02/2024] [Indexed: 04/25/2024]
Abstract
Etomidate is a nonbarbiturate sedative derived from imidazole. Prolonged and excessive use of etomidate can lead to the suppression of adrenocortical function, myoclonus, and even death. This report describes a rare case of a 47-year-old man who died from acute intoxication after oral ingestion of liquid containing etomidate. The cause of death was conclusively attributed to etomidate based on a comprehensive investigation, including autopsy, histopathological examination, toxicological analysis, and biochemical analysis. This is the first reported case of a fatality solely resulting from the oral ingestion of etomidate, which can provide valuable insights for future forensic investigations involving etomidate poisoning. Therefore, it is imperative to share this case with the scientific community.
Collapse
Affiliation(s)
- Yinyu Chen
- Department of Forensic Medicine, Hainan Provincial Tropical Forensic Engineering Research Center, Hainan Provincial Academician Workstation (tropical forensic medicine), Hainan Medical University, Haikou, China
| | - Jiaqi Liu
- Department of Neurology, the First Affiliated Hospital, International School of Public Health and One Health, Hainan Medical University, Haikou, China
| | - Tao Song
- Department of Forensic Medicine, Hainan Provincial Tropical Forensic Engineering Research Center, Hainan Provincial Academician Workstation (tropical forensic medicine), Hainan Medical University, Haikou, China
| | - Xing Zou
- Department of Forensic Medicine, Hainan Provincial Tropical Forensic Engineering Research Center, Hainan Provincial Academician Workstation (tropical forensic medicine), Hainan Medical University, Haikou, China
| | - Leilei Li
- Department of Forensic Medicine, Hainan Provincial Tropical Forensic Engineering Research Center, Hainan Provincial Academician Workstation (tropical forensic medicine), Hainan Medical University, Haikou, China
| | - Qianyun Nie
- Department of Pathology, School of Basic Medicine and Life Sciences, Hainan Medical University, Haikou, 571199, China.
- Department of Pathology, The First Affiliated Hospital of Hainan Medical University, Haikou, 570102, China.
| | - Peng Zhang
- Department of Forensic Medicine, Hainan Provincial Tropical Forensic Engineering Research Center, Hainan Provincial Academician Workstation (tropical forensic medicine), Hainan Medical University, Haikou, China.
| |
Collapse
|
5
|
Okudaira N, Akimoto M, Susa T, Akimoto M, Hisaki H, Iizuka M, Okinaga H, Almunia JA, Ogiso N, Okazaki T, Tamamori‐Adachi M. Accumulation of senescent cells in the adrenal gland induces hypersecretion of corticosterone via IL1β secretion. Aging Cell 2024; 23:e14206. [PMID: 38769821 PMCID: PMC11488315 DOI: 10.1111/acel.14206] [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: 04/17/2024] [Accepted: 05/02/2024] [Indexed: 05/22/2024] Open
Abstract
Aging progresses through the interaction of metabolic processes, including changes in the immune and endocrine systems. Glucocorticoids (GCs), which are regulated by the hypothalamic-pituitary-adrenal (HPA) axis, play an important role in regulating metabolism and immune responses. However, the age-related changes in the secretion mechanisms of GCs remain elusive. Here, we found that corticosterone (CORT) secretion follows a circadian rhythm in young mice, whereas it oversecreted throughout the day in aged mice >18 months old, resulting in the disappearance of diurnal variation. Furthermore, senescent cells progressively accumulated in the zF of the adrenal gland as mice aged beyond 18 months. This accumulation was accompanied by an increase in the number of Ad4BP/SF1 (SF1), a key transcription factor, strongly expressing cells (SF1-high positive: HP). Removal of senescent cells with senolytics, dasatinib, and quercetin resulted in the reduction of the number of SF1-HP cells and recovery of CORT diurnal oscillation in 24-month-old mice. Similarly, administration of a neutralizing antibody against IL1β, which was found to be strongly expressed in the adrenocortical cells of the zF, resulted in a marked decrease in SF1-HP cells and restoration of the CORT circadian rhythm. Our findings suggest that the disappearance of CORT diurnal oscillation is a characteristic of aging individuals and is caused by the secretion of IL1β, one of the SASPs, from senescent cells that accumulate in the zF of the adrenal cortex. These findings provide a novel insight into aging. Age-related hypersecretory GCs could be a potential therapeutic target for aging-related diseases.
Collapse
Affiliation(s)
- Noriyuki Okudaira
- Department of BiochemistryTeikyo University School of MedicineTokyoJapan
| | - Mi‐Ho Akimoto
- Department of BiochemistryTeikyo University School of MedicineTokyoJapan
| | - Takao Susa
- Department of BiochemistryTeikyo University School of MedicineTokyoJapan
| | - Miho Akimoto
- Department of BiochemistryTeikyo University School of MedicineTokyoJapan
| | - Harumi Hisaki
- Department of BiochemistryTeikyo University School of MedicineTokyoJapan
| | - Masayoshi Iizuka
- Department of BiochemistryTeikyo University School of MedicineTokyoJapan
- Medical Education CentreTeikyo University School of MedicineTokyoJapan
| | - Hiroko Okinaga
- Department of Internal MedicineTeikyo University School of MedicineTokyoJapan
| | - Julio A. Almunia
- Department of Laboratory of Experimental AnimalsNational Center for Geriatrics and Gerontology (NCGG)ObuAichiJapan
| | - Noboru Ogiso
- Department of Laboratory of Experimental AnimalsNational Center for Geriatrics and Gerontology (NCGG)ObuAichiJapan
| | - Tomoki Okazaki
- Department of BiochemistryTeikyo University School of MedicineTokyoJapan
| | | |
Collapse
|
6
|
Jäger MC, González-Ruiz V, Joos FL, Winter DV, Boccard J, Degenhardt T, Brand S, Rudaz S, Thompson GR, Odermatt A. Assessment of the potential risk of oteseconazole and two other tetrazole antifungals to inhibit adrenal steroidogenesis and peripheral metabolism of corticosteroids. Front Pharmacol 2024; 15:1394846. [PMID: 39175536 PMCID: PMC11338861 DOI: 10.3389/fphar.2024.1394846] [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: 03/02/2024] [Accepted: 07/15/2024] [Indexed: 08/24/2024] Open
Abstract
The triazole antifungals posaconazole and itraconazole can cause pseudohyperaldosteronism with hypertension and hypokalemia, edema, and gynecomastia by inhibiting steroid synthesis and metabolism. Mechanisms underlying pseudohyperaldosteronism include inhibition of adrenal 11β-hydroxylase cytochrome-P450 (CYP) 11B1 and 17α-hydroxylase (CYP17A1) as well as peripherally expressed 11β-hydroxysteroid dehydrogenase type 2 (11β-HSD2). To enhance specificity for fungal CYP51, tetrazoles have been developed. This study employed H295R adrenocortical cells and enzyme activity assays to assess the potential risk of oteseconazole and two other tetrazoles, VT-1598 and quilseconazole, to inhibit adrenal steroidogenesis or 11β-HSD2. Steroidomic footprint analyses of H295R cell supernatants using untargeted liquid-chromatography-high-resolution mass-spectrometry (LC-HRMS) indicated overall patterns common to oteseconazole, quilseconazole and itraconazole, as well as similarities between VT-1598 and isavuconazole. Additionally, more specific features of the steroid signatures were observed. Targeted quantification of nine adrenal steroids in supernatants from treated H295R cells revealed an overall inhibition of adrenal steroidogenesis by the three tetrazoles, itraconazole and isavuconazole, providing an explanation for their similar steroidomic pattern. Applying recombinant enzymes indicated that this effect is not due to direct inhibition of steroidogenic enzymes because no or only weak inhibition could be observed. Moreover, oteseconazole and the two other tetrazoles did not inhibit 11β-HSD2, suggesting that they do not pose a risk of pseudohyperaldosteronism. Furthermore, oteseconazole did not alter steroid concentrations in a recent clinical study. Nevertheless, follow-up studies should assess the mechanism underlying the observed overall steroidogenesis inhibition by tetrazoles, itraconazole and isavuconazole, and whether concentrations achievable in a subgroup of susceptible patients might cause adrenal insufficiency and hyperplasia.
Collapse
Affiliation(s)
- Marie-Christin Jäger
- Swiss Centre for Applied Human Toxicology (SCAHT), University of Basel, Basel, Switzerland
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Víctor González-Ruiz
- Swiss Centre for Applied Human Toxicology (SCAHT), University of Basel, Basel, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland
| | - Friedrich L. Joos
- Swiss Centre for Applied Human Toxicology (SCAHT), University of Basel, Basel, Switzerland
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Denise V. Winter
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Julien Boccard
- Swiss Centre for Applied Human Toxicology (SCAHT), University of Basel, Basel, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland
| | - Thorsten Degenhardt
- Mycovia Pharmaceuticals Inc., Imperial Business Park, Durham, NC, United States
| | - Steve Brand
- Mycovia Pharmaceuticals Inc., Imperial Business Park, Durham, NC, United States
| | - Serge Rudaz
- Swiss Centre for Applied Human Toxicology (SCAHT), University of Basel, Basel, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland
| | - George R. Thompson
- Department of Internal Medicine, Division of Infectious Diseases, University of California–Davis Health, Sacramento, CA, United States
| | - Alex Odermatt
- Swiss Centre for Applied Human Toxicology (SCAHT), University of Basel, Basel, Switzerland
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| |
Collapse
|
7
|
Su FC, Wei YC, Sun CY, Hsu HJ, Lee CC, Chen YT, Pan HC, Hsu CK, Liu YA, Chen CY. Endocrine-Disrupting Chemicals Exposure and Neurocognitive Function in the General Population: A Community-Based Study. TOXICS 2024; 12:514. [PMID: 39058166 PMCID: PMC11281080 DOI: 10.3390/toxics12070514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 07/12/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024]
Abstract
BACKGROUND Endocrine-disrupting chemicals (EDCs) are pervasive in everyday environments. The impacts of these chemicals, along with EDC-related lifestyle and dietary habits on neurocognitive function, are not well understood. METHODS The Chang Gung Community Medicine Research Center conducted a cross-sectional study involving 887 participants. From this initial cohort, 120 individuals were selected based on their EDC exposure scores for detailed analysis. Among these, 67 participants aged 55 years or older were further chosen to undergo cognitive impairment assessments using the Ascertain Dementia-8 (AD-8) questionnaire. RESULTS These 67 older participants did not significantly differ in age, albuminuria, or estimated glomerular filtration rate compared to those with lower impairment scores. This study revealed that mono-(2-ethylhexyl) phthalate (MEHP) levels (8.511 vs. 6.432 µg/g creatinine, p = 0.038) were associated with greater risk of cognitive impairment (AD-8 ≥ 2). Statistical models adjusting for age, gender, and diabetes indicated that MEHP levels positively correlated with AD-8 scores, achieving statistical significance in more comprehensive models (β ± SE: 0.160 ± 0.076, p = 0.042). Logistic regression analysis underscored a significant positive association between high MEHP levels and higher AD-8 scores (odds ratio: 1.217, p = 0.006). Receiver operating characteristic curves highlighted the association of high MEHP levels and EDC exposure scores for significant cognitive impairment, with areas under the curve of 66.3% and 66.6%, respectively. CONCLUSION Exposure to EDCs, specifically di-(2-ethylhexyl) phthalate, the precursor to MEHP, may be associated with neurocognitive impairment in middle-aged and older adults.
Collapse
Affiliation(s)
- Feng-Chieh Su
- Department of Neurology, Chang Gung Memorial Hospital, Keelung 204, Taiwan; (F.-C.S.); (Y.-C.W.)
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; (C.-Y.S.); (H.-J.H.); (C.-C.L.); (Y.-T.C.); (H.-C.P.); (C.-K.H.); (Y.-A.L.)
| | - Yi-Chia Wei
- Department of Neurology, Chang Gung Memorial Hospital, Keelung 204, Taiwan; (F.-C.S.); (Y.-C.W.)
- School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
- Community Medicine Research Center, Chang Gung Memorial Hospital, Keelung 204, Taiwan
| | - Chiao-Yin Sun
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; (C.-Y.S.); (H.-J.H.); (C.-C.L.); (Y.-T.C.); (H.-C.P.); (C.-K.H.); (Y.-A.L.)
- Community Medicine Research Center, Chang Gung Memorial Hospital, Keelung 204, Taiwan
- Department of Nephrology, Chang Gung Memorial Hospital, Keelung 204, Taiwan
| | - Heng-Jung Hsu
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; (C.-Y.S.); (H.-J.H.); (C.-C.L.); (Y.-T.C.); (H.-C.P.); (C.-K.H.); (Y.-A.L.)
- Community Medicine Research Center, Chang Gung Memorial Hospital, Keelung 204, Taiwan
- Department of Nephrology, Chang Gung Memorial Hospital, Keelung 204, Taiwan
| | - Chin-Chan Lee
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; (C.-Y.S.); (H.-J.H.); (C.-C.L.); (Y.-T.C.); (H.-C.P.); (C.-K.H.); (Y.-A.L.)
- Community Medicine Research Center, Chang Gung Memorial Hospital, Keelung 204, Taiwan
- Department of Nephrology, Chang Gung Memorial Hospital, Keelung 204, Taiwan
| | - Yih-Ting Chen
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; (C.-Y.S.); (H.-J.H.); (C.-C.L.); (Y.-T.C.); (H.-C.P.); (C.-K.H.); (Y.-A.L.)
- Community Medicine Research Center, Chang Gung Memorial Hospital, Keelung 204, Taiwan
- Department of Nephrology, Chang Gung Memorial Hospital, Keelung 204, Taiwan
| | - Heng-Chih Pan
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; (C.-Y.S.); (H.-J.H.); (C.-C.L.); (Y.-T.C.); (H.-C.P.); (C.-K.H.); (Y.-A.L.)
- Community Medicine Research Center, Chang Gung Memorial Hospital, Keelung 204, Taiwan
- Department of Nephrology, Chang Gung Memorial Hospital, Keelung 204, Taiwan
| | - Cheng-Kai Hsu
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; (C.-Y.S.); (H.-J.H.); (C.-C.L.); (Y.-T.C.); (H.-C.P.); (C.-K.H.); (Y.-A.L.)
- Department of Nephrology, Chang Gung Memorial Hospital, Keelung 204, Taiwan
| | - Yun-An Liu
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; (C.-Y.S.); (H.-J.H.); (C.-C.L.); (Y.-T.C.); (H.-C.P.); (C.-K.H.); (Y.-A.L.)
- Community Medicine Research Center, Chang Gung Memorial Hospital, Keelung 204, Taiwan
- Department of Nephrology, Chang Gung Memorial Hospital, Keelung 204, Taiwan
| | - Chun-Yu Chen
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; (C.-Y.S.); (H.-J.H.); (C.-C.L.); (Y.-T.C.); (H.-C.P.); (C.-K.H.); (Y.-A.L.)
