1
|
Kaštelan S, Hat K, Tomić Z, Matejić T, Gotovac N. Sex Differences in the Lacrimal Gland: Implications for Dry Eye Disease. Int J Mol Sci 2025; 26:3833. [PMID: 40332492 PMCID: PMC12028224 DOI: 10.3390/ijms26083833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Revised: 04/10/2025] [Accepted: 04/16/2025] [Indexed: 05/08/2025] Open
Abstract
Sexual dimorphism significantly impacts the lacrimal gland's structure, function, and ageing processes, playing an important role in dry eye disease (DED) pathophysiology. This multifactorial disorder, characterised by tear film instability, inflammation, and visual impairment, disproportionately affects women, especially after menopause. It highlights the interplay between sex steroid hormones, lacrimal gland function, and environmental factors. Systemic and local androgens are vital for maintaining lacrimal gland health and tear production, while the role of oestrogens remains less clear. Evidence suggests dose and context-dependent effects on inflammation and glandular function. Histopathological and molecular studies reveal significant sex differences in the lacrimal gland, with women exhibiting more pronounced age-related degenerative changes, including fibrosis and acinar atrophy, contributing to their increased susceptibility to DED. Despite these findings, the underlying mechanisms connecting sex steroid hormones, receptor expression, and local tissue regulation to these disparities remain poorly understood, highlighting the need for further research. This review synthesises the current knowledge of sex-specific differences in the lacrimal gland, emphasising the importance of integrating systemic and local biomarkers, histological data, and molecular insights into personalised therapeutic strategies. By tailoring treatments to patients' unique hormonal and molecular profiles, personalised medicine has the potential to transform DED management, addressing unmet clinical needs and improving outcomes.
Collapse
Affiliation(s)
- Snježana Kaštelan
- Department of Ophthalmology, Clinical Hospital Dubrava, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Koraljka Hat
- Department of Maxillofacial Surgery, Clinical Hospital Dubrava, University of Zagreb School of Dental Medicine, 10000 Zagreb, Croatia
| | - Zora Tomić
- Health Centre of the Croatian Department of Internal Affairs, 10000 Zagreb, Croatia
| | - Tomislav Matejić
- Surgery Clinic, Clinical Hospital Sveti Duh, 10000 Zagreb, Croatia
| | - Nikola Gotovac
- Department of Clinical Radiology, General Hospital Požega, Faculty of Dental Medicine and Health, J. J. Strossmayer University of Osijek, 31000 Osijek, Croatia
| |
Collapse
|
2
|
Manna PR, Yang S, Reddy PH. Transcriptomic analysis reveals suppression of steroidogenic acute regulatory protein in gender-specific differences in Alzheimer's disease. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167667. [PMID: 39809368 DOI: 10.1016/j.bbadis.2025.167667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 01/05/2025] [Accepted: 01/08/2025] [Indexed: 01/16/2025]
Abstract
Alzheimer's disease (AD)-related dementia preferentially impacts two-thirds of women and one-third of men. The steroidogenic acute regulatory (StAR) protein mediates the biosynthesis of neurosteroids that sustain diverse neuronal activities. Aging, involving neurosteroidal imbalance, is the predominant risk factor for AD causing dementia. Transcriptomic analysis, including clinical cognitive diagnosis (cogdx) stages, displays marked attenuation of StAR in brains of AD women than those of AD men, compared with cognitively normal (Non-AD) subjects. The present data provide the first evidence and new insights into the mechanism exemplifying the suppression of StAR in gender-specific differences in AD.
Collapse
Affiliation(s)
- Pulak R Manna
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.
| | - Shengping Yang
- Department of Biostatistics, Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA 70808, USA
| | - P Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Neurology, Departments of School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Public Health Department of Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Speech, Language and Hearing Sciences, School Health Professions, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| |
Collapse
|
3
|
Manna PR, Yang S, Manna C, Waters H, Islam MA, Reddy AP, Rawat P, Reddy PH. Steroidogenic acute regulatory protein mediated variations of gender-specific sex neurosteroids in Alzheimer's disease: Relevance to hormonal and neuronal imbalance. Neurosci Biobehav Rev 2025; 169:105969. [PMID: 39631487 DOI: 10.1016/j.neubiorev.2024.105969] [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: 05/13/2024] [Revised: 07/24/2024] [Accepted: 12/01/2024] [Indexed: 12/07/2024]
Abstract
The steroidogenic acute regulatory (StAR) protein mediates the rate-liming step in neuro/steroid biosynthesis. Multifaceted and delicate changes during aging, disrupting hormonal and neuronal homeostasis, constitute human senescence, an inevitable phenomenon that attributes to increased morbidity and mortality. Aging, along with progressive decreases in bioactive neurosteroids, is the primary risk factor for Alzheimer's disease (AD), which preferentially impacts two-thirds of women and one-third of men. AD is neuropathologically characterized by the accumulation of extracellular amyloid-β and intracellular phosphorylated Tau containing neurofibrillary tangles, resulting in dementia. Postmortem brains pertaining to gender-specific AD patients exhibit varied suppression of StAR and sex neurosteroid levels compared with age-matched cognitively healthy subjects, in which the attenuation of StAR is inversely correlated with the AD pathological markers. Interestingly, retinoid signaling upregulates StAR-motivated neurosteroid biosynthesis and reinstates various neurodegenerative vulnerabilities that promote AD pathogenesis. This review summarizes current understanding of StAR-driven alterations of sex neurosteroids in gender-specific AD risks and provides biochemical and molecular insights into therapeutic interventions for preventing and/or alleviating dementia for healthy aging.
Collapse
Affiliation(s)
- Pulak R Manna
- Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.
| | - Shengping Yang
- Department of Biostatistics, Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA 70808, USA
| | - Chayan Manna
- Baylor College of Medicine, Ben Taub Research Center, 1 Baylor Plaza, Houston, TX 77030, USA
| | - Hope Waters
- Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Md Ariful Islam
- Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Arubala P Reddy
- Nutritional Sciences Department, College of Human Sciences, Texas Tech University, Lubbock, TX 79409, USA
| | - Priyanka Rawat
- Nutritional Sciences Department, College of Human Sciences, Texas Tech University, Lubbock, TX 79409, USA
| | - P Hemachandra Reddy
- Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Nutritional Sciences Department, College of Human Sciences, Texas Tech University, Lubbock, TX 79409, USA; Neurology, Departments of School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Public Health Department of Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Speech, Language and Hearing Sciences, School Health Professions, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.
| |
Collapse
|
4
|
Zhang H, Liu A, Bo W, Zhang M, Wang H, Feng X, Wu Y. Upregulation of HSD11B1 promotes cortisol production and inhibits NK cell activation in pancreatic adenocarcinoma. Mol Immunol 2024; 175:10-19. [PMID: 39276709 DOI: 10.1016/j.molimm.2024.08.005] [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: 01/21/2024] [Revised: 08/12/2024] [Accepted: 08/22/2024] [Indexed: 09/17/2024]
Abstract
Cortisol is a glucocorticoid hormone that has immunosuppressive function. Elevated basal cortisol levels are present in patients with some kinds of cancers, but its role in the microenvironment of pancreatic adenocarcinoma (PAAD) remains unclear. This study analyzed the expression of genes involved in cortisol generation by using high-throughput sequencing data from TCGA portal and found HSD11B1 was significantly upregulated in patients with PAAD. The correlations between HSD11B1 level and the expression of 23 immunosuppressive receptors were analyzed by Spearman's correlation analysis. The function of HSD11B1 was examined in primary NK cells and PAAD cell lines. The levels of cortisol in medium and cell lysates were detected by ELISA. In vitro killing assay was used to evaluate the cytotoxicity of NK cells. Cell surface levels of CD96, Tim-3, PD-1, TIGIT, CTLA-4, NKp46, NKp30, NKD2G and LFA-1A, and intracellular levels of CD107a and IFN-γ were examined by flow cytometry. We observed that patients with higher HSD11B1 level had shorter survival time. HSD11B1 is positively correlated with the mRNA levels of 11 immunosuppressive receptors in PAAD. Higher HSD11B1 level relates to reduced abundance of activated NK cells in the tumors. HSD11B1 overexpressed NK cells exhibit exhausted phenotype with increased cortisol production, reduced viability, and reduced cytotoxicity against cancer cells. Overexpression of HSD11B1 did not change the viability of tumor cells but upregulated cortisol production. Targeting HSD11B1 by a specific inhibitor improved the NK cells responsiveness. In conclusion, HSD11B1 is upregulated in patients with PAAD, and higher HSD11B1 level is related to poor prognosis. Upregulation of HSD11B1 in NK and tumor cells increased the production and secretion of cortisol and induces NK cell exhaustion.
Collapse
Affiliation(s)
- Hui Zhang
- Department of Hepatopancreatobiliary Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Aixiang Liu
- Department of Hepatopancreatobiliary Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Wentao Bo
- Department of Hepatopancreatobiliary Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Mingyi Zhang
- Department of Hepatopancreatobiliary Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Haiqing Wang
- Department of Hepatopancreatobiliary Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Xielin Feng
- Department of Hepatopancreatobiliary Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Ying Wu
- Department of Hepatopancreatobiliary Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China; Department of Medical Oncology, Daytime Medical Treatment Area, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China.
| |
Collapse
|
5
|
Lin YC, Papadopoulos V. Expression of pregnenolone-synthesizing enzymes CYP11A1 and CYP1B1 in the human, rat, and mouse brain. Steroids 2024; 212:109521. [PMID: 39395524 DOI: 10.1016/j.steroids.2024.109521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 10/07/2024] [Accepted: 10/09/2024] [Indexed: 10/14/2024]
Abstract
The central nervous system (CNS) is capable of synthesizing steroids for modulating essential functions such as neurotransmission, neuroplasticity, and neuroinflammation. These locally synthesized steroids, called neurosteroids, are produced through the conversion of cholesterol into the major steroid precursor pregnenolone, followed by downstream metabolism to form various steroids such as progesterone and allopregnanolone. Given that changes in neurosteroids are implicated in many neurological and psychiatric disorders, understanding the neurosteroidogenesis pathway is crucial. Recent studies have demonstrated an alternative pathway for the biosynthesis of pregnenolone, which is classically produced by CYP11A1 but was found instead to be made by CYP1B1 in human glial cells. However, numerous studies have demonstrated Cyp11a1 expression and activity in rodent brain tissue and brain cells. To elucidate whether species differences exist for the pregnenolone synthesis enzyme in human and rodent brains, we sought to directly compare the expression levels of CYP11A1 and CYP1B1 in human, rat, and mouse CNS tissues. We found that CYP1B1 mRNA expression was significantly higher than that of CYP11A1 in almost all CNS brain regions in human, rat, and mouse. The exception is in the mouse cerebral cortex, where Cyp11a1 RNA was more abundant than Cyp1b1. However, Cyp11a1 protein was clearly detectable in rodent CNS while completely undetectable in human brain. In contrast, the presence of CYP1B1 protein can be observed in both human and rodent brains. These results suggest that CYP1B1 is likely the dominant pregnenolone synthesis enzyme in the human brain, while rodent brains may use both Cyp11a1 and Cyp1b1.
Collapse
Affiliation(s)
- Yiqi Christina Lin
- Department of Pharmacology and Pharmaceutical Sciences, Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA, USA
| | - Vassilios Papadopoulos
- Department of Pharmacology and Pharmaceutical Sciences, Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
6
|
Vallée M. Advances in steroid research from the pioneering neurosteroid concept to metabolomics: New insights into pregnenolone function. Front Neuroendocrinol 2024; 72:101113. [PMID: 37993022 DOI: 10.1016/j.yfrne.2023.101113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 11/13/2023] [Accepted: 11/19/2023] [Indexed: 11/24/2023]
Abstract
Advances in neuroendocrinology have led to major discoveries since the 19th century, identifying adaptive loops for maintaining homeostasis. One of the most remarkable discoveries was the concept of neurosteroids, according to which the brain is not only a target but also a source of steroid production. The identification of new membrane steroid targets now underpins the neuromodulatory effects of neurosteroids such as pregnenolone, which is involved in functions mediated by the GPCR CB1 receptor. Structural analysis of steroids is a key feature of their interactions with the phospholipid membrane, receptors and resulting activity. Therefore, mass spectrometry-based methods have been developed to elucidate the metabolic pathways of steroids, the ultimate approach being metabolomics, which allows the identification of a large number of metabolites in a single sample. This approach should enable us to make progress in understanding the role of neurosteroids in the functioning of physiological and pathological processes.
Collapse
Affiliation(s)
- Monique Vallée
- University Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000 Bordeaux, France.
| |
Collapse
|
7
|
Sandor LF, Ragacs R, Gyori DS. Local Effects of Steroid Hormones within the Bone Microenvironment. Int J Mol Sci 2023; 24:17482. [PMID: 38139309 PMCID: PMC10744126 DOI: 10.3390/ijms242417482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 12/05/2023] [Accepted: 12/11/2023] [Indexed: 12/24/2023] Open
Abstract
Steroid hormone production via the adrenal cortex, gonads, and placenta (so-called glandular steroidogenesis) is responsible for the endocrine control of the body's homeostasis and is organized by a feedback regulatory mechanism based on the hypothalamus-pituitary-steroidogenic gland axis. On the other hand, recently discovered extraglandular steroidogenesis occurring locally in different tissues is instead linked to paracrine or autocrine signaling, and it is independent of the control by the hypothalamus and pituitary glands. Bone cells, such as bone-forming osteoblasts, osteoblast-derived osteocytes, and bone-resorbing osteoclasts, respond to steroid hormones produced by both glandular and extraglandular steroidogenesis. Recently, new techniques to identify steroid hormones, as well as synthetic steroids and steroidogenesis inhibitors, have been introduced, which greatly empowered steroid hormone research. Based on recent literature and new advances in the field, here we review the local role of steroid hormones in regulating bone homeostasis and skeletal lesion formation. The novel idea of extraglandular steroidogenesis occurring within the skeletal system raises the possibility of the development of new therapies for the treatment of bone diseases.