- Community Medicine Research Center, Chang Gung Memorial Hospital, Keelung 204, Taiwan
- Department of Nephrology, Chang Gung Memorial Hospital, Keelung 204, Taiwan
| |
Collapse
|
8
|
Farag AA, Bayoumi H, Radwaan SE, El Gazzar WB, Youssef HS, Nasr HE, Badr AM, Mansour HM, Elalfy A, Sayed AEDH, Kharboush TG, Aboelkomsan EAF, Sliem RE. Melatonin counteracts polyethylene microplastics induced adreno-cortical damage in male albino rats. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 279:116499. [PMID: 38805828 DOI: 10.1016/j.ecoenv.2024.116499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 05/03/2024] [Accepted: 05/21/2024] [Indexed: 05/30/2024]
Abstract
There are various substances that can disrupt the homeostatic mechanisms of the body, defined as endocrine-disrupting chemicals (EDCs). The persistent nature of microplastics (MPs) is a cause for concern due to their ability to accumulate in food chains and widespread use, making their toxic effects particularly alarming. The potential of MPs for disrupting the endocrine system was observed in multiple tissues. Moreover, the adrenal gland is known to be extremely sensitive to EDCs, while with the effect of MPs on the adrenal gland has not previously been studied. This study aimed to highlight the potential polyethylene microplastics (PE-MPs) induced adreno-toxic effects rather than exploring the implicated mechanisms and concluding if melatonin (Mel) can afford protection against PE-MPs induced adreno-toxicity. To fulfill the goal, six groups of rats were used; control, Mel, PE-MPs (3.75 mg/kg), PE-MPs (15 mg/kg), PE-MPs (3.75 mg/kg) +Mel, and PE-MPs (15 mg/kg) +Mel. PE-MPs induced toxic changes in the adrenal cortex, which was evident by increased adrenal weight, histopathological examination, and ultrastructural changes detected by electron microscope. A reduction in serum cortisol and an increase in serum adrenocorticotropic hormone resulted from the adreno-toxic effects of PE-MPs. Mechanisms may include the reduction of steroidogenesis-related genes, as PE-MPs drastically reduce mRNA levels of StAR, Nr0b1, Cyp11A1, as well as Cyp11B1. Also, oxidative stress that results from PE-MPs is associated with higher rates of lipid peroxidation and decreased superoxide dismutase and glutathione. PE-MPs inflammatory effect was illustrated by elevated expression of IL-1β and NF-ķB, detected by immunohistochemical staining, in addition to increased expression of caspase-3 and mRNA of Bax, markers of proapoptotic activity. The impacts of PE-MPs were relatively dose-related, with the higher dose showing more significant toxicity than the lower one. Mel treatment was associated with a substantial amelioration of PE-MPs-induced toxic changes. Collectively, this study fills the knowledge gap about the MPs-induced adrenal cortex and elucidates various related toxic mechanisms. It also supports Mel's potential protective activity through antioxidant, anti-inflammatory, anti-apoptotic, and gene transcription regulatory effects.
Collapse
Affiliation(s)
- Amina A Farag
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Benha University, Benha City 13518, Egypt
| | - Heba Bayoumi
- Department of Histology and Cell Biology, Faculty of Medicine, Benha University, Benha City 13518, Egypt
| | - Shaimaa E Radwaan
- Department of Zoology, Faculty of Science, Benha University, Benha City 13518, Egypt
| | - Walaa Bayoumie El Gazzar
- Department of Anatomy, Physiology and Biochemistry, Faculty of Medicine, the Hashemite University, Zarqa 13133, Jordan; Department of Medical Biochemistry and Molecular biology, Faculty of Medicine, Benha University, Benha city 13518, Egypt
| | - Heba S Youssef
- Department of Physiology, Faculty of Medicine, Benha University, Benha City 13518, Egypt
| | - Hend Elsayed Nasr
- Department of Medical Biochemistry and Molecular biology, Faculty of Medicine, Benha University, Benha city 13518, Egypt
| | - Amira M Badr
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Heba M Mansour
- Department of Pharmacology and Toxicology, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology, October City 12573, Egypt
| | - Amira Elalfy
- Department of Histology and Cell Biology, Faculty of Medicine, Benha University, Benha City 13518, Egypt
| | - Alaa El-Din Hamid Sayed
- Department of Zoology, Faculty of Science, Assiut University, 71516, Egypt; Molecular Biology Research & Studies Institute, Assiut University, 71516 Assiut, Egypt.
| | - Tayseir G Kharboush
- Department of Pharmacology and therapeutics, Faculty of Medicine, Benha University, Benha City 13518, Egypt
| | | | - Rania E Sliem
- Department of Zoology, Faculty of Science, Benha University, Benha City 13518, Egypt
| |
Collapse
|
9
|
Pötzl B, Kürzinger L, Kendl S, Stopper H, Kurlbaum M, Fassnacht M, Dischinger U. Disruptive effects of plasticizers bisphenol A, F, and S on steroidogenesis of adrenocortical cells. Front Endocrinol (Lausanne) 2024; 15:1387133. [PMID: 38966215 PMCID: PMC11222671 DOI: 10.3389/fendo.2024.1387133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 06/03/2024] [Indexed: 07/06/2024] Open
Abstract
Introduction Endocrine disrupting chemicals (EDCs) are known to interfere with endocrine homeostasis. Their impact on the adrenal cortex and steroidogenesis has not yet been sufficiently elucidated. This applies in particular to the ubiquitously available bisphenols A (BPA), F (BPF), and S (BPS). Methods NCI-H295R adrenocortical cells were exposed to different concentrations (1nM-1mM) of BPA, BPF, BPS, and an equimolar mixture of them (BPmix). After 72 hours, 15 endogenous steroids were measured using LC-MS/MS. Ratios of substrate and product of CYP-regulated steps were calculated to identify most influenced steps of steroidogenesis. mRNA expression of steroidogenic enzymes was determined by real-time PCR. Results Cell viability remained unaffected at bisphenol concentrations lower than 250 µM. All tested bisphenols and their combination led to extensive alterations in the quantified steroid levels. The most profound fold changes (FC) in steroid concentrations after exposure to BPA (>10µM) were seen for androstenedione, e.g. a 0.37±0.11-fold decrease at 25µM (p≤0.0001) compared to vehicle-treated controls. For BPF, levels of 17-hydroxyprogesterone were significantly increased by 25µM (FC 2.57±0.49, p≤0.001) and 50µM (FC 2.65±0.61, p≤0.0001). BPS treatment led to a dose-dependent decrease of 11-deoxycorticosterone at >1µM (e.g. FC 0.24±0.14, p≤0.0001 at 10µM). However, when combining all three bisphenols, additive effects were detected: e.g. 11-deoxycortisosterone was decreased at doses >10µM (FC 0.27±0.04, p≤0.0001, at 25µM), whereas 21-deoxycortisol was increased by 2.92±0.20 (p≤0.01) at 10µM, and by 3.21±0.45 (p≤0.001) at 50µM. While every measured androgen (DHEA, DHEAS, androstenedione, testosterone, DHT) was lowered in all experiments, estradiol levels were significantly increased by BPA, BPF, BPS, and BPmix (e.g. FC 3.60±0.54, p≤0.0001 at 100µM BPF). Calculated substrate-product ratios indicated an inhibition of CYP17A1-, and CYP21A2 mediated conversions, whereas CYP11B1 and CYP19A1 showed higher activity in the presence of bisphenols. Based on these findings, most relevant mRNA expression of CYP genes were analysed. mRNA levels of StAR, CYP11B1, and CYP17A1 were significantly increased by BPF, BPS, and BPmix. Discussion In cell culture, bisphenols interfere with steroidogenesis at non-cytotoxic levels, leading to compound-specific patterns of significantly altered hormone levels. These results justify and call for additional in-vivo studies to evaluate effects of EDCs on adrenal gland functionality.
Collapse
Affiliation(s)
- Benedikt Pötzl
- Division of Endocrinology and Diabetology, Department of Internal Medicine I, University Hospital of Würzburg, Würzburg, Germany
| | - Lydia Kürzinger
- Division of Endocrinology and Diabetology, Department of Internal Medicine I, University Hospital of Würzburg, Würzburg, Germany
| | - Sabine Kendl
- Central Laboratory, Core Unit Clinical Mass Spectrometry, University Hospital, University of Würzburg, Würzburg, Germany
| | - Helga Stopper
- Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany
| | - Max Kurlbaum
- Division of Endocrinology and Diabetology, Department of Internal Medicine I, University Hospital of Würzburg, Würzburg, Germany
- Central Laboratory, Core Unit Clinical Mass Spectrometry, University Hospital, University of Würzburg, Würzburg, Germany
| | - Martin Fassnacht
- Division of Endocrinology and Diabetology, Department of Internal Medicine I, University Hospital of Würzburg, Würzburg, Germany
- Central Laboratory, Core Unit Clinical Mass Spectrometry, University Hospital, University of Würzburg, Würzburg, Germany
| | - Ulrich Dischinger
- Division of Endocrinology and Diabetology, Department of Internal Medicine I, University Hospital of Würzburg, Würzburg, Germany
| |
Collapse
|
10
|
Feng Y, Chang P, Liu J, Zhang WS. Effects and mechanisms of perioperative medications on the hypothalamic pituitary adrenal response to surgical injury: A narrative review. J Clin Anesth 2024; 94:111367. [PMID: 38232466 DOI: 10.1016/j.jclinane.2023.111367] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 11/30/2023] [Accepted: 12/18/2023] [Indexed: 01/19/2024]
Abstract
The adrenal gland is a vital endocrine organ, and adrenal steroid synthesis and secretion are closely regulated by the hypothalamic-pituitary-adrenal (HPA) axis in response to various stimuli. Surgery or trauma can activate the HPA axis and induce the secretion of cortisol. Different cortisol responses vary with the grade of surgery. Perioperative medications have the potential to decrease the cortisol level in the body, and both excessive and insufficient cortisol levels after surgery are disadvantageous. The effect of perioperative medications on the HPA response to surgery can be divided into three levels: "adrenal insufficiency (AI)", "stress response inhibition", and "uncertainty". The clinical presentation of AI includes fatigue, nausea, vomiting, abdominal pain, muscle cramps, hypotension, hypovolemic shock and prerenal failure, which may result in fatal consequences. Stress response inhibition can reduce postoperative complications, such as pain and cognitive dysfunction. This is protective to patients during perioperative and postoperative periods. The aim of the present review is to shed light on current evidence regarding the exact effects and mechanisms of perioperative medications on the HPA response to surgical injury and provide the applicable guidance on clinical anesthesia.
Collapse
Affiliation(s)
- Yan Feng
- Department of Anesthesiology, Affiliated Hospital of North Sichuan Medical College, China; Department of Anesthesiology, West China Hospital, Sichuan University, China; Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, China
| | - Pan Chang
- Department of Anesthesiology, West China Hospital, Sichuan University, China; Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, China
| | - Jin Liu
- Department of Anesthesiology, West China Hospital, Sichuan University, China; Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, China
| | - Wen-Sheng Zhang
- Department of Anesthesiology, West China Hospital, Sichuan University, China; Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, China.
| |
Collapse
|
11
|
Peng K, Liu Q, Wang N, Wang L, Duan X, Ding D. Association between smoking and alcohol drinking and benign adrenal tumors: a Mendelian randomization study. Endocrine 2024; 84:1206-1215. [PMID: 38409624 DOI: 10.1007/s12020-024-03714-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 01/23/2024] [Indexed: 02/28/2024]
Abstract
BACKGROUND In recent years, the detection rate of adrenal tumors has increased, but it is unclear whether smoking and alcohol drinking are risk factors for benign adrenal tumors. The objective of this study is to employ Mendelian randomization (MR) analysis to explore the causal relationship between smoking, alcohol drinking and susceptibility to benign adrenal tumors. METHODS We acquired large-scale data from publicly accessible databases on genome-wide association studies (GWAS) pertaining to smoking, alcohol drinking and benign adrenal tumors. A total of 11 sets of instrumental variables (IVs) and 281 associated single nucleotide polymorphic (SNP) loci were identified. The Mendelian randomization analyses were conducted using inverse variance weighting (IVW), MR-Egger regression and weighted median estimation (WME) methods, in addition to sensitivity analyses. RESULTS There is no causal relationship between smoking status, alcohol drinking status, alcohol intake frequency, alcohol taken with meals, alcohol consumption and benign adrenal tumors, while pack years of smoking and cigarettes per day are risk factors for benign adrenal tumors. The IVW analysis revealed that both the pack years of smoking and cigarettes per day were positively associated with an increased risk of benign adrenal tumors (OR = 2.853, 95%CI = 1.384-5.878, p = 0.004; OR = 1.543, 95%CI = 1.147-2.076, p = 0.004). Two SNPs (rs8042849 in the analysis of pack years of smoking and rs8034191 in the analysis of cigarettes per day) significantly drove the observed causal effects. CONCLUSION Two-sample Mendelian randomization analysis showed a causal effect between smoking but not alcohol consumption and benign adrenal tumors.
Collapse
Affiliation(s)
- Kun Peng
- Department of Urology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, 450003, China
| | - Qingyuan Liu
- Department of Urology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, 450003, China
| | - Ning Wang
- Department of Urology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, 450003, China
| | - Lingdian Wang
- Department of Urology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, 450003, China
| | - Xiaoyu Duan
- Department of Urology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, 450003, China
| | - Degang Ding
- Department of Urology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, 450003, China.
| |
Collapse
|
12
|
Kędzierski J, Jäger MC, Naeem S, Odermatt A, Smieško M. In silico and in vitro assessment of drugs potentially causing adverse effects by inhibiting CYP17A1. Toxicol Appl Pharmacol 2024; 486:116945. [PMID: 38688424 DOI: 10.1016/j.taap.2024.116945] [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: 01/18/2024] [Revised: 04/11/2024] [Accepted: 04/25/2024] [Indexed: 05/02/2024]
Abstract
Cytochrome P450 enzymes (CYPs) play a crucial role in the metabolism and synthesis of various compound classes. While drug-metabolizing CYP enzymes are frequently investigated as anti-targets, the inhibition of CYP enzymes involved in adrenal steroidogenesis is not well studied. The steroidogenic enzyme CYP17A1 is a dual-function enzyme catalyzing hydroxylase and lyase reactions relevant for the biosynthesis of adrenal glucocorticoids and androgens. Inhibition of CYP17A1-hydroxylase leads to pseudohyperaldosteronism with subsequent excessive mineralocorticoid receptor activation, hypertension and hypokalemia. In contrast, specific inhibition of the lyase function might be beneficial for the treatment of prostate cancer by decreasing adrenal androgen levels. This study combined in silico and in vitro methods to identify drugs inhibiting CYP17A1. The most potent CYP17A1 inhibitors identified are serdemetan, mocetinostat, nolatrexed, liarozole, and talarozole. While some of these drugs are currently under investigation for the treatment of various cancers, their potential for the treatment of prostate cancer is yet to be explored. The DrugBank database was screened for CYP17A1 inhibitors, to increase the awareness for the risk of drug-induced pseudohyperaldosteronism and to highlight drugs so far unknown for their potential to cause side effects resulting from CYP17A1 inhibition.