Collapse
Affiliation(s)
| | | | - David S. Gyori
- Department of Physiology, School of Medicine, Semmelweis University, 1085 Budapest, Hungary
| |
Collapse
|
8
|
Faviana P, Boldrini L, Gronchi L, Galli L, Erba P, Gentile C, Lippolis PV, Marchetti E, Di Stefano I, Sammarco E, Chapman AD, Bardi M. Steroid Hormones as Modulators of Emotional Regulation in Male Urogenital Cancers. Int J Behav Med 2023; 30:836-848. [PMID: 36459332 PMCID: PMC10713796 DOI: 10.1007/s12529-022-10139-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/25/2022] [Indexed: 12/03/2022]
Abstract
BACKGROUND Tumors develop within an organism operating in a specific social and physical environment. Cortisol and dehydroepiandrosterone (DHEA), two of the most abundant steroid hormones in humans, are involved in both emotional regulation and the tumor progression. Several studies reported preclinical findings that DHEA can have preventive and therapeutic efficacy in treating major age-associated diseases, including cancer, although the mechanisms of action are not yet defined. The main aim of current study was to investigate the relationship between psychological and physiological emotional regulation and cancer development. METHOD This study assessed the quality of life of urogenital cancer male patients using several validated tools, including the Functional Assessment of Cancer Therapy-General and the Profile of Mood States. Saliva samples were collected to monitor peripheral activity of both cortisol and DHEA. It was hypothesized that patients with a better quality of life would have higher levels of the DHEA/cortisol ratios. RESULTS We found that the quality of life was positively related to DHEA, but not cortisol levels. Negative mood increases were related to lower levels of DHEA. Logistic regression of the predictors of metastases indicated three main independent factors involved: DHEA, age, and cortisol. In other words, the higher the DHEA levels in comparison to cortisol levels, controlling for age, the lower the probability of metastases. CONCLUSION Our results appear to support the hypothesis that emotional dysregulation mediated by DHEA/cortisol activity is a key factor in the probability of metastasis in urogenital cancers.
Collapse
Affiliation(s)
- Pinuccia Faviana
- Department of Surgical, Medical, Molecular Pathology and Critical Area, University of Pisa, Via Roma, 57, Pisa, Italy.
| | - Laura Boldrini
- Department of Surgical, Medical, Molecular Pathology and Critical Area, University of Pisa, Via Roma, 57, Pisa, Italy
| | - Lisa Gronchi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma, 57, Pisa, Italy
| | - Luca Galli
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma, 57, Pisa, Italy
| | - Paola Erba
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma, 57, Pisa, Italy
| | - Carlo Gentile
- Istituto Europeo Di Oncologia, Via Ripamonti 435, I-20132, Milan, Italy
| | | | - Elio Marchetti
- Department of Surgical, Medical, Molecular Pathology and Critical Area, University of Pisa, Via Roma, 57, Pisa, Italy
| | - Iosè Di Stefano
- Department of Surgical, Medical, Molecular Pathology and Critical Area, University of Pisa, Via Roma, 57, Pisa, Italy
| | - Enrico Sammarco
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma, 57, Pisa, Italy
| | - Alex D Chapman
- Department of Psychology and Neuroscience, Randolph-Macon College, Ashland, VA, 23005, USA
| | - Massimo Bardi
- Department of Psychology and Neuroscience, Randolph-Macon College, Ashland, VA, 23005, USA
| |
Collapse
|
9
|
Lin YC, Cheung G, Zhang Z, Papadopoulos V. Mitochondrial cytochrome P450 1B1 is involved in pregnenolone synthesis in human brain cells. J Biol Chem 2023; 299:105035. [PMID: 37442234 PMCID: PMC10413356 DOI: 10.1016/j.jbc.2023.105035] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 07/02/2023] [Accepted: 07/03/2023] [Indexed: 07/15/2023] Open
Abstract
Neurosteroids, which are steroids synthesized by the nervous system, can exert neuromodulatory and neuroprotective effects via genomic and nongenomic pathways. The neurosteroid and major steroid precursor pregnenolone has therapeutical potential in various diseases, such as psychiatric and pain disorders, and may play important roles in myelination, neuroinflammation, neurotransmission, and neuroplasticity. Although pregnenolone is synthesized by CYP11A1 in peripheral steroidogenic organs, our recent study showed that pregnenolone must be synthesized by another mitochondrial cytochrome P450 (CYP450) enzyme other than CYP11A1 in human glial cells. Therefore, we sought to identify the CYP450 responsible for pregnenolone production in the human brain. Upon screening for CYP450s expressed in the human brain that have mitochondrial localization, we identified three enzyme candidates: CYP27A1, CYP1A1, and CYP1B1. We found that inhibition of CYP27A1 through inhibitors and siRNA knockdown did not negatively affect pregnenolone synthesis in human glial cells. Meanwhile, treatment of human glial cells with CYP1A1/CYP1B1 inhibitors significantly reduced pregnenolone production in the presence of 22(R)-hydroxycholesterol. We performed siRNA knockdown of CYP1A1 or CYP1B1 in human glial cells and found that only CYP1B1 knockdown significantly decreased pregnenolone production. Furthermore, overexpression of mitochondria-targeted CYP1B1 significantly increased pregnenolone production under basal conditions and in the presence of hydroxycholesterols and low-density lipoprotein. Inhibition of CYP1A1 and/or CYP1B1 via inhibitors or siRNA knockdown did not significantly reduce pregnenolone synthesis in human adrenal cortical cells, implying that CYP1B1 is not a major pregnenolone-producing enzyme in the periphery. These data suggest that mitochondrial CYP1B1 is involved in pregnenolone synthesis in human glial cells.
Collapse
Affiliation(s)
- Yiqi Christina Lin
- Department of Pharmacology and Pharmaceutical Sciences, Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, California, USA
| | - Garett Cheung
- Department of Pharmacology and Pharmaceutical Sciences, Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, California, USA
| | - Zeyu Zhang
- Department of Pharmacology and Pharmaceutical Sciences, Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, California, USA
| | - Vassilios Papadopoulos
- Department of Pharmacology and Pharmaceutical Sciences, Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, California, USA.
| |
Collapse
|
10
|
Manna PR, Kshirsagar S, Pradeepkiran JA, Rawat P, Kumar S, Reddy AP, Reddy PH. Protective function of StAR in amyloid-β accumulated hippocampal neurotoxicity and neurosteroidogenesis: Mechanistic insights into Alzheimer's disease. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166738. [PMID: 37142132 DOI: 10.1016/j.bbadis.2023.166738] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 04/25/2023] [Accepted: 04/27/2023] [Indexed: 05/06/2023]
Abstract
The steroidogenic acute regulatory (StAR) protein principally mediates steroid hormone biosynthesis by governing the transport of intramitochondrial cholesterol. Neurosteroids progressively decrease during aging, the key risk factor for Alzheimer's disease (AD), which is triggered by brain-region specific accumulation of amyloid beta (Aβ) precursor protein (APP), a key pathological factor. We demonstrate that hippocampal neuronal cells overexpressing wild-type (WtAPP) and mutant APP (mAPP) plasmids, conditions mimetic to AD, resulted in decreases in StAR mRNA, free cholesterol, and pregnenolone levels. The magnitude of suppression of the steroidogenic response was more pronounced with mAPP than that of WtAPP. While mAPP-waned assorted anomalies correlate to AD pathology, deterioration of APP/Aβ laden StAR expression and neurosteroid biosynthesis was enhanced by retinoid signaling. An abundance of mitochondrially targeted StAR expression partially restored APP/Aβ accumulated diverse neurodegenerative vulnerabilities. Immunofluorescence analyses revealed that overexpression of StAR diminishes mAPP provoked Aβ aggregation. Co-expression of StAR and mAPP in hippocampal neurons substantially reversed the declines in mAPP mediated cell survival, mitochondrial oxygen consumption rate, and ATP production. Concurrently, induction of mAPP induced Aβ loading showed an increase in cholesterol esters, but decrease in free cholesterol, concomitant with pregnenolone biosynthesis, events that were inversely regulated by StAR. Moreover, retinoid signaling was found to augment cholesterol content for facilitating neurosteroid biosynthesis in an AD mimetic condition. These findings provide novel insights into the molecular events by which StAR acts to protect mAPP-induced hippocampal neurotoxicity, mitochondrial dysfunction, and neurosteroidogenesis, and these measures are fundamental for ameliorating and/or delaying dementia in individuals with AD.
Collapse
Affiliation(s)
- Pulak R Manna
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.
| | - Sudhir Kshirsagar
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | | | - Priyanka Rawat
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Subodh Kumar
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Center of Emphasis in Neuroscience, Department of Molecular and Translational Medicine, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX 79905, USA
| | - Arubala P Reddy
- Nutritional Sciences Department, College of Human Sciences, Texas Tech University, 1301 Akron Ave, Lubbock, TX 79409, USA
| | - P Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Nutritional Sciences Department, College of Human Sciences, Texas Tech University, 1301 Akron Ave, Lubbock, TX 79409, USA; Neurology, Departments of School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Public Health Department of Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Speech, Language and Hearing Sciences, School Health Professions, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.
| |
Collapse
|
11
|
Manna PR, Bose C, Reddy PH. Downregulation of StAR driven neurosteroid biosynthesis as a distinctive feature in the brains of Alzheimer's disease patients. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166757. [PMID: 37209874 DOI: 10.1016/j.bbadis.2023.166757] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 05/11/2023] [Accepted: 05/12/2023] [Indexed: 05/22/2023]
Affiliation(s)
- Pulak R Manna
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.
| | - Chhanda Bose
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - P Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Neurology, Departments of School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Public Health Department of Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Speech, Language and Hearing Sciences, School Health Professions, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Nutritional Sciences Department, College of Human Sciences, Texas Tech University, 1301 Akron Ave, Lubbock, TX 79409, USA.
| |
Collapse
|
12
|
Abaffy T, Lu HY, Matsunami H. Sex steroid hormone synthesis, metabolism, and the effects on the mammalian olfactory system. Cell Tissue Res 2023; 391:19-42. [PMID: 36401093 PMCID: PMC9676892 DOI: 10.1007/s00441-022-03707-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 11/03/2022] [Indexed: 11/21/2022]
Abstract
Sex steroid hormones influence olfactory-mediated social behaviors, and it is generally hypothesized that these effects result from circulating hormones and/or neurosteroids synthesized in the brain. However, it is unclear whether sex steroid hormones are synthesized in the olfactory epithelium or the olfactory bulb, and if they can modulate the activity of the olfactory sensory neurons. Here, we review important discoveries related to the metabolism of sex steroids in the mouse olfactory epithelium and olfactory bulb, along with potential areas of future research. We summarize current knowledge regarding the expression, neuroanatomical distribution, and biological activity of the steroidogenic enzymes, sex steroid receptors, and proteins that are important to the metabolism of these hormones and reflect on their potential to influence early olfactory processing. We also review evidence related to the effects of sex steroid hormones on the development and activity of olfactory sensory neurons. By better understanding how these hormones are metabolized and how they act both at the periphery and olfactory bulb level, we can better appreciate the complexity of the olfactory system and discover potential similarities and differences in early olfactory processing between sexes.
Collapse
Affiliation(s)
- Tatjana Abaffy
- Molecular Genetics and Microbiology Department, Duke University Medical Center, Durham, NC 27710 USA
| | - Hsiu-Yi Lu
- Molecular Genetics and Microbiology Department, Duke University Medical Center, Durham, NC 27710 USA
| | - Hiroaki Matsunami
- Molecular Genetics and Microbiology Department, Duke University Medical Center, Durham, NC 27710 USA
| |
Collapse
|
13
|
Gatta E, Camussi D, Auta J, Guidotti A, Pandey SC. Neurosteroids (allopregnanolone) and alcohol use disorder: From mechanisms to potential pharmacotherapy. Pharmacol Ther 2022; 240:108299. [PMID: 36323379 PMCID: PMC9810076 DOI: 10.1016/j.pharmthera.2022.108299] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 10/14/2022] [Accepted: 10/17/2022] [Indexed: 11/06/2022]
Abstract
Alcohol Use Disorder (AUD) is a multifaceted relapsing disorder that is commonly comorbid with psychiatric disorders, including anxiety. Alcohol exposure produces a plethora of effects on neurobiology. Currently, therapeutic strategies are limited, and only a few treatments - disulfiram, acamprosate, and naltrexone - are available. Given the complexity of this disorder, there is a great need for the identification of novel targets to develop new pharmacotherapy. The GABAergic system, the primary inhibitory system in the brain, is one of the well-known targets for alcohol and is responsible for the anxiolytic effects of alcohol. Interestingly, GABAergic neurotransmission is fine-tuned by neuroactive steroids that exert a regulatory role on several endocrine systems involved in neuropsychiatric disorders including AUD. Mounting evidence indicates that alcohol alters the biosynthesis of neurosteroids, whereas acute alcohol increases and chronic alcohol decreases allopregnanolone levels. Our recent work highlighted that chronic alcohol-induced changes in neurosteroid levels are mediated by epigenetic modifications, e.g., DNA methylation, affecting key enzymes involved in neurosteroid biosynthesis. These changes were associated with changes in GABAA receptor subunit expression, suggesting an imbalance between excitatory and inhibitory signaling in AUD. This review will recapitulate the role of neurosteroids in the regulation of the neuroendocrine system, highlight their role in the observed allostatic load in AUD, and develop a framework from mechanisms to potential pharmacotherapy.
Collapse
Affiliation(s)
- Eleonora Gatta
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, Psychiatric Institute, University of Illinois at Chicago, USA
| | - Diletta Camussi
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, Psychiatric Institute, University of Illinois at Chicago, USA
| | - James Auta
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, Psychiatric Institute, University of Illinois at Chicago, USA
| | - Alessandro Guidotti
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, Psychiatric Institute, University of Illinois at Chicago, USA
| | - Subhash C Pandey
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, Psychiatric Institute, University of Illinois at Chicago, USA; Jesse Brown Veterans Affairs Medical Center Chicago, IL 60612, USA.
| |
Collapse
|
14
|
Steroid hormone regulation of immune responses in cancer. IMMUNOMETABOLISM (COBHAM (SURREY, ENGLAND)) 2022; 4:e00012. [PMID: 36337733 PMCID: PMC9622373 DOI: 10.1097/in9.0000000000000012] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 09/23/2022] [Indexed: 01/24/2023]
Abstract
Steroid hormones are derived from cholesterol and can be classified into sex hormones (estrogens, androgens, progesterone) that are primarily synthesized in the gonads and adrenal hormones (glucocorticoids and mineralocorticoids) that are primarily synthesized in the adrenal gland. Although, it has long been known that steroid hormones have potent effects on the immune system, recent studies have led to renewed interest in their role in regulating anti-tumor immunity. Extra-glandular cells, such as epithelial cells and immune cells, have been shown to synthesize glucocorticoids and thereby modulate immune responses in the tumor microenvironment. Additionally, new insight into the role of androgens on immune cell responses have shed light on mechanisms underpinning the observed sex bias in cancer survival outcomes. Here, we review the role of steroid hormones, specifically glucocorticoids and androgens, in regulating anti-tumor immunity and discuss how their modulation could pave the way for designing novel therapeutic strategies to improve anti-tumor immune responses.
Collapse
|
15
|
Zheng JL, Zhu QL, Hu XC, Parsons D, Lawson R, Hogstrand C. Transgenerational effects of zinc in zebrafish following early life stage exposure. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 828:154443. [PMID: 35278549 DOI: 10.1016/j.scitotenv.2022.154443] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 03/01/2022] [Accepted: 03/06/2022] [Indexed: 06/14/2023]
Abstract
Although toxic effects of zinc (Zn) have been well established in the different developmental stages in fish, long-lasting effects of Zn exposure during embryonic development have not been explored. Exposure to an environmentally relevant Zn concentration of 10 μM (650 μg/L) during the first five days after fertilization did not affect survival, body weight, malformations or overall hatching success of F0 and F1 larvae. Zn exposure did, however, result in delayed hatching in both the F0 and F1 generations and caused significant changes in homeostasis of Zn and selenium (Se) in F0 and F1 fish. This was especially pronounced when F1 embryos from Zn-exposed parents were treated with 30 μM (2000 μg/L) Zn. In the F0 generation, skewed sex ratio towards males and changes in homeostasis of Zn, Se and manganese (Mn) in the brain, gill, liver and gonad of adult fish were also observed. These changes were associated with altered expression of Zn- and Mn-regulatory genes and sex differentiation genes in F0 and F1 fish. The present study suggests that fish may carry memory from embryo-larval Zn exposure into adulthood and further to the next generation. The present study shows that ecotoxicological risk of an exposure to Zn during embryo-larval development may persist long after recovery and may also manifest in the F1 generation.
Collapse
Affiliation(s)
- Jia-Lang Zheng
- National Engineering Research Center of Marine Facilities Aquaculture, College of Marine Science and Technology, Zhejiang Ocean University, Zhoushan 316022, PR China; King's College London, Faculty of Life Sciences and Medicine, Department of Nutritional Sciences, Metal Metabolism Group, London, UK
| | - Qing-Ling Zhu
- National Engineering Research Center of Marine Facilities Aquaculture, College of Marine Science and Technology, Zhejiang Ocean University, Zhoushan 316022, PR China; King's College London, Faculty of Life Sciences and Medicine, Department of Nutritional Sciences, Metal Metabolism Group, London, UK
| | - Xiu-Chuan Hu
- King's College London, Faculty of Life Sciences and Medicine, Department of Nutritional Sciences, Metal Metabolism Group, London, UK
| | - Douglas Parsons
- King's College London, Faculty of Life Sciences and Medicine, Department of Nutritional Sciences, Metal Metabolism Group, London, UK
| | - Rebecca Lawson
- King's College London, Faculty of Life Sciences and Medicine, Department of Nutritional Sciences, Metal Metabolism Group, London, UK
| | - Christer Hogstrand
- King's College London, Faculty of Life Sciences and Medicine, Department of Nutritional Sciences, Metal Metabolism Group, London, UK.
| |
Collapse
|
16
|
Lin YC, Cheung G, Porter E, Papadopoulos V. The neurosteroid pregnenolone is synthesized by a mitochondrial P450 enzyme other than CYP11A1 in human glial cells. J Biol Chem 2022; 298:102110. [PMID: 35688208 PMCID: PMC9278081 DOI: 10.1016/j.jbc.2022.102110] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 06/03/2022] [Accepted: 06/04/2022] [Indexed: 11/03/2022] Open
Abstract
Neurosteroids, modulators of neuronal and glial cell functions, are synthesized in the nervous system from cholesterol. In peripheral steroidogenic tissues, cholesterol is converted to the major steroid precursor pregnenolone by the CYP11A1 enzyme. Although pregnenolone is one of the most abundant neurosteroids in the brain, expression of CYP11A1 is difficult to detect. We found that human glial cells produced pregnenolone, detectable by mass spectrometry and ELISA, despite the absence of observable immunoreactive CYP11A1 protein. Unlike testicular and adrenal cortical cells, pregnenolone production in glial cells was not inhibited by CYP11A1 inhibitors DL-aminoglutethimide and ketoconazole. Furthermore, addition of hydroxycholesterols increased pregnenolone synthesis, suggesting desmolase activity that was not blocked by DL-aminoglutethimide or ketoconazole. We explored three different possibilities for an alternative pathway for glial cell pregnenolone synthesis: (1) regulation by reactive oxygen species, (2) metabolism via a different CYP11A1 isoform, and (3) metabolism via another CYP450 enzyme. First, we found oxidants and antioxidants had no significant effects on pregnenolone synthesis, suggesting it is not regulated by reactive oxygen species. Second, overexpression of CYP11A1 isoform b did not alter synthesis, indicating use of another CYP11A1 isoform is unlikely. Finally, we show nitric oxide and iron chelators deferoxamine and deferiprone significantly inhibited pregnenolone production, indicating involvement of another CYP450 enzyme. Ultimately, knockdown of endoplasmic reticulum cofactor NADPH-cytochrome P450 reductase had no effect, while knockdown of mitochondrial CYP450 cofactor ferredoxin reductase inhibited pregnenolone production. These data suggest that pregnenolone is synthesized by a mitochondrial cytochrome P450 enzyme other than CYP11A1 in human glial cells.
Collapse
|
17
|
Raux PL, Drutel G, Revest JM, Vallée M. New perspectives on the role of the neurosteroid pregnenolone as an endogenous regulator of type-1 cannabinoid receptor (CB1R) activity and function. J Neuroendocrinol 2022; 34:e13034. [PMID: 34486765 DOI: 10.1111/jne.13034] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 08/06/2021] [Accepted: 08/06/2021] [Indexed: 12/14/2022]
Abstract
Pregnenolone is a steroid with specific characteristics, being the first steroid to be synthesised from cholesterol at all sites of steroidogenesis, including the brain. For many years, pregnenolone was defined as an inactive precursor of all steroids because no specific target had been discovered. However, over the last decade, it has become a steroid of interest because it has been recognised as being a biomarker for brain-related disorders through the development of metabolomic approaches and advanced analytical methods. In addition, physiological roles for pregnenolone emerged when specific targets were discovered. In this review, we highlight the discovery of the selective interaction of pregnenolone with the type-1 cannabinoid receptor (CB1R). After describing the specific characteristic of CB1Rs, we discuss the newly discovered mechanisms of their regulation by pregnenolone. In particular, we describe the action of pregnenolone as a negative allosteric modulator and a specific signalling inhibitor of the CB1R. These particular characteristics of pregnenolone provide a great strategic opportunity for therapeutic development in CB1-related disorders. Finally, we outline new perspectives using innovative genetic tools for the discovery of original regulatory mechanisms of pregnenolone on CB1-related functions.
Collapse
Affiliation(s)
- Pierre-Louis Raux
- INSERM U1215, Neurocentre Magendie, Group "Physiopathology and Therapeutic Approaches of Stress-Related Disease", Bordeaux, France
- University of Bordeaux, Bordeaux, France
| | - Guillaume Drutel
- INSERM U1215, Neurocentre Magendie, Group "Physiopathology and Therapeutic Approaches of Stress-Related Disease", Bordeaux, France
- University of Bordeaux, Bordeaux, France
| | - Jean-Michel Revest
- INSERM U1215, Neurocentre Magendie, Group "Physiopathology and Therapeutic Approaches of Stress-Related Disease", Bordeaux, France
- University of Bordeaux, Bordeaux, France
| | - Monique Vallée
- INSERM U1215, Neurocentre Magendie, Group "Physiopathology and Therapeutic Approaches of Stress-Related Disease", Bordeaux, France
- University of Bordeaux, Bordeaux, France
| |
Collapse
|
18
|
Kalarani A, Vinodha V, Moses IR. Inter-relations of brain neurosteroids and monoamines towards reproduction in fish. REPRODUCTION AND BREEDING 2021. [DOI: 10.1016/j.repbre.2021.09.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
|
19
|
Chakraborty S, Pramanik J, Mahata B. Revisiting steroidogenesis and its role in immune regulation with the advanced tools and technologies. Genes Immun 2021; 22:125-140. [PMID: 34127827 PMCID: PMC8277576 DOI: 10.1038/s41435-021-00139-3] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 05/03/2021] [Accepted: 05/21/2021] [Indexed: 12/19/2022]
Abstract
Historically tools and technologies facilitated scientific discoveries. Steroid hormone research is not an exception. Unfortunately, the dramatic advancement of the field faded this research area and flagged it as a solved topic. However, it should have been the opposite. The area should glitter with its strong foundation and attract next-generation scientists. Over the past century, a myriad of new facts on biochemistry, molecular biology, cell biology, physiology and pathology of the steroid hormones was discovered. Several innovations were made and translated into life-saving treatment strategies such as synthetic steroids, and inhibitors of steroidogenesis and steroid signaling. Steroid molecules exhibit their diverse effects on cell metabolism, salt and water balance, development and function of the reproductive system, pregnancy, and immune-cell function. Despite vigorous research, the molecular basis of the immunomodulatory effect of steroids is still mysterious. The recent excitement on local extra-glandular steroidogenesis in regulating inflammation and immunity is revitalizing the topic with a new perspective. Therefore, here we review the role of steroidogenesis in regulating inflammation and immunity, discuss the unresolved questions, and how this area can bring another golden age of steroid hormone research with the development of new tools and technologies and advancement of the scientific methods.
Collapse
Affiliation(s)
| | - Jhuma Pramanik
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Bidesh Mahata
- Department of Pathology, University of Cambridge, Cambridge, UK.
| |
Collapse
|
20
|
Lin YC, Papadopoulos V. Neurosteroidogenic enzymes: CYP11A1 in the central nervous system. Front Neuroendocrinol 2021; 62:100925. [PMID: 34015388 DOI: 10.1016/j.yfrne.2021.100925] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 05/07/2021] [Accepted: 05/14/2021] [Indexed: 01/08/2023]
Abstract
Neurosteroids, steroid hormones synthesized locally in the nervous system, have important neuromodulatory and neuroprotective effects in the central nervous system. Progress in neurosteroid research has led to the successful translation of allopregnanolone into an approved therapy for postpartum depression. However, there is insufficient evidence to support the assumption that steroidogenesis is exactly the same between the nervous system and the periphery. This review focuses on CYP11A1, the only enzyme currently known to catalyze the first reaction in steroidogenesis to produce pregnenolone, the precursor to all other steroids. Although CYP11A1 mRNA has been found in brain of many mammals, the presence of CYP11A1 protein has been difficult to detect, particularly in humans. Here, we highlight the discrepancies in the current evidence for CYP11A1 in the central nervous system and propose new directions for understanding neurosteroidogenesis, which will be crucial for developing neurosteroid-based therapies for the future.
Collapse
Affiliation(s)
- Yiqi Christina Lin
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, United States
| | - Vassilios Papadopoulos
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, United States.
| |
Collapse
|
21
|
Dattilo MA, Benzo Y, Herrera LM, Prada JG, Lopez PF, Caruso CM, Lasaga M, García CI, Paz C, Maloberti PM. Regulation and role of Acyl-CoA synthetase 4 in glial cells. J Steroid Biochem Mol Biol 2021; 208:105792. [PMID: 33246155 DOI: 10.1016/j.jsbmb.2020.105792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 10/23/2020] [Accepted: 11/14/2020] [Indexed: 10/22/2022]
Abstract
Acyl-CoA synthetase 4 (Acsl4), an enzyme involved in arachidonic acid (AA) metabolism, participates in physiological and pathological processes such as steroidogenesis and cancer. The role of Acsl4 in neurons and in nervous system development has also been documented but little is known regarding its functionality in glial cells. In turn, several processes in glial cells, including neurosteroidogenesis, stellation and AA uptake, are regulated by cyclic adenosine monophosphate (cAMP) signal. In this context, the aim of this work was to analyze the expression and functional role of Acsl4 in primary rat astrocyte cultures and in the C6 glioma cell line by chemical inhibition and stable silencing, respectively. Results show that Acsl4 expression was regulated by cAMP in both models and that cAMP stimulation of steroidogenic acute regulatory protein mRNA levels was reduced by Acsl4 inhibition or silencing. Also, Acsl4 inhibition reduced progesterone synthesis stimulated by cAMP and also affected cAMP-induced astrocyte stellation, decreasing process elongation and increasing branching complexity. Similar effects were observed for Acsl4 silencing on cAMP-induced C6 cell morphological shift. Moreover, Acsl4 inhibition and silencing reduced proliferation and migration of both cell types. Acsl4 silencing in C6 cells reduced the capacity for colony proliferation and neurosphere formation, the latter ability also being abolished by Acsl4 inhibition. In sum, this work presents novel evidence of Acsl4 involvement in neurosteroidogenesis and the morphological changes of glial cells promoted by cAMP. Furthermore, Acsl4 participates in migration and proliferation, also affecting cell self-renewal. Altogether, these findings provide insights into Acsl4 functions in glial cells.