Collapse
Affiliation(s)
- Jacek Kędzierski
- Computational Pharmacy, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, Basel 4056, Switzerland; Swiss Centre for Human Applied Toxicology, University of Basel, Missionsstrasse 64, Basel 4055, Switzerland.
| | - Marie-Christin Jäger
- Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, Basel 4056, Switzerland; Swiss Centre for Human Applied Toxicology, University of Basel, Missionsstrasse 64, Basel 4055, Switzerland.
| | - Sadaf Naeem
- Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, Basel 4056, Switzerland; Department of Biochemistry, University of Karachi, KU, Circular Road, Karachi, Pakistan
| | - Alex Odermatt
- Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, Basel 4056, Switzerland; Swiss Centre for Human Applied Toxicology, University of Basel, Missionsstrasse 64, Basel 4055, Switzerland.
| | - Martin Smieško
- Computational Pharmacy, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, Basel 4056, Switzerland; Swiss Centre for Human Applied Toxicology, University of Basel, Missionsstrasse 64, Basel 4055, Switzerland.
| |
Collapse
|
13
|
Liu J, Yang T, Li Y, Li S, Li Y, Xu S, Xia W. Associations of maternal exposure to 2,4-dichlorophenoxyacetic acid during early pregnancy with steroid hormones among one-month-old infants. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 912:169414. [PMID: 38114038 DOI: 10.1016/j.scitotenv.2023.169414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 12/12/2023] [Accepted: 12/13/2023] [Indexed: 12/21/2023]
Abstract
BACKGROUND Exposure to 2,4-dichlorophenoxyacetic acid (2,4-D), a widely used hormonal herbicide, may disrupt steroid hormone homeostasis. However, evidence from population-based studies is limited, especially for one-month-old infants whose steroid hormones are in a state of adjustment to extrauterine life and can be important indicators of endocrine development. This study aimed to explore the associations between maternal 2,4-D exposure during early pregnancy and infant steroid hormone levels. METHODS The 885 mother-infant pairs were from a birth cohort in Wuhan, China. Maternal exposure to 2,4-D was determined in urine samples from early pregnancy, and nine steroid hormones were determined in infant urine. The associations of maternal 2,4-D exposure with infant steroid hormones and their product-to-precursor ratios were estimated based on generalized linear models, and bioinformatic analysis was conducted with public databases to explore the potential mechanisms involved. RESULTS The detection frequency of 2,4-D was 99.32 %, and the detection frequency of steroid hormones ranged from 98.42 % to 100.00 %. After adjusting for covariates, an interquartile range increase in 2,4-D concentrations was associated with a 7.84 % decrease in 11-deoxycortisol (95 % confidence interval, CI: -14.12 %, -1.10 %), an 8.09 % decrease in corticosterone (95 % CI: -14.56 %, -1.14 %), an 8.67 % decrease in cortisol (95 % CI: -14.43 %, -2.52 %), a 13.00 % decrease in cortisone (95 % CI: -20.64 %, -4.62 %), and an 11.17 % decrease in aldosterone (95 % CI: -19.62 %, -1.83 %). Maternal 2,4-D was also associated with lower infant cortisol/17α-OH-progesterone, cortisol/pregnenolone, and aldosterone/pregnenolone ratios. In bioinformatic analysis, pathways/biological processes related to steroid hormone synthesis and secretion were enriched from target genes of 2,4-D exposure. CONCLUSIONS Maternal urinary 2,4-D during early pregnancy was associated with lower infant urinary 11-deoxycortisol, corticosterone, cortisol, cortisone, and aldosterone, reflecting that 2,4-D exposure may interfere with infant steroid hormone homeostasis. Further efforts are still needed to study the relevant health effects of exposure to 2,4-D, particularly for vulnerable populations.
Collapse
Affiliation(s)
- Jiangtao Liu
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Tingting Yang
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ying Li
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Avenue, Shenzhen 518055, Guangdong Province, China
| | - Shulan Li
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yuanyuan Li
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shunqing Xu
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wei Xia
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
14
|
Kim HS, Cheon YP, Lee SH. Effects of Nonylphenol on the Secretion of Catecholamines and Adrenocortical Hormones from Short-Term Incubated Rat Adrenal Glands. Dev Reprod 2023; 27:213-220. [PMID: 38292238 PMCID: PMC10824570 DOI: 10.12717/dr.2023.27.4.213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 10/13/2023] [Accepted: 11/18/2023] [Indexed: 02/01/2024]
Abstract
Previously, we showed that a chronic-low-dose nonylphenol (NP) exposure resulted in histological changes with sexually dimorphic pattern in rat adrenal glands. We hypothesized that such structural changes are closely related to the hormonal secretory patterns. To test this hypothesis, we developed the short-term adrenal incubation method, and measured the levels of catecholamines and cortical steroids using the high-performance liquid chromatography with electrochemical detection (HPLC-ECD) and specific enzyme-linked immunosorbent assay, respectively. The norepinephrine (NE) levels in media from NP-treated female adrenal, except 100 pM NP, were significantly increased [control (CTL) vs 1 nM NP, p<0.001; vs 10 nM NP, p<0.05; vs 100 nM NP, p<0.001; vs 1 μM NP, p<0.01]. The NE secretion from male adrenal was higher when treated with 100 nM and 1 μM NP (CTL vs 100 nM NP, p<0.05; vs 1 μM NP, p<0.05, respectively). The aldosterone level in the female adrenal media treated with 100 pM NP was significantly decreased, on the other hand, that of media treated with 10 nM NP was significantly increased (CTL vs 100 pM NP, p<0.05; vs 10 nM NP, p<0.01). In male adrenal media, the aldosterone levels of 10 nM, 100 nM and 1 μM NP-treated media were significantly declined (CTL vs 10 nM NP, p<0.001; vs 100 nM NP, p<0.001; vs 1 μM NP, p<0.001). These results showed the NP treatment altered secretory pattern of aldosterone from adrenals of both sexes, showing sexual dimorphism. It may be helpful for understanding possible adrenal pathophysiology, and endocrine disrupting chemicals-related sexually dimorphic phenomena in adrenals.
Collapse
Affiliation(s)
- Hee-Su Kim
- Department of Biotechnology, Sangmyung
University, Seoul 03016, Korea
| | - Yong-Pil Cheon
- Division of Developmental Biology and
Physiology, School of Biological Sciences and Chemistry, Sungshin
University, Seoul 02844, Korea
| | - Sung-Ho Lee
- Department of Biotechnology, Sangmyung
University, Seoul 03016, Korea
| |
Collapse
|
15
|
Jäger MC, Kędzierski J, Gell V, Wey T, Kollár J, Winter DV, Schuster D, Smieško M, Odermatt A. Virtual screening and biological evaluation to identify pharmaceuticals potentially causing hypertension and hypokalemia by inhibiting steroid 11β-hydroxylase. Toxicol Appl Pharmacol 2023; 475:116638. [PMID: 37499767 DOI: 10.1016/j.taap.2023.116638] [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: 04/04/2023] [Revised: 07/21/2023] [Accepted: 07/24/2023] [Indexed: 07/29/2023]
Abstract
Several drugs were found after their market approval to unexpectedly inhibit adrenal 11β-hydroxylase (CYP11B1)-dependent cortisol synthesis. Known side-effects of CYP11B1 inhibition include hypertension and hypokalemia, due to a feedback activation of adrenal steroidogenesis, leading to supraphysiological concentrations of 11-deoxycortisol and 11-deoxycorticosterone that can activate the mineralocorticoid receptor. This results in potassium excretion and sodium and water retention, ultimately causing hypertension. With the risk known but usually not addressed in preclinical evaluation, this study aimed to identify drugs and drug candidates inhibiting CYP11B1. Two conceptually different virtual screening methods were combined, a pharmacophore based and an induced fit docking approach. Cell-free and cell-based CYP11B1 activity measurements revealed several inhibitors with IC50 values in the nanomolar range. Inhibitors include retinoic acid metabolism blocking agents (RAMBAs), azole antifungals, α2-adrenoceptor ligands, and a farnesyltransferase inhibitor. The active compounds share a nitrogen atom embedded in an aromatic ring system. Structure activity analysis identified the free electron pair of the nitrogen atom as a prerequisite for the drug-enzyme interaction, with its pKa value as an indicator of inhibitory potency. Another important parameter is drug lipophilicity, exemplified by etomidate. Changing its ethyl ester moiety to a more hydrophilic carboxylic acid group dramatically decreased the inhibitory potential, most likely due to less efficient cellular uptake. The presented work successfully combined different in silico and in vitro methods to identify several previously unknown CYP11B1 inhibitors. This workflow facilitates the identification of compounds that inhibit CYP11B1 and therefore pose a risk for inducing hypertension and hypokalemia.
Collapse
Affiliation(s)
- Marie-Christin Jäger
- Swiss Centre for Applied Human Toxicology (SCAHT), University of Basel, Missionsstrasse 64, 4055 Basel, Switzerland; Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland.
| | - Jacek Kędzierski
- Swiss Centre for Applied Human Toxicology (SCAHT), University of Basel, Missionsstrasse 64, 4055 Basel, Switzerland; Division of Computational Pharmacy, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 61, 4056 Basel, Switzerland.
| | - Victoria Gell
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland; Division of Computational Pharmacy, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 61, 4056 Basel, Switzerland.
| | - Tim Wey
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland.
| | - Jakub Kollár
- Institute of Pharmacy, Department of Pharmaceutical and Medicinal Chemistry, Paracelsus Medical University, Strubergasse 21, 5020 Salzburg, Austria.
| | - Denise V Winter
- Swiss Centre for Applied Human Toxicology (SCAHT), University of Basel, Missionsstrasse 64, 4055 Basel, Switzerland; Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland.
| | - Daniela Schuster
- Institute of Pharmacy, Department of Pharmaceutical and Medicinal Chemistry, Paracelsus Medical University, Strubergasse 21, 5020 Salzburg, Austria.
| | - Martin Smieško
- Swiss Centre for Applied Human Toxicology (SCAHT), University of Basel, Missionsstrasse 64, 4055 Basel, Switzerland; Division of Computational Pharmacy, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 61, 4056 Basel, Switzerland.
| | - Alex Odermatt
- Swiss Centre for Applied Human Toxicology (SCAHT), University of Basel, Missionsstrasse 64, 4055 Basel, Switzerland; Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland.
| |
Collapse
|
16
|
Yang F, Li Y, Xie Y, Yao W, Ren F. Diethyl phosphate disrupts hypothalamus-pituitary-adrenal axis endocrine hormones via nuclear receptors GR and Nur77: Integration of evidences from in vivo, in vitro and in silico approaches. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 844:157015. [PMID: 35777568 DOI: 10.1016/j.scitotenv.2022.157015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 05/25/2022] [Accepted: 06/23/2022] [Indexed: 06/15/2023]
Abstract
Plenty of population epidemiology and cohort studies have found dialkyl phosphates (DAPs) in the urine were related to endocrine hormone disorders. However, we did not know whether these effects were caused by parent organophosphorus pesticides (OPs) or metabolite DAPs, especially the non-specific metabolite diethyl phosphate (DEP), which was the metabolic end product of most widely used diethyl OPs. In this study, animal experiments (in vivo), cell experiments (in vitro), small molecule-protein binding interaction experiments and computer molecular simulations (in silico) were used to explore the disturbing effects and molecular mechanisms of DEP on the hypothalamic-pituitary-adrenal (HPA) axis endocrine hormones. The animal experiments showed that chronic DEP exposure significantly disturbed the serum contents of HPA axis hormones in adult male rats. The target genes of glucocorticoid receptor (GR) in rat liver, including 11β-hsd1 and Pepck1 and PEPCK protein expressions, were down-regulated. Moreover, the gluconeogenic abilities of rats were impaired. However, it did not affect the expression of GR in the rat hypothalamus. These results indicated that the physiological functions of glucocorticoids and GR were damaged. Furthermore, spectroscopy experiments, cell experiments, molecular docking and molecular dynamics simulations also suggested that DEP can bind to nuclear receptors GR and Nur77, affecting their transcription factor functions, and the transcriptional expression levels of their downstream target genes were reduced. The biosynthesis and secretion of adrenocorticotropic hormone and glucocorticoids were blocked. Therefore, DEP can inhibit the production and physiological functions of HPA axis endocrine hormones by disrupting these related proteins and antagonizing nuclear receptors. These results were considered to provide a theoretical basis for strictly controlling the residue limits of OPs and their metabolites in foods, agricultural products and the environment. They also revealed new targets for evaluating the toxicities and risks of pesticide metabolites.
Collapse
Affiliation(s)
- Fangwei Yang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, No. 17 Qinghua East Road, Haidian District, Beijing 100083, China; Key Laboratory of Functional Dairy, Co-constructed by Ministry of Education and Beijing Government, and Beijing Laboratory of Food Quality and Safety, China Agricultural University, No. 17 Qinghua East Road, Haidian District, Beijing 100083, China; School of Food Science and Technology, Jiangnan University, No.1800 Lihu Avenue, Binhu District, Wuxi, Jiangsu Province 214122, China
| | - Yixuan Li
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, No. 17 Qinghua East Road, Haidian District, Beijing 100083, China; Key Laboratory of Functional Dairy, Co-constructed by Ministry of Education and Beijing Government, and Beijing Laboratory of Food Quality and Safety, China Agricultural University, No. 17 Qinghua East Road, Haidian District, Beijing 100083, China
| | - Yunfei Xie
- School of Food Science and Technology, Jiangnan University, No.1800 Lihu Avenue, Binhu District, Wuxi, Jiangsu Province 214122, China
| | - Weirong Yao
- School of Food Science and Technology, Jiangnan University, No.1800 Lihu Avenue, Binhu District, Wuxi, Jiangsu Province 214122, China
| | - Fazheng Ren
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, No. 17 Qinghua East Road, Haidian District, Beijing 100083, China; Key Laboratory of Functional Dairy, Co-constructed by Ministry of Education and Beijing Government, and Beijing Laboratory of Food Quality and Safety, China Agricultural University, No. 17 Qinghua East Road, Haidian District, Beijing 100083, China.
| |
Collapse
|
17
|
Dasmahapatra AK, Tchounwou PB. Histopathological evaluation of the interrenal gland (adrenal homolog) of Japanese medaka (Oryzias latipes) exposed to graphene oxide. ENVIRONMENTAL TOXICOLOGY 2022; 37:2460-2482. [PMID: 35809259 PMCID: PMC9463118 DOI: 10.1002/tox.23610] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 05/31/2022] [Accepted: 06/14/2022] [Indexed: 05/23/2023]
Abstract
Due to unique physicochemical properties and wide industrial and biomedical applications, graphene oxide (GO) is ubiquitous in the aquatic ecosystem. Using Japanese medaka (Oryzias latipes) fish as a model, we previously demonstrated minimal endocrine disrupting (ED) effects of GO on reproductive organs, and thyroids. Current study investigated the ED-effects of GO on the interrenal gland (IRG) of medaka. Breeding pairs of adult male and female fish were exposed to 0 mg/L (control) or 20 mg/L GO by continuous immersion for 96 h, or to 0 or 100 μg/g GO by intraperitoneal administration. Also, 1 day post-hatch (dph) larvae were exposed to different concentrations of GO (2.5-20 mg/L) for 96 h. IRG was evaluated by immunohistochemical techniques after 21 days depuration in adults and 6 weeks in larvae. IRG cells were counted and the nuclear area was measured in hematoxylin-eosin stained sections using ImageJ software. We found that IRG is distributed adjacent to the posterior cardinal vein and its branches within the head kidney. Columnar/oval shaped periodic acid-Schiff negative, tyrosine hydroxylase positive cells are arranged either in a single, or in groups, sometimes encircling a sinusoid, or in a straight chord, laying adjacent to the endothelium of the cardinal vein, and having eosinophilic cytoplasm with round/oval basophilic nuclei. GO effect on nuclei and cell population in IRG was inconsistent; depending on exposure route, sex, and/or age of the fish. Also, because of its high adsorptive property and sharp edges, GO probably agglomerated on IRG, and induced physical injury, and ED effects.
Collapse
Affiliation(s)
- Asok K. Dasmahapatra
- RCMI Center for Environmental HealthJackson State UniversityJacksonMississippiUSA
- Department of Biomolecular Sciences, Environmental Toxicology DivisionUniversity of MississippiOxfordMississippiUSA
| | - Paul B. Tchounwou
- RCMI Center for Environmental HealthJackson State UniversityJacksonMississippiUSA
| |
Collapse
|
18
|
Guarnotta V, Amodei R, Frasca F, Aversa A, Giordano C. Impact of Chemical Endocrine Disruptors and Hormone Modulators on the Endocrine System. Int J Mol Sci 2022; 23:ijms23105710. [PMID: 35628520 PMCID: PMC9145289 DOI: 10.3390/ijms23105710] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/14/2022] [Accepted: 05/17/2022] [Indexed: 11/16/2022] Open
Abstract
There is growing concern regarding the health and safety issues of endocrine-disrupting chemicals (EDCs). Long-term exposure to EDCs has alarming adverse health effects through both hormone-direct and hormone-indirect pathways. Non-chemical agents, including physical agents such as artificial light, radiation, temperature, and stress exposure, are currently poorly investigated, even though they can seriously affect the endocrine system, by modulation of hormonal action. Several mechanisms have been suggested to explain the interference of EDCs with hormonal activity. However, difficulty in quantifying the exposure, low standardization of studies, and the presence of confounding factors do not allow the establishment of a causal relationship between endocrine disorders and exposure to specific toxic agents. In this review, we focus on recent findings on the effects of EDCs and hormone system modulators on the endocrine system, including the thyroid, parathyroid glands, adrenal steroidogenesis, beta-cell function, and male and female reproductive function.