Collapse
Affiliation(s)
- Melina A Dattilo
- Universidad de Buenos Aires-CONICET, Instituto de Investigaciones Biomédicas (INBIOMED), Buenos Aires, Argentina; Universidad de Buenos Aires, Facultad de Medicina, Departamento de Bioquímica Humana, Buenos Aires, Argentina
| | - Yanina Benzo
- Universidad de Buenos Aires-CONICET, Instituto de Investigaciones Biomédicas (INBIOMED), Buenos Aires, Argentina; Universidad de Buenos Aires, Facultad de Medicina, Departamento de Bioquímica Humana, Buenos Aires, Argentina
| | - Lucia M Herrera
- Universidad de Buenos Aires-CONICET, Instituto de Investigaciones Biomédicas (INBIOMED), Buenos Aires, Argentina
| | - Jesica G Prada
- Universidad de Buenos Aires-CONICET, Instituto de Investigaciones Biomédicas (INBIOMED), Buenos Aires, Argentina
| | - Paula F Lopez
- Universidad de Buenos Aires-CONICET, Instituto de Investigaciones Biomédicas (INBIOMED), Buenos Aires, Argentina
| | - Carla M Caruso
- Universidad de Buenos Aires-CONICET, Instituto de Investigaciones Biomédicas (INBIOMED), Buenos Aires, Argentina; Universidad de Buenos Aires, Facultad de Medicina, Departamento de Biología Celular e Histología, Buenos Aires, Argentina
| | - Mercedes Lasaga
- Universidad de Buenos Aires-CONICET, Instituto de Investigaciones Biomédicas (INBIOMED), Buenos Aires, Argentina; Universidad de Buenos Aires, Facultad de Medicina, Departamento de Biología Celular e Histología, Buenos Aires, Argentina
| | - Corina I García
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Bioquímica Humana, Buenos Aires, Argentina; Fundación Instituto Leloir and Instituto de Investigaciones Bioquímicas de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Cristina Paz
- Universidad de Buenos Aires-CONICET, Instituto de Investigaciones Biomédicas (INBIOMED), Buenos Aires, Argentina; Universidad de Buenos Aires, Facultad de Medicina, Departamento de Bioquímica Humana, Buenos Aires, Argentina
| | - Paula M Maloberti
- Universidad de Buenos Aires-CONICET, Instituto de Investigaciones Biomédicas (INBIOMED), Buenos Aires, Argentina; Universidad de Buenos Aires, Facultad de Medicina, Departamento de Bioquímica Humana, Buenos Aires, Argentina.
| |
Collapse
|
22
|
Vahidinia Z, Karimian M, Joghataei MT. Neurosteroids and their receptors in ischemic stroke: From molecular mechanisms to therapeutic opportunities. Pharmacol Res 2020; 160:105163. [DOI: 10.1016/j.phrs.2020.105163] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 08/17/2020] [Accepted: 08/17/2020] [Indexed: 01/09/2023]
|
23
|
Extra-adrenal glucocorticoid biosynthesis: implications for autoimmune and inflammatory disorders. Genes Immun 2020; 21:150-168. [PMID: 32203088 PMCID: PMC7276297 DOI: 10.1038/s41435-020-0096-6] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 03/05/2020] [Accepted: 03/09/2020] [Indexed: 12/11/2022]
Abstract
Glucocorticoid synthesis is a complex, multistep process that starts with cholesterol being delivered to the inner membrane of mitochondria by StAR and StAR-related proteins. Here its side chain is cleaved by CYP11A1 producing pregnenolone. Pregnenolone is converted to cortisol by the enzymes 3-βHSD, CYP17A1, CYP21A2 and CYP11B1. Glucocorticoids play a critical role in the regulation of the immune system and exert their action through the glucocorticoid receptor (GR). Although corticosteroids are primarily produced in the adrenal gland, they can also be produced in a number of extra-adrenal tissue including the immune system, skin, brain, and intestine. Glucocorticoid production is regulated by ACTH, CRH, and cytokines such as IL-1, IL-6 and TNFα. The bioavailability of cortisol is also dependent on its interconversion to cortisone which is inactive, by 11βHSD1/2. Local and systemic glucocorticoid biosynthesis can be stimulated by ultraviolet B, explaining its immunosuppressive activity. In this review, we want to emphasize that dysregulation of extra-adrenal glucocorticoid production can play a key role in a variety of autoimmune diseases including multiple sclerosis (MS), lupus erythematosus (LE), rheumatoid arthritis (RA), and skin inflammatory disorders such as psoriasis and atopic dermatitis (AD). Further research on local glucocorticoid production and its bioavailability may open doors into new therapies for autoimmune diseases.
Collapse
|
24
|
Shen X, Yan H, Zhang L, Yuan Z, Liu W, Wu Y, Liu Q, Luo X, Liu Y. Transcriptomic analyses reveal novel genes with sexually dimorphic expression in Takifugu rubripes brain during gonadal sex differentiation. Genes Genomics 2020; 42:425-439. [DOI: 10.1007/s13258-019-00914-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 12/30/2019] [Indexed: 12/29/2022]
|
25
|
Arunkumar R, Gorusupudi A, Bernstein PS. The macular carotenoids: A biochemical overview. Biochim Biophys Acta Mol Cell Biol Lipids 2020; 1865:158617. [PMID: 31931175 DOI: 10.1016/j.bbalip.2020.158617] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 01/04/2020] [Accepted: 01/07/2020] [Indexed: 12/12/2022]
Abstract
Among the more than 750 carotenoids identified in nature, only lutein, zeaxanthin, meso-zeaxanthin, and their oxidative metabolites are selectively accumulated in the macula lutea region of the human retina. These retinal carotenoids are collectively referred to as the macular pigment (MP) and are obtained only through dietary sources such as green leafy vegetables and yellow and orange fruits and vegetables. Lutein- and zeaxanthin-specific binding proteins (StARD3 and GSTP1, respectively) mediate the highly selective uptake of MP into the retina. Meso-zeaxanthin is rarely present in the diet, and its unique presence in the human eye results from metabolic conversion from dietary lutein by the RPE65 enzyme. The MP carotenoids filter high-intensity, short-wavelength visible light and are powerful antioxidants in a region vulnerable to light-induced oxidative stress. This review focuses on MP chemistry, absorption, metabolism, transport, and distribution with special emphasis on animal models used for MP study. This article is part of a Special Issue entitled Carotenoids recent advances in cell and molecular biology edited by Johannes von Lintig and Loredana Quadro.
Collapse
Affiliation(s)
- Ranganathan Arunkumar
- Department of Ophthalmology and Visual Science, Moran Eye Center, University of Utah School of Medicine, 65 Mario Capecchi Drive, Salt Lake City, UT 84132, USA
| | - Aruna Gorusupudi
- Department of Ophthalmology and Visual Science, Moran Eye Center, University of Utah School of Medicine, 65 Mario Capecchi Drive, Salt Lake City, UT 84132, USA
| | - Paul S Bernstein
- Department of Ophthalmology and Visual Science, Moran Eye Center, University of Utah School of Medicine, 65 Mario Capecchi Drive, Salt Lake City, UT 84132, USA.
| |
Collapse
|
26
|
Abstract
The term neurosteroid refers to rapid membrane actions of steroid hormones and their derivatives that can modulate physiological functions and behavior via their interactions with ligand-gated ion channels. This chapter will highlight recent advances pertaining to the modulatory effects of a select group of neurosteroids that are primarily potent positive allosteric modulators of γ-aminobutyric acidA receptors (GABAARs). Nanomolar concentrations of neurosteroids, which occur in vivo, potentiate phasic and tonic forms of GABAAR-mediated inhibition, indicating that both synaptic and extrasynaptic GABAARs possess sensitivity to neurosteroids and contribute to the overall ability of neurosteroids to modulate central nervous system excitability. Common effects of alcohol and neurosteroids at GABAARs have stimulated research on the ability of neurosteroids to modulate alcohol's acute and chronic effects. Background on neurosteroid pharmacology and biosynthetic enzymes will be provided as it relates to experimental findings. Data will be summarized on alcohol and neurosteroid interactions across neuroanatomical regions and models of intoxication, consumption, dependence, and withdrawal. Evidence supports independent regulation of neurosteroid synthesis between periphery and brain as well as across brain regions following acute alcohol administration and during withdrawal. Local mechanisms for fine-tuning neuronal excitability via manipulation of neurosteroid synthesis exert predicted behavioral and electrophysiological responses on GABAAR-mediated inhibition. Collectively, targeting neurosteroidogenesis may be a beneficial treatment strategy for alcohol use disorders.
Collapse
|
27
|
González SL, Meyer L, Raggio MC, Taleb O, Coronel MF, Patte-Mensah C, Mensah-Nyagan AG. Allopregnanolone and Progesterone in Experimental Neuropathic Pain: Former and New Insights with a Translational Perspective. Cell Mol Neurobiol 2019; 39:523-537. [PMID: 30187261 PMCID: PMC11469882 DOI: 10.1007/s10571-018-0618-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 08/31/2018] [Indexed: 02/06/2023]
Abstract
In the last decades, an active and stimulating area of research has been devoted to explore the role of neuroactive steroids in pain modulation. Despite challenges, these studies have clearly contributed to unravel the multiple and complex actions and potential mechanisms underlying steroid effects in several experimental conditions that mimic human chronic pain states. Based on the available data, this review focuses mainly on progesterone and its reduced derivative allopregnanolone (also called 3α,5α-tetrahydroprogesterone) which have been shown to prevent or even reverse the complex maladaptive changes and pain behaviors that arise in the nervous system after injury or disease. Because the characterization of new related molecules with improved specificity and enhanced pharmacological profiles may represent a crucial step to develop more efficient steroid-based therapies, we have also discussed the potential of novel synthetic analogs of allopregnanolone as valuable molecules for the treatment of neuropathic pain.
Collapse
Affiliation(s)
- Susana Laura González
- Departamento de Bioquímica Humana, Facultad de Medicina, Universidad de Buenos Aires, Paraguay 2155, C1121ABG, Buenos Aires, Argentina.
- Laboratorio de Nocicepción y Dolor Neuropático, Instituto de Biología y Medicina Experimental, CONICET, Vuelta de Obligado 2490, C1428ADN, Buenos Aires, Argentina.
| | - Laurence Meyer
- Biopathologie de la Myéline, Neuroprotection et Stratégies Thérapeutiques, INSERM U1119, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Bâtiment 3 de la Faculté de Médicine, 11 rue Humann, 67 000, Strasbourg, France
| | - María Celeste Raggio
- Laboratorio de Nocicepción y Dolor Neuropático, Instituto de Biología y Medicina Experimental, CONICET, Vuelta de Obligado 2490, C1428ADN, Buenos Aires, Argentina
| | - Omar Taleb
- Biopathologie de la Myéline, Neuroprotection et Stratégies Thérapeutiques, INSERM U1119, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Bâtiment 3 de la Faculté de Médicine, 11 rue Humann, 67 000, Strasbourg, France
| | - María Florencia Coronel
- Laboratorio de Nocicepción y Dolor Neuropático, Instituto de Biología y Medicina Experimental, CONICET, Vuelta de Obligado 2490, C1428ADN, Buenos Aires, Argentina
| | - Christine Patte-Mensah
- Biopathologie de la Myéline, Neuroprotection et Stratégies Thérapeutiques, INSERM U1119, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Bâtiment 3 de la Faculté de Médicine, 11 rue Humann, 67 000, Strasbourg, France
| | - Ayikoe Guy Mensah-Nyagan
- Biopathologie de la Myéline, Neuroprotection et Stratégies Thérapeutiques, INSERM U1119, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Bâtiment 3 de la Faculté de Médicine, 11 rue Humann, 67 000, Strasbourg, France.
| |
Collapse
|
28
|
Joshi S, Kapur J. Neurosteroid regulation of GABA A receptors: A role in catamenial epilepsy. Brain Res 2019; 1703:31-40. [PMID: 29481795 PMCID: PMC6107446 DOI: 10.1016/j.brainres.2018.02.031] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 09/08/2017] [Accepted: 02/20/2018] [Indexed: 12/31/2022]
Abstract
The female reproductive hormones progesterone and estrogen regulate network excitability. Fluctuations in the circulating levels of these hormones during the menstrual cycle cause frequent seizures during certain phases of the cycle in women with epilepsy. This seizure exacerbation, called catamenial epilepsy, is a dominant form of drug-refractory epilepsy in women of reproductive age. Progesterone, through its neurosteroid derivative allopregnanolone, increases γ-aminobutyric acid type-A receptor (GABAR)-mediated inhibition in the brain and keeps seizures under control. Catamenial seizures are believed to be a neurosteroid withdrawal symptom, and it was hypothesized that exogenous administration of progesterone to maintain its levels high during luteal phase will treat catamenial seizures. However, in a multicenter, double-blind, phase III clinical trial, progesterone treatment did not suppress catamenial seizures. The expression of GABARs with reduced neurosteroid sensitivity in epileptic animals may explain the failure of the progesterone clinical trial. The expression of neurosteroid-sensitive δ subunit-containing GABARs is reduced, and the expression of α4γ2 subunit-containing GABARs is upregulated, which alters the inhibition of dentate granule cells in epilepsy. These changes reduce the endogenous neurosteroid control of seizures and contribute to catamenial seizures.
Collapse
Affiliation(s)
- Suchitra Joshi
- Department of Neurology, University of Virginia, Charlottesville, VA 22908, United States.
| | - Jaideep Kapur
- Department of Neurology, University of Virginia, Charlottesville, VA 22908, United States; Department of Neuroscience, University of Virginia, Charlottesville, VA 22908, United States
| |
Collapse
|
29
|
Peek CE, Cohen RE. Seasonal regulation of steroidogenic enzyme expression within the green anole lizard (Anolis carolinensis) brain and gonad. Gen Comp Endocrinol 2018; 268:88-95. [PMID: 30077794 DOI: 10.1016/j.ygcen.2018.08.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 07/12/2018] [Accepted: 08/01/2018] [Indexed: 12/12/2022]
Abstract
Steroid hormones, such as testosterone and estradiol, are necessary for reproductive behavior. Seasonally breeding animals have increased sex steroid hormone levels during the breeding compared to non-breeding season, with increased reproductive behaviors and altered brain morphology in breeding individuals. Similar to other seasonally breeding animals, green anole lizards (Anolis carolinensis) have high sex steroid hormone levels and increased reproductive behaviors in the breeding season. Relatively less is known regarding the regulation of steroidogenesis in reptiles and this experiment examined whether enzymes involved in sex steroid hormone synthesis vary seasonally within the brain and gonads in wild-caught anole lizards. Specifically, we examined mRNA expression of steroidogenic acute regulatory protein (StAR), P450 17α-hydroxylase/C17-20lyase (Cyp17α1), 17 beta-hydroxysteroid dehydrogenase type 3 (17βHSD 3), and aromatase (Cyp19α1). We found that the mRNA for each of these genes was expressed in the lizard brain. Interestingly, Cyp19α1 mRNA expression in the brain was increased during the non-breeding season, potentially revealing a role for aromatase expression in the non-breeding brain. In the anole gonads, StAR mRNA expression levels were increased in both males and females during the breeding season, while the mRNA expression levels of CYP17α1 and 17βHSD 3 are increased when StAR mRNA expression was decreased, suggesting that the enzymes in the steroidogenic pathway are potentially regulated independently of StAR. This work reveals the seasonal regulation of steroidogenesis in the reptilian brain and gonad, although more work is necessary to determine the regulatory mechanisms that control these expression patterns.
Collapse
Affiliation(s)
- Christine E Peek
- Department of Biological Sciences, Minnesota State University, Mankato, Mankato, MN 56001-6062, USA
| | - Rachel E Cohen
- Department of Biological Sciences, Minnesota State University, Mankato, Mankato, MN 56001-6062, USA.
| |
Collapse
|
30
|
Leicaj ML, Pasquini LA, Lima A, Gonzalez Deniselle MC, Pasquini JM, De Nicola AF, Garay LI. Changes in neurosteroidogenesis during demyelination and remyelination in cuprizone-treated mice. J Neuroendocrinol 2018; 30:e12649. [PMID: 30303567 DOI: 10.1111/jne.12649] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 09/20/2018] [Accepted: 10/01/2018] [Indexed: 12/28/2022]
Abstract
Changes of neurosteroids may be involved in the pathophysiology of multiple sclerosis (MS). The present study investigated whether changes of neurosteroidogenesis also occurred in the grey and white matter regions of the brain in mice subjected to cuprizone-induced demyelination. Accordingly, we compared the expression of neurosteroidogenic proteins, including steroidogenic acute regulatory protein (StAR), voltage-dependent anion channel (VDAC) and 18 kDa translocator protein (TSPO), as well as neurosteroidogenic enzymes, including the side chain cleavage enzyme (P450scc), 3β-hydroxysteroid dehydrogenase/isomerase and 5α-reductase (5α-R), during the demyelination and remyelination periods. Using immunohistochemistry and a quantitative polymerase chain reaction, we demonstrated a decreased expression of StAR, P450scc and 5α-R with respect to an increase astrocytic and microglial reaction and elevated levels of tumor necrosis factor (TNF)α during the cuprizone demyelination period in the hippocampus, cortex and corpus callosum. These parameters, as well as the glial reaction, were normalised after 2 weeks of spontaneous remyelination in regions containing grey matter. Conversely, persistent elevated levels of TNFα and low levels of StAR and P450scc were observed during remyelination in corpus callosum white matter. We conclude that neurosteroidogenesis/myelination status and glial reactivity are inversely related in the hippocampus and neocortex. Establishing a cause and effect relationship for the measured variables remains a future challenge for understanding the pathophysiology of MS.
Collapse
Affiliation(s)
- María L Leicaj
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biologia y Medicina Experimental and National Research Council (CONICET), Buenos Aires, Argentina
| | - Laura A Pasquini
- Department of Biological Chemistry, School of Pharmacy and Biochemistry, Institute of Chemistry and Biological Physicochemistry (IQUIFIB), University of Buenos Aires and National Research Council (CONICET), Buenos Aires, Argentina
| | - Analia Lima
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biologia y Medicina Experimental and National Research Council (CONICET), Buenos Aires, Argentina
| | - Maria C Gonzalez Deniselle
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biologia y Medicina Experimental and National Research Council (CONICET), Buenos Aires, Argentina
- Department of Physiological Sciences, University of Buenos Aires, Buenos Aires, Argentina
| | - Juana M Pasquini
- Department of Biological Chemistry, School of Pharmacy and Biochemistry, Institute of Chemistry and Biological Physicochemistry (IQUIFIB), University of Buenos Aires and National Research Council (CONICET), Buenos Aires, Argentina
| | - Alejandro F De Nicola
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biologia y Medicina Experimental and National Research Council (CONICET), Buenos Aires, Argentina
- Department of Human Biochemistry, University of Buenos Aires, Buenos Aires, Argentina
| | - Laura I Garay
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biologia y Medicina Experimental and National Research Council (CONICET), Buenos Aires, Argentina
- Department of Human Biochemistry, University of Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
31
|
Zhang X, Guan T, Yang B, Chi Z, Wang ZY, Gu HF. A novel role for zinc transporter 8 in the facilitation of zinc accumulation and regulation of testosterone synthesis in Leydig cells of human and mouse testicles. Metabolism 2018; 88:40-50. [PMID: 30236453 DOI: 10.1016/j.metabol.2018.09.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 09/05/2018] [Accepted: 09/11/2018] [Indexed: 11/22/2022]
Abstract
OBJECTIVE Zinc is intimately involved in testosterone production. Zinc transporter 8 (ZnT8) is found to be localized in insulin secretory granules as a β-cell specific Zn transporter. The effect of ZnT8 and related zinc accumulation in steroidogenesis, however, is still unknown. The present study aimed to explore whether ZnT8 plays a role in the facilitation of zinc accumulation and regulation of testosterone synthesis in testicles. METHODS Leydig cells were isolated from the testicles of human, CD-1 suckling and ZnT8-KO mice. Zn accumulation in mitochondria was induced by hCG stimulation. Transfection of hZnT8-EGFP and RNA interfere of mZnT8 were done in MLTC-1 cells. ZnT8 expression and its co-localization with steroidogenic acute regulatory (StAR) protein were analyzed with RT-PCR, Western blot and dual-fluorescent staining protocols. Serum testosterone levels in mice were determined with chemiluminescent enzyme immunoassay. RESULTS ZnT8 was found to be presented in Leydig cells and up-regulated in suckling mouse Leydig cells and MLTC-1 cells after hCG administration, by which zinc accumulation occurred in mitochondria. ZnT8 gene silencing or knockout inhibited stimulated progesterone and testosterone production, reduced stimulated zinc accumulation and down-regulated phosphorylated steroidogenic acute regulatory (StAR) expression in Leydig cells. Furthermore, an inhibitor (H89) of PKA blocked hCG-stimulated progesterone caused by ZnT8 over-expression and zinc treatment. CONCLUSION The present study provided the first evidence that ZnT8 transports Zn into Leydig cell mitochondria with gonadotropin stimulation and suggests that ZnT8 may play a role in testosterone production via the PKA signaling pathway.
Collapse
Affiliation(s)
- Xiuli Zhang
- Department of Nephrology, The second People's Hospital, Shenzhen, The first Affiliated Hospital of Shenzhen University, Guangdong 518000, PR China; Department of Pathophysiology, China Medical University, Shenyang, Liaoning 110001, PR China
| | - Tingwen Guan
- Department of Pathophysiology, China Medical University, Shenyang, Liaoning 110001, PR China
| | - Boxuan Yang
- Department of Pathophysiology, China Medical University, Shenyang, Liaoning 110001, PR China
| | - Zhihong Chi
- Department of Pathophysiology, China Medical University, Shenyang, Liaoning 110001, PR China.
| | - Zhan-You Wang
- College of Life and Health Sciences, China Medical University, Shenyang, Liaoning 110001, PR China
| | - Harvest F Gu
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, PR China.
| |
Collapse
|
32
|
Cisternas CD, Garcia-Segura LM, Cambiasso MJ. Hormonal and genetic factors interact to control aromatase expression in the developing brain. J Neuroendocrinol 2018; 30. [PMID: 28891264 DOI: 10.1111/jne.12535] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 09/05/2017] [Accepted: 09/05/2017] [Indexed: 02/03/2023]
Abstract
Brain expression of the enzyme P450-aromatase has been studied extensively. Subsequent to the aromatisation hypothesis having established brain aromatase as a key factor to convert gonadal testosterone to oestradiol, several studies have investigated the regulation of aromatase during the critical period of brain sexual differentiation. We review previous and recent findings concerning regulation of aromatase. The role of gonadal hormones, sex chromosome genes and neurosteroids is analysed in terms of their contribution to aromatase expression, as well as implications for the organisational effect of steroids during development.
Collapse
Affiliation(s)
- C D Cisternas
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina
- Departamento de Biología Bucal, Facultad de Odontología, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - L M Garcia-Segura
- Instituto Cajal, CSIC, Madrid, Spain
- Ciber de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - M J Cambiasso
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina
- Departamento de Biología Bucal, Facultad de Odontología, Universidad Nacional de Córdoba, Córdoba, Argentina
| |
Collapse
|
33
|
Huang YC, Hung CF, Lin PY, Lee Y, Wu CC, Hsu ST, Chen CC, Chong MY, Lin CH, Wang LJ. Gender differences in susceptibility to schizophrenia: Potential implication of neurosteroids. Psychoneuroendocrinology 2017; 84:87-93. [PMID: 28686904 DOI: 10.1016/j.psyneuen.2017.06.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 06/21/2017] [Accepted: 06/23/2017] [Indexed: 11/28/2022]
Abstract
Past research has indicated gender differences in the clinical characteristics and course of schizophrenia. In this study, we investigated whether gender differences in the manifestation of schizophrenia are correlated with neurosteroids, including dehydroepiandrosterone (DHEA), dehydroepiandrosterone sulfate (DHEA-S), and pregnenolone. We further explored the potential relationship between the aforementioned neurosteroids and psychopathology. We recruited 65 schizophrenic patients (36 males and 29 females) and 103 healthy control subjects (47 males and 56 females) and obtained blood samples from the subjects in the morning while in a fasting state to determine the serum levels of DHEA, DHEA-S, and pregnenolone. The psychopathology and mood symptoms of patients with schizophrenia were evaluated using the Positive and Negative Syndrome Scale (PANSS) and 17-item Hamilton Depression Rating Scale, respectively. Compared to the male control subjects, male patients with schizophrenia had significantly lower serum levels of DHEA and pregnenolone. In males with schizophrenia, the serum levels of DHEA and DHEA-S were associated with the age of onset and the duration of illness, while pregnenolone levels were associated with general symptoms of the PANSS. However, none of the neurosteroid levels were different between the female patients with schizophrenia and the female controls, and no significant correlation between neurosteroid levels and psychopathology evaluations was found among the schizophrenic females. Neurosteroids, including DHEA, DHEA-S, and pregnenolone, are involved in the pathophysiology of schizophrenia in male patients, but not in female ones. Therefore, our findings suggest that neurosteroids may be associated with gender differences in susceptibility to schizophrenia.
Collapse
Affiliation(s)
- Yu-Chi Huang
- Department of Psychiatry, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan; Chung Shan Medical University School of Medicine, Taichung, Taiwan
| | - Chi-Fa Hung
- Department of Psychiatry, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Pao-Yen Lin
- Department of Psychiatry, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan; Institute for Translational Research in Biomedical Sciences, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Yu Lee
- Department of Psychiatry, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Chih-Ching Wu
- Molecular Medicine Research Center, Chang Gung University, Tao-Yuan, Taiwan; Department of Medical Biotechnology and Laboratory Science, Chang Gung University, Tao-Yuan, Taiwan
| | - Su-Ting Hsu
- Kaohsiung Municipal Kai-Syuan Psychiatric Hospital, Kaohsiung, Taiwan
| | - Chien-Chih Chen
- Department of Psychiatry, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Mian-Yoon Chong
- Department of Psychiatry, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Chieh-Hsin Lin
- Department of Psychiatry, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Liang-Jen Wang
- Department of Child and Adolescent Psychiatry, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan.
| |
Collapse
|
34
|
Santos da Rosa JG, Alcântara Barcellos HHD, Fagundes M, Variani C, Rossini M, Kalichak F, Koakoski G, Acosta Oliveira T, Idalencio R, Frandoloso R, Piato AL, José Gil Barcellos L. Muscarinic receptors mediate the endocrine-disrupting effects of an organophosphorus insecticide in zebrafish. ENVIRONMENTAL TOXICOLOGY 2017; 32:1964-1972. [PMID: 28371364 DOI: 10.1002/tox.22424] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Revised: 03/20/2017] [Accepted: 03/20/2017] [Indexed: 06/07/2023]
Abstract
The glucocorticoid cortisol, the end product of hypothalamus-pituitary-interrenal axis in zebrafish (Danio rerio), is synthesized via steroidogenesis and promotes important physiological regulations in response to a stressor. The failure of this axis leads to inability to cope with environmental challenges preventing adaptive processes in order to restore homeostasis. Pesticides and agrichemicals are widely used, and may constitute an important class of environmental pollutants when reach aquatic ecosystems and nontarget species. These chemical compounds may disrupt hypothalamus-pituitary-interrenal axis by altering synthesis, structure or function of its constituents. We present evidence that organophosphorus exposure disrupts stress response by altering the expression of key genes of the neural steroidogenesis, causing downregulation of star, hsp70, and pomc genes. This appears to be mediated via muscarinic receptors, since the muscarinic antagonist scopolamine blocked these effects.