Collapse
Affiliation(s)
- Valentina Guarnotta
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties “G. D’Alessandro” (PROMISE), Section of Endocrinology, University of Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy; (V.G.); (R.A.)
| | - Roberta Amodei
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties “G. D’Alessandro” (PROMISE), Section of Endocrinology, University of Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy; (V.G.); (R.A.)
| | - Francesco Frasca
- Endocrinology Section, Department of Clinical and Experimental Medicine, Garibaldi Nesima Hospital, University of Catania, 95122 Catania, Italy;
| | - Antonio Aversa
- Department of Experimental and Clinical Medicine, Section of Endocrinology, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy;
| | - Carla Giordano
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties “G. D’Alessandro” (PROMISE), Section of Endocrinology, University of Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy; (V.G.); (R.A.)
- Correspondence: ; Tel.: +39-0916552110
| |
Collapse
|
19
|
Akimoto T, Kobayashi S, Nakayama A, Isobe A, Abe K, Hatakeyama T, Ohta R, Yanagisawa R, Koike E, Suzuki N, Kawaguchi M. Toxicological effects of Tris (1,3-dichloro-2-propyl) phosphate exposure in adult male rats differ depending on the history of exposure in the neonatal period. J Appl Toxicol 2022; 42:1503-1509. [PMID: 35274318 DOI: 10.1002/jat.4316] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 02/04/2022] [Accepted: 03/02/2022] [Indexed: 11/06/2022]
Abstract
There is increasing concern about multiple high concentration exposure to toxins in disaster and emergency situations. However, conventional toxicology testing methods may not adequately address these situations. Thus, we assessed whether the toxic effects of exposure in the adulthood differ depending on the presence or absence of neonatal exposure to Tris (1,3-dichloro-2-propyl) phosphate (TDCIPP) in male rats to investigate the effects of exposure history of chemicals. In the neonatal stage [postnatal days (PNDs) 1-7], animals were treated with either sesame oil (5 ml/kg/day) as a control or TDCIPP (250 mg/kg/day) dissolved in sesame oil. In adulthood (PND 101-107), animals were treated with either sesame oil (5 ml/kg/day) or TDCIPP (650 mg/kg/day). One day after the final administration, dissection was performed, and body and organ weight, hematology, blood biochemistry and histopathology were examined. The results demonstrated that the toxic effects of TDCIPP exposure in adulthood on adrenal gland size, serum iron content, and unsaturated iron binding capacity were enhanced by TDCIPP exposure in the neonatal stage. From these findings, it was indicated that the toxic effects of TDCIPP exposure in the adult stage are affected by pediatric exposure. These results suggest that the toxic effects of high-dose and long-term unsteady exposure to chemicals in large-scale disasters may change based on the exposure history of chemicals.
Collapse
Affiliation(s)
- Takahiro Akimoto
- Lab of Animal Behavior and Environmental Science, School of Agriculture, Meiji University, Kawasaki, Kanagawa, Japan
| | - Shohei Kobayashi
- Lab of Animal Behavior and Environmental Science, School of Agriculture, Meiji University, Kawasaki, Kanagawa, Japan.,Organization for the Strategic Coordination of Research and Intellectual Property, Meiji University, Kawasaki, Kanagawa, Japan
| | - Airi Nakayama
- Lab of Animal Behavior and Environmental Science, School of Agriculture, Meiji University, Kawasaki, Kanagawa, Japan
| | - Anna Isobe
- Lab of Animal Behavior and Environmental Science, School of Agriculture, Meiji University, Kawasaki, Kanagawa, Japan
| | - Koudai Abe
- Lab of Animal Behavior and Environmental Science, School of Agriculture, Meiji University, Kawasaki, Kanagawa, Japan
| | - Taichi Hatakeyama
- Lab of Animal Behavior and Environmental Science, School of Agriculture, Meiji University, Kawasaki, Kanagawa, Japan.,Organization for the Strategic Coordination of Research and Intellectual Property, Meiji University, Kawasaki, Kanagawa, Japan
| | - Ryo Ohta
- Hatano Research Institute, Food and Drug Safety Center, Hadano, Kanagawa, Japan
| | - Rie Yanagisawa
- Health and Environmental Risk Division, National Institute for Environmental Studies, Ibaraki, Japan
| | - Eiko Koike
- Health and Environmental Risk Division, National Institute for Environmental Studies, Ibaraki, Japan
| | - Noriyuki Suzuki
- Planning Division, National Institute for Environmental Studies, Ibaraki, Japan
| | - Maiko Kawaguchi
- Lab of Animal Behavior and Environmental Science, School of Agriculture, Meiji University, Kawasaki, Kanagawa, Japan
| |
Collapse
|
20
|
Vassiliadi DA, Vassiliou AG, Ilias I, Tsagarakis S, Kotanidou A, Dimopoulou I. Pituitary-Adrenal Responses and Glucocorticoid Receptor Expression in Critically Ill Patients with COVID-19. Int J Mol Sci 2021; 22:11473. [PMID: 34768903 PMCID: PMC8584241 DOI: 10.3390/ijms222111473] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 10/18/2021] [Accepted: 10/21/2021] [Indexed: 12/15/2022] Open
Abstract
The hypothalamus-pituitary-adrenal (HPA) axis was described as the principal component of the stress response 85 years ago, along with the acute-phase reaction, and the defense response at the tissue level. The orchestration of these processes is essential since systemic inflammation is a double-edged sword; whereas inflammation that is timely and of appropriate magnitude is beneficial, exuberant systemic inflammation incites tissue damage with potentially devastating consequences. Apart from its beneficial cardiovascular and metabolic effects, cortisol exerts a significant immunoregulatory role, a major attribute being that it restrains the excessive inflammatory reaction, thereby preventing unwanted tissue damage. In this review, we will discuss the role of the HPA axis in the normal stress response and in critical illness, especially in critically ill patients with coronavirus disease 2019 (COVID-19). Finally, a chapter will be dedicated to the findings from clinical studies in critical illness and COVID-19 on the expression of the mediator of glucocorticoid actions, the glucocorticoid receptor (GCR).
Collapse
Affiliation(s)
- Dimitra A. Vassiliadi
- Department of Endocrinology, Diabetes and Metabolism, National Expertise Centre for Rare Endocrine Diseases, Evangelismos Hospital, 106 76 Athens, Greece; (D.A.V.); (S.T.)
| | - Alice G. Vassiliou
- First Department of Critical Care Medicine & Pulmonary Services, School of Medicine, National & Kapodistrian University of Athens, Evangelismos Hospital, 106 76 Athens, Greece; (A.G.V.); (A.K.)
| | - Ioannis Ilias
- Department of Endocrinology, Helena Venizelos Hospital, 115 21 Athens, Greece;
| | - Stylianos Tsagarakis
- Department of Endocrinology, Diabetes and Metabolism, National Expertise Centre for Rare Endocrine Diseases, Evangelismos Hospital, 106 76 Athens, Greece; (D.A.V.); (S.T.)
| | - Anastasia Kotanidou
- First Department of Critical Care Medicine & Pulmonary Services, School of Medicine, National & Kapodistrian University of Athens, Evangelismos Hospital, 106 76 Athens, Greece; (A.G.V.); (A.K.)
| | - Ioanna Dimopoulou
- First Department of Critical Care Medicine & Pulmonary Services, School of Medicine, National & Kapodistrian University of Athens, Evangelismos Hospital, 106 76 Athens, Greece; (A.G.V.); (A.K.)
| |
Collapse
|
21
|
Rousseau K, Dufour S, Sachs LM. Interdependence of Thyroid and Corticosteroid Signaling in Vertebrate Developmental Transitions. Front Ecol Evol 2021. [DOI: 10.3389/fevo.2021.735487] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Post-embryonic acute developmental processes mainly allow the transition from one life stage in a specific ecological niche to the next life stage in a different ecological niche. Metamorphosis, an emblematic type of these post-embryonic developmental processes, has occurred repeatedly and independently in various phylogenetic groups throughout metazoan evolution, such as in cnidarian, insects, molluscs, tunicates, or vertebrates. This review will focus on metamorphoses and developmental transitions in vertebrates, including typical larval metamorphosis in anuran amphibians, larval and secondary metamorphoses in teleost fishes, egg hatching in sauropsids and birth in mammals. Two neuroendocrine axes, the hypothalamic-pituitary-thyroid and the hypothalamic-pituitary-adrenal/interrenal axes, are central players in the regulation of these life transitions. The review will address the molecular and functional evolution of these axes and their interactions. Mechanisms of integration of internal and environmental cues, and activation of these neuroendocrine axes represent key questions in an “eco-evo-devo” perspective of metamorphosis. The roles played by developmental transitions in the innovation, adaptation, and plasticity of life cycles throughout vertebrates will be discussed. In the current context of global climate change and habitat destruction, the review will also address the impact of environmental factors, such as global warming and endocrine disruptors on hypothalamic-pituitary-thyroid and hypothalamic-pituitary-adrenal/interrenal axes, and regulation of developmental transitions.
Collapse
|
22
|
Faruk EM, Alasmari WA, Fouad H, Nafea OE, Hasan RAA. Extracellular vesicles derived from bone marrow mesenchymal stem cells repair functional and structural rat adrenal gland damage induced by fluoride. Life Sci 2021; 270:119122. [PMID: 33508294 DOI: 10.1016/j.lfs.2021.119122] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 01/10/2021] [Accepted: 01/18/2021] [Indexed: 02/07/2023]
Abstract
The adrenal glands have striking morpho-biochemical features that render them vulnerable to the effects of toxins. AIMS This study was conducted to explore the therapeutic utility of extracellular vesicles derived from bone marrow mesenchymal stem cells (BMSC-EVs) against fluoride-induced adrenal toxicity. MATERIALS AND METHODS The work included isolation and further identification of BMSC-EVs by transmission electron microscopy and flow cytometric analysis. Adrenal toxicity in rats was induced by oral administration of 300 ppm of sodium fluoride (NaF) in drinking water for 60 days followed by a single dose injection of BMSC-EVs. The effects of BMSC-EVs against NaF was evaluated by adrenal oxidant/antioxidant biomarkers, hormonal assay of plasma adrenocorticotrophic hormone (ACTH) and corticosterone (CORT) and mRNA gene expression quantitation for adrenal cortical steroidogenic pathway-encoding genes. Histopathological examination of the adrenal tissue was performed. KEY FINDINGS BMSC-EVs were effectively isolated and characterized. NaF exposure decreased adrenal superoxide dismutase and catalase activities, increased adrenal malondialdehyde levels, elevated plasma ACTH, diminished CORT concentrations and downregulated the adrenal cortical steroidogenic pathway-encoding genes. In addition, NaF-induced marked adrenal histopathological lesions. SIGNIFICANCE BMSC-EVs treatment repaired damaged adrenal tissue and recovered its function greatly following NaF consumption. BMSC-EVs reversed the toxic effects of NaF and reprogramed injured adrenal cells by activating regenerative processes.
Collapse
Affiliation(s)
- Eman Mohamed Faruk
- Department of Histology and Cell Biology, Faculty of Medicine, Benha University, Benha, Egypt
| | | | - Hanan Fouad
- Medical Biochemistry and Molecular Biology, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Ola Elsayed Nafea
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Zagazig University, Zagazig, Egypt; Department of Clinical Pharmacy, College of Pharmacy, Taif University, Taif, Saudi Arabia.
| | - Rehab Abd Allah Hasan
- Department of Histology and Cell Biology, Faculty of Medicine for Girls (AFMG), Al-Azhar University, Cairo, Egypt
| |
Collapse
|
23
|
Graceli JB, Dettogni RS, Merlo E, Niño O, da Costa CS, Zanol JF, Ríos Morris EA, Miranda-Alves L, Denicol AC. The impact of endocrine-disrupting chemical exposure in the mammalian hypothalamic-pituitary axis. Mol Cell Endocrinol 2020; 518:110997. [PMID: 32841708 DOI: 10.1016/j.mce.2020.110997] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 08/14/2020] [Accepted: 08/17/2020] [Indexed: 12/15/2022]
Abstract
The hypothalamic-pituitary axis (HP axis) plays a critical and integrative role in the endocrine system control to maintain homeostasis. The HP axis is responsible for the hormonal events necessary to regulate the thyroid, adrenal glands, gonads, somatic growth, among other functions. Endocrine-disrupting chemicals (EDCs) are a worldwide public health concern. There is growing evidence that exposure to EDCs such as bisphenol A (BPA), some phthalates, polychlorinated biphenyls (PCBs), polybrominated diphenyl ethers (PBDEs) and biphenyls (PBBs), dichlorodiphenyltrichloroethane (DDT), tributyltin (TBT), and atrazine (ATR), is associated with HP axis abnormalities. EDCs act on hormone receptors and their downstream signaling pathways and can interfere with hormone synthesis, metabolism, and actions. Because the HP axis function is particularly sensitive to endogenous hormonal changes, disruptions by EDCs can alter HP axis proper function, leading to important endocrine irregularities. Here, we review the evidence that EDCs could directly affect the mammalian HP axis function.
Collapse
Affiliation(s)
- Jones B Graceli
- Department of Morphology, Health Sciences Center, Federal University of Espirito Santo. Av. Marechal Campos, 1468, CEP: 290440-090 Vitória, ES, Brazil.
| | - Raquel S Dettogni
- Department of Morphology, Health Sciences Center, Federal University of Espirito Santo. Av. Marechal Campos, 1468, CEP: 290440-090 Vitória, ES, Brazil.
| | - Eduardo Merlo
- Department of Morphology, Health Sciences Center, Federal University of Espirito Santo. Av. Marechal Campos, 1468, CEP: 290440-090 Vitória, ES, Brazil.
| | - Oscar Niño
- Department of Morphology, Health Sciences Center, Federal University of Espirito Santo. Av. Marechal Campos, 1468, CEP: 290440-090 Vitória, ES, Brazil.
| | - Charles S da Costa
- Department of Morphology, Health Sciences Center, Federal University of Espirito Santo. Av. Marechal Campos, 1468, CEP: 290440-090 Vitória, ES, Brazil.
| | - Jordana F Zanol
- Department of Morphology, Health Sciences Center, Federal University of Espirito Santo. Av. Marechal Campos, 1468, CEP: 290440-090 Vitória, ES, Brazil.
| | - Eduardo A Ríos Morris
- Laboratory of Experimental Endocrinology-LEEx, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Brazil. Graduate Program in Endocrinology, Faculty of Medicine, Federal University of Rio de Janeiro, Brazil.
| | - Leandro Miranda-Alves
- Laboratory of Experimental Endocrinology-LEEx, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Brazil. Graduate Program in Endocrinology, Faculty of Medicine, Federal University of Rio de Janeiro, Brazil. Graduate Program in Pharmacology and Medicinal Chemistry, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Brazil.
| | - Anna C Denicol
- Department of Animal Science, University of California, Davis, One Shields Avenue Davis, CA, 95616, USA.
| |
Collapse
|
24
|
Di Lorenzo M, Sciarrillo R, Rosati L, Sellitti A, Barra T, De Luca A, Laforgia V, De Falco M. Effects of alkylphenols mixture on the adrenal gland of the lizard Podarcis sicula. CHEMOSPHERE 2020; 258:127239. [PMID: 32535440 DOI: 10.1016/j.chemosphere.2020.127239] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 05/25/2020] [Accepted: 05/26/2020] [Indexed: 05/20/2023]
Abstract
Alkylphenols (AP) are widespread environmental compounds belonging to the large family of substances known as Endocrine Disrupting Chemicals (EDCs). The present study was carried out to assess the effects of Octylphenol (OP) alone and in combination with Nonylphenol (NP) on the hypothalamus-pituitary-adrenal gland (HPA) axis of the lizard Podarcis sicula. Lizards are good bioindicators due to their features such as wide distribution, large population and good sensitivity to contaminants. Results obtained showed a time and dose-dependent stimulation of the HPA together with a high variation of both catecholamine plasma levels and greater vascularization and hypertrophy of steroidogenic cord of adrenal gland after both OP and OP + NP treatments. Interestingly, the OP + NP mixture treatment has provoked a state of stress of the adrenal gland which in fact appeared to be characterized by the presence of a marked macrophage infiltration which can be seen especially close to the connective capsule surrounding the gland. This macrophage infiltration could be an evidence of a particularly pronounced inflammatory state to indicate, probably, an animal's response to a non-physiological situation.