Collapse
Affiliation(s)
- João Gabriel Santos da Rosa
- Programa de Pós-graduação em Farmacologia, Universidade Federal de Santa Maria (UFSM), Av. Roraima, 1000, Cidade Universitária, Camobi, Santa Maria, RS, 97105-900, Brazil
| | - Heloísa Helena de Alcântara Barcellos
- Programa de Pós-graduação em Farmacologia, Universidade Federal de Santa Maria (UFSM), Av. Roraima, 1000, Cidade Universitária, Camobi, Santa Maria, RS, 97105-900, Brazil
- Faculdade de Agronomia e Medicina Veterinária, Universidade de Passo Fundo (UPF), BR 285, São José, Passo Fundo, RS, 99052-900, Brazil
| | - Michele Fagundes
- Faculdade de Agronomia e Medicina Veterinária, Universidade de Passo Fundo (UPF), BR 285, São José, Passo Fundo, RS, 99052-900, Brazil
| | - Cristiane Variani
- Faculdade de Agronomia e Medicina Veterinária, Universidade de Passo Fundo (UPF), BR 285, São José, Passo Fundo, RS, 99052-900, Brazil
| | - Mainara Rossini
- Faculdade de Agronomia e Medicina Veterinária, Universidade de Passo Fundo (UPF), BR 285, São José, Passo Fundo, RS, 99052-900, Brazil
| | - Fabiana Kalichak
- Programa de Pós-graduação em Farmacologia, Universidade Federal de Santa Maria (UFSM), Av. Roraima, 1000, Cidade Universitária, Camobi, Santa Maria, RS, 97105-900, Brazil
| | - Gessi Koakoski
- Programa de Pós-graduação em Farmacologia, Universidade Federal de Santa Maria (UFSM), Av. Roraima, 1000, Cidade Universitária, Camobi, Santa Maria, RS, 97105-900, Brazil
| | - Thiago Acosta Oliveira
- Programa de Pós-graduação em Farmacologia, Universidade Federal de Santa Maria (UFSM), Av. Roraima, 1000, Cidade Universitária, Camobi, Santa Maria, RS, 97105-900, Brazil
| | - Renan Idalencio
- Programa de Pós-graduação em Farmacologia, Universidade Federal de Santa Maria (UFSM), Av. Roraima, 1000, Cidade Universitária, Camobi, Santa Maria, RS, 97105-900, Brazil
- Faculdade de Agronomia e Medicina Veterinária, Universidade de Passo Fundo (UPF), BR 285, São José, Passo Fundo, RS, 99052-900, Brazil
| | - Rafael Frandoloso
- Faculdade de Agronomia e Medicina Veterinária, Universidade de Passo Fundo (UPF), BR 285, São José, Passo Fundo, RS, 99052-900, Brazil
- Programa de Pós-graduação em Bioexperimentação, Universidade de Passo Fundo (UPF), BR 285, São José, Passo Fundo, RS, 99052-900, Brazil
| | - Angelo L Piato
- Programa de Pós-Graduação em Farmacologia e Terapêutica, ICBS, Universidade Federal do Rio Grande do Sul (UFRGS), Rua Sarmento Leite 500, sala 305, Centro Histórico, Porto Alegre, RS, 90050-170, Brazil
| | - Leonardo José Gil Barcellos
- Programa de Pós-graduação em Farmacologia, Universidade Federal de Santa Maria (UFSM), Av. Roraima, 1000, Cidade Universitária, Camobi, Santa Maria, RS, 97105-900, Brazil
- Faculdade de Agronomia e Medicina Veterinária, Universidade de Passo Fundo (UPF), BR 285, São José, Passo Fundo, RS, 99052-900, Brazil
- Programa de Pós-graduação em Bioexperimentação, Universidade de Passo Fundo (UPF), BR 285, São José, Passo Fundo, RS, 99052-900, Brazil
| |
Collapse
|
35
|
Mukherjee A, Haldar C. Effect of Naltrexone on photoperiodic regulation of testicular steroidogenesis in adult golden hamster, Mesocricetus auratus. Gen Comp Endocrinol 2016; 239:89-96. [PMID: 26449162 DOI: 10.1016/j.ygcen.2015.10.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2015] [Revised: 09/14/2015] [Accepted: 10/03/2015] [Indexed: 11/27/2022]
Abstract
Photoperiodic regulation of testicular steroidogenesis through modulation of MT1R expression and local melatonin content is well established. However, additional mediators besides local melatonergic system in photoperiodic control of testicular steroidogenesis in golden hamster have not been studied in detail. Endogenous opioid peptides (EOP) are known to regulate reproduction via acting at multiple levels of the hypothalamus-pituitary-gonadal (HPG) axis. The presence of β-endorphin, a naturally occurring opioid peptide, and its receptor (μ-opioid receptor, μOR) has been reported in rat testes; however the functional significance of photoperiodic regulation μOR in testicular steroidogenesis is not clear. In the present study, we assessed the effect of Naltrexone (Nal), a μOR antagonist, in photoperiodic regulation of testicular steroidogenesis. Immunohistochemical (IHC) localization and expression of μOR along with the expression of steroidogenic markers in testes was analyzed through western blot analyses. IHC suggest immunoreactivity for μOR in Leydig cells with strong immunoreactivity under SD (short-day) condition, whereas weak immunoreactivity was observed under LD (long-day). The expression of μOR was significantly decreased following Nal administration in both the photoperiodic conditions. The localization and differential photoperiodic regulation of μOR in Leydig cells suggests its involvement in testicular steroidogenesis. Further, Nal administration significantly increased the expression of steroidogenic markers (AR, StAR, P450SCC, LH-R, 3β-HSD and 17-HSD) and plasma testosterone concentration under SD condition as compared to SD-control. We may therefore suggest that photoperiod differentially regulates the expression of μOR which thereby mediates the inhibitory effect of melatonin on testicular steroidogenesis.
Collapse
Affiliation(s)
- Arun Mukherjee
- Pineal Research Lab., Department of Zoology, Banaras Hindu University, Varanasi 221005, India.
| | - Chandana Haldar
- Pineal Research Lab., Department of Zoology, Banaras Hindu University, Varanasi 221005, India.
| |
Collapse
|
36
|
Selvaraj V, Tu LN. Current status and future perspectives: TSPO in steroid neuroendocrinology. J Endocrinol 2016; 231:R1-R30. [PMID: 27422254 DOI: 10.1530/joe-16-0241] [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: 07/08/2016] [Accepted: 07/15/2016] [Indexed: 12/21/2022]
Abstract
The mitochondrial translocator protein (TSPO), previously known as the peripheral benzodiazepine receptor (PBR), has received significant attention both as a diagnostic biomarker and as a therapeutic target for different neuronal disease pathologies. Recently, its functional basis believed to be mediating mitochondrial cholesterol import for steroid hormone production has been refuted by studies examining both in vivo and in vitro genetic Tspo-deficient models. As a result, there now exists a fundamental gap in the understanding of TSPO function in the nervous system, and its putative pharmacology in neurosteroid production. In this review, we discuss several recent findings in steroidogenic cells that are in direct contradiction to previous studies, and necessitate a re-examination of the purported role for TSPO in de novo neurosteroid biosynthesis. We critically examine the pharmacological effects of different TSPO-binding drugs with particular focus on studies that measure neurosteroid levels. We highlight the basis of key misconceptions regarding TSPO that continue to pervade the literature, and the need for interpretation with caution to avoid negative impacts. We also summarize the emerging perspectives that point to new directions that need to be investigated for understanding the molecular function of TSPO, only after which the true potential of this therapeutic target in medicine may be realized.
Collapse
Affiliation(s)
- Vimal Selvaraj
- Department of Animal ScienceCornell University, Ithaca, New York, USA
| | - Lan N Tu
- Department of Animal ScienceCornell University, Ithaca, New York, USA
| |
Collapse
|
37
|
Coronel MF, Labombarda F, González SL. Neuroactive steroids, nociception and neuropathic pain: A flashback to go forward. Steroids 2016; 110:77-87. [PMID: 27091763 DOI: 10.1016/j.steroids.2016.04.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Revised: 03/29/2016] [Accepted: 04/11/2016] [Indexed: 11/26/2022]
Abstract
The present review discusses the potential role of neurosteroids/neuroactive steroids in the regulation of nociceptive and neuropathic pain, and recapitulates the current knowledge on the main mechanisms involved in the reduction of pain, especially those occurring at the dorsal horn of the spinal cord, a crucial site for nociceptive processing. We will make special focus on progesterone and its derivative allopregnanolone, which have been shown to exert remarkable actions in order to prevent or reverse the maladaptive changes and pain behaviors that arise after nervous system damage in various experimental neuropathic conditions.
Collapse
Affiliation(s)
- María F Coronel
- Laboratorio de Nocicepción y Dolor Neuropático, Instituto de Biología y Medicina Experimental, CONICET, Vuelta de Obligado 2490, C1428ADN, Buenos Aires, Argentina; Facultad de Ciencias Biomédicas, Universidad Austral, Presidente Perón 1500, B1629AHJ, Pilar, Buenos Aires, Argentina
| | - Florencia Labombarda
- Laboratorio de Bioquímica Neuroendócrina, Instituto de Biología y Medicina Experimental, CONICET, Vuelta de Obligado 2490, C1428ADN, Buenos Aires, Argentina; Departamento de Bioquímica Humana, Facultad de Medicina, Universidad de Buenos Aires, Paraguay 2155, C1121ABG, Buenos Aires, Argentina
| | - Susana L González
- Laboratorio de Nocicepción y Dolor Neuropático, Instituto de Biología y Medicina Experimental, CONICET, Vuelta de Obligado 2490, C1428ADN, Buenos Aires, Argentina; Departamento de Bioquímica Humana, Facultad de Medicina, Universidad de Buenos Aires, Paraguay 2155, C1121ABG, Buenos Aires, Argentina.
| |
Collapse
|
38
|
Chu Q, Chi ZH, Zhang X, Liang D, Wang X, Zhao Y, Zhang L, Zhang P. A potential role for zinc transporter 7 in testosterone synthesis in mouse Leydig tumor cells. Int J Mol Med 2016; 37:1619-26. [PMID: 27121848 DOI: 10.3892/ijmm.2016.2576] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 04/14/2016] [Indexed: 11/06/2022] Open
Abstract
Previous studies have demonstrated that zinc (Zn) is an essential trace element which is involved in male reproduction. The zinc transporter (ZnT) family, SLC30a, is involved in the maintenance of Zn homeostasis and in mediating intracellular signaling events; however, relatively little is known regarding the effect of ZnTs on testosterone synthesis. Thus, in the present study, we aimed to determine the effect of Zn transporter 7 (ZnT7) on testosterone synthesis in male CD-1 mice and mouse Leydig cells. The findings of the present study revealed that the concentrations of Zn in the testes and Leydig cells were significantly lower in mice fed a Zn-deficient diet compared with the control mice fed a Zn-adequate diet. In addition, ZnT7 was principally expressed and colocalized with steroidogenic acute regulatory protein (StAR) in the Leydig cells of male CD-1 mice. ZnT7 expression was downregulated in the mice fed a Zn-deficient diet, which led to decreases in the expression of the enzymes involved in testosterone synthesis namely cholesterol side‑chain cleavage enzyme (P450scc) and 3β-hydroxysteroid dehydrogenase/D5-D4 isomerase (3β-HSD) as well as decreased serum testosterone levels. These results suggested that Znt7 may be involved in testosterone synthesis in the mouse testes. To examine this hypothesis, we used the mouse Leydig tumor cell line (MLTC-1 cell line) in which the ZnT7 gene had been silenced, in order to gauge the impact of changes in ZnT7 expression on testosterone secretion and the enzymes involved in testosterone synthesis. The results demonstrated that ZnT7 gene silencing downregulated the expression of StAR, P450scc and 3β-HSD as well as progesterone concentrations in the human chorionic gonadotrophin (hCG)-stimulated MLTC-1 cells. Taken together, these findings reveal that ZnT7 may play an important role in the regulation of testosterone synthesis by modulating steroidogenic enzymes, and may represent a therapeutic target in testosterone deficiency.
Collapse
Affiliation(s)
- Qingqing Chu
- Department of Pathophysiology, China Medical University, Shenyang, Liaoning, P.R. China
| | - Zhi-Hong Chi
- Department of Pathophysiology, China Medical University, Shenyang, Liaoning, P.R. China
| | - Xiuli Zhang
- Department of Nephrology, Benxi Center Hospital, China Medical University, Benxi, Liaoning, P.R. China
| | - Dan Liang
- Troops of 95935 Unit, Harbin, Heilongjiang, P.R. China
| | - Xuemei Wang
- School of Stomatology of China Medical University, Shenyang, Liaoning, P.R. China
| | - Yue Zhao
- Department of Pathophysiology, China Medical University, Shenyang, Liaoning, P.R. China
| | - Li Zhang
- School of Stomatology of China Medical University, Shenyang, Liaoning, P.R. China
| | - Ping Zhang
- Department of Histology and Embryology, Liaoning Medical College, Jinzhou, Liaoning, P.R. China
| |
Collapse
|
39
|
do Rego JL, Vaudry H. Comparative aspects of neurosteroidogenesis: From fish to mammals. Gen Comp Endocrinol 2016; 227:120-9. [PMID: 26079790 DOI: 10.1016/j.ygcen.2015.05.014] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Revised: 05/27/2015] [Accepted: 05/28/2015] [Indexed: 11/24/2022]
Abstract
It is now clearly established that the central and peripheral nervous systems have the ability to synthesize de novo steroids referred to as neurosteroids. The major evidence for biosynthesis of neuroactive steroids by nervous tissues is based on the expression of enzymes implicated in the formation of steroids in neural cells. The aim of the present review is to summarize the current knowledge regarding the presence of steroidogenic enzymes in the brain of vertebrates and to highlight the very considerable contribution of Professor Kazuyoshi Tsutsui in this domain. The data indicate that expression of steroid-producing enzymes in the brain appeared early during vertebrate evolution and has been preserved from fish to mammals.