Collapse
Affiliation(s)
- Mariana Di Lorenzo
- Department of Biology, University of Naples Federico II, Via Cinthia, 80126, Naples, Italy
| | - Rosaria Sciarrillo
- Department of Biological and Environmental Sciences, University of Sannio, Via Port'Arsa 11, 82100, Benevento, Italy
| | - Luigi Rosati
- Department of Biology, University of Naples Federico II, Via Cinthia, 80126, Naples, Italy
| | - Anna Sellitti
- Department of Biology, University of Naples Federico II, Via Cinthia, 80126, Naples, Italy
| | - Teresa Barra
- Department of Biology, University of Naples Federico II, Via Cinthia, 80126, Naples, Italy
| | - Antonio De Luca
- Department of Mental and Physical Health and Preventive Medicine, Section of Human Anatomy, University of Campania "Luigi Vanvitelli", Via Costantinopoli 16, 80138, Naples, Italy
| | - Vincenza Laforgia
- Department of Biology, University of Naples Federico II, Via Cinthia, 80126, Naples, Italy; National Institute of Biostructures and Biosystems (INBB), Rome, Italy
| | - Maria De Falco
- Department of Biology, University of Naples Federico II, Via Cinthia, 80126, Naples, Italy; National Institute of Biostructures and Biosystems (INBB), Rome, Italy.
| |
Collapse
|
25
|
Di Lorenzo M, Barra T, Rosati L, Valiante S, Capaldo A, De Falco M, Laforgia V. Adrenal gland response to endocrine disrupting chemicals in fishes, amphibians and reptiles: A comparative overview. Gen Comp Endocrinol 2020; 297:113550. [PMID: 32679158 DOI: 10.1016/j.ygcen.2020.113550] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 07/09/2020] [Indexed: 12/18/2022]
Abstract
The adrenal gland is an essential component of the body stress response; it is formed by two portions: a steroidogenic and a chromaffin tissue. Despite the anatomy of adrenal gland is different among classes of vertebrates, the hormones produced are almost the same. During stress, these hormones contribute to body homeostasis and maintenance of ion balance. The adrenal gland is very sensitive to toxic compounds, many of which behave like endocrine-disruptor chemicals (EDCs). They contribute to alter the endocrine system in wildlife and humans and are considered as possible responsible of the decline of several vertebrate ectotherms. Considering that EDCs regularly can be found in all environmental matrices, the aim of this review is to collect information about the impact of these chemical compounds on the adrenal gland of fishes, amphibians and reptiles. In particular, this review shows the different behavior of these "sentinel species" when they are exposed to stress condition. The data supplied in this review can help to further elucidate the role of EDCs and their harmful impact on the survival of these vertebrates.
Collapse
Affiliation(s)
- Mariana Di Lorenzo
- Department of Biology, University of Naples Federico II, Via Cinthia, 80126 Naples, Italy.
| | - Teresa Barra
- Department of Biology, University of Naples Federico II, Via Cinthia, 80126 Naples, Italy
| | - Luigi Rosati
- Department of Biology, University of Naples Federico II, Via Cinthia, 80126 Naples, Italy
| | - Salvatore Valiante
- Department of Biology, University of Naples Federico II, Via Cinthia, 80126 Naples, Italy
| | - Anna Capaldo
- Department of Biology, University of Naples Federico II, Via Cinthia, 80126 Naples, Italy
| | - Maria De Falco
- Department of Biology, University of Naples Federico II, Via Cinthia, 80126 Naples, Italy; National Institute of Biostructures and Biosystems (INBB), Rome, Italy
| | - Vincenza Laforgia
- Department of Biology, University of Naples Federico II, Via Cinthia, 80126 Naples, Italy; National Institute of Biostructures and Biosystems (INBB), Rome, Italy
| |
Collapse
|
26
|
Abstract
There is a trend to reduce the space allowance per animal in cattle feedlot, despite its potential negative impact on animal welfare. Aiming to evaluate the effects of space allowance per animal in outdoor feedlots on beef cattle welfare, a total of 1350 Nellore bulls (450 pure and 900 crossbred) were confined for 12 weeks using three space allowances: 6 (SA6), 12 (SA12) and 24 (SA24) m2/animal (n = 450 per treatment). Bulls were housed in three pens per treatment (n = 150 per pen). The first 6 weeks in the feedlot were defined as 'dry' and the last as 'rainy' period, according to the accumulated precipitation. Animal-based (body cleanliness, health indicators and maintenance behaviour) and environmental-based indicators (mud depth and air dust concentration) were assessed weekly during the feedlot period. Most of the health indicators (nasal and ocular discharge, hoof and locomotion alterations, diarrhoea, bloated rumen and breathing difficulty) were assessed in a subset of 15 animals randomly selected from each pen. Coughs and sneezes were counted in each pen. Maintenance behaviours (number of animals lying and attending the feed bunk) were recorded with scan sampling and instantaneous recording at 20-min intervals. Postmortem assessments were carried out in all animals by recording the frequencies of macroscopic signs of bronchitis, pulmonary emphysema, nephritis and urinary cyst and by measuring the weight and cortical and medullar areas of adrenal glands (n = 30 per pen). Compared with SA12 and SA24, SA6 showed a greater number of sneezes per minute during the dry period and a greater percentage of animals with locomotion alterations during the rainy period. Coughing, diarrhoea and nasal discharge affected a larger number of animals in the SA6 relative to the other two groups. During the rainy period, there was a lower percentage of animals with nasal and ocular discharge, and a greater percentage of animals with abnormal hoof and lying. A lower percentage of animals in SA6 and SA12 (but not SA24) attended the feed bunk during the rainy relative to the dry period. A mud depth score of 0 (no mud) was most frequent in SA24 pens, followed by SA12 and then SA6. Adrenal gland weight and cortical area were lower in SA24 animals compared with those in SA6 and SA12. The results show that decreasing the space allowance for beef cattle in outdoor feedlots degrades the feedlot environment and impoverishes animal welfare.
Collapse
|
27
|
Yilmaz B, Terekeci H, Sandal S, Kelestimur F. Endocrine disrupting chemicals: exposure, effects on human health, mechanism of action, models for testing and strategies for prevention. Rev Endocr Metab Disord 2020; 21:127-147. [PMID: 31792807 DOI: 10.1007/s11154-019-09521-z] [Citation(s) in RCA: 349] [Impact Index Per Article: 69.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Endocrine Disrupting Chemicals (EDCs) are a global problem for environmental and human health. They are defined as "an exogenous chemical, or mixture of chemicals, that can interfere with any aspect of hormone action". It is estimated that there are about 1000 chemicals with endocrine-acting properties. EDCs comprise pesticides, fungicides, industrial chemicals, plasticizers, nonylphenols, metals, pharmaceutical agents and phytoestrogens. Human exposure to EDCs mainly occurs by ingestion and to some extent by inhalation and dermal uptake. Most EDCs are lipophilic and bioaccumulate in the adipose tissue, thus they have a very long half-life in the body. It is difficult to assess the full impact of human exposure to EDCs because adverse effects develop latently and manifest at later ages, and in some people do not present. Timing of exposure is of importance. Developing fetus and neonates are the most vulnerable to endocrine disruption. EDCs may interfere with synthesis, action and metabolism of sex steroid hormones that in turn cause developmental and fertility problems, infertility and hormone-sensitive cancers in women and men. Some EDCs exert obesogenic effects that result in disturbance in energy homeostasis. Interference with hypothalamo-pituitary-thyroid and adrenal axes has also been reported. In this review, potential EDCs, their effects and mechanisms of action, epidemiological studies to analyze their effects on human health, bio-detection and chemical identification methods, difficulties in extrapolating experimental findings and studying endocrine disruptors in humans and recommendations for endocrinologists, individuals and policy makers will be discussed in view of the relevant literature.
Collapse
Affiliation(s)
- Bayram Yilmaz
- Department of Physiology, Faculty of Medicine, Yeditepe University, Istanbul, Turkey
| | - Hakan Terekeci
- Department of Internal Medicine, Faculty of Medicine, Yeditepe University, Istanbul, Turkey
| | - Suleyman Sandal
- Department of Physiology, Faculty of Medicine, Inonu University, Malatya, Turkey
| | - Fahrettin Kelestimur
- Department of Endocrinology, Faculty of Medicine, Yeditepe University, Istanbul, Turkey.
| |
Collapse
|
28
|
Untangling the association between environmental endocrine disruptive chemicals and the etiology of male genitourinary cancers. Biochem Pharmacol 2019; 172:113743. [PMID: 31812676 DOI: 10.1016/j.bcp.2019.113743] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 12/02/2019] [Indexed: 02/06/2023]
Abstract
Endocrine disrupting chemicals disrupt normal physiological function of endogenous hormones, their receptors, and signaling pathways of the endocrine system. Most endocrine disrupting chemicals exhibit estrogen/androgen agonistic and antagonistic activities that impinge upon hormone receptors and related pathways. Humans are exposed to endocrine disrupting chemicals through food, water and air, affecting the synthesis, release, transport, metabolism, binding, function and elimination of naturally occurring hormones. The urogenital organs function as sources of steroid hormones, are targeted end organs, and participate within systemic feedback loops within the endocrine system. The effects of endocrine disruptors can ultimately alter cellular homeostasis leading to a broad range of health effects, including malignancy. Human cancer is characterized by uncontrolled cell proliferation, mechanisms opposing cell-death, development of immortality, induction of angiogenesis, and promotion of invasion/metastasis. While hormonal malignancies of the male genitourinary organs are the second most common types of cancer, the molecular effects of endocrine disrupting chemicals in hormone-driven cancers has yet to be fully explored. In this commentary, we examine the molecular evidence for the involvement of endocrine disrupting chemicals in the genesis and progression of hormone-driven cancers in the prostate, testes, and bladder. We also report on challenges that have to be overcome to drive our understanding of these chemicals and explore the potential avenues of discovery that could ultimately allow the development of tools to prevent cancer in populations where exposure is inevitable.
Collapse
|
29
|
Affiliation(s)
- Stefan R Bornstein
- Department of Medicine, University Hospital Carl Gustav Carus, Dresden, Germany.
- Division of Endocrinology, Diabetes and Nutritional Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK.
| | - Tobias D Bornstein
- Department of Dermatology, University Hospital Heidelberg, Heidelberg, Germany
| | - Cynthia L Andoniadou
- Department of Medicine, University Hospital Carl Gustav Carus, Dresden, Germany
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, UK
| |
Collapse
|
30
|
Xu L, Wu W, Cheng R, Sun L, Jiang Z, Zhang L, Zhang Z, Su Y, Huang X. Toxic effects of triptolide on adrenal steroidogenesis in H295R cells and female rats. J Biochem Mol Toxicol 2019; 33:e22394. [DOI: 10.1002/jbt.22394] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 06/08/2019] [Accepted: 08/26/2019] [Indexed: 12/25/2022]
Affiliation(s)
- Ling‐Yan Xu
- Jiangsu Key Laboratory of Drug ScreeningChina Pharmaceutical University Nanjing China
- Department of Clinical Pharmacology, Sir Run Run HospitalNanjing Medical University Nanjing China
| | - Wei Wu
- Jiangsu Key Laboratory of Drug ScreeningChina Pharmaceutical University Nanjing China
- Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical UniversityMinistry of Education Nanjing China
| | - Rui Cheng
- Jiangsu Key Laboratory of Drug ScreeningChina Pharmaceutical University Nanjing China
- Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical UniversityMinistry of Education Nanjing China
| | - Li‐Xin Sun
- Jiangsu Key Laboratory of Drug ScreeningChina Pharmaceutical University Nanjing China
- Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical UniversityMinistry of Education Nanjing China
| | - Zhen‐Zhou Jiang
- Jiangsu Key Laboratory of Drug ScreeningChina Pharmaceutical University Nanjing China
- Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical UniversityMinistry of Education Nanjing China
| | - Lu‐Yong Zhang
- Center for Drug Screening and Pharmacodynamic Evaluation, School of PharmacyGuangdong Pharmaceutical University Guangdong China
| | - Zun‐Jian Zhang
- Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical UniversityMinistry of Education Nanjing China
| | - Yu‐Wen Su
- Department of Clinical Pharmacology, Sir Run Run HospitalNanjing Medical University Nanjing China
- School of PharmacyNanjing Medical University Nanjing China
| | - Xin Huang
- Jiangsu Key Laboratory of Drug ScreeningChina Pharmaceutical University Nanjing China
- Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical UniversityMinistry of Education Nanjing China
| |
Collapse
|
31
|
Yang FW, Fang B, Pang GF, Ren FZ. Organophosphorus pesticide triazophos: A new endocrine disruptor chemical of hypothalamus-pituitary-adrenal axis. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2019; 159:91-97. [PMID: 31400790 DOI: 10.1016/j.pestbp.2019.05.021] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 03/25/2019] [Accepted: 05/30/2019] [Indexed: 06/10/2023]
Abstract
The organophosphorus pesticide, triazophos (TAP) was banned to use in agriculture in several countries due to its high toxicity. However, TAP was still widely used and frequently detected in foods. Recently, many studies reported the endocrine-disrupting effect of pesticides, especially the hypothalamic-pituitary-thyroid and hypothalamic-pituitary-gonadal axis. In this study, adult male Wistar rats were exposed to TAP at the dose of 0.164 and 1.64 mg/kg bodyweight (~1/500th and 1/50th of LD50) for 24 weeks and serum contents of hormones were measured. TAP exposure significantly reduced serum contents of adrenocorticotropic hormone, corticosterone and epinephrine in rats (p < .05), leading to the delay in glucose homeostasis during the insulin tolerance test and decrease in serum contents of total cholesterol, triglyceride and low density lipoprotein. Molecular docking results suggested TAP may be an antagonist of glucocorticoid receptor which decreased significantly in the liver of rats, resulting in the decreased expression of 11β-hydroxysteroid dehydrogenase 1 and PEPCK1. This study revealed that TAP is a potential endocrine disruptor, especially in the hypothalamus-pituitary-adrenal system and may disturb the metabolism by affecting glucocorticoid receptor. This study provided new evidence about the toxicity of TAP and it was necessary to strictly control the usage of TAP in food.