Collapse
Affiliation(s)
- Jean Luc do Rego
- Institute for Research and Innovation in Biomedicine (IRIB), Institut National de la Santé et de la Recherche Médicale (INSERM), University of Rouen, 76821 Mont-Saint-Aignan, France
| | - Hubert Vaudry
- Institute for Research and Innovation in Biomedicine (IRIB), Institut National de la Santé et de la Recherche Médicale (INSERM), University of Rouen, 76821 Mont-Saint-Aignan, France; Neurotrophic Factors and Neuronal Differentiation Team, Inserm U982, University of Rouen, 76821 Mont-Saint-Aignan, France.
| |
Collapse
|
40
|
Porcu P, Barron AM, Frye CA, Walf AA, Yang SY, He XY, Morrow AL, Panzica GC, Melcangi RC. Neurosteroidogenesis Today: Novel Targets for Neuroactive Steroid Synthesis and Action and Their Relevance for Translational Research. J Neuroendocrinol 2016; 28:12351. [PMID: 26681259 PMCID: PMC4769676 DOI: 10.1111/jne.12351] [Citation(s) in RCA: 130] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2015] [Revised: 12/12/2015] [Accepted: 12/12/2015] [Indexed: 12/19/2022]
Abstract
Neuroactive steroids are endogenous neuromodulators synthesised in the brain that rapidly alter neuronal excitability by binding to membrane receptors, in addition to the regulation of gene expression via intracellular steroid receptors. Neuroactive steroids induce potent anxiolytic, antidepressant, anticonvulsant, sedative, analgesic and amnesic effects, mainly through interaction with the GABAA receptor. They also exert neuroprotective, neurotrophic and antiapoptotic effects in several animal models of neurodegenerative diseases. Neuroactive steroids regulate many physiological functions, such as the stress response, puberty, the ovarian cycle, pregnancy and reward. Their levels are altered in several neuropsychiatric and neurological diseases and both preclinical and clinical studies emphasise a therapeutic potential of neuroactive steroids for these diseases, whereby symptomatology ameliorates upon restoration of neuroactive steroid concentrations. However, direct administration of neuroactive steroids has several challenges, including pharmacokinetics, low bioavailability, addiction potential, safety and tolerability, which limit its therapeutic use. Therefore, modulation of neurosteroidogenesis to restore the altered endogenous neuroactive steroid tone may represent a better therapeutic approach. This review summarises recent approaches that target the neuroactive steroid biosynthetic pathway at different levels aiming to promote neurosteroidogenesis. These include modulation of neurosteroidogenesis through ligands of the translocator protein 18 kDa and the pregnane xenobiotic receptor, as well as targeting of specific neurosteroidogenic enzymes such as 17β-hydroxysteroid dehydrogenase type 10 or P450 side chain cleavage. Enhanced neurosteroidogenesis through these targets may be beneficial not only for neurodegenerative diseases, such as Alzheimer's disease and age-related dementia, but also for neuropsychiatric diseases, including alcohol use disorders.
Collapse
Affiliation(s)
- Patrizia Porcu
- Neuroscience Institute, National Research Council of Italy (CNR), Cagliari, Italy
| | - Anna M. Barron
- Molecular Imaging Center, National Institute of Radiological Sciences, Anagawa, Inage-ku, Chiba, Japan
| | - Cheryl Anne Frye
- Institute of Arctic Biology, The University of Alaska–Fairbanks, Fairbanks, AK, USA
- The University at Albany, Albany, NY, USA
| | - Alicia A. Walf
- Institute of Arctic Biology, The University of Alaska–Fairbanks, Fairbanks, AK, USA
- The University at Albany, Albany, NY, USA
- Department of Cognitive Science, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Song-Yu Yang
- Department of Developmental Biochemistry, NYS Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| | - Xue-Ying He
- Department of Developmental Biochemistry, NYS Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| | - A. Leslie Morrow
- Departments of Psychiatry and Pharmacology, Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Gian Carlo Panzica
- Department of Neuroscience, University of Turin, and NICO - Neuroscience Institute Cavalieri Ottolenghi, Orbassano, Italy
| | - Roberto C. Melcangi
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
41
|
Cisternas CD, Tome K, Caeiro XE, Dadam FM, Garcia-Segura LM, Cambiasso MJ. Sex chromosome complement determines sex differences in aromatase expression and regulation in the stria terminalis and anterior amygdala of the developing mouse brain. Mol Cell Endocrinol 2015; 414:99-110. [PMID: 26231585 DOI: 10.1016/j.mce.2015.07.027] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Revised: 07/24/2015] [Accepted: 07/25/2015] [Indexed: 10/23/2022]
Abstract
Aromatase, which converts testosterone in estradiol, is involved in the generation of brain sex dimorphisms. Here we used the "four core genotypes" mouse model, in which the effect of gonadal sex and sex chromosome complement is dissociated, to determine if sex chromosomes influence the expression of brain aromatase. The brain of 16 days old XY mouse embryos showed higher aromatase expression in the stria terminalis and the anterior amygdaloid area than the brain of XX embryos, independent of gonadal sex. Furthermore, estradiol or dihydrotestosterone increased aromatase expression in cultures of anterior amygdala neurons derived from XX embryos, but not in those derived from XY embryos. This effect was also independent of gonadal sex. The expression of other steroidogenic molecules, estrogen receptor-α and androgen receptor was not influenced by sex chromosomes. In conclusion, sex chromosomes determine sex dimorphisms in aromatase expression and regulation in the developing mouse brain.
Collapse
Affiliation(s)
- Carla D Cisternas
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina; Departamento de Biología Bucal, Facultad de Odontología - Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Karina Tome
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Ximena E Caeiro
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Florencia M Dadam
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina
| | | | - María J Cambiasso
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina; Departamento de Biología Bucal, Facultad de Odontología - Universidad Nacional de Córdoba, Córdoba, Argentina.
| |
Collapse
|
42
|
Gao L, Rabbitt EH, Condon JC, Renthal NE, Johnston JM, Mitsche MA, Chambon P, Xu J, O'Malley BW, Mendelson CR. Steroid receptor coactivators 1 and 2 mediate fetal-to-maternal signaling that initiates parturition. J Clin Invest 2015; 125:2808-24. [PMID: 26098214 DOI: 10.1172/jci78544] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Accepted: 05/06/2015] [Indexed: 12/22/2022] Open
Abstract
The precise mechanisms that lead to parturition are incompletely defined. Surfactant protein-A (SP-A), which is secreted by fetal lungs into amniotic fluid (AF) near term, likely provides a signal for parturition; however, SP-A-deficient mice have only a relatively modest delay (~12 hours) in parturition, suggesting additional factors. Here, we evaluated the contribution of steroid receptor coactivators 1 and 2 (SRC-1 and SRC-2), which upregulate SP-A transcription, to the parturition process. As mice lacking both SRC-1 and SRC-2 die at birth due to respiratory distress, we crossed double-heterozygous males and females. Parturition was severely delayed (~38 hours) in heterozygous dams harboring SRC-1/-2-deficient embryos. These mothers exhibited decreased myometrial NF-κB activation, PGF2α, and expression of contraction-associated genes; impaired luteolysis; and elevated circulating progesterone. These manifestations also occurred in WT females bearing SRC-1/-2 double-deficient embryos, indicating that a fetal-specific defect delayed labor. SP-A, as well as the enzyme lysophosphatidylcholine acyltransferase-1 (LPCAT1), required for synthesis of surfactant dipalmitoylphosphatidylcholine, and the proinflammatory glycerophospholipid platelet-activating factor (PAF) were markedly reduced in SRC-1/-2-deficient fetal lungs near term. Injection of PAF or SP-A into AF at 17.5 days post coitum enhanced uterine NF-κB activation and contractile gene expression, promoted luteolysis, and rescued delayed parturition in SRC-1/-2-deficient embryo-bearing dams. These findings reveal that fetal lungs produce signals to initiate labor when mature and that SRC-1/-2-dependent production of SP-A and PAF is crucial for this process.
Collapse
|
43
|
Guennoun R, Labombarda F, Gonzalez Deniselle MC, Liere P, De Nicola AF, Schumacher M. Progesterone and allopregnanolone in the central nervous system: response to injury and implication for neuroprotection. J Steroid Biochem Mol Biol 2015; 146:48-61. [PMID: 25196185 DOI: 10.1016/j.jsbmb.2014.09.001] [Citation(s) in RCA: 155] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Revised: 08/29/2014] [Accepted: 09/02/2014] [Indexed: 01/26/2023]
Abstract
Progesterone is a well-known steroid hormone, synthesized by ovaries and placenta in females, and by adrenal glands in both males and females. Several tissues are targets of progesterone and the nervous system is a major one. Progesterone is also locally synthesized by the nervous system and qualifies, therefore, as a neurosteroid. In addition, the nervous system has the capacity to bio-convert progesterone into its active metabolite allopregnanolone. The enzymes required for progesterone and allopregnanolone synthesis are widely distributed in brain and spinal cord. Increased local biosynthesis of pregnenolone, progesterone and 5α-dihydroprogesterone may be a part of an endogenous neuroprotective mechanism in response to nervous system injuries. Progesterone and allopregnanolone neuroprotective effects have been widely recognized. Multiple receptors or associated proteins may contribute to the progesterone effects: classical nuclear receptors (PR), membrane progesterone receptor component 1 (PGRMC1), membrane progesterone receptors (mPR), and γ-aminobutyric acid type A (GABAA) receptors after conversion to allopregnanolone. In this review, we will succinctly describe progesterone and allopregnanolone biosynthetic pathways and enzyme distribution in brain and spinal cord. Then, we will summarize our work on progesterone receptor distribution and cellular expression in brain and spinal cord; neurosteroid stimulation after nervous system injuries (spinal cord injury, traumatic brain injury, and stroke); and on progesterone and allopregnanolone neuroprotective effects in different experimental models including stroke and spinal cord injury. We will discuss in detail the neuroprotective effects of progesterone on the nervous system via PR, and of allopregnanolone via its modulation of GABAA receptors.
Collapse
Affiliation(s)
- R Guennoun
- UMR 788, Inserm and University Paris-Sud, 80 rue du Général Leclerc, 94276 Bicêtre, Kremlin-Bicêtre, France.
| | - F Labombarda
- Instituto de Biologia y Medicina Experimental and University of Buenos Aires, Argentina
| | | | - P Liere
- UMR 788, Inserm and University Paris-Sud, 80 rue du Général Leclerc, 94276 Bicêtre, Kremlin-Bicêtre, France
| | - A F De Nicola
- Instituto de Biologia y Medicina Experimental and University of Buenos Aires, Argentina
| | - M Schumacher
- UMR 788, Inserm and University Paris-Sud, 80 rue du Général Leclerc, 94276 Bicêtre, Kremlin-Bicêtre, France
| |
Collapse
|
44
|
GABAA receptor-acting neurosteroids: a role in the development and regulation of the stress response. Front Neuroendocrinol 2015; 36:28-48. [PMID: 24929099 PMCID: PMC4349499 DOI: 10.1016/j.yfrne.2014.06.001] [Citation(s) in RCA: 108] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Revised: 05/26/2014] [Accepted: 06/01/2014] [Indexed: 12/22/2022]
Abstract
Regulation of hypothalamic-pituitary-adrenocortical (HPA) axis activity by stress is a fundamental survival mechanism and HPA-dysfunction is implicated in psychiatric disorders. Adverse early life experiences, e.g. poor maternal care, negatively influence brain development and programs an abnormal stress response by encoding long-lasting molecular changes, which may extend to the next generation. How HPA-dysfunction leads to the development of affective disorders is complex, but may involve GABAA receptors (GABAARs), as they curtail stress-induced HPA axis activation. Of particular interest are endogenous neurosteroids that potently modulate the function of GABAARs and exhibit stress-protective properties. Importantly, neurosteroid levels rise rapidly during acute stress, are perturbed in chronic stress and are implicated in the behavioural changes associated with early-life adversity. We will appraise how GABAAR-active neurosteroids may impact on HPA axis development and the orchestration of the stress-evoked response. The significance of these actions will be discussed in the context of stress-associated mood disorders.
Collapse
|
45
|
Fokidis HB, Adomat HH, Kharmate G, Hosseini-Beheshti E, Guns ES, Soma KK. Regulation of local steroidogenesis in the brain and in prostate cancer: lessons learned from interdisciplinary collaboration. Front Neuroendocrinol 2015; 36:108-29. [PMID: 25223867 DOI: 10.1016/j.yfrne.2014.08.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Revised: 08/28/2014] [Accepted: 08/28/2014] [Indexed: 11/16/2022]
Abstract
Sex steroids play critical roles in the regulation of the brain and many other organs. Traditionally, researchers have focused on sex steroid signaling that involves travel from the gonads via the circulation to intracellular receptors in target tissues. This classic concept has been challenged, however, by the growing number of cases in which steroids are synthesized locally and act locally within diverse tissues. For example, the brain and prostate carcinoma were previously considered targets of gonadal sex steroids, but under certain circumstances, these tissues can upregulate their steroidogenic potential, particularly when circulating sex steroid concentrations are low. We review some of the similarities and differences between local sex steroid synthesis in the brain and prostate cancer. We also share five lessons that we have learned during the course of our interdisciplinary collaboration, which brought together neuroendocrinologists and cancer biologists. These lessons have important implications for future research in both fields.
Collapse
Affiliation(s)
- H Bobby Fokidis
- Department of Biology, Rollins College, Winter Park, FL 37289, USA; Department of Psychology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; Vancouver Prostate Centre, Vancouver, BC V6H 3Z6, Canada.
| | - Hans H Adomat
- Vancouver Prostate Centre, Vancouver, BC V6H 3Z6, Canada
| | | | | | - Emma S Guns
- Vancouver Prostate Centre, Vancouver, BC V6H 3Z6, Canada; Department of Urological Sciences, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| | - Kiran K Soma
- Department of Psychology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; Brain Research Centre, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; Department of Zoology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada.
| |
Collapse
|
46
|
Lee WC, Kim MT, Ko KT, Lee WK, Kim SY, Kim HY, Yang DY. Relationship between Serum Testosterone and Cardiovascular Disease Risk Determined Using the Framingham Risk Score in Male Patients with Sexual Dysfunction. World J Mens Health 2014; 32:139-44. [PMID: 25606562 PMCID: PMC4298816 DOI: 10.5534/wjmh.2014.32.3.139] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Revised: 10/11/2014] [Accepted: 10/14/2014] [Indexed: 11/20/2022] Open
Abstract
Purpose The aim of the present study aimed to evaluate the effect of testosterone on cardiovascular disease by using the Framingham Risk Score (FRS) in patients with sexual dysfunction. Materials and Methods A total of 308 men with sexual dysfunction were enrolled in this study. Clinical assessments included the 15-item International Index of Erectile Function (IIEF), blood pressure measurement, and clinical laboratory indexes. The FRS, which predicts the incidence rate of cardiovascular diseases in the next 10 years, was calculated on the basis of age, gender, total cholesterol, smoking status, high density lipoprotein cholesterol, and systolic blood pressure. Results The mean age of the 308 enrolled patients was 49.42±10.73 years, and the patients' mean body mass index (kg/m2) was 25.07±3.14. The mean total IIEF score was 28.44±18.06. The median total testosterone concentration was 3.2 ng/mL (interquartile range [IQR]: 2.3~3.2 ng/mL). The median calculated free and bioavailable testosterone concentrations were 0.052 ng/mL (IQR 0.039~0.070 ng/mL) and 1.30 ng/mL (IQR: 1.00~1.76 ng/mL), respectively. The mean FRS was 10.47±6.45. The FRS tended to show a negative correlation with the total and calculated free testosterone levels, but this was not significant (p=0.064 and p=0.074, respectively). In the multiple linear regression analysis, a significant negative correlation was observed between the total testosterone level and the FRS (p=0.048). Conclusions The results suggest that the testosterone level is related to the FRS and that a high testosterone level may decrease the risk of cardiovascular disease.
Collapse
Affiliation(s)
- Wan Chul Lee
- Department of Urology, Hallym University Kangdong Sacred Heart Hospital, Seoul, Korea
| | - Ma Tae Kim
- Department of Urology, Hallym University Kangdong Sacred Heart Hospital, Seoul, Korea
| | - Kyung Tae Ko
- Department of Urology, Hallym University Kangdong Sacred Heart Hospital, Seoul, Korea
| | - Won Ki Lee
- Department of Urology, Hallym University Chuncheon Sacred Heart Hospital, Chuncheon, Korea
| | - Sung Yong Kim
- Department of Urology, Hallym University Kangdong Sacred Heart Hospital, Seoul, Korea
| | - Ha Young Kim
- Department of Urology, Hallym University Kangdong Sacred Heart Hospital, Seoul, Korea
| | - Dae Yul Yang
- Department of Urology, Hallym University Kangdong Sacred Heart Hospital, Seoul, Korea
| |
Collapse
|
47
|
El Hajj Chehadeh S, Dreumont N, Willekens J, Canabady-Rochelle L, Jeannesson E, Alberto JM, Daval JL, Guéant JL, Leininger-Muller B. Early methyl donor deficiency alters cAMP signaling pathway and neurosteroidogenesis in the cerebellum of female rat pups. Am J Physiol Endocrinol Metab 2014; 307:E1009-19. [PMID: 25294213 PMCID: PMC4254983 DOI: 10.1152/ajpendo.00364.2014] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Early deficiency of the methyl donors folate and vitamin B12 produces hyperhomocysteinemia and cognitive and motor disorders in 21-day-old rat pups from dams fed a diet deficient in methyl donors during gestation and lactation. These disorders are associated with impaired neurogenesis and altered synaptic plasticity in cerebellum. We aimed to investigate whether these disorders could be related to impaired expression of neurosteroidogenesis-associated proteins, key regulator receptors, and some steroid content in the cerebellum. The methyl donor deficiency produced a decreased concentration of folate and vitamin B12, along with accumulation of homocysteine in Purkinje cells in both sexes, whereas the S-adenosylmethionine/S-adenosylhomocysteine ratio was reduced only in females. The transcription level and protein expression of StAR, aromatase, ERα, ERβ, and LH receptors were decreased only in females, with a marked effect in Purkinje cells, as shown by immunohistochemistry. Consistently, reduced levels of estradiol and pregnenolone were measured in cerebellar extracts of females only. The decreased expression levels of the transcriptional factors CREB, phospho-CREB, and SF-1, the lesser increase of cAMP concentration, and the lower level of phospho-PKC in the cerebellum of deficient females suggest that the activation of neurosteroidogenesis via cAMP-mediated signaling pathways associated with LHR activation would be altered. In conclusion, a gestational methyl donor deficiency impairs neurosteroidogenesis in cerebellum in a sex-dependent manner.
Collapse
Affiliation(s)
- Sarah El Hajj Chehadeh
- Institut National de la Sante et de la Recherche Medicale, U-954, Nutrition-Génétique et Exposition Aux Risques Environnementaux, Faculté de Médecine, Université de Lorraine, Vandoeuvre les Nancy, France
| | - Natacha Dreumont
- Institut National de la Sante et de la Recherche Medicale, U-954, Nutrition-Génétique et Exposition Aux Risques Environnementaux, Faculté de Médecine, Université de Lorraine, Vandoeuvre les Nancy, France
| | - Jérèmy Willekens
- Institut National de la Sante et de la Recherche Medicale, U-954, Nutrition-Génétique et Exposition Aux Risques Environnementaux, Faculté de Médecine, Université de Lorraine, Vandoeuvre les Nancy, France
| | - Laetitia Canabady-Rochelle
- Institut National de la Sante et de la Recherche Medicale, U-954, Nutrition-Génétique et Exposition Aux Risques Environnementaux, Faculté de Médecine, Université de Lorraine, Vandoeuvre les Nancy, France
| | - Elise Jeannesson
- Institut National de la Sante et de la Recherche Medicale, U-954, Nutrition-Génétique et Exposition Aux Risques Environnementaux, Faculté de Médecine, Université de Lorraine, Vandoeuvre les Nancy, France
| | - Jean-Marc Alberto
- Institut National de la Sante et de la Recherche Medicale, U-954, Nutrition-Génétique et Exposition Aux Risques Environnementaux, Faculté de Médecine, Université de Lorraine, Vandoeuvre les Nancy, France
| | - Jean-Luc Daval
- Institut National de la Sante et de la Recherche Medicale, U-954, Nutrition-Génétique et Exposition Aux Risques Environnementaux, Faculté de Médecine, Université de Lorraine, Vandoeuvre les Nancy, France
| | - Jean-Louis Guéant
- Institut National de la Sante et de la Recherche Medicale, U-954, Nutrition-Génétique et Exposition Aux Risques Environnementaux, Faculté de Médecine, Université de Lorraine, Vandoeuvre les Nancy, France
| | - Brigitte Leininger-Muller
- Institut National de la Sante et de la Recherche Medicale, U-954, Nutrition-Génétique et Exposition Aux Risques Environnementaux, Faculté de Médecine, Université de Lorraine, Vandoeuvre les Nancy, France
| |
Collapse
|
48
|
Mukherjee A, Haldar C. Photoperiodic regulation of melatonin membrane receptor (MT1R) expression and steroidogenesis in testis of adult golden hamster, Mesocricetus auratus. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2014; 140:374-80. [DOI: 10.1016/j.jphotobiol.2014.08.022] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Revised: 08/27/2014] [Accepted: 08/28/2014] [Indexed: 12/15/2022]
|
49
|
Roland AV, Moenter SM. Reproductive neuroendocrine dysfunction in polycystic ovary syndrome: insight from animal models. Front Neuroendocrinol 2014; 35:494-511. [PMID: 24747343 PMCID: PMC4175187 DOI: 10.1016/j.yfrne.2014.04.002] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Revised: 03/15/2014] [Accepted: 04/09/2014] [Indexed: 12/23/2022]
Abstract
Polycystic ovary syndrome (PCOS) is a common endocrinopathy with elusive origins. A clinically heterogeneous disorder, PCOS is likely to have multiple etiologies comprised of both genetic and environmental factors. Reproductive neuroendocrine dysfunction involving increased frequency and amplitude of gonadotropin-releasing hormone (GnRH) release, as reflected by pulsatile luteinizing hormone (LH) secretion, is an important pathophysiologic component in PCOS. Whether this defect is primary or secondary to other changes in PCOS is unclear, but it contributes significantly to ongoing reproductive dysfunction. This review highlights recent work in animal models, with a particular emphasis on the mouse, demonstrating the ability of pre- and postnatal steroidal and metabolic factors to drive changes in GnRH/LH pulsatility and GnRH neuron function consistent with the observed abnormalities in PCOS. This work has begun to elucidate how a complex interplay of ovarian, metabolic, and neuroendocrine factors culminates in this syndrome.
Collapse
Affiliation(s)
- Alison V Roland
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Suzanne M Moenter
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
50
|
Frye C, Koonce C, Walf A. Role of pregnane xenobiotic receptor in the midbrain ventral tegmental area for estradiol- and 3α,5α-THP-facilitated lordosis of female rats. Psychopharmacology (Berl) 2014; 231:3365-74. [PMID: 24435323 PMCID: PMC4102666 DOI: 10.1007/s00213-013-3406-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2013] [Accepted: 12/05/2013] [Indexed: 12/17/2022]
Abstract
RATIONALE Progesterone and its metabolite, 5α-pregnan-3α-ol-20-one (3α,5α-THP), have actions in the ventral tegmental area (VTA) that are required for lordosis, a characteristic mating posture of female rodents. 17β-estradiol (estradiol) co-varies with progestogens over natural cycles, enhances production of 3α,5α-THP, and is required for successful reproductive behavior. OBJECTIVES A question of interest is the role of pregnane xenobiotic receptor (PXR), a nuclear receptor that regulates enzymes needed for the production of 3α,5α-THP, for estradiol-mediated lordosis. The hypothesis tested was that if PXR is involved in estradiol-mediated biosynthesis of 3α,5α-THP and reproductive behavior, knocking down expression of PXR in the VTA of estradiol-primed, but not vehicle-primed, rats should decrease lordosis and midbrain 3α,5α-THP; effects may be attenuated by 3α,5α-THP administered to the VTA. METHODS Ovariectomized rats were administered subcutaneous injections of oil vehicle or estradiol. Rats were then administered PXR antisense oligonucleotides (PXR AS-ODNs; which are expected to locally knock down expression of PXR), or control (saline), infusions to the VTA. Rats were administered 3α,5α-THP or vehicle via infusions to the VTA. Reproductive behavior (paced mating task) of rats was determined in addition to exploratory (open field), affective (elevated plus maze), and pro-social (social interaction task) behavior. RESULTS Reproductive behavior (i.e., increased lordosis) was enhanced with estradiol-priming and infusions of 3α,5α-THP to the VTA. Infusions of PXR AS-ODNs to the VTA attenuated responses in estradiol-, but not vehicle-, primed rats, compared to control infusions. CONCLUSIONS PXR may be involved in a neuroregulatory response involving biosynthesis of 3α,5α-THP in the midbrain VTA of estradiol-primed rats.
Collapse
Affiliation(s)
- C.A. Frye
- Dept. of Psychology, The University at Albany-SUNY, Life Sciences 01058, 1400 Washington Ave., Albany, NY USA 12222,Dept. of Biological Sciences, The University at Albany-SUNY, Life Sciences 01058, 1400 Washington Ave., Albany, NY USA 12222,The Centers for Neuroscience, The University at Albany-SUNY, Life Sciences 01058, 1400 Washington Ave., Albany, NY USA 12222,The Centers for Life Sciences Research, The University at Albany-SUNY, Life Sciences 01058, 1400 Washington Ave., Albany, NY USA 12222,Department of Chemistry, Institute for Arctic Biology, The University of Alaska–Fairbanks, Fairbanks, Alaska USA 99775,IDeA Network of Biomedical Excellence, The University of Alaska–Fairbanks, Fairbanks, Alaska USA 99775
| | - C.J. Koonce
- Dept. of Psychology, The University at Albany-SUNY, Life Sciences 01058, 1400 Washington Ave., Albany, NY USA 12222,Department of Chemistry, Institute for Arctic Biology, The University of Alaska–Fairbanks, Fairbanks, Alaska USA 99775
| | - A.A. Walf
- Dept. of Psychology, The University at Albany-SUNY, Life Sciences 01058, 1400 Washington Ave., Albany, NY USA 12222,Department of Chemistry, Institute for Arctic Biology, The University of Alaska–Fairbanks, Fairbanks, Alaska USA 99775,IDeA Network of Biomedical Excellence, The University of Alaska–Fairbanks, Fairbanks, Alaska USA 99775
| |
Collapse
|