Collapse
Affiliation(s)
- Fang-Wei Yang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Bing Fang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China.
| | - Guo-Fang Pang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Fa-Zheng Ren
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; Key Laboratory of Functional Dairy, Co-constructed by Ministry of Education and Beijing Government, Beijing Laboratory of Food Quality and Safety, China Agricultural University, Beijing 100083, China
| |
Collapse
|
32
|
Akram M, Patt M, Kaserer T, Temml V, Waratchareeyakul W, Kratschmar DV, Haupenthal J, Hartmann RW, Odermatt A, Schuster D. Identification of the fungicide epoxiconazole by virtual screening and biological assessment as inhibitor of human 11β-hydroxylase and aldosterone synthase. J Steroid Biochem Mol Biol 2019; 192:105358. [PMID: 30965118 DOI: 10.1016/j.jsbmb.2019.04.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Revised: 04/05/2019] [Accepted: 04/06/2019] [Indexed: 12/22/2022]
Abstract
Humans are constantly exposed to a multitude of environmental chemicals that may disturb endocrine functions. It is crucial to identify such chemicals and uncover their mode-of-action to avoid adverse health effects. 11β-hydroxylase (CYP11B1) and aldosterone synthase (CYP11B2) catalyze the formation of cortisol and aldosterone, respectively, in the adrenal cortex. Disruption of their synthesis by exogenous chemicals can contribute to cardio-metabolic diseases, chronic kidney disease, osteoporosis, and immune-related disorders. This study applied in silico screening and in vitro evaluation for the discovery of xenobiotics inhibiting CYP11B1 and CYP11B2. Several databases comprising environmentally relevant pollutants, chemicals in body care products, food additives and drugs were virtually screened using CYP11B1 and CYP11B2 pharmacophore models. A first round of biological testing used hamster cells overexpressing human CYP11B1 or CYP11B2 to analyze 25 selected virtual hits. Three compounds inhibited CYP11B1 and CYP11B2 with IC50 values below 3 μM. The most potent inhibitor was epoxiconazole (IC50 value of 623 nM for CYP11B1 and 113 nM for CYP11B2, respectively); flurprimidol and ancymidol were moderate inhibitors. In a second round, these three compounds were tested in human adrenal H295R cells endogenously expressing CYP11B1 and CYP11B2, confirming the potent inhibition by epoxiconazole and the more moderate effects by flurprimidol and ancymidol. Thus, the in silico screening, prioritization of chemicals for initial biological tests and use of H295R cells to provide initial mechanistic information is a promising strategy to identify potential endocrine disruptors inhibiting corticosteroid synthesis. A critical assessment of human exposure levels and in vivo evaluation of potential corticosteroid disrupting effects by epoxiconazole is required.
Collapse
Affiliation(s)
- Muhammad Akram
- Institute of Pharmacy / Pharmaceutical Chemistry and Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innrain 80-82, 6020, Innsbruck, Austria; Department of Medicinal and Pharmaceutical Chemistry, Institute of Pharmacy, Paracelsus Medical University, Strubergasse 22, 5020, Salzburg, Austria.
| | - Melanie Patt
- Swiss Centre for Applied Human Toxicology and Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland.
| | - Teresa Kaserer
- Institute of Pharmacy / Pharmaceutical Chemistry and Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innrain 80-82, 6020, Innsbruck, Austria.
| | - Veronika Temml
- Institute of Pharmacy / Pharmaceutical Chemistry and Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innrain 80-82, 6020, Innsbruck, Austria.
| | - Watcharee Waratchareeyakul
- Department of Chemistry, Faculty of Science and Technology, Rambhai Barni Rajabhat University, 22000, Chanthaburi, Thailand.
| | - Denise V Kratschmar
- Swiss Centre for Applied Human Toxicology and Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland.
| | - Joerg Haupenthal
- Department of Drug Design and Optimization, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Universitätscampus E8 1, 66123, Saarbrücken, Germany.
| | - Rolf W Hartmann
- Department of Drug Design and Optimization, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Universitätscampus E8 1, 66123, Saarbrücken, Germany; Department of Pharmaceutical and Medicinal Chemistry, Saarland University, Campus C2.3, 66123, Saarbrücken, Germany.
| | - Alex Odermatt
- Swiss Centre for Applied Human Toxicology and Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland.
| | - Daniela Schuster
- Institute of Pharmacy / Pharmaceutical Chemistry and Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innrain 80-82, 6020, Innsbruck, Austria; Department of Medicinal and Pharmaceutical Chemistry, Institute of Pharmacy, Paracelsus Medical University, Strubergasse 22, 5020, Salzburg, Austria.
| |
Collapse
|
33
|
Yang FW, Li YX, Ren FZ, Luo J, Pang GF. Assessment of the endocrine-disrupting effects of organophosphorus pesticide triazophos and its metabolites on endocrine hormones biosynthesis, transport and receptor binding in silico. Food Chem Toxicol 2019; 133:110759. [PMID: 31421215 DOI: 10.1016/j.fct.2019.110759] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 07/31/2019] [Accepted: 08/12/2019] [Indexed: 02/06/2023]
Abstract
Triazophos (TAP) was a widely used organophosphorus insecticide in developing countries. TAP could produce specific metabolites triazophos-oxon (TAPO) and 1-phenyl-3-hydroxy-1,2,4-triazole (PHT) and non-specific metabolites diethylthiophosphate (DETP) and diethylphosphate (DEP). The objective of this study involved computational approaches to discover potential mechanisms of molecular interaction of TAP and its major metabolites with endocrine hormone-related proteins using molecular docking in silico. We found that TAP, TAPO and DEP showed high binding affinity with more proteins and enzymes than PHT and DETP. TAP might interfere with the endocrine function of the adrenal gland, and TAP might also bind strongly with glucocorticoid receptors and thyroid hormone receptors. TAPO might disrupt the normal binding of androgen receptor, estrogen receptor, progesterone receptor and adrenergic receptor to their natural hormone ligands. DEP might affect biosynthesis of steroid hormones and thyroid hormones. Meanwhile, DEP might disrupt the binding and transport of thyroid hormones in the blood and the normal binding of thyroid hormones to their receptors. These results suggested that TAP and DEP might have endocrine disrupting activities and were potential endocrine disrupting chemicals. Our results provided further reference for the comprehensive evaluation of toxicity of organophosphorus chemicals and their metabolites.
Collapse
Affiliation(s)
- Fang-Wei Yang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
| | - Yi-Xuan Li
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
| | - Fa-Zheng Ren
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China; Key Laboratory of Functional Dairy, Co-constructed by Ministry of Education and Beijing Government, Beijing Laboratory of Food Quality and Safety, China Agricultural University, Beijing, 100083, China
| | - Jie Luo
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China; College of Food Science and Technology, Hunan Agricultural University, Changsha, 410114, China
| | - Guo-Fang Pang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China; Chinese Academy of Inspection and Quarantine, Beijing, 100176, China.
| |
Collapse
|
34
|
Beck KR, Thompson GR, Odermatt A. Drug-induced endocrine blood pressure elevation. Pharmacol Res 2019; 154:104311. [PMID: 31212012 DOI: 10.1016/j.phrs.2019.104311] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 06/08/2019] [Accepted: 06/10/2019] [Indexed: 11/16/2022]
Abstract
Patients with uncontrolled hypertension are at risk for cardiovascular complications. The majority of them suffers from unidentified forms of hypertension and a fraction has so-called secondary hypertension with an identifiable cause. The patient's medications, its use of certain herbal supplements and over-the-counter agents represent potential causal factors for secondary hypertension that are often overlooked. The current review focuses on drugs that are likely to elevate blood pressure by affecting the human endocrine system at the level of steroid synthesis or metabolism, mineralocorticoid receptor activity, or by affecting the catecholaminergic system. Drugs with known adverse effects but where benefits outweigh their risks, drug candidates and market withdrawals are reviewed. Finally, potential therapeutic strategies are discussed.
Collapse
Affiliation(s)
- Katharina R Beck
- Swiss Centre for Applied Human Toxicology and Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - George R Thompson
- Department of Internal Medicine, Division of Infectious Diseases and the Department of Medical Microbiology and Immunology, University of California Davis Medical Center, Davis, California, USA
| | - Alex Odermatt
- Swiss Centre for Applied Human Toxicology and Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland.
| |
Collapse
|
35
|
Lauretta R, Sansone A, Sansone M, Romanelli F, Appetecchia M. Endocrine Disrupting Chemicals: Effects on Endocrine Glands. Front Endocrinol (Lausanne) 2019; 10:178. [PMID: 30984107 PMCID: PMC6448049 DOI: 10.3389/fendo.2019.00178] [Citation(s) in RCA: 172] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 03/01/2019] [Indexed: 12/13/2022] Open
Abstract
In recent years, endocrine disrupting chemicals have gained interest in human physiopathology and more and more studies aimed to explain how these chemicals compounds affect endocrine system. In human populations, the majority of the studies point toward an association between exposure to endocrine disrupting chemicals and the disorders affecting endocrine axis. A great number of endocrine disrupting chemicals seem to be able to interfere with the physiology of hypothalamus-pituitary-gonadal axis; however, every endocrine axis may be a target for each EDCs and their action is not limited to a single axis or organ. Several compounds may also have a negative impact on energy metabolic homeostasis altering adipose tissue and promoting obesity, metabolic syndrome, and diabetes. Different mechanism have been proposed to explain these associations but their complexity together with the degree of occupational or environmental exposure, the low standardization of the studies, and the presence of confounding factors have prevented to establish causal relationship between the endocrine disorders and exposure to specific toxicants so far. This manuscript aims to review the state of art of scientific literature regarding the effects of endocrine-disrupting chemicals (EDCs) on endocrine system.
Collapse
Affiliation(s)
- Rosa Lauretta
- Endocrinology Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Andrea Sansone
- Section of Medical Pathophysiology, Food Science and Endocrinology, Department of Experimental Medicine, Sapienza Università di Roma, Rome, Italy
| | - Massimiliano Sansone
- Section of Medical Pathophysiology, Food Science and Endocrinology, Department of Experimental Medicine, Sapienza Università di Roma, Rome, Italy
| | - Francesco Romanelli
- Section of Medical Pathophysiology, Food Science and Endocrinology, Department of Experimental Medicine, Sapienza Università di Roma, Rome, Italy
| | - Marialuisa Appetecchia
- Endocrinology Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
- *Correspondence: Marialuisa Appetecchia
| |
Collapse
|
36
|
Díaz-Aguila Y, Cuevas-Romero E, Castelán F, Martínez-Gómez M, Rodríguez-Antolín J, Nicolás-Toledo L. Chronic stress and high sucrose intake cause distinctive morphometric effects in the adrenal glands of post-weaned rats. Biotech Histochem 2018; 93:565-574. [DOI: 10.1080/10520295.2018.1499961] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Affiliation(s)
- Y Díaz-Aguila
- Doctorate Biological Sciences, Autonomous University of Tlaxcala, Tlaxcala, México
| | - E Cuevas-Romero
- Tlaxcala Center of Behavioral Biology, Autonomous University of Tlaxcala, Tlaxcala, México
| | - F Castelán
- Tlaxcala Center of Behavioral Biology, Autonomous University of Tlaxcala, Tlaxcala, México
- Department of Cell Biology and Physiology, Institute of Research Biomedical, National Autonomous University of México, México-CDMX, México
| | - M Martínez-Gómez
- Tlaxcala Center of Behavioral Biology, Autonomous University of Tlaxcala, Tlaxcala, México
- Department of Cell Biology and Physiology, Institute of Research Biomedical, National Autonomous University of México, México-CDMX, México
| | - J Rodríguez-Antolín
- Tlaxcala Center of Behavioral Biology, Autonomous University of Tlaxcala, Tlaxcala, México
| | - L Nicolás-Toledo
- Tlaxcala Center of Behavioral Biology, Autonomous University of Tlaxcala, Tlaxcala, México
| |
Collapse
|
37
|
Pyun BJ, Yang H, Sohn E, Yu SY, Lee D, Jung DH, Ko BS, Lee HW. Tetragonia tetragonioides (Pall.) Kuntze Regulates Androgen Production in a Letrozole-Induced Polycystic Ovary Syndrome Model. Molecules 2018; 23:molecules23051173. [PMID: 29757997 PMCID: PMC6099488 DOI: 10.3390/molecules23051173] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Revised: 05/04/2018] [Accepted: 05/11/2018] [Indexed: 11/16/2022] Open
Abstract
Tetragonia tetragonioides (Pall.) Kuntze (TTK) is a medicinal plant traditionally used to treat various diseases such as diabetic, inflammatory, and female-related disorders. Polycystic ovary syndrome (PCOS) is a common endocrinological disorder in women of reproductive age, and hyperandrogenism is a prominent feature of PCOS resulting in anovulation and infertility. In this study, we investigated the effects of a TTK extract on androgen generation and regulation of steroidogenic enzymes in vitro and in vivo. Human adrenocortical NCI-H295R cells were used to assess the effects of TTK extract on production of dehydroepiandrosterone and testosterone, as well as the protein expression of steroidogenic enzymes. Further, a letrozole-induced PCOS rat model was used in vivo to assess whether dietary administration of TTK extract restores normal hormones and reduces PCOS symptoms. TTK extract significantly inhibited forskolin (FOR)-induced androgen production in NCI-H295R cells and serum luteinizing hormone, testosterone, and follicular cysts, but not estradiol, were reduced in letrozole-induced PCOS rats orally administered the TTK extract. In addition, TTK extract inhibits androgen biosynthesis through the ERK-CREB signaling pathway, which regulates CYP17A1 or HSD3B2 expression. TTK extract could be utilized for the prevention and treatment of hyperandrogenism and other types of PCOS.
Collapse
Affiliation(s)
- Bo-Jeong Pyun
- Herbal Medicine Research Division, Korea Institute of Oriental Medicine (KIOM), Daejeon 34054, Korea.
| | - Hyun Yang
- Herbal Medicine Research Division, Korea Institute of Oriental Medicine (KIOM), Daejeon 34054, Korea.
| | - Eunjin Sohn
- Herbal Medicine Research Division, Korea Institute of Oriental Medicine (KIOM), Daejeon 34054, Korea.
| | - Song Yi Yu
- Herbal Medicine Research Division, Korea Institute of Oriental Medicine (KIOM), Daejeon 34054, Korea.
| | - Dongoh Lee
- Herbal Medicine Research Division, Korea Institute of Oriental Medicine (KIOM), Daejeon 34054, Korea.
| | - Dong Ho Jung
- Herbal Medicine Research Division, Korea Institute of Oriental Medicine (KIOM), Daejeon 34054, Korea.
| | - Byoung Seob Ko
- Herbal Medicine Research Division, Korea Institute of Oriental Medicine (KIOM), Daejeon 34054, Korea.
| | - Hye Won Lee
- Herbal Medicine Research Division, Korea Institute of Oriental Medicine (KIOM), Daejeon 34054, Korea.
| |
Collapse
|
38
|
Islin J, Munkboel CH, Styrishave B. Steroidogenic disruptive effects of the serotonin-noradrenaline reuptake inhibitors duloxetine, venlafaxine and tramadol in the H295R cell assay and in a recombinant CYP17 assay. Toxicol In Vitro 2018; 47:63-71. [DOI: 10.1016/j.tiv.2017.10.029] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 10/11/2017] [Accepted: 10/30/2017] [Indexed: 01/03/2023]
|
39
|
Botteri Principato NL, Suarez JD, Laws SC, Klinefelter GR. The use of purified rat Leydig cells complements the H295R screen to detect chemical-induced alterations in testosterone production. Biol Reprod 2018; 98:239-249. [PMID: 29272331 PMCID: PMC6691738 DOI: 10.1093/biolre/iox177] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 12/18/2017] [Indexed: 12/18/2022] Open
Abstract
Exposure to endocrine disrupting chemicals has been associated with compromised testosterone production leading to abnormal male reproductive development and altered spermatogenesis. In vitro high-throughput screening (HTS) assays are needed to evaluate risk to testosterone production, yet the main steroidogenesis assay currently utilized is a human adrenocortical carcinoma cell line, H295R, which does not synthesize gonadal steroids at the same level as the gonads, thus limiting assay sensitivity. Here, we propose a complementary assay using a highly purified rat Leydig cell assay to evaluate the potential for chemical-induced alterations in testosterone production by the testis. We evaluated a subset of chemicals that failed to decrease testosterone production in the HTS H295R assay. The chemicals examined fit into one of two categories based on changes in substrates upstream of testosterone in the adrenal steroidogenic pathway (17α-hydroxyprogesterone and 11-deoxycorticosterone) that we predicted should have elicited a decrease in testosterone production. We found that 85% of 20 test chemicals examined inhibited Leydig cell testosterone production in our assay. Importantly, we adopted a 96-well format to increase throughput and efficiency of the Leydig cell assay. We identified a selection criterion based on the AC50 values for 17α-hydroxyprogesterone and 11-deoxycorticosterone generated from the HTS H295R assay that will help prioritize chemicals for further testing in the Leydig cell screen. We hypothesize that the greater dynamic range of testosterone production and sensitivity of the Leydig cell assay permits the detection of small, yet significant, chemical-induced changes not detected by the HTS H295R assay.
Collapse
Affiliation(s)
- Nicole L. Botteri Principato
- Oak Ridge Institute for Science and Education, Research Participation Program, Oak Ridge, Tennessee, USA
- Endocrine Toxicity Branch, Toxicity Assessment Division, National Health and Environmental Effects Research Laboratory, Office of Research and Development, US Environmental Protection Agency, Research Triangle Park, North Carolina, USA
| | - Juan D. Suarez
- Reproductive Toxicity Branch, Toxicity Assessment Division, National Health and Environmental Effects Research Laboratory, Office of Research and Development, US Environmental Protection Agency,Research Triangle Park, North Carolina, USA
| | - Susan C. Laws
- Endocrine Toxicity Branch, Toxicity Assessment Division, National Health and Environmental Effects Research Laboratory, Office of Research and Development, US Environmental Protection Agency, Research Triangle Park, North Carolina, USA
| | - Gary R. Klinefelter
- Reproductive Toxicity Branch, Toxicity Assessment Division, National Health and Environmental Effects Research Laboratory, Office of Research and Development, US Environmental Protection Agency,Research Triangle Park, North Carolina, USA
| |
Collapse
|
40
|
Fommei E, Turci R, Ripoli A, Balzan S, Bianchi F, Morelli L, Coi A. Evidence for persistent organochlorine pollutants in the human adrenal cortex. J Appl Toxicol 2017; 37:1091-1097. [PMID: 28332723 DOI: 10.1002/jat.3460] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 01/26/2017] [Accepted: 01/31/2017] [Indexed: 02/05/2023]
Abstract
Environmental pollutants may act as endocrine disruptors in animals. Organochlorine pesticides (OCPs) and polychlorinated biphenyls (PCBs) enter the food chain and may accumulate in the fatty animal tissues, including adrenals. To our knowledge, no previous study has investigated their presence in the human normal adrenal (NA) cortex and aldosterone-producing adenomas (APA). Surgical fragments of APA from 11 patients and NA from 8 kidney donors were analyzed for 16 PCBs congeners and 10 OCPs. A Matrix Solid-Phase Dispersion (MSPD) method for simultaneous determination of the target compounds in cortex homogenates was developed. A gas-chromatography triple quadrupole mass spectrometry (Triple Quad GC-MS) system was used for the analysis. Data were analyzed using Random Forest and Wilcoxon's rank-sum test. OCPs and PCBs were found in specimens from both types. A subset of pollutants characterized APA more than NA. Higher concentrations (μg g-1 ) in APA were observed for α-, β-, and γ- Hexachlorocyclohexane (HCH) (1.48 ± 3.32 vs. 0.17 ± 0.19, P = 0.028; 2.81 ± 2.10 vs. 0.96 ± 0.98, P = 0.011; 2.16 ± 4.85 vs. 0.17 ± 0.26, P = 0.004, respectively), as well as for Hexachlorobenzene (HCB) and for PCBs 28, 52 and 101 (3.41 ± 3.11 vs. 0.97 ± 1.06, P = 0.021; 2.34 ± 4.68 vs. 0.25 ± 0.22, P = 0.039; 0.58 ± 1.19 vs. 0.06 ± 0.02, P = 0.002; 0.26 ± 0.43 vs. 0.05 ± 0.00, P = 0.001, respectively). Environmental organochlorine pollutants were shown to be present in the human normal and abnormal adrenal cortex, deserving future investigation on their possible role as adrenal endocrine disruptors in human disease. Copyright © 2017 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Enza Fommei
- Fondazione Toscana "Gabriele Monasterio" (FTGM), Pisa, Italy
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Roberta Turci
- International Center of Toxicology Information, Istituti Clinici Scientifici Maugeri, Pavia, Italy
| | - Andrea Ripoli
- Fondazione Toscana "Gabriele Monasterio" (FTGM), Pisa, Italy
| | - Silvana Balzan
- Institute of Clinical Physiology, National Research Council (CNR), Pisa, Italy
| | - Fabrizio Bianchi
- Institute of Clinical Physiology, National Research Council (CNR), Pisa, Italy
| | - Luca Morelli
- Department of Transplantation and New Technologies, University of Pisa, Italy
| | - Alessio Coi
- Institute of Clinical Physiology, National Research Council (CNR), Pisa, Italy
| |
Collapse
|
41
|
Mutsuga M, Asaoka Y, Imura N, Miyoshi T, Togashi Y. Aminoglutethimide-induced lysosomal changes in adrenal gland in mice. ACTA ACUST UNITED AC 2017; 69:424-429. [DOI: 10.1016/j.etp.2017.04.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 03/16/2017] [Accepted: 04/03/2017] [Indexed: 10/19/2022]
|
42
|
Pottinger TG. Modulation of the stress response in wild fish is associated with variation in dissolved nitrate and nitrite. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2017; 225:550-558. [PMID: 28318786 DOI: 10.1016/j.envpol.2017.03.021] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 02/24/2017] [Accepted: 03/09/2017] [Indexed: 06/06/2023]
Abstract
Disruption of non-reproductive endocrine systems in wildlife by chemicals has received little attention but represents a potentially significant problem. Nitrate is a major anthropogenic contaminant in the freshwater aquatic environment and has been identified as a potential disrupter of endocrine function in aquatic animals. This study was conducted to investigate the relationship between the function of the neuroendocrine stress axis in fish and inorganic N loading along reaches of rivers receiving cumulative point source and diffuse chemical inputs. To accomplish this, the responsiveness of the stress axis, quantified as the rate of release of cortisol to water across the gills during exposure to a standardised stressor, was measured in three-spined sticklebacks (Gasterosteus aculeatus L.) resident at three sites on each of four rivers in north-west England. The magnitude of the stress response in fish captured at the sites furthest downstream on all rivers was more than twice that of fish captured at upstream sites. Site-specific variation in stress axis reactivity was better explained by between-site variation in concentrations of dissolved nitrate, nitrite, and ammonia than by the concentration of wastewater treatment works effluent. An increase in the magnitude of the stress response was seen among sticklebacks at sites where long-term averaged concentrations of NH3-N, NO3-N and NO2-N exceeded 0.6, 4.0 and 0.1 mg/L respectively. These data suggest that either (i) inorganic N is a better surrogate than wastewater effluent concentration for an unknown factor or factors affecting stress axis function in fish, or (ii) dissolved inorganic N directly exerts a disruptive influence on the function of the neuroendocrine stress axis in fish, supporting concerns that nitrate is an endocrine-modulating chemical.
Collapse
Affiliation(s)
- Tom G Pottinger
- Centre for Ecology & Hydrology, Lancaster Environment Centre, Library Avenue, Bailrigg, Lancaster LA1 4AP, UK.
| |
Collapse
|
43
|
Steroid profiling in H295R cells to identify chemicals potentially disrupting the production of adrenal steroids. Toxicology 2017; 381:51-63. [DOI: 10.1016/j.tox.2017.02.010] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Revised: 02/09/2017] [Accepted: 02/16/2017] [Indexed: 12/16/2022]
|
44
|
Yazawa T, Imamichi Y, Miyamoto K, Khan MRI, Uwada J, Umezawa A, Taniguchi T. Induction of steroidogenic cells from adult stem cells and pluripotent stem cells [Review]. Endocr J 2016; 63:943-951. [PMID: 27681884 DOI: 10.1507/endocrj.ej16-0373] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Steroid hormones are mainly produced in adrenal glands and gonads. Because steroid hormones play vital roles in various physiological processes, replacement of deficient steroid hormones by hormone replacement therapy (HRT) is necessary for patients with adrenal and gonadal failure. In addition to HRT, tissue regeneration using stem cells is predicted to provide novel therapy. Among various stem cell types, mesenchymal stem cells can be differentiated into steroidogenic cells following ectopic expression of nuclear receptor (NR) 5A subfamily proteins, steroidogenic factor-1 (also known as adrenal 4 binding protein) and liver receptor homolog-1, with the aid of cAMP signaling. Conversely, these approaches cannot be applied to pluripotent stem cells, such as embryonic stem cells and induced pluripotent stem cells, because of poor survival following cytotoxic expression of NR5A subfamily proteins. However, if pluripotent stem cells are first differentiated through mesenchymal lineage, they can also be differentiated into steroidogenic cells via NR5A subfamily protein expression. This approach offers a potential suitable cells for future regenerative medicine and gene therapy for diseases caused by steroidogenesis deficiencies. It represents a powerful tool to investigate the molecular mechanisms involved in steroidogenesis. This article highlights our own and current research on the induction of steroidogenic cells from various stem cells. We also discuss the future direction of their clinical application.
Collapse
Affiliation(s)
- Takashi Yazawa
- Department of Biochemistry, Asahikawa Medical University, Asahikawa 078-8510, Japan
| | | | | | | | | | | | | |
Collapse
|
45
|
Oskarsson A, Ullerås E, Ohlsson Andersson Å. Acetaminophen Increases Aldosterone Secretion While Suppressing Cortisol and Androgens: A Possible Link to Increased Risk of Hypertension. Am J Hypertens 2016; 29:1158-64. [PMID: 27217499 DOI: 10.1093/ajh/hpw055] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 05/09/2016] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Acetaminophen (paracetamol) is a widely used analgesic and antipyretic drug. Potential side effects are of public health concern, and liver toxicity from acute overdose is well known. More recently, a regular use of acetaminophen has been associated with an increased risk of hypertension. METHODS We investigated effects of acetaminophen on steroidogenesis as a possible mechanism for the hypertensive action by using the human adrenocortical cell line, H295R. Cells were treated with 0.1, 0.5, and 1mM of acetaminophen for 24 hours, and secretion of steroids and gene expression of key steps in the steroidogenesis were investigated. RESULTS Progesterone and aldosterone secretion were increased dose dependently, while secretion of 17α-OH-progesterone and cortisol as well as dehydroepiandrosterone and androstenedione was decreased. CYP17α-hydroxylase activity, assessed by the ratio 17α-OH-progesterone/progesterone, and CYP17-lyase activity, assessed by the ratio androstenedione/17α-OH-progesterone, were both dose-dependently decreased by acetaminophen. No effects were revealed on cell viability. Treatment of cells with 0.5mM of acetaminophen did not cause any effects on the expression of 10 genes in the steroidogenic pathways. CONCLUSIONS The pattern of steroid secretion caused by acetaminophen can be explained by inhibition of CYP17A1 enzyme activity. A decreased secretion of glucocorticoids and androgens, as demonstrated by acetaminophen, would, in an in vivo situation, induce adrenocorticotropic hormone release via negative feedback in the hypothalamic-pituitary-adrenal axis and result in an upregulation of aldosterone secretion. Our results suggest a novel possible mechanism for acetaminophen-induced hypertension, which needs to be further elucidated in clinical investigations.
Collapse
Affiliation(s)
- Agneta Oskarsson
- Department of Biomedical Sciences and Veterinary Public Health, Swedish University of Agricultural Sciences, Uppsala, Sweden;
| | - Erik Ullerås
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Åsa Ohlsson Andersson
- Department of Biomedical Sciences and Veterinary Public Health, Swedish University of Agricultural Sciences, Uppsala, Sweden
| |
Collapse
|
46
|
Merlo E, Podratz PL, Sena GC, de Araújo JFP, Lima LCF, Alves ISS, Gama-de-Souza LN, Pelição R, Rodrigues LCM, Brandão PAA, Carneiro MTWD, Pires RGW, Martins-Silva C, Alarcon TA, Miranda-Alves L, Silva IV, Graceli JB. The Environmental Pollutant Tributyltin Chloride Disrupts the Hypothalamic-Pituitary-Adrenal Axis at Different Levels in Female Rats. Endocrinology 2016; 157:2978-95. [PMID: 27267847 DOI: 10.1210/en.2015-1896] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Tributyltin chloride (TBT) is an environmental contaminant that is used as a biocide in antifouling paints. TBT has been shown to induce endocrine-disrupting effects. However, studies evaluating the effects of TBT on the hypothalamus-pituitary-adrenal (HPA) axis are especially rare. The current study demonstrates that exposure to TBT is critically responsible for the improper function of the mammalian HPA axis as well as the development of abnormal morphophysiology in the pituitary and adrenal glands. Female rats were treated with TBT, and their HPA axis morphophysiology was assessed. High CRH and low ACTH expression and high plasma corticosterone levels were detected in TBT rats. In addition, TBT leads to an increased in the inducible nitric oxide synthase protein expression in the hypothalamus of TBT rats. Morphophysiological abnormalities, including increases in inflammation, a disrupted cellular redox balance, apoptosis, and collagen deposition in the pituitary and adrenal glands, were observed in TBT rats. Increases in adiposity and peroxisome proliferator-activated receptor-γ protein expression in the adrenal gland were observed in TBT rats. Together, these data provide in vivo evidence that TBT leads to functional dissociation between CRH, ACTH, and costicosterone, which could be associated an inflammation and increased of inducible nitric oxide synthase expression in hypothalamus. Thus, TBT exerts toxic effects at different levels on the HPA axis function.
Collapse
Affiliation(s)
- Eduardo Merlo
- Department of Morphology (E.M., P.L.P., G.C.S., J.F.P.d.A., I.S.S.A., L.N.G.-d.S., I.V.S., J.B.G.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Biophysics and Physiology (L.C.F.L.), Federal University of Minas Gerais, Vitória ES, 29040090 Brazil; Department of Physiological Sciences (R.P., L.C.M.R., R.G.W.P., C.M.-S., T.A.A.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Chemistry (P.A.A.B., M.T.W.D.C.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Experimental Endocrinology Research Group (L.M.-A.), Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil; and Postgraduate Program in Endocrinology (L.M.-A.), School of Medicine, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil
| | - Priscila L Podratz
- Department of Morphology (E.M., P.L.P., G.C.S., J.F.P.d.A., I.S.S.A., L.N.G.-d.S., I.V.S., J.B.G.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Biophysics and Physiology (L.C.F.L.), Federal University of Minas Gerais, Vitória ES, 29040090 Brazil; Department of Physiological Sciences (R.P., L.C.M.R., R.G.W.P., C.M.-S., T.A.A.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Chemistry (P.A.A.B., M.T.W.D.C.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Experimental Endocrinology Research Group (L.M.-A.), Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil; and Postgraduate Program in Endocrinology (L.M.-A.), School of Medicine, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil
| | - Gabriela C Sena
- Department of Morphology (E.M., P.L.P., G.C.S., J.F.P.d.A., I.S.S.A., L.N.G.-d.S., I.V.S., J.B.G.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Biophysics and Physiology (L.C.F.L.), Federal University of Minas Gerais, Vitória ES, 29040090 Brazil; Department of Physiological Sciences (R.P., L.C.M.R., R.G.W.P., C.M.-S., T.A.A.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Chemistry (P.A.A.B., M.T.W.D.C.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Experimental Endocrinology Research Group (L.M.-A.), Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil; and Postgraduate Program in Endocrinology (L.M.-A.), School of Medicine, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil
| | - Julia F P de Araújo
- Department of Morphology (E.M., P.L.P., G.C.S., J.F.P.d.A., I.S.S.A., L.N.G.-d.S., I.V.S., J.B.G.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Biophysics and Physiology (L.C.F.L.), Federal University of Minas Gerais, Vitória ES, 29040090 Brazil; Department of Physiological Sciences (R.P., L.C.M.R., R.G.W.P., C.M.-S., T.A.A.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Chemistry (P.A.A.B., M.T.W.D.C.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Experimental Endocrinology Research Group (L.M.-A.), Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil; and Postgraduate Program in Endocrinology (L.M.-A.), School of Medicine, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil
| | - Leandro C F Lima
- Department of Morphology (E.M., P.L.P., G.C.S., J.F.P.d.A., I.S.S.A., L.N.G.-d.S., I.V.S., J.B.G.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Biophysics and Physiology (L.C.F.L.), Federal University of Minas Gerais, Vitória ES, 29040090 Brazil; Department of Physiological Sciences (R.P., L.C.M.R., R.G.W.P., C.M.-S., T.A.A.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Chemistry (P.A.A.B., M.T.W.D.C.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Experimental Endocrinology Research Group (L.M.-A.), Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil; and Postgraduate Program in Endocrinology (L.M.-A.), School of Medicine, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil
| | - Izabela S S Alves
- Department of Morphology (E.M., P.L.P., G.C.S., J.F.P.d.A., I.S.S.A., L.N.G.-d.S., I.V.S., J.B.G.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Biophysics and Physiology (L.C.F.L.), Federal University of Minas Gerais, Vitória ES, 29040090 Brazil; Department of Physiological Sciences (R.P., L.C.M.R., R.G.W.P., C.M.-S., T.A.A.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Chemistry (P.A.A.B., M.T.W.D.C.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Experimental Endocrinology Research Group (L.M.-A.), Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil; and Postgraduate Program in Endocrinology (L.M.-A.), School of Medicine, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil
| | - Letícia N Gama-de-Souza
- Department of Morphology (E.M., P.L.P., G.C.S., J.F.P.d.A., I.S.S.A., L.N.G.-d.S., I.V.S., J.B.G.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Biophysics and Physiology (L.C.F.L.), Federal University of Minas Gerais, Vitória ES, 29040090 Brazil; Department of Physiological Sciences (R.P., L.C.M.R., R.G.W.P., C.M.-S., T.A.A.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Chemistry (P.A.A.B., M.T.W.D.C.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Experimental Endocrinology Research Group (L.M.-A.), Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil; and Postgraduate Program in Endocrinology (L.M.-A.), School of Medicine, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil
| | - Renan Pelição
- Department of Morphology (E.M., P.L.P., G.C.S., J.F.P.d.A., I.S.S.A., L.N.G.-d.S., I.V.S., J.B.G.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Biophysics and Physiology (L.C.F.L.), Federal University of Minas Gerais, Vitória ES, 29040090 Brazil; Department of Physiological Sciences (R.P., L.C.M.R., R.G.W.P., C.M.-S., T.A.A.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Chemistry (P.A.A.B., M.T.W.D.C.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Experimental Endocrinology Research Group (L.M.-A.), Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil; and Postgraduate Program in Endocrinology (L.M.-A.), School of Medicine, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil
| | - Lívia C M Rodrigues
- Department of Morphology (E.M., P.L.P., G.C.S., J.F.P.d.A., I.S.S.A., L.N.G.-d.S., I.V.S., J.B.G.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Biophysics and Physiology (L.C.F.L.), Federal University of Minas Gerais, Vitória ES, 29040090 Brazil; Department of Physiological Sciences (R.P., L.C.M.R., R.G.W.P., C.M.-S., T.A.A.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Chemistry (P.A.A.B., M.T.W.D.C.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Experimental Endocrinology Research Group (L.M.-A.), Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil; and Postgraduate Program in Endocrinology (L.M.-A.), School of Medicine, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil
| | - Poliane A A Brandão
- Department of Morphology (E.M., P.L.P., G.C.S., J.F.P.d.A., I.S.S.A., L.N.G.-d.S., I.V.S., J.B.G.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Biophysics and Physiology (L.C.F.L.), Federal University of Minas Gerais, Vitória ES, 29040090 Brazil; Department of Physiological Sciences (R.P., L.C.M.R., R.G.W.P., C.M.-S., T.A.A.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Chemistry (P.A.A.B., M.T.W.D.C.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Experimental Endocrinology Research Group (L.M.-A.), Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil; and Postgraduate Program in Endocrinology (L.M.-A.), School of Medicine, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil
| | - Maria T W D Carneiro
- Department of Morphology (E.M., P.L.P., G.C.S., J.F.P.d.A., I.S.S.A., L.N.G.-d.S., I.V.S., J.B.G.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Biophysics and Physiology (L.C.F.L.), Federal University of Minas Gerais, Vitória ES, 29040090 Brazil; Department of Physiological Sciences (R.P., L.C.M.R., R.G.W.P., C.M.-S., T.A.A.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Chemistry (P.A.A.B., M.T.W.D.C.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Experimental Endocrinology Research Group (L.M.-A.), Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil; and Postgraduate Program in Endocrinology (L.M.-A.), School of Medicine, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil
| | - Rita G W Pires
- Department of Morphology (E.M., P.L.P., G.C.S., J.F.P.d.A., I.S.S.A., L.N.G.-d.S., I.V.S., J.B.G.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Biophysics and Physiology (L.C.F.L.), Federal University of Minas Gerais, Vitória ES, 29040090 Brazil; Department of Physiological Sciences (R.P., L.C.M.R., R.G.W.P., C.M.-S., T.A.A.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Chemistry (P.A.A.B., M.T.W.D.C.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Experimental Endocrinology Research Group (L.M.-A.), Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil; and Postgraduate Program in Endocrinology (L.M.-A.), School of Medicine, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil
| | - Cristina Martins-Silva
- Department of Morphology (E.M., P.L.P., G.C.S., J.F.P.d.A., I.S.S.A., L.N.G.-d.S., I.V.S., J.B.G.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Biophysics and Physiology (L.C.F.L.), Federal University of Minas Gerais, Vitória ES, 29040090 Brazil; Department of Physiological Sciences (R.P., L.C.M.R., R.G.W.P., C.M.-S., T.A.A.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Chemistry (P.A.A.B., M.T.W.D.C.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Experimental Endocrinology Research Group (L.M.-A.), Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil; and Postgraduate Program in Endocrinology (L.M.-A.), School of Medicine, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil
| | - Tamara A Alarcon
- Department of Morphology (E.M., P.L.P., G.C.S., J.F.P.d.A., I.S.S.A., L.N.G.-d.S., I.V.S., J.B.G.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Biophysics and Physiology (L.C.F.L.), Federal University of Minas Gerais, Vitória ES, 29040090 Brazil; Department of Physiological Sciences (R.P., L.C.M.R., R.G.W.P., C.M.-S., T.A.A.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Chemistry (P.A.A.B., M.T.W.D.C.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Experimental Endocrinology Research Group (L.M.-A.), Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil; and Postgraduate Program in Endocrinology (L.M.-A.), School of Medicine, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil
| | - Leandro Miranda-Alves
- Department of Morphology (E.M., P.L.P., G.C.S., J.F.P.d.A., I.S.S.A., L.N.G.-d.S., I.V.S., J.B.G.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Biophysics and Physiology (L.C.F.L.), Federal University of Minas Gerais, Vitória ES, 29040090 Brazil; Department of Physiological Sciences (R.P., L.C.M.R., R.G.W.P., C.M.-S., T.A.A.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Chemistry (P.A.A.B., M.T.W.D.C.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Experimental Endocrinology Research Group (L.M.-A.), Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil; and Postgraduate Program in Endocrinology (L.M.-A.), School of Medicine, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil
| | - Ian V Silva
- Department of Morphology (E.M., P.L.P., G.C.S., J.F.P.d.A., I.S.S.A., L.N.G.-d.S., I.V.S., J.B.G.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Biophysics and Physiology (L.C.F.L.), Federal University of Minas Gerais, Vitória ES, 29040090 Brazil; Department of Physiological Sciences (R.P., L.C.M.R., R.G.W.P., C.M.-S., T.A.A.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Chemistry (P.A.A.B., M.T.W.D.C.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Experimental Endocrinology Research Group (L.M.-A.), Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil; and Postgraduate Program in Endocrinology (L.M.-A.), School of Medicine, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil
| | - Jones B Graceli
- Department of Morphology (E.M., P.L.P., G.C.S., J.F.P.d.A., I.S.S.A., L.N.G.-d.S., I.V.S., J.B.G.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Biophysics and Physiology (L.C.F.L.), Federal University of Minas Gerais, Vitória ES, 29040090 Brazil; Department of Physiological Sciences (R.P., L.C.M.R., R.G.W.P., C.M.-S., T.A.A.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Department of Chemistry (P.A.A.B., M.T.W.D.C.), Federal University of Espírito Santo, Vitória ES, 29040090 Brazil; Experimental Endocrinology Research Group (L.M.-A.), Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil; and Postgraduate Program in Endocrinology (L.M.-A.), School of Medicine, Federal University of Rio de Janeiro, Vitória ES, 29040090 Brazil
| |
Collapse
|
47
|
Ahmad S, Khan MF, Parvez S, Akhtar M, Raisuddin S. Molecular docking reveals the potential of phthalate esters to inhibit the enzymes of the glucocorticoid biosynthesis pathway. J Appl Toxicol 2016; 37:265-277. [DOI: 10.1002/jat.3355] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 05/08/2016] [Accepted: 05/10/2016] [Indexed: 02/06/2023]
Affiliation(s)
- Shahzad Ahmad
- Department of Medical Elementology and Toxicology; Jamia Hamdard (Hamdard University); New Delhi 110062 India
| | - Mohemmed Faraz Khan
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy; Jamia Hamdard (Hamdard University); New Delhi 110062 India
| | - Suhel Parvez
- Department of Medical Elementology and Toxicology; Jamia Hamdard (Hamdard University); New Delhi 110062 India
| | - Mohammad Akhtar
- Department of Pharmacology, Faculty of Pharmacy; Jamia Hamdard (Hamdard University); New Delhi 110062 India
| | - Sheikh Raisuddin
- Department of Medical Elementology and Toxicology; Jamia Hamdard (Hamdard University); New Delhi 110062 India
| |
Collapse
|
48
|
Odermatt A, Strajhar P, Engeli RT. Disruption of steroidogenesis: Cell models for mechanistic investigations and as screening tools. J Steroid Biochem Mol Biol 2016; 158:9-21. [PMID: 26807866 DOI: 10.1016/j.jsbmb.2016.01.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Revised: 12/31/2015] [Accepted: 01/20/2016] [Indexed: 02/03/2023]
Abstract
In the modern world, humans are exposed during their whole life to a large number of synthetic chemicals. Some of these chemicals have the potential to disrupt endocrine functions and contribute to the development and/or progression of major diseases. Every year approximately 1000 novel chemicals, used in industrial production, agriculture, consumer products or as pharmaceuticals, are reaching the market, often with limited safety assessment regarding potential endocrine activities. Steroids are essential endocrine hormones, and the importance of the steroidogenesis pathway as a target for endocrine disrupting chemicals (EDCs) has been recognized by leading scientists and authorities. Cell lines have a prominent role in the initial stages of toxicity assessment, i.e. for mechanistic investigations and for the medium to high throughput analysis of chemicals for potential steroidogenesis disrupting activities. Nevertheless, the users have to be aware of the limitations of the existing cell models in order to apply them properly, and there is a great demand for improved cell-based testing systems and protocols. This review intends to provide an overview of the available cell lines for studying effects of chemicals on gonadal and adrenal steroidogenesis, their use and limitations, as well as the need for future improvements of cell-based testing systems and protocols.
Collapse
Affiliation(s)
- Alex Odermatt
- Swiss Center for Human Toxicology and Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, Pharmacenter, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland.
| | - Petra Strajhar
- Swiss Center for Human Toxicology and Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, Pharmacenter, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland
| | - Roger T Engeli
- Swiss Center for Human Toxicology and Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, Pharmacenter, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland
| |
Collapse
|
49
|
Annane D. The Role of ACTH and Corticosteroids for Sepsis and Septic Shock: An Update. Front Endocrinol (Lausanne) 2016; 7:70. [PMID: 27379022 PMCID: PMC4913096 DOI: 10.3389/fendo.2016.00070] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 06/08/2016] [Indexed: 02/05/2023] Open
Abstract
Sepsis is a common disorder associated with high morbidity and mortality. It is now defined as an abnormal host response to infection, resulting in life-threatening dysfunction of organs. There is evidence from in vitro and in vivo experiments in various animal models and in patients that endotoxin or sepsis may directly and indirectly alter the hypothalamic-pituitary-adrenal response to severe infection. These alterations may include necrosis or hemorrhage or inflammatory mediator-mediated decreased ACTH synthesis, steroidogenesis, cortisol delivery to tissues, clearance from plasma, and decreased sensitivity of tissues to cortisol. Disruption of the hypothalamic-pituitary-adrenal axis may translate in patients with sepsis into cardiovascular and other organ dysfunction, and eventually an increase in the risk of death. Exogenous administration of corticosteroids at moderate dose, i.e., <400 mg of hydrocortisone or equivalent for >96 h, may help reversing sepsis-associated shock and organ dysfunction. Corticosteroids may also shorten the duration of stay in the ICU. Except for increased blood glucose and sodium levels, treatment with corticosteroids was rather well tolerated in the context of clinical trials. The benefit of treatment on survival remains controversial. Based on available randomized controlled trials, the likelihood of survival benefit is greater in septic shock versus sepsis patients, in sepsis with acute respiratory distress syndrome or with community-acquired pneumonia versus patients without these conditions, and in patients with a blunted cortisol response to 250 μg of ACTH test versus those with normal response.
Collapse
Affiliation(s)
- Djillali Annane
- General Intensive Care Unit, Raymond Poincaré Hospital (AP-HP), Garches, France
- Laboratory of Infection and Inflammation, U1173, University of Versailles Saint-Quentin-en-Yvelines University, INSERM, Garches, France
- *Correspondence: Djillali Annane,
| |
Collapse
|
50
|
Gu H, Zhang M, Cai M, Liu J. Combined Use of Etomidate and Dexmedetomidine Produces an Additive Effect in Inhibiting the Secretion of Human Adrenocortical Hormones. Med Sci Monit 2015; 21:3528-35. [PMID: 26568275 PMCID: PMC4654590 DOI: 10.12659/msm.894728] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Background The direct effects of etomidate were investigated on the secretion of cortisol and its precursors by dispersed cells from the adrenal cortex of human of animals. Dexmedetomidine (DEX) is an anesthetic agent that may interfere with cortisol secretion via an unknown mechanism, such as involving inhibition of 11β-hydroxylase and the cholesterol side-chain cleavage enzyme system. The aim of this study was to determine whether dexmedetomidine (DEX) has a similar inhibitory effect on adrenocortical function, and whether combined use of etomidate (ETO) and DEX could produce a synergistic action in inhibiting the secretion of human adrenocortical hormones. Material/Methods Human adrenocortical cells were exposed to different concentrations of ETO and DEX. The dose-effect model between the ETO concentration and the mean secretion of cortisone (CORT) and aldosterone (ALDO) per hour was estimated. Results Hill’s equation well-described the dose-effect correlation between the ETO concentration and the amount of ALDO and CORT secretion. When the DEX concentration was introduced into the model by using E0 (basal secretion) as the covariate, the goodness of fit of the ETO-CORT dose-effect model was improved significantly and the objective function value was reduced by 4.55 points (P<0.05). The parameters of the final ETO-ALDO pharmacodynamics model were EC50=9.74, Emax=1.20, E0=1.33, and γ=18.5; the parameters of the final ETO-CORT pharmacodynamics model were EC50=9.49, Emax=8.16, E0=8.57, and γ=37.0. In the presence of DEX, E0 was 8.57–0.0247×(CDEX–4.6), and the other parameters remained unchanged. All parameters but γ were natural logarithm conversion values. Conclusions Combined use of DEX and ETO reduced ETO’s inhibitory E0 (basal secretion) of CORT from human adrenocortical cells in a dose-dependent manner, suggesting that combined use of ETO and DEX produced an additive effect in inhibiting the secretion of human adrenocortical hormones.
Collapse
Affiliation(s)
- Hongbin Gu
- Department of Anesthesiology, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, China (mainland)
| | - Mazhong Zhang
- Department of Anesthesiology, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, China (mainland)
| | - Meihua Cai
- Department of Anesthesiology, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, China (mainland)
| | - Jinfen Liu
- Department of Cardiothoracic Surgery, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, China (mainland)
| |
Collapse
